WO2002096927A2 - Ribozyme based treatment of female reproductive diseases - Google Patents
Ribozyme based treatment of female reproductive diseases Download PDFInfo
- Publication number
- WO2002096927A2 WO2002096927A2 PCT/US2002/017674 US0217674W WO02096927A2 WO 2002096927 A2 WO2002096927 A2 WO 2002096927A2 US 0217674 W US0217674 W US 0217674W WO 02096927 A2 WO02096927 A2 WO 02096927A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nucleic acid
- acid molecule
- vegf
- rna
- cell
- Prior art date
Links
- 238000011282 treatment Methods 0.000 title claims abstract description 60
- 108091092562 ribozyme Proteins 0.000 title abstract description 131
- 108090000994 Catalytic RNA Proteins 0.000 title abstract description 128
- 102000053642 Catalytic RNA Human genes 0.000 title abstract description 128
- 208000011404 female reproductive system disease Diseases 0.000 title description 2
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 590
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 566
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 566
- 230000002255 enzymatic effect Effects 0.000 claims abstract description 162
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims abstract description 144
- 230000000692 anti-sense effect Effects 0.000 claims abstract description 92
- 230000014509 gene expression Effects 0.000 claims abstract description 61
- 108091008605 VEGF receptors Proteins 0.000 claims abstract description 56
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 claims abstract description 55
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 42
- 230000033115 angiogenesis Effects 0.000 claims abstract description 42
- 102000040650 (ribonucleotides)n+m Human genes 0.000 claims abstract description 36
- 201000009273 Endometriosis Diseases 0.000 claims abstract description 32
- 206010012689 Diabetic retinopathy Diseases 0.000 claims abstract description 24
- 108091027757 Deoxyribozyme Proteins 0.000 claims abstract description 19
- 230000005747 tumor angiogenesis Effects 0.000 claims abstract description 19
- 108010044467 Isoenzymes Proteins 0.000 claims abstract description 13
- 241000251131 Sphyrna Species 0.000 claims abstract description 12
- 201000011510 cancer Diseases 0.000 claims abstract description 11
- 238000000034 method Methods 0.000 claims description 168
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 138
- 210000004027 cell Anatomy 0.000 claims description 96
- 102100033177 Vascular endothelial growth factor receptor 2 Human genes 0.000 claims description 91
- 101000851007 Homo sapiens Vascular endothelial growth factor receptor 2 Proteins 0.000 claims description 86
- 230000000694 effects Effects 0.000 claims description 80
- 238000003776 cleavage reaction Methods 0.000 claims description 65
- 230000007017 scission Effects 0.000 claims description 61
- 150000001875 compounds Chemical class 0.000 claims description 51
- 239000000203 mixture Substances 0.000 claims description 45
- -1 cationic lipid Chemical class 0.000 claims description 44
- 230000004048 modification Effects 0.000 claims description 34
- 238000012986 modification Methods 0.000 claims description 34
- 239000003814 drug Substances 0.000 claims description 33
- 108010053099 Vascular Endothelial Growth Factor Receptor-2 Proteins 0.000 claims description 31
- 239000003153 chemical reaction reagent Substances 0.000 claims description 30
- 101100372762 Rattus norvegicus Flt1 gene Proteins 0.000 claims description 29
- 101100381481 Caenorhabditis elegans baz-2 gene Proteins 0.000 claims description 28
- 150000002632 lipids Chemical group 0.000 claims description 27
- 241000282414 Homo sapiens Species 0.000 claims description 26
- GURKHSYORGJETM-WAQYZQTGSA-N irinotecan hydrochloride (anhydrous) Chemical compound Cl.C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 GURKHSYORGJETM-WAQYZQTGSA-N 0.000 claims description 26
- 230000005764 inhibitory process Effects 0.000 claims description 24
- 239000002502 liposome Substances 0.000 claims description 24
- 229940079593 drug Drugs 0.000 claims description 23
- 241000124008 Mammalia Species 0.000 claims description 20
- 239000002246 antineoplastic agent Substances 0.000 claims description 20
- 229940127089 cytotoxic agent Drugs 0.000 claims description 20
- 210000004962 mammalian cell Anatomy 0.000 claims description 16
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 claims description 14
- 230000001965 increasing effect Effects 0.000 claims description 14
- 150000003904 phospholipids Chemical class 0.000 claims description 14
- 238000002560 therapeutic procedure Methods 0.000 claims description 14
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 claims description 13
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Natural products O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 claims description 13
- 206010009944 Colon cancer Diseases 0.000 claims description 12
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 12
- XLXSAKCOAKORKW-AQJXLSMYSA-N gonadorelin Chemical compound C([C@@H](C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)NCC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 XLXSAKCOAKORKW-AQJXLSMYSA-N 0.000 claims description 12
- 201000001441 melanoma Diseases 0.000 claims description 12
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 claims description 12
- 239000000579 Gonadotropin-Releasing Hormone Substances 0.000 claims description 11
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 11
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 11
- 206010038389 Renal cancer Diseases 0.000 claims description 11
- 101000857870 Squalus acanthias Gonadoliberin Proteins 0.000 claims description 11
- 239000003937 drug carrier Substances 0.000 claims description 11
- 229940035638 gonadotropin-releasing hormone Drugs 0.000 claims description 11
- 201000010982 kidney cancer Diseases 0.000 claims description 11
- 201000005202 lung cancer Diseases 0.000 claims description 11
- 208000020816 lung neoplasm Diseases 0.000 claims description 11
- 206010006187 Breast cancer Diseases 0.000 claims description 10
- 208000026310 Breast neoplasm Diseases 0.000 claims description 10
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 10
- 239000003085 diluting agent Substances 0.000 claims description 10
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 10
- 201000002528 pancreatic cancer Diseases 0.000 claims description 10
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 10
- 108010042407 Endonucleases Proteins 0.000 claims description 9
- 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 claims description 9
- 210000005260 human cell Anatomy 0.000 claims description 9
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 claims description 9
- 229940045145 uridine Drugs 0.000 claims description 9
- 108010037003 Buserelin Proteins 0.000 claims description 8
- PYMDEDHDQYLBRT-DRIHCAFSSA-N Buserelin acetate Chemical compound CC(O)=O.CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](COC(C)(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 PYMDEDHDQYLBRT-DRIHCAFSSA-N 0.000 claims description 8
- 239000000556 agonist Substances 0.000 claims description 8
- 230000002401 inhibitory effect Effects 0.000 claims description 8
- RGLRXNKKBLIBQS-XNHQSDQCSA-N leuprolide acetate Chemical compound CC(O)=O.CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 RGLRXNKKBLIBQS-XNHQSDQCSA-N 0.000 claims description 8
- 239000002126 C01EB10 - Adenosine Substances 0.000 claims description 7
- 229960005305 adenosine Drugs 0.000 claims description 7
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 claims description 7
- FHIDNBAQOFJWCA-UAKXSSHOSA-N 5-fluorouridine Chemical group O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 FHIDNBAQOFJWCA-UAKXSSHOSA-N 0.000 claims description 6
- VVIAGPKUTFNRDU-UHFFFAOYSA-N 6S-folinic acid Natural products C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-UHFFFAOYSA-N 0.000 claims description 6
- MIKUYHXYGGJMLM-GIMIYPNGSA-N Crotonoside Natural products C1=NC2=C(N)NC(=O)N=C2N1[C@H]1O[C@@H](CO)[C@H](O)[C@@H]1O MIKUYHXYGGJMLM-GIMIYPNGSA-N 0.000 claims description 6
- NYHBQMYGNKIUIF-UHFFFAOYSA-N D-guanosine Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(O)C1O NYHBQMYGNKIUIF-UHFFFAOYSA-N 0.000 claims description 6
- 229930012538 Paclitaxel Natural products 0.000 claims description 6
- 229940088954 camptosar Drugs 0.000 claims description 6
- 229960004562 carboplatin Drugs 0.000 claims description 6
- VVIAGPKUTFNRDU-ABLWVSNPSA-N folinic acid Chemical group C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-ABLWVSNPSA-N 0.000 claims description 6
- 235000008191 folinic acid Nutrition 0.000 claims description 6
- 239000011672 folinic acid Substances 0.000 claims description 6
- 229940029575 guanosine Drugs 0.000 claims description 6
- 229960001691 leucovorin Drugs 0.000 claims description 6
- 229960001592 paclitaxel Drugs 0.000 claims description 6
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 claims description 6
- OVYNGSFVYRPRCG-KQYNXXCUSA-N 2'-O-methylguanosine Chemical class CO[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=C(N)NC2=O)=C2N=C1 OVYNGSFVYRPRCG-KQYNXXCUSA-N 0.000 claims description 5
- 150000001768 cations Chemical class 0.000 claims description 5
- RFCQJGFZUQFYRF-ZOQUXTDFSA-N 2'-O-methylcytidine Chemical class CO[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)N=C(N)C=C1 RFCQJGFZUQFYRF-ZOQUXTDFSA-N 0.000 claims description 4
- SXUXMRMBWZCMEN-ZOQUXTDFSA-N 2'-O-methyluridine Chemical class CO[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 SXUXMRMBWZCMEN-ZOQUXTDFSA-N 0.000 claims description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 claims description 4
- 108010069236 Goserelin Proteins 0.000 claims description 4
- 108010000817 Leuprolide Proteins 0.000 claims description 4
- 108010021717 Nafarelin Proteins 0.000 claims description 4
- 229960005064 buserelin acetate Drugs 0.000 claims description 4
- FPUGCISOLXNPPC-IOSLPCCCSA-N cordysinin B Chemical class CO[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(N)=C2N=C1 FPUGCISOLXNPPC-IOSLPCCCSA-N 0.000 claims description 4
- POZRVZJJTULAOH-LHZXLZLDSA-N danazol Chemical compound C1[C@]2(C)[C@H]3CC[C@](C)([C@](CC4)(O)C#C)[C@@H]4[C@@H]3CCC2=CC2=C1C=NO2 POZRVZJJTULAOH-LHZXLZLDSA-N 0.000 claims description 4
- 229960000766 danazol Drugs 0.000 claims description 4
- 238000002651 drug therapy Methods 0.000 claims description 4
- 229960003690 goserelin acetate Drugs 0.000 claims description 4
- 229960004768 irinotecan Drugs 0.000 claims description 4
- 229960004338 leuprorelin Drugs 0.000 claims description 4
- BLCLNMBMMGCOAS-UHFFFAOYSA-N n-[1-[[1-[[1-[[1-[[1-[[1-[[1-[2-[(carbamoylamino)carbamoyl]pyrrolidin-1-yl]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-3-[(2-methylpropan-2-yl)oxy]-1-oxopropan-2-yl]amino]-3-(4-hydroxyphenyl)-1-oxopropan-2-yl]amin Chemical compound C1CCC(C(=O)NNC(N)=O)N1C(=O)C(CCCN=C(N)N)NC(=O)C(CC(C)C)NC(=O)C(COC(C)(C)C)NC(=O)C(NC(=O)C(CO)NC(=O)C(CC=1C2=CC=CC=C2NC=1)NC(=O)C(CC=1NC=NC=1)NC(=O)C1NC(=O)CC1)CC1=CC=C(O)C=C1 BLCLNMBMMGCOAS-UHFFFAOYSA-N 0.000 claims description 4
- FSBTYDWUUWLHBD-UDXTWCDOSA-N nafarelin acetate hydrate Chemical compound O.CC(O)=O.C([C@@H](C(=O)N[C@H](CC=1C=C2C=CC=CC2=CC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NCC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 FSBTYDWUUWLHBD-UDXTWCDOSA-N 0.000 claims description 4
- 108091008104 nucleic acid aptamers Proteins 0.000 claims description 4
- 229940127234 oral contraceptive Drugs 0.000 claims description 4
- 239000003539 oral contraceptive agent Substances 0.000 claims description 4
- 229940086546 synarel Drugs 0.000 claims description 4
- 190000008236 Carboplatin Chemical group 0.000 claims 3
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims 2
- 102000004533 Endonucleases Human genes 0.000 claims 1
- 208000006265 Renal cell carcinoma Diseases 0.000 claims 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims 1
- 108090000623 proteins and genes Proteins 0.000 abstract description 51
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 abstract description 39
- 229940124676 vascular endothelial growth factor receptor Drugs 0.000 abstract description 36
- 201000010099 disease Diseases 0.000 abstract description 32
- 108020004459 Small interfering RNA Proteins 0.000 abstract description 31
- 206010036618 Premenstrual syndrome Diseases 0.000 abstract description 28
- 108091023037 Aptamer Proteins 0.000 abstract description 25
- 208000002780 macular degeneration Diseases 0.000 abstract description 17
- 206010064930 age-related macular degeneration Diseases 0.000 abstract description 16
- 230000016087 ovulation Effects 0.000 abstract description 16
- 206010014733 Endometrial cancer Diseases 0.000 abstract description 14
- 206010014759 Endometrial neoplasm Diseases 0.000 abstract description 14
- 206010027339 Menstruation irregular Diseases 0.000 abstract description 14
- 230000004064 dysfunction Effects 0.000 abstract description 14
- 201000003914 endometrial carcinoma Diseases 0.000 abstract description 14
- 208000031169 hemorrhagic disease Diseases 0.000 abstract description 14
- 201000004681 Psoriasis Diseases 0.000 abstract description 7
- 208000035475 disorder Diseases 0.000 abstract description 7
- 230000001850 reproductive effect Effects 0.000 abstract description 6
- 206010021143 Hypoxia Diseases 0.000 abstract description 4
- 230000007954 hypoxia Effects 0.000 abstract description 4
- 206010039073 rheumatoid arthritis Diseases 0.000 abstract description 4
- 201000007914 proliferative diabetic retinopathy Diseases 0.000 abstract description 3
- 230000029663 wound healing Effects 0.000 abstract description 3
- 238000003745 diagnosis Methods 0.000 abstract description 2
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 139
- 239000002773 nucleotide Substances 0.000 description 134
- 125000003729 nucleotide group Chemical group 0.000 description 126
- 102000009524 Vascular Endothelial Growth Factor A Human genes 0.000 description 117
- 230000027455 binding Effects 0.000 description 70
- 108091034117 Oligonucleotide Proteins 0.000 description 44
- 239000000243 solution Substances 0.000 description 38
- 108020004414 DNA Proteins 0.000 description 36
- 239000000758 substrate Substances 0.000 description 33
- 230000002238 attenuated effect Effects 0.000 description 31
- 241001465754 Metazoa Species 0.000 description 30
- 230000000295 complement effect Effects 0.000 description 29
- 210000001519 tissue Anatomy 0.000 description 29
- 108020004999 messenger RNA Proteins 0.000 description 27
- 102000004169 proteins and genes Human genes 0.000 description 26
- 108091028043 Nucleic acid sequence Proteins 0.000 description 25
- 235000018102 proteins Nutrition 0.000 description 25
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 24
- 238000003786 synthesis reaction Methods 0.000 description 23
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 23
- 235000000346 sugar Nutrition 0.000 description 22
- 239000013598 vector Substances 0.000 description 22
- 230000015572 biosynthetic process Effects 0.000 description 21
- 238000001727 in vivo Methods 0.000 description 20
- 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 19
- 230000000670 limiting effect Effects 0.000 description 19
- 239000002777 nucleoside Substances 0.000 description 19
- 238000009472 formulation Methods 0.000 description 17
- 238000000338 in vitro Methods 0.000 description 17
- 229920000642 polymer Polymers 0.000 description 17
- 206010029113 Neovascularisation Diseases 0.000 description 16
- 102000005962 receptors Human genes 0.000 description 16
- 108020003175 receptors Proteins 0.000 description 16
- 101710163270 Nuclease Proteins 0.000 description 15
- 230000003197 catalytic effect Effects 0.000 description 15
- 230000008878 coupling Effects 0.000 description 15
- 238000010168 coupling process Methods 0.000 description 15
- 238000005859 coupling reaction Methods 0.000 description 15
- 230000009467 reduction Effects 0.000 description 15
- 239000000126 substance Substances 0.000 description 15
- 102100034343 Integrase Human genes 0.000 description 14
- 239000013604 expression vector Substances 0.000 description 14
- 229910019142 PO4 Inorganic materials 0.000 description 13
- 238000002347 injection Methods 0.000 description 13
- 239000007924 injection Substances 0.000 description 13
- 230000003993 interaction Effects 0.000 description 13
- 239000010452 phosphate Substances 0.000 description 13
- 230000004044 response Effects 0.000 description 13
- 239000003981 vehicle Substances 0.000 description 13
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 12
- 125000003118 aryl group Chemical group 0.000 description 12
- 238000003556 assay Methods 0.000 description 12
- 230000006870 function Effects 0.000 description 12
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 12
- 238000012552 review Methods 0.000 description 12
- 238000007920 subcutaneous administration Methods 0.000 description 12
- 108010001267 Protein Subunits Proteins 0.000 description 11
- 125000000217 alkyl group Chemical group 0.000 description 11
- 125000003636 chemical group Chemical group 0.000 description 11
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 11
- 125000003835 nucleoside group Chemical group 0.000 description 11
- 108090000765 processed proteins & peptides Proteins 0.000 description 11
- 230000008685 targeting Effects 0.000 description 11
- 230000001225 therapeutic effect Effects 0.000 description 11
- 230000004614 tumor growth Effects 0.000 description 11
- 101710203526 Integrase Proteins 0.000 description 10
- 206010027476 Metastases Diseases 0.000 description 10
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 10
- 230000003213 activating effect Effects 0.000 description 10
- 238000004113 cell culture Methods 0.000 description 10
- 230000001413 cellular effect Effects 0.000 description 10
- 230000002357 endometrial effect Effects 0.000 description 10
- 230000007246 mechanism Effects 0.000 description 10
- 238000011160 research Methods 0.000 description 10
- 230000011664 signaling Effects 0.000 description 10
- 208000035657 Abasia Diseases 0.000 description 9
- 108700026244 Open Reading Frames Proteins 0.000 description 9
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 9
- 239000004480 active ingredient Substances 0.000 description 9
- 230000001772 anti-angiogenic effect Effects 0.000 description 9
- 238000006243 chemical reaction Methods 0.000 description 9
- 210000002889 endothelial cell Anatomy 0.000 description 9
- 230000009368 gene silencing by RNA Effects 0.000 description 9
- 239000002609 medium Substances 0.000 description 9
- PSGAAPLEWMOORI-PEINSRQWSA-N medroxyprogesterone acetate Chemical compound C([C@@]12C)CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2CC[C@]2(C)[C@@](OC(C)=O)(C(C)=O)CC[C@H]21 PSGAAPLEWMOORI-PEINSRQWSA-N 0.000 description 9
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 9
- 238000002360 preparation method Methods 0.000 description 9
- 239000008223 sterile water Substances 0.000 description 9
- 238000006467 substitution reaction Methods 0.000 description 9
- 238000001356 surgical procedure Methods 0.000 description 9
- 238000012360 testing method Methods 0.000 description 9
- 230000035897 transcription Effects 0.000 description 9
- 238000013518 transcription Methods 0.000 description 9
- 102000053602 DNA Human genes 0.000 description 8
- 102100031780 Endonuclease Human genes 0.000 description 8
- 230000015556 catabolic process Effects 0.000 description 8
- 239000003795 chemical substances by application Substances 0.000 description 8
- 210000004087 cornea Anatomy 0.000 description 8
- 230000003247 decreasing effect Effects 0.000 description 8
- 238000006731 degradation reaction Methods 0.000 description 8
- 125000005647 linker group Chemical group 0.000 description 8
- 238000010172 mouse model Methods 0.000 description 8
- 102000004196 processed proteins & peptides Human genes 0.000 description 8
- 210000001525 retina Anatomy 0.000 description 8
- 239000000725 suspension Substances 0.000 description 8
- 208000024891 symptom Diseases 0.000 description 8
- 102000004190 Enzymes Human genes 0.000 description 7
- 108090000790 Enzymes Proteins 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 7
- 241000699670 Mus sp. Species 0.000 description 7
- 239000000074 antisense oligonucleotide Substances 0.000 description 7
- 238000012230 antisense oligonucleotides Methods 0.000 description 7
- 238000013459 approach Methods 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 230000001419 dependent effect Effects 0.000 description 7
- 150000002148 esters Chemical class 0.000 description 7
- 239000000796 flavoring agent Substances 0.000 description 7
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 7
- 239000010931 gold Substances 0.000 description 7
- 229910052737 gold Inorganic materials 0.000 description 7
- 230000012010 growth Effects 0.000 description 7
- 229910052739 hydrogen Inorganic materials 0.000 description 7
- 239000001257 hydrogen Substances 0.000 description 7
- 238000002513 implantation Methods 0.000 description 7
- 239000003112 inhibitor Substances 0.000 description 7
- 230000000977 initiatory effect Effects 0.000 description 7
- 210000004072 lung Anatomy 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 150000003833 nucleoside derivatives Chemical class 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 239000011780 sodium chloride Substances 0.000 description 7
- 230000000638 stimulation Effects 0.000 description 7
- OISVCGZHLKNMSJ-UHFFFAOYSA-N 2,6-dimethylpyridine Chemical compound CC1=CC=CC(C)=N1 OISVCGZHLKNMSJ-UHFFFAOYSA-N 0.000 description 6
- 108091027075 5S-rRNA precursor Proteins 0.000 description 6
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 6
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 6
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 6
- BAVYZALUXZFZLV-UHFFFAOYSA-N Methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 description 6
- 102000002067 Protein Subunits Human genes 0.000 description 6
- 241000700159 Rattus Species 0.000 description 6
- 108020004682 Single-Stranded DNA Proteins 0.000 description 6
- 229910052770 Uranium Inorganic materials 0.000 description 6
- 230000006427 angiogenic response Effects 0.000 description 6
- 230000004663 cell proliferation Effects 0.000 description 6
- 229960004397 cyclophosphamide Drugs 0.000 description 6
- NAGJZTKCGNOGPW-UHFFFAOYSA-K dioxido-sulfanylidene-sulfido-$l^{5}-phosphane Chemical compound [O-]P([O-])([S-])=S NAGJZTKCGNOGPW-UHFFFAOYSA-K 0.000 description 6
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 6
- 239000003446 ligand Substances 0.000 description 6
- 230000036961 partial effect Effects 0.000 description 6
- 239000008194 pharmaceutical composition Substances 0.000 description 6
- 239000000546 pharmaceutical excipient Substances 0.000 description 6
- 229920001223 polyethylene glycol Polymers 0.000 description 6
- 239000003755 preservative agent Substances 0.000 description 6
- 239000006228 supernatant Substances 0.000 description 6
- 239000003765 sweetening agent Substances 0.000 description 6
- 230000009885 systemic effect Effects 0.000 description 6
- 239000003826 tablet Substances 0.000 description 6
- 229940124597 therapeutic agent Drugs 0.000 description 6
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 6
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 6
- 239000013603 viral vector Substances 0.000 description 6
- UHDGCWIWMRVCDJ-UHFFFAOYSA-N 1-beta-D-Xylofuranosyl-NH-Cytosine Natural products O=C1N=C(N)C=CN1C1C(O)C(O)C(CO)O1 UHDGCWIWMRVCDJ-UHFFFAOYSA-N 0.000 description 5
- 108020004491 Antisense DNA Proteins 0.000 description 5
- UHDGCWIWMRVCDJ-PSQAKQOGSA-N Cytidine Natural products O=C1N=C(N)C=CN1[C@@H]1[C@@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-PSQAKQOGSA-N 0.000 description 5
- 230000007022 RNA scission Effects 0.000 description 5
- 239000007983 Tris buffer Substances 0.000 description 5
- 102000016549 Vascular Endothelial Growth Factor Receptor-2 Human genes 0.000 description 5
- 125000003342 alkenyl group Chemical group 0.000 description 5
- 125000000304 alkynyl group Chemical group 0.000 description 5
- 239000003816 antisense DNA Substances 0.000 description 5
- 210000004369 blood Anatomy 0.000 description 5
- 239000008280 blood Substances 0.000 description 5
- 238000001516 cell proliferation assay Methods 0.000 description 5
- 238000007385 chemical modification Methods 0.000 description 5
- 239000007859 condensation product Substances 0.000 description 5
- UHDGCWIWMRVCDJ-ZAKLUEHWSA-N cytidine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-ZAKLUEHWSA-N 0.000 description 5
- 229940104302 cytosine Drugs 0.000 description 5
- 235000014113 dietary fatty acids Nutrition 0.000 description 5
- 231100000673 dose–response relationship Toxicity 0.000 description 5
- 230000003828 downregulation Effects 0.000 description 5
- 229930195729 fatty acid Natural products 0.000 description 5
- 239000000194 fatty acid Substances 0.000 description 5
- 150000004665 fatty acids Chemical class 0.000 description 5
- 235000013355 food flavoring agent Nutrition 0.000 description 5
- 235000003599 food sweetener Nutrition 0.000 description 5
- 239000001963 growth medium Substances 0.000 description 5
- 239000007943 implant Substances 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 150000008300 phosphoramidites Chemical class 0.000 description 5
- 102000040430 polynucleotide Human genes 0.000 description 5
- 108091033319 polynucleotide Proteins 0.000 description 5
- 239000002157 polynucleotide Substances 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 230000001105 regulatory effect Effects 0.000 description 5
- 125000002652 ribonucleotide group Chemical group 0.000 description 5
- 239000000375 suspending agent Substances 0.000 description 5
- 238000007910 systemic administration Methods 0.000 description 5
- 230000005026 transcription initiation Effects 0.000 description 5
- 230000005030 transcription termination Effects 0.000 description 5
- 229940035893 uracil Drugs 0.000 description 5
- 102100027962 2-5A-dependent ribonuclease Human genes 0.000 description 4
- 108010000834 2-5A-dependent ribonuclease Proteins 0.000 description 4
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 4
- 206010002091 Anaesthesia Diseases 0.000 description 4
- 108090000644 Angiozyme Proteins 0.000 description 4
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 4
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 4
- 102000001493 Cyclophilins Human genes 0.000 description 4
- 108010068682 Cyclophilins Proteins 0.000 description 4
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 4
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 4
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 4
- 208000006552 Lewis Lung Carcinoma Diseases 0.000 description 4
- SECXISVLQFMRJM-UHFFFAOYSA-N N-Methylpyrrolidone Chemical compound CN1CCCC1=O SECXISVLQFMRJM-UHFFFAOYSA-N 0.000 description 4
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 4
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 4
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 4
- 108091028664 Ribonucleotide Proteins 0.000 description 4
- 238000009825 accumulation Methods 0.000 description 4
- 239000003377 acid catalyst Substances 0.000 description 4
- 125000003545 alkoxy group Chemical group 0.000 description 4
- 125000002877 alkyl aryl group Chemical group 0.000 description 4
- 230000003281 allosteric effect Effects 0.000 description 4
- 235000001014 amino acid Nutrition 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 230000037005 anaesthesia Effects 0.000 description 4
- 238000010171 animal model Methods 0.000 description 4
- 239000007900 aqueous suspension Substances 0.000 description 4
- 206010003246 arthritis Diseases 0.000 description 4
- 210000004204 blood vessel Anatomy 0.000 description 4
- 150000001720 carbohydrates Chemical class 0.000 description 4
- 235000014633 carbohydrates Nutrition 0.000 description 4
- 210000003169 central nervous system Anatomy 0.000 description 4
- 239000003086 colorant Substances 0.000 description 4
- 239000003433 contraceptive agent Substances 0.000 description 4
- 125000004093 cyano group Chemical group *C#N 0.000 description 4
- 238000013461 design Methods 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 239000002270 dispersing agent Substances 0.000 description 4
- 210000004696 endometrium Anatomy 0.000 description 4
- 210000001508 eye Anatomy 0.000 description 4
- 238000007429 general method Methods 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 229940088597 hormone Drugs 0.000 description 4
- 239000005556 hormone Substances 0.000 description 4
- 238000011081 inoculation Methods 0.000 description 4
- 229960003786 inosine Drugs 0.000 description 4
- 230000003834 intracellular effect Effects 0.000 description 4
- 238000007918 intramuscular administration Methods 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 210000004379 membrane Anatomy 0.000 description 4
- 230000001394 metastastic effect Effects 0.000 description 4
- 206010061289 metastatic neoplasm Diseases 0.000 description 4
- YACKEPLHDIMKIO-UHFFFAOYSA-N methylphosphonic acid Chemical group CP(O)(O)=O YACKEPLHDIMKIO-UHFFFAOYSA-N 0.000 description 4
- 238000002156 mixing Methods 0.000 description 4
- 231100000252 nontoxic Toxicity 0.000 description 4
- 230000003000 nontoxic effect Effects 0.000 description 4
- 229920001184 polypeptide Polymers 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 239000002336 ribonucleotide Substances 0.000 description 4
- 150000003839 salts Chemical group 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 239000012085 test solution Substances 0.000 description 4
- 231100000419 toxicity Toxicity 0.000 description 4
- 230000001988 toxicity Effects 0.000 description 4
- 230000014616 translation Effects 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- 239000000080 wetting agent Substances 0.000 description 4
- JUDOLRSMWHVKGX-UHFFFAOYSA-N 1,1-dioxo-1$l^{6},2-benzodithiol-3-one Chemical compound C1=CC=C2C(=O)SS(=O)(=O)C2=C1 JUDOLRSMWHVKGX-UHFFFAOYSA-N 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 229930024421 Adenine Natural products 0.000 description 3
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 3
- 241000710929 Alphavirus Species 0.000 description 3
- 201000004569 Blindness Diseases 0.000 description 3
- 101000909256 Caldicellulosiruptor bescii (strain ATCC BAA-1888 / DSM 6725 / Z-1320) DNA polymerase I Proteins 0.000 description 3
- 241000702421 Dependoparvovirus Species 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 102100024785 Fibroblast growth factor 2 Human genes 0.000 description 3
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 3
- 102000012673 Follicle Stimulating Hormone Human genes 0.000 description 3
- 108010079345 Follicle Stimulating Hormone Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102100029880 Glycodelin Human genes 0.000 description 3
- 108090001102 Hammerhead ribozyme Proteins 0.000 description 3
- 208000032843 Hemorrhage Diseases 0.000 description 3
- 101000585553 Homo sapiens Glycodelin Proteins 0.000 description 3
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical class O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 3
- 108091092195 Intron Proteins 0.000 description 3
- 102000009151 Luteinizing Hormone Human genes 0.000 description 3
- 108010073521 Luteinizing Hormone Proteins 0.000 description 3
- 206010027458 Metastases to lung Diseases 0.000 description 3
- 239000000020 Nitrocellulose Substances 0.000 description 3
- JCXJVPUVTGWSNB-UHFFFAOYSA-N Nitrogen dioxide Chemical compound O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 description 3
- 208000022873 Ocular disease Diseases 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 101000902592 Pyrococcus furiosus (strain ATCC 43587 / DSM 3638 / JCM 8422 / Vc1) DNA polymerase Proteins 0.000 description 3
- 230000006819 RNA synthesis Effects 0.000 description 3
- 206010038934 Retinopathy proliferative Diseases 0.000 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- NOSIYYJFMPDDSA-UHFFFAOYSA-N acepromazine Chemical compound C1=C(C(C)=O)C=C2N(CCCN(C)C)C3=CC=CC=C3SC2=C1 NOSIYYJFMPDDSA-UHFFFAOYSA-N 0.000 description 3
- 229960005054 acepromazine Drugs 0.000 description 3
- 125000002015 acyclic group Chemical group 0.000 description 3
- 229960000643 adenine Drugs 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 125000005103 alkyl silyl group Chemical group 0.000 description 3
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 3
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 3
- 230000000740 bleeding effect Effects 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 125000002837 carbocyclic group Chemical group 0.000 description 3
- 125000004432 carbon atom Chemical group C* 0.000 description 3
- 125000002091 cationic group Chemical group 0.000 description 3
- 108091036078 conserved sequence Proteins 0.000 description 3
- 230000002254 contraceptive effect Effects 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 229940063223 depo-provera Drugs 0.000 description 3
- 125000002147 dimethylamino group Chemical group [H]C([H])([H])N(*)C([H])([H])[H] 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- 239000003651 drinking water Substances 0.000 description 3
- 235000020188 drinking water Nutrition 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 229940011871 estrogen Drugs 0.000 description 3
- 239000000262 estrogen Substances 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 229940028334 follicle stimulating hormone Drugs 0.000 description 3
- 239000000499 gel Substances 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 239000003102 growth factor Substances 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 230000002452 interceptive effect Effects 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 230000004807 localization Effects 0.000 description 3
- 229940040129 luteinizing hormone Drugs 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 238000012423 maintenance Methods 0.000 description 3
- 231100000682 maximum tolerated dose Toxicity 0.000 description 3
- 229960002985 medroxyprogesterone acetate Drugs 0.000 description 3
- 239000003226 mitogen Substances 0.000 description 3
- 229920001220 nitrocellulos Polymers 0.000 description 3
- 238000002515 oligonucleotide synthesis Methods 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 239000001301 oxygen Substances 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 150000004713 phosphodiesters Chemical class 0.000 description 3
- 230000004962 physiological condition Effects 0.000 description 3
- 230000036470 plasma concentration Effects 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 229920000768 polyamine Polymers 0.000 description 3
- 239000000186 progesterone Substances 0.000 description 3
- 229960003387 progesterone Drugs 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 229940063222 provera Drugs 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 150000003290 ribose derivatives Chemical class 0.000 description 3
- 125000006413 ring segment Chemical group 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 229940113082 thymine Drugs 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- PUPZLCDOIYMWBV-UHFFFAOYSA-N (+/-)-1,3-Butanediol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 2
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 2
- FYADHXFMURLYQI-UHFFFAOYSA-N 1,2,4-triazine Chemical class C1=CN=NC=N1 FYADHXFMURLYQI-UHFFFAOYSA-N 0.000 description 2
- LKUDPHPHKOZXCD-UHFFFAOYSA-N 1,3,5-trimethoxybenzene Chemical compound COC1=CC(OC)=CC(OC)=C1 LKUDPHPHKOZXCD-UHFFFAOYSA-N 0.000 description 2
- MPCAJMNYNOGXPB-UHFFFAOYSA-N 1,5-anhydrohexitol Chemical class OCC1OCC(O)C(O)C1O MPCAJMNYNOGXPB-UHFFFAOYSA-N 0.000 description 2
- NEOJKYRRLHDYII-TURQNECASA-N 1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-(2-oxopropyl)pyrimidine-2,4-dione Chemical compound O=C1NC(=O)C(CC(=O)C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NEOJKYRRLHDYII-TURQNECASA-N 0.000 description 2
- SGKGZYGMLGVQHP-ZOQUXTDFSA-N 1-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6-methylpyrimidine-2,4-dione Chemical compound CC1=CC(=O)NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 SGKGZYGMLGVQHP-ZOQUXTDFSA-N 0.000 description 2
- CPKVUHPKYQGHMW-UHFFFAOYSA-N 1-ethenylpyrrolidin-2-one;molecular iodine Chemical compound II.C=CN1CCCC1=O CPKVUHPKYQGHMW-UHFFFAOYSA-N 0.000 description 2
- MCTWTZJPVLRJOU-UHFFFAOYSA-N 1-methyl-1H-imidazole Chemical compound CN1C=CN=C1 MCTWTZJPVLRJOU-UHFFFAOYSA-N 0.000 description 2
- GFYLSDSUCHVORB-IOSLPCCCSA-N 1-methyladenosine Chemical compound C1=NC=2C(=N)N(C)C=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O GFYLSDSUCHVORB-IOSLPCCCSA-N 0.000 description 2
- WJNGQIYEQLPJMN-IOSLPCCCSA-N 1-methylinosine Chemical compound C1=NC=2C(=O)N(C)C=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O WJNGQIYEQLPJMN-IOSLPCCCSA-N 0.000 description 2
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 2
- 102000007445 2',5'-Oligoadenylate Synthetase Human genes 0.000 description 2
- 108010086241 2',5'-Oligoadenylate Synthetase Proteins 0.000 description 2
- IQZWKGWOBPJWMX-UHFFFAOYSA-N 2-Methyladenosine Natural products C12=NC(C)=NC(N)=C2N=CN1C1OC(CO)C(O)C1O IQZWKGWOBPJWMX-UHFFFAOYSA-N 0.000 description 2
- UYZFIFAQZNYZNT-VUBKMPIKSA-N 2-[[2-[(2r,3s,4r,5r)-3,4-dihydroxy-5-(4-oxo-2-sulfanylidenepyrimidin-1-yl)oxolan-2-yl]-2-hydroxyethyl]amino]acetic acid Chemical compound O[C@@H]1[C@H](O)[C@@H](C(CNCC(O)=O)O)O[C@H]1N1C(=S)NC(=O)C=C1 UYZFIFAQZNYZNT-VUBKMPIKSA-N 0.000 description 2
- IZHVBANLECCAGF-UHFFFAOYSA-N 2-hydroxy-3-(octadecanoyloxy)propyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)COC(=O)CCCCCCCCCCCCCCCCC IZHVBANLECCAGF-UHFFFAOYSA-N 0.000 description 2
- XWKFPIODWVPXLX-UHFFFAOYSA-N 2-methyl-5-methylpyridine Natural products CC1=CC=C(C)N=C1 XWKFPIODWVPXLX-UHFFFAOYSA-N 0.000 description 2
- IQZWKGWOBPJWMX-IOSLPCCCSA-N 2-methyladenosine Chemical compound C12=NC(C)=NC(N)=C2N=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O IQZWKGWOBPJWMX-IOSLPCCCSA-N 0.000 description 2
- VZQXUWKZDSEQRR-SDBHATRESA-N 2-methylthio-N(6)-(Delta(2)-isopentenyl)adenosine Chemical compound C12=NC(SC)=NC(NCC=C(C)C)=C2N=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O VZQXUWKZDSEQRR-SDBHATRESA-N 0.000 description 2
- RHFUOMFWUGWKKO-XVFCMESISA-N 2-thiocytidine Chemical compound S=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 RHFUOMFWUGWKKO-XVFCMESISA-N 0.000 description 2
- GJTBSTBJLVYKAU-XVFCMESISA-N 2-thiouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=S)NC(=O)C=C1 GJTBSTBJLVYKAU-XVFCMESISA-N 0.000 description 2
- RDPUKVRQKWBSPK-UHFFFAOYSA-N 3-Methylcytidine Natural products O=C1N(C)C(=N)C=CN1C1C(O)C(O)C(CO)O1 RDPUKVRQKWBSPK-UHFFFAOYSA-N 0.000 description 2
- RDPUKVRQKWBSPK-ZOQUXTDFSA-N 3-methylcytidine Chemical compound O=C1N(C)C(=N)C=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 RDPUKVRQKWBSPK-ZOQUXTDFSA-N 0.000 description 2
- VPLZGVOSFFCKFC-UHFFFAOYSA-N 3-methyluracil Chemical compound CN1C(=O)C=CNC1=O VPLZGVOSFFCKFC-UHFFFAOYSA-N 0.000 description 2
- ZLOIGESWDJYCTF-UHFFFAOYSA-N 4-Thiouridine Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=S)C=C1 ZLOIGESWDJYCTF-UHFFFAOYSA-N 0.000 description 2
- BCZUPRDAAVVBSO-MJXNYTJMSA-N 4-acetylcytidine Chemical compound C1=CC(C(=O)C)(N)NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 BCZUPRDAAVVBSO-MJXNYTJMSA-N 0.000 description 2
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 2
- LZINOQJQXIEBNN-UHFFFAOYSA-N 4-hydroxybutyl dihydrogen phosphate Chemical compound OCCCCOP(O)(O)=O LZINOQJQXIEBNN-UHFFFAOYSA-N 0.000 description 2
- GCNTZFIIOFTKIY-UHFFFAOYSA-N 4-hydroxypyridine Chemical compound OC1=CC=NC=C1 GCNTZFIIOFTKIY-UHFFFAOYSA-N 0.000 description 2
- ZLOIGESWDJYCTF-XVFCMESISA-N 4-thiouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=S)C=C1 ZLOIGESWDJYCTF-XVFCMESISA-N 0.000 description 2
- UVGCZRPOXXYZKH-QADQDURISA-N 5-(carboxyhydroxymethyl)uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(C(O)C(O)=O)=C1 UVGCZRPOXXYZKH-QADQDURISA-N 0.000 description 2
- VSCNRXVDHRNJOA-PNHWDRBUSA-N 5-(carboxymethylaminomethyl)uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(CNCC(O)=O)=C1 VSCNRXVDHRNJOA-PNHWDRBUSA-N 0.000 description 2
- ZAYHVCMSTBRABG-UHFFFAOYSA-N 5-Methylcytidine Natural products O=C1N=C(N)C(C)=CN1C1C(O)C(O)C(CO)O1 ZAYHVCMSTBRABG-UHFFFAOYSA-N 0.000 description 2
- AGFIRQJZCNVMCW-UAKXSSHOSA-N 5-bromouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(Br)=C1 AGFIRQJZCNVMCW-UAKXSSHOSA-N 0.000 description 2
- RJUNHHFZFRMZQQ-FDDDBJFASA-N 5-methoxyaminomethyl-2-thiouridine Chemical compound S=C1NC(=O)C(CNOC)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 RJUNHHFZFRMZQQ-FDDDBJFASA-N 0.000 description 2
- ZXIATBNUWJBBGT-JXOAFFINSA-N 5-methoxyuridine Chemical compound O=C1NC(=O)C(OC)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 ZXIATBNUWJBBGT-JXOAFFINSA-N 0.000 description 2
- SNNBPMAXGYBMHM-JXOAFFINSA-N 5-methyl-2-thiouridine Chemical compound S=C1NC(=O)C(C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 SNNBPMAXGYBMHM-JXOAFFINSA-N 0.000 description 2
- ZXQHKBUIXRFZBV-FDDDBJFASA-N 5-methylaminomethyluridine Chemical compound O=C1NC(=O)C(CNC)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 ZXQHKBUIXRFZBV-FDDDBJFASA-N 0.000 description 2
- ZAYHVCMSTBRABG-JXOAFFINSA-N 5-methylcytidine Chemical compound O=C1N=C(N)C(C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 ZAYHVCMSTBRABG-JXOAFFINSA-N 0.000 description 2
- OGHAROSJZRTIOK-KQYNXXCUSA-O 7-methylguanosine Chemical compound C1=2N=C(N)NC(=O)C=2[N+](C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OGHAROSJZRTIOK-KQYNXXCUSA-O 0.000 description 2
- 244000215068 Acacia senegal Species 0.000 description 2
- 235000006491 Acacia senegal Nutrition 0.000 description 2
- 108020005544 Antisense RNA Proteins 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 241000416162 Astragalus gummifer Species 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 229930003347 Atropine Natural products 0.000 description 2
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 2
- 208000016216 Choristoma Diseases 0.000 description 2
- 101710177611 DNA polymerase II large subunit Proteins 0.000 description 2
- 101710184669 DNA polymerase II small subunit Proteins 0.000 description 2
- 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 2
- 102100023387 Endoribonuclease Dicer Human genes 0.000 description 2
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- 108091027874 Group I catalytic intron Proteins 0.000 description 2
- 208000012766 Growth delay Diseases 0.000 description 2
- 229920000084 Gum arabic Polymers 0.000 description 2
- 241000724709 Hepatitis delta virus Species 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000907904 Homo sapiens Endoribonuclease Dicer Proteins 0.000 description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 2
- RKUNBYITZUJHSG-UHFFFAOYSA-N Hyosciamin-hydrochlorid Natural products CN1C(C2)CCC1CC2OC(=O)C(CO)C1=CC=CC=C1 RKUNBYITZUJHSG-UHFFFAOYSA-N 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 206010022095 Injection Site reaction Diseases 0.000 description 2
- 229930010555 Inosine Natural products 0.000 description 2
- YQEZLKZALYSWHR-UHFFFAOYSA-N Ketamine Chemical compound C=1C=CC=C(Cl)C=1C1(NC)CCCCC1=O YQEZLKZALYSWHR-UHFFFAOYSA-N 0.000 description 2
- NNJVILVZKWQKPM-UHFFFAOYSA-N Lidocaine Chemical compound CCN(CC)CC(=O)NC1=C(C)C=CC=C1C NNJVILVZKWQKPM-UHFFFAOYSA-N 0.000 description 2
- 241000699660 Mus musculus Species 0.000 description 2
- 108010021466 Mutant Proteins Proteins 0.000 description 2
- 102000008300 Mutant Proteins Human genes 0.000 description 2
- RSPURTUNRHNVGF-IOSLPCCCSA-N N(2),N(2)-dimethylguanosine Chemical compound C1=NC=2C(=O)NC(N(C)C)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O RSPURTUNRHNVGF-IOSLPCCCSA-N 0.000 description 2
- VQAYFKKCNSOZKM-IOSLPCCCSA-N N(6)-methyladenosine Chemical compound C1=NC=2C(NC)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O VQAYFKKCNSOZKM-IOSLPCCCSA-N 0.000 description 2
- VQAYFKKCNSOZKM-UHFFFAOYSA-N NSC 29409 Natural products C1=NC=2C(NC)=NC=NC=2N1C1OC(CO)C(O)C1O VQAYFKKCNSOZKM-UHFFFAOYSA-N 0.000 description 2
- 241000221960 Neurospora Species 0.000 description 2
- VZQXUWKZDSEQRR-UHFFFAOYSA-N Nucleosid Natural products C12=NC(SC)=NC(NCC=C(C)C)=C2N=CN1C1OC(CO)C(O)C1O VZQXUWKZDSEQRR-UHFFFAOYSA-N 0.000 description 2
- 108700020796 Oncogene Proteins 0.000 description 2
- 102000010292 Peptide Elongation Factor 1 Human genes 0.000 description 2
- 108010077524 Peptide Elongation Factor 1 Proteins 0.000 description 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- 239000004952 Polyamide Substances 0.000 description 2
- 229920000153 Povidone-iodine Polymers 0.000 description 2
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 2
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 2
- 208000017442 Retinal disease Diseases 0.000 description 2
- 206010038923 Retinopathy Diseases 0.000 description 2
- 108090000621 Ribonuclease P Proteins 0.000 description 2
- 102000004167 Ribonuclease P Human genes 0.000 description 2
- 108010083644 Ribonucleases Proteins 0.000 description 2
- 102000006382 Ribonucleases Human genes 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- 229920002472 Starch Polymers 0.000 description 2
- 241000529895 Stercorarius Species 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 241000282898 Sus scrofa Species 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 2
- 229920001615 Tragacanth Polymers 0.000 description 2
- 108020004566 Transfer RNA Proteins 0.000 description 2
- 235000010489 acacia gum Nutrition 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 150000001408 amides Chemical class 0.000 description 2
- 235000012538 ammonium bicarbonate Nutrition 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000001028 anti-proliverative effect Effects 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- RKUNBYITZUJHSG-SPUOUPEWSA-N atropine Chemical compound O([C@H]1C[C@H]2CC[C@@H](C1)N2C)C(=O)C(CO)C1=CC=CC=C1 RKUNBYITZUJHSG-SPUOUPEWSA-N 0.000 description 2
- 229960000396 atropine Drugs 0.000 description 2
- 230000003190 augmentative effect Effects 0.000 description 2
- 230000037429 base substitution Effects 0.000 description 2
- HMFHBZSHGGEWLO-TXICZTDVSA-N beta-D-ribose Chemical group OC[C@H]1O[C@@H](O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-TXICZTDVSA-N 0.000 description 2
- 229920002988 biodegradable polymer Polymers 0.000 description 2
- 239000004621 biodegradable polymer Substances 0.000 description 2
- 229920001222 biopolymer Polymers 0.000 description 2
- 230000036770 blood supply Effects 0.000 description 2
- 229910000019 calcium carbonate Inorganic materials 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 238000001816 cooling Methods 0.000 description 2
- 230000000875 corresponding effect Effects 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- 230000003111 delayed effect Effects 0.000 description 2
- 238000010511 deprotection reaction Methods 0.000 description 2
- 238000006642 detritylation reaction Methods 0.000 description 2
- 235000005911 diet Nutrition 0.000 description 2
- 230000037213 diet Effects 0.000 description 2
- ZPTBLXKRQACLCR-XVFCMESISA-N dihydrouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)CC1 ZPTBLXKRQACLCR-XVFCMESISA-N 0.000 description 2
- 238000012377 drug delivery Methods 0.000 description 2
- 208000023965 endometrium neoplasm Diseases 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 238000001415 gene therapy Methods 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 229910052736 halogen Inorganic materials 0.000 description 2
- 150000002367 halogens Chemical class 0.000 description 2
- 125000005842 heteroatom Chemical group 0.000 description 2
- BXWNKGSJHAJOGX-UHFFFAOYSA-N hexadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO BXWNKGSJHAJOGX-UHFFFAOYSA-N 0.000 description 2
- FBPFZTCFMRRESA-UHFFFAOYSA-N hexane-1,2,3,4,5,6-hexol Chemical compound OCC(O)C(O)C(O)C(O)CO FBPFZTCFMRRESA-UHFFFAOYSA-N 0.000 description 2
- 150000002430 hydrocarbons Chemical group 0.000 description 2
- 230000007062 hydrolysis Effects 0.000 description 2
- 238000006460 hydrolysis reaction Methods 0.000 description 2
- 238000009802 hysterectomy Methods 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 230000008863 intramolecular interaction Effects 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 229960003299 ketamine Drugs 0.000 description 2
- 238000002357 laparoscopic surgery Methods 0.000 description 2
- 239000000787 lecithin Substances 0.000 description 2
- 235000010445 lecithin Nutrition 0.000 description 2
- 229940067606 lecithin Drugs 0.000 description 2
- 229960004194 lidocaine Drugs 0.000 description 2
- 229940057995 liquid paraffin Drugs 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 230000010534 mechanism of action Effects 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 150000002739 metals Chemical class 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 229920000609 methyl cellulose Polymers 0.000 description 2
- 239000001923 methylcellulose Substances 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 0.000 description 2
- 230000009149 molecular binding Effects 0.000 description 2
- 125000001624 naphthyl group Chemical group 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 239000000346 nonvolatile oil Substances 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 235000019198 oils Nutrition 0.000 description 2
- 239000004006 olive oil Substances 0.000 description 2
- 235000008390 olive oil Nutrition 0.000 description 2
- 238000005580 one pot reaction Methods 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 230000003204 osmotic effect Effects 0.000 description 2
- 230000003647 oxidation Effects 0.000 description 2
- 238000007254 oxidation reaction Methods 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 239000008177 pharmaceutical agent Substances 0.000 description 2
- 230000003285 pharmacodynamic effect Effects 0.000 description 2
- 239000008196 pharmacological composition Substances 0.000 description 2
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 229920002401 polyacrylamide Polymers 0.000 description 2
- 229920002647 polyamide Polymers 0.000 description 2
- 229920000570 polyether Polymers 0.000 description 2
- 238000010149 post-hoc-test Methods 0.000 description 2
- 238000012809 post-inoculation Methods 0.000 description 2
- 230000032361 posttranscriptional gene silencing Effects 0.000 description 2
- 229960001621 povidone-iodine Drugs 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 2
- MWWATHDPGQKSAR-UHFFFAOYSA-N propyne Chemical compound CC#C MWWATHDPGQKSAR-UHFFFAOYSA-N 0.000 description 2
- UBQKCCHYAOITMY-UHFFFAOYSA-N pyridin-2-ol Chemical compound OC1=CC=CC=N1 UBQKCCHYAOITMY-UHFFFAOYSA-N 0.000 description 2
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 2
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 2
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 230000033458 reproduction Effects 0.000 description 2
- 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 2
- RHFUOMFWUGWKKO-UHFFFAOYSA-N s2C Natural products S=C1N=C(N)C=CN1C1C(O)C(O)C(CO)O1 RHFUOMFWUGWKKO-UHFFFAOYSA-N 0.000 description 2
- 229920006395 saturated elastomer Polymers 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 239000001593 sorbitan monooleate Substances 0.000 description 2
- 235000011069 sorbitan monooleate Nutrition 0.000 description 2
- 229940035049 sorbitan monooleate Drugs 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 239000008107 starch Substances 0.000 description 2
- 235000019698 starch Nutrition 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 239000005451 thionucleotide Substances 0.000 description 2
- 150000003587 threonine derivatives Chemical class 0.000 description 2
- 239000003053 toxin Substances 0.000 description 2
- 231100000765 toxin Toxicity 0.000 description 2
- 108700012359 toxins Proteins 0.000 description 2
- 230000010474 transient expression Effects 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- 125000002221 trityl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1C([*])(C1=C(C(=C(C(=C1[H])[H])[H])[H])[H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 2
- 229930195735 unsaturated hydrocarbon Natural products 0.000 description 2
- RVCNQQGZJWVLIP-VPCXQMTMSA-N uridin-5-yloxyacetic acid Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(OCC(O)=O)=C1 RVCNQQGZJWVLIP-VPCXQMTMSA-N 0.000 description 2
- 210000004291 uterus Anatomy 0.000 description 2
- 230000002792 vascular Effects 0.000 description 2
- 210000003556 vascular endothelial cell Anatomy 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 230000007998 vessel formation Effects 0.000 description 2
- QAOHCFGKCWTBGC-QHOAOGIMSA-N wybutosine Chemical compound C1=NC=2C(=O)N3C(CC[C@H](NC(=O)OC)C(=O)OC)=C(C)N=C3N(C)C=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O QAOHCFGKCWTBGC-QHOAOGIMSA-N 0.000 description 2
- QAOHCFGKCWTBGC-UHFFFAOYSA-N wybutosine Natural products C1=NC=2C(=O)N3C(CCC(NC(=O)OC)C(=O)OC)=C(C)N=C3N(C)C=2N1C1OC(CO)C(O)C1O QAOHCFGKCWTBGC-UHFFFAOYSA-N 0.000 description 2
- WCNMEQDMUYVWMJ-JPZHCBQBSA-N wybutoxosine Chemical compound C1=NC=2C(=O)N3C(CC([C@H](NC(=O)OC)C(=O)OC)OO)=C(C)N=C3N(C)C=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O WCNMEQDMUYVWMJ-JPZHCBQBSA-N 0.000 description 2
- 229960001600 xylazine Drugs 0.000 description 2
- BPICBUSOMSTKRF-UHFFFAOYSA-N xylazine Chemical compound CC1=CC=CC(C)=C1NC1=NCCCS1 BPICBUSOMSTKRF-UHFFFAOYSA-N 0.000 description 2
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- ASJSAQIRZKANQN-CRCLSJGQSA-N 2-deoxy-D-ribose Chemical compound OC[C@@H](O)[C@@H](O)CC=O ASJSAQIRZKANQN-CRCLSJGQSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- KUQZVISZELWDNZ-UHFFFAOYSA-N 3-aminopropyl dihydrogen phosphate Chemical compound NCCCOP(O)(O)=O KUQZVISZELWDNZ-UHFFFAOYSA-N 0.000 description 1
- HYCSHFLKPSMPGO-UHFFFAOYSA-N 3-hydroxypropyl dihydrogen phosphate Chemical compound OCCCOP(O)(O)=O HYCSHFLKPSMPGO-UHFFFAOYSA-N 0.000 description 1
- UPXRTVAIJMUAQR-UHFFFAOYSA-N 4-(9h-fluoren-9-ylmethoxycarbonylamino)-1-[(2-methylpropan-2-yl)oxycarbonyl]pyrrolidine-2-carboxylic acid Chemical compound C1C(C(O)=O)N(C(=O)OC(C)(C)C)CC1NC(=O)OCC1C2=CC=CC=C2C2=CC=CC=C21 UPXRTVAIJMUAQR-UHFFFAOYSA-N 0.000 description 1
- 229940090248 4-hydroxybenzoic acid Drugs 0.000 description 1
- TYJOQICPGZGYDT-UHFFFAOYSA-N 4-methylsulfonylbenzenesulfonyl chloride Chemical compound CS(=O)(=O)C1=CC=C(S(Cl)(=O)=O)C=C1 TYJOQICPGZGYDT-UHFFFAOYSA-N 0.000 description 1
- XYVLZAYJHCECPN-UHFFFAOYSA-N 6-aminohexyl phosphate Chemical compound NCCCCCCOP(O)(O)=O XYVLZAYJHCECPN-UHFFFAOYSA-N 0.000 description 1
- XYVLZAYJHCECPN-UHFFFAOYSA-L 6-aminohexyl phosphate Chemical compound NCCCCCCOP([O-])([O-])=O XYVLZAYJHCECPN-UHFFFAOYSA-L 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- DLFVBJFMPXGRIB-UHFFFAOYSA-N Acetamide Chemical compound CC(N)=O DLFVBJFMPXGRIB-UHFFFAOYSA-N 0.000 description 1
- 102100022712 Alpha-1-antitrypsin Human genes 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- 208000003120 Angiofibroma Diseases 0.000 description 1
- 102100029470 Apolipoprotein E Human genes 0.000 description 1
- 101710095339 Apolipoprotein E Proteins 0.000 description 1
- 235000003911 Arachis Nutrition 0.000 description 1
- 244000105624 Arachis hypogaea Species 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- KXDHJXZQYSOELW-UHFFFAOYSA-M Carbamate Chemical compound NC([O-])=O KXDHJXZQYSOELW-UHFFFAOYSA-M 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 102000003908 Cathepsin D Human genes 0.000 description 1
- 108090000258 Cathepsin D Proteins 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 102000012272 Cholestanetriol 26-monooxygenase Human genes 0.000 description 1
- 108010022102 Cholestanetriol 26-monooxygenase Proteins 0.000 description 1
- 102100038447 Claudin-4 Human genes 0.000 description 1
- 108090000601 Claudin-4 Proteins 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 108010035532 Collagen Proteins 0.000 description 1
- 102000008186 Collagen Human genes 0.000 description 1
- 102100036213 Collagen alpha-2(I) chain Human genes 0.000 description 1
- 101710126238 Collagen alpha-2(I) chain Proteins 0.000 description 1
- 108010028780 Complement C3 Proteins 0.000 description 1
- 102000016918 Complement C3 Human genes 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- 108010061635 Cystatin B Proteins 0.000 description 1
- 102100026891 Cystatin-B Human genes 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- XULFJDKZVHTRLG-JDVCJPALSA-N DOSPA trifluoroacetate Chemical compound [O-]C(=O)C(F)(F)F.CCCCCCCC\C=C/CCCCCCCCOCC(C[N+](C)(C)CCNC(=O)C(CCCNCCCN)NCCCN)OCCCCCCCC\C=C/CCCCCCCC XULFJDKZVHTRLG-JDVCJPALSA-N 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 102100029952 Double-strand-break repair protein rad21 homolog Human genes 0.000 description 1
- 102100029791 Double-stranded RNA-specific adenosine deaminase Human genes 0.000 description 1
- 102000010778 Dual Specificity Phosphatase 1 Human genes 0.000 description 1
- 108010038537 Dual Specificity Phosphatase 1 Proteins 0.000 description 1
- 108010093099 Endoribonucleases Proteins 0.000 description 1
- 102000002494 Endoribonucleases Human genes 0.000 description 1
- 206010015150 Erythema Diseases 0.000 description 1
- 101000686777 Escherichia phage T7 T7 RNA polymerase Proteins 0.000 description 1
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical class OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 1
- 108060002716 Exonuclease Proteins 0.000 description 1
- 206010015866 Extravasation Diseases 0.000 description 1
- 108010060374 FSH Receptors Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000008857 Ferritin Human genes 0.000 description 1
- 108050000784 Ferritin Proteins 0.000 description 1
- 238000008416 Ferritin Methods 0.000 description 1
- 102000018343 Follicle stimulating hormone receptors Human genes 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 102000034286 G proteins Human genes 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 240000005702 Galium aparine Species 0.000 description 1
- 235000014820 Galium aparine Nutrition 0.000 description 1
- 102000004878 Gelsolin Human genes 0.000 description 1
- 108090001064 Gelsolin Proteins 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108091029499 Group II intron Proteins 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 206010019695 Hepatic neoplasm Diseases 0.000 description 1
- 108091027305 Heteroduplex Proteins 0.000 description 1
- 241001272567 Hominoidea Species 0.000 description 1
- 101000584942 Homo sapiens Double-strand-break repair protein rad21 homolog Proteins 0.000 description 1
- 101000865408 Homo sapiens Double-stranded RNA-specific adenosine deaminase Proteins 0.000 description 1
- 101000612134 Homo sapiens Procollagen C-endopeptidase enhancer 1 Proteins 0.000 description 1
- 101000808011 Homo sapiens Vascular endothelial growth factor A Proteins 0.000 description 1
- 108010070875 Human Immunodeficiency Virus tat Gene Products Proteins 0.000 description 1
- 102000016844 Immunoglobulin-like domains Human genes 0.000 description 1
- 108050006430 Immunoglobulin-like domains Proteins 0.000 description 1
- 102000004374 Insulin-like growth factor binding protein 3 Human genes 0.000 description 1
- 108090000965 Insulin-like growth factor binding protein 3 Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 206010073324 Keratinising squamous cell carcinoma of nasopharynx Diseases 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 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
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 108090000004 Leadzyme Proteins 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- 108010076502 Matrix Metalloproteinase 11 Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- SLEHROROQDYRAW-KQYNXXCUSA-N N(2)-methylguanosine Chemical compound C1=NC=2C(=O)NC(NC)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O SLEHROROQDYRAW-KQYNXXCUSA-N 0.000 description 1
- 102100026779 Nascent polypeptide-associated complex subunit alpha, muscle-specific form Human genes 0.000 description 1
- 208000002454 Nasopharyngeal Carcinoma Diseases 0.000 description 1
- 206010061306 Nasopharyngeal cancer Diseases 0.000 description 1
- 240000007817 Olea europaea Species 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 208000034038 Pathologic Neovascularization Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 208000000450 Pelvic Pain Diseases 0.000 description 1
- 229940049937 Pgp inhibitor Drugs 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 229920002730 Poly(butyl cyanoacrylate) Polymers 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 108010021757 Polynucleotide 5'-Hydroxyl-Kinase Proteins 0.000 description 1
- 102000008422 Polynucleotide 5'-hydroxyl-kinase Human genes 0.000 description 1
- 239000004721 Polyphenylene oxide Substances 0.000 description 1
- 208000006787 Port-Wine Stain Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 102100041026 Procollagen C-endopeptidase enhancer 1 Human genes 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 102100036197 Prosaposin Human genes 0.000 description 1
- 101710152403 Prosaposin Proteins 0.000 description 1
- 101710115194 Protease inhibitor 1 Proteins 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 108010013845 RNA Polymerase I Proteins 0.000 description 1
- 102000017143 RNA Polymerase I Human genes 0.000 description 1
- 108010009460 RNA Polymerase II Proteins 0.000 description 1
- 102000009572 RNA Polymerase II Human genes 0.000 description 1
- 108091008103 RNA aptamers Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 208000007135 Retinal Neovascularization Diseases 0.000 description 1
- 206010038933 Retinopathy of prematurity Diseases 0.000 description 1
- 102000003861 Ribosomal protein S6 Human genes 0.000 description 1
- 108090000221 Ribosomal protein S6 Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 229920005654 Sephadex Polymers 0.000 description 1
- 239000012507 Sephadex™ Substances 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 102000049867 Steroidogenic acute regulatory protein Human genes 0.000 description 1
- 108010018411 Steroidogenic acute regulatory protein Proteins 0.000 description 1
- 102000017303 Stromelysin-3 Human genes 0.000 description 1
- 206010042265 Sturge-Weber Syndrome Diseases 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 241001365914 Taira Species 0.000 description 1
- 244000121910 Talauma ovata Species 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 102000011409 Transcobalamins Human genes 0.000 description 1
- 108010023603 Transcobalamins Proteins 0.000 description 1
- 208000026911 Tuberous sclerosis complex Diseases 0.000 description 1
- 206010054094 Tumour necrosis Diseases 0.000 description 1
- 108091034131 VA RNA Proteins 0.000 description 1
- 108010053096 Vascular Endothelial Growth Factor Receptor-1 Proteins 0.000 description 1
- 102100033178 Vascular endothelial growth factor receptor 1 Human genes 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 208000013058 Weber syndrome Diseases 0.000 description 1
- SIIZPVYVXNXXQG-KGXOGWRBSA-N [(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-4-[[(3s,4r)-5-(6-aminopurin-9-yl)-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-3-hydroxyoxolan-2-yl]methyl [(2r,4r,5r)-2-(6-aminopurin-9-yl)-4-hydroxy-5-(phosphonooxymethyl)oxolan-3-yl] hydrogen phosphate Polymers C1=NC2=C(N)N=CN=C2N1[C@@H]1O[C@H](COP(O)(=O)OC2[C@@H](O[C@H](COP(O)(O)=O)[C@H]2O)N2C3=NC=NC(N)=C3N=C2)[C@@H](O)[C@H]1OP(O)(=O)OCC([C@@H](O)[C@H]1O)OC1N1C(N=CN=C2N)=C2N=C1 SIIZPVYVXNXXQG-KGXOGWRBSA-N 0.000 description 1
- JCAQMQLAHNGVPY-UUOKFMHZSA-N [(2r,3s,4r,5r)-3,4-dihydroxy-5-(2,2,4-trioxo-1h-imidazo[4,5-c][1,2,6]thiadiazin-7-yl)oxolan-2-yl]methyl dihydrogen phosphate Chemical compound O[C@@H]1[C@H](O)[C@@H](COP(O)(O)=O)O[C@H]1N1C(NS(=O)(=O)NC2=O)=C2N=C1 JCAQMQLAHNGVPY-UUOKFMHZSA-N 0.000 description 1
- WERKSKAQRVDLDW-ANOHMWSOSA-N [(2s,3r,4r,5r)-2,3,4,5,6-pentahydroxyhexyl] (z)-octadec-9-enoate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO WERKSKAQRVDLDW-ANOHMWSOSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000011149 active material Substances 0.000 description 1
- UDMBCSSLTHHNCD-KQYNXXCUSA-N adenosine 5'-monophosphate Chemical group C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H]1O UDMBCSSLTHHNCD-KQYNXXCUSA-N 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 150000001338 aliphatic hydrocarbons Chemical class 0.000 description 1
- 125000002947 alkylene group Chemical group 0.000 description 1
- 239000012637 allosteric effector Substances 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 229940059260 amidate Drugs 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 150000008064 anhydrides Chemical class 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000002001 anti-metastasis Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 210000001142 back Anatomy 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 235000013871 bee wax Nutrition 0.000 description 1
- 239000012166 beeswax Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 229940092714 benzenesulfonic acid Drugs 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 125000005841 biaryl group Chemical group 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 239000000227 bioadhesive Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000008236 biological pathway Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 208000034158 bleeding Diseases 0.000 description 1
- 230000008499 blood brain barrier function Effects 0.000 description 1
- 230000017531 blood circulation Effects 0.000 description 1
- 210000001218 blood-brain barrier Anatomy 0.000 description 1
- 230000036471 bradycardia Effects 0.000 description 1
- 208000006218 bradycardia Diseases 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 201000007293 brain stem infarction Diseases 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- UDSAIICHUKSCKT-UHFFFAOYSA-N bromophenol blue Chemical compound C1=C(Br)C(O)=C(Br)C=C1C1(C=2C=C(Br)C(O)=C(Br)C=2)C2=CC=CC=C2S(=O)(=O)O1 UDSAIICHUKSCKT-UHFFFAOYSA-N 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 244000309464 bull Species 0.000 description 1
- 235000010216 calcium carbonate Nutrition 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 125000002057 carboxymethyl group Chemical group [H]OC(=O)C([H])([H])[*] 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000012876 carrier material Substances 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 238000006555 catalytic reaction Methods 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 229960000541 cetyl alcohol Drugs 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000002144 chemical decomposition reaction Methods 0.000 description 1
- 230000000973 chemotherapeutic effect Effects 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 239000003240 coconut oil Substances 0.000 description 1
- 235000019864 coconut oil Nutrition 0.000 description 1
- 229920001436 collagen Polymers 0.000 description 1
- 201000004196 common wart Diseases 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 230000000536 complexating effect Effects 0.000 description 1
- 229940124301 concurrent medication Drugs 0.000 description 1
- 229940124558 contraceptive agent Drugs 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 238000011461 current therapy Methods 0.000 description 1
- 125000006165 cyclic alkyl group Chemical group 0.000 description 1
- 229940097362 cyclodextrins Drugs 0.000 description 1
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- 230000003412 degenerative effect Effects 0.000 description 1
- 230000004340 degenerative myopia Effects 0.000 description 1
- 238000002716 delivery method Methods 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-N dithiophosphoric acid Chemical class OP(O)(S)=S NAGJZTKCGNOGPW-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 230000000565 effect on metastases Effects 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007515 enzymatic degradation Effects 0.000 description 1
- 231100000321 erythema Toxicity 0.000 description 1
- 125000004185 ester group Chemical group 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- NPUKDXXFDDZOKR-LLVKDONJSA-N etomidate Chemical compound CCOC(=O)C1=CN=CN1[C@H](C)C1=CC=CC=C1 NPUKDXXFDDZOKR-LLVKDONJSA-N 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 102000013165 exonuclease Human genes 0.000 description 1
- 230000036251 extravasation Effects 0.000 description 1
- 210000003414 extremity Anatomy 0.000 description 1
- 210000000744 eyelid Anatomy 0.000 description 1
- 210000000887 face Anatomy 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 230000005021 gait Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229940074045 glyceryl distearate Drugs 0.000 description 1
- 125000003976 glyceryl group Chemical group [H]C([*])([H])C(O[H])([H])C(O[H])([H])[H] 0.000 description 1
- 239000002748 glycoprotein P inhibitor Substances 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- PHNWGDTYCJFUGZ-UHFFFAOYSA-L hexyl phosphate Chemical compound CCCCCCOP([O-])([O-])=O PHNWGDTYCJFUGZ-UHFFFAOYSA-L 0.000 description 1
- 230000003054 hormonal effect Effects 0.000 description 1
- 239000003688 hormone derivative Substances 0.000 description 1
- 102000058223 human VEGFA Human genes 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000000222 hyperoxic effect Effects 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 230000008073 immune recognition Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 208000000509 infertility Diseases 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 231100000535 infertility Toxicity 0.000 description 1
- 231100000546 inhibition of ovulation Toxicity 0.000 description 1
- 229940102223 injectable solution Drugs 0.000 description 1
- 229940102213 injectable suspension Drugs 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 230000002262 irrigation Effects 0.000 description 1
- 238000003973 irrigation Methods 0.000 description 1
- 230000000622 irritating effect Effects 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 210000004731 jugular vein Anatomy 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 229960004184 ketamine hydrochloride Drugs 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 238000002350 laparotomy Methods 0.000 description 1
- 108020001756 ligand binding domains Proteins 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 239000003589 local anesthetic agent Substances 0.000 description 1
- 229960005015 local anesthetics Drugs 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 210000004914 menses Anatomy 0.000 description 1
- 230000005906 menstruation Effects 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 210000004925 microvascular endothelial cell Anatomy 0.000 description 1
- 230000002297 mitogenic effect Effects 0.000 description 1
- 238000000329 molecular dynamics simulation Methods 0.000 description 1
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 238000013425 morphometry Methods 0.000 description 1
- 230000037023 motor activity Effects 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 108010037351 nascent-polypeptide-associated complex Proteins 0.000 description 1
- 208000028133 nasopharyngeal squamous cell carcinoma Diseases 0.000 description 1
- 201000011216 nasopharynx carcinoma Diseases 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000014399 negative regulation of angiogenesis Effects 0.000 description 1
- 201000003142 neovascular glaucoma Diseases 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 230000003982 neuronal uptake Effects 0.000 description 1
- 231100000344 non-irritating Toxicity 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- GYCKQBWUSACYIF-UHFFFAOYSA-N o-hydroxybenzoic acid ethyl ester Natural products CCOC(=O)C1=CC=CC=C1O GYCKQBWUSACYIF-UHFFFAOYSA-N 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 230000014207 opsonization Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 230000027758 ovulation cycle Effects 0.000 description 1
- 125000004043 oxo group Chemical group O=* 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 201000002513 peritoneal mesothelioma Diseases 0.000 description 1
- UEZVMMHDMIWARA-UHFFFAOYSA-M phosphonate Chemical compound [O-]P(=O)=O UEZVMMHDMIWARA-UHFFFAOYSA-M 0.000 description 1
- PTMHPRAIXMAOOB-UHFFFAOYSA-L phosphoramidate Chemical compound NP([O-])([O-])=O PTMHPRAIXMAOOB-UHFFFAOYSA-L 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 108090000917 podocalyxin Proteins 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Chemical class 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 230000023603 positive regulation of transcription initiation, DNA-dependent Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 238000012802 pre-warming Methods 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000000135 prohibitive effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000004076 pyridyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 238000011555 rabbit model Methods 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 108091008601 sVEGFR Proteins 0.000 description 1
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 1
- 229940081974 saccharin Drugs 0.000 description 1
- 235000019204 saccharin Nutrition 0.000 description 1
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 1
- 231100000241 scar Toxicity 0.000 description 1
- 210000001423 scleral cell Anatomy 0.000 description 1
- 229910052711 selenium Inorganic materials 0.000 description 1
- 239000011669 selenium Substances 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 201000010153 skin papilloma Diseases 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- WXMKPNITSTVMEF-UHFFFAOYSA-M sodium benzoate Chemical compound [Na+].[O-]C(=O)C1=CC=CC=C1 WXMKPNITSTVMEF-UHFFFAOYSA-M 0.000 description 1
- 239000004299 sodium benzoate Substances 0.000 description 1
- 235000010234 sodium benzoate Nutrition 0.000 description 1
- 229910001467 sodium calcium phosphate Inorganic materials 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- YZHUMGUJCQRKBT-UHFFFAOYSA-M sodium chlorate Chemical compound [Na+].[O-]Cl(=O)=O YZHUMGUJCQRKBT-UHFFFAOYSA-M 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 238000013222 sprague-dawley male rat Methods 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- IIACRCGMVDHOTQ-UHFFFAOYSA-M sulfamate Chemical compound NS([O-])(=O)=O IIACRCGMVDHOTQ-UHFFFAOYSA-M 0.000 description 1
- 229940124530 sulfonamide Drugs 0.000 description 1
- 150000003456 sulfonamides Chemical class 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 238000011191 terminal modification Methods 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- AVBGNFCMKJOFIN-UHFFFAOYSA-N triethylammonium acetate Chemical compound CC(O)=O.CCN(CC)CC AVBGNFCMKJOFIN-UHFFFAOYSA-N 0.000 description 1
- 125000004953 trihalomethyl group Chemical group 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 208000009999 tuberous sclerosis Diseases 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 231100000588 tumorigenic Toxicity 0.000 description 1
- 230000000381 tumorigenic effect Effects 0.000 description 1
- 230000007306 turnover Effects 0.000 description 1
- 125000004417 unsaturated alkyl group Chemical group 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 230000006444 vascular growth Effects 0.000 description 1
- 230000008728 vascular permeability Effects 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 210000004127 vitreous body Anatomy 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000037314 wound repair Effects 0.000 description 1
- 229960004175 xylazine hydrochloride Drugs 0.000 description 1
- NLIVDORGVGAOOJ-MAHBNPEESA-M xylene cyanol Chemical compound [Na+].C1=C(C)C(NCC)=CC=C1C(\C=1C(=CC(OS([O-])=O)=CC=1)OS([O-])=O)=C\1C=C(C)\C(=[NH+]/CC)\C=C/1 NLIVDORGVGAOOJ-MAHBNPEESA-M 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
- C12N15/1138—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 against receptors or cell surface proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
- A61P15/08—Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
- A61P15/12—Drugs for genital or sexual disorders; Contraceptives for climacteric disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
- A61P15/18—Feminine contraceptives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/02—Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/06—Antipsoriatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/02—Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin
-
- 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
-
- 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
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/31—Chemical structure of the backbone
- C12N2310/315—Phosphorothioates
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/31—Chemical structure of the backbone
- C12N2310/317—Chemical structure of the backbone with an inverted bond, e.g. a cap structure
-
- 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
- C12N2310/33—Chemical structure of the base
- C12N2310/332—Abasic residue
Definitions
- This invention relates to methods and reagents for the treatment of diseases or conditions relating to the levels of expression of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor(s).
- VEGF vascular endothelial growth factor
- the instant mvention features nucleic-acid based molecules and methods that modulate the expression of vascular endothelial growth factor and/or vascular endothelial growth factor receptors, such as VEGFR1 and or VEGFR2, that are useful in preventing, treating, controlling and/or diagnosing disorders and conditions related to angiogenesis, including but not limited to cancer, tumor angiogenesis, or ocular indications such as diabetic retinopathy, or age related macular degeneration, proliferative diabetic retinopathy, hypoxia-induced angiogenesis, rheumatoid arthritis, psoriasis, wound healing, endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual
- VEGF also referred to as vascular permeability factor (VPF) and vasculotropin
- VPF vascular permeability factor
- vasculotropin is a potent and highly specific mitogen of vascular endothelial cells (for a review see Ferrara, 1993 Trends Cardiovas. Med. 3, 244; Neufeld et al, 1994, Prog. Growth Factor Res. 5, 89).
- VEGF-induced neovascularization is implicated in various pathological conditions such as tumor angiogenesis, or ocular indications such as diabetic retinopathy, or age related macular degeneration, proliferative diabetic retinopathy, hypoxia-induced angiogenesis, rheumatoid arthritis, psoriasis, wound healing and others.
- VEGF an endothelial cell-specific mitogen
- VEGF belongs to the platelet-derived growth factor (PDGF) family of growth factors with approximately 18% homology with the A and B chain of PDGF at the amino acid level. Additionally, VEGF contains the eight conserved cysteine residues common to all growth factors belonging to the PDGF family (Neufeld et al, supra). VEGF protein is believed to exist predominantly as disulfide-lihked homodimers; monomers of VEGF have been shown to be inactive (Plouet et al, 1989 EMBO J. 8, 3801).
- VEGF exerts its influence on vascular endothelial cells by binding to specific high- affinity cell surface receptors.
- Covalent cross-linking experiments with 125 I-labeled VEGF protein have led to the identification of three high molecular weight complexes of 225, 195 and 175 kDa presumed to be VEGF and VEGF receptor complexes (Vaisman et al, 1990 J Biol. Chem. 265, 19461). Based on these studies VEGF-specific receptors of 180, 150 and 130 kDa molecular mass were predicted. In endothelial cells, receptors of 150 and 130 kDa have been identified.
- the VEGF receptors belong to the superfamily of receptor tyrosine kinases (RTKs) characterized by a conserved cytoplasmic catalytic kinase domain and a hydrophilic kinase sequence.
- RTKs receptor tyrosine kinases
- the extracellular domains of the VEGF receptors consist of seven immunoglobulin-like domains that are thought to be involved in VEGF binding functions.
- VEGF vascular endothelial growth factor receptor 1
- FEGFR1 farnesoid RI-1
- KDR kinase-insert-domain-containing receptor
- VEGF expression has been associated with several pathological states such as tumor angiogenesis, several forms of blindness, rheumatoid arthritis, psoriasis and others.
- pathological states such as tumor angiogenesis, several forms of blindness, rheumatoid arthritis, psoriasis and others.
- VEGF is both necessary and sufficient for neovascularization.
- Takashita et al, 1995 J. Clin. Invest. 93, 662 demonstrated that a single injection of VEGF augmented collateral vessel development in a rabbit model of ischemia.
- VEGF also can induce neovascularization when injected into the cornea.
- Expression of the VEGF gene in CHO cells is sufficient to confer tumorigenic potential to the cells.
- Kim et al, supra and Millauer et al, supra used monoclonal antibodies against VEGF or a dominant negative form of VEGFR2 receptor to inhibit tumor-induced neovascularization.
- VEGF and its receptors are associated with regions of new vascular growth (Millauer et al., 1993 Cell 72, 835; Shalaby et al., 1993 J. Clin. Invest. 91, 2235). Furthermore, transgenic mice lacking either of the VEGF receptors are defective in blood vessel formation and these mice do not survive; VEGFR2 appears to be required for differentiation of endothelial cells, while VEGFRl appears to be required at later stages of vessel formation (Shalaby et al., 1995 Nature 376, 62; Fung et al., 1995 Nature 376, 66). Thus, these receptors apparently need to be present to properly signal endothelial cells or their precursors to respond to vascularization-promoting stimuli.
- VEGF family may also be involved with both the etiology and maintenance of peritoneal endometriosis.
- Peritoneal endometriosis is a significant debilitating gynecological problem of widespread prevalence. It is now generally accepted that the pathogenesis of peritoneal endometriosis involves the implantation of exfoliated endometrmm. Maintenance of exfoliated endometrial tissue is dependent upon the generation and maintenance of an extensive blood supply both within and surrounding the ectopic tissue.
- Endometriosis is a disease affecting an estimated 77 million women and teenagers worldwide. Endometriosis is a leading cause of infertility, chronic pelvic pain and hysterectomy. Endometriosis can be characterized when endometrial tissue (the tissue inside the uterus which builds up and is shed each month during menses) is found outside the uterus, in other areas of the body. The endometrial tissue can respond to hormonal commands each month and break down and bleed. However, unlike the endometrium, these tissue deposits have no way of leaving the body. The result is internal bleeding, degeneration of blood and tissue shed from the growths, inflammation of the surrounding areas, expression of irritating enzymes and formation of scar tissue.
- endometrial tissue the tissue inside the uterus which builds up and is shed each month during menses
- Endometriosis can only be confirmed through surgery such as laparoscopy, but can be suspected based on symptoms, physical findings and diagnostic tests. Endometriosis can be treated in many different ways, both surgically and medically. Most commonly, surgery will be performed during which the disease will be excised, ablated, fulgarated, cauterized or otherwise removed, and adhesions will also be freed. Surgeries include but are not limited to laparoscopy; laparotomy; presacral and uterosacral and various levels of hysterectomies, where some or all of the reproductive organs are removed. Often, this method will only relieve the symptoms associated with growths on the reproductive organs, not the bowels or kidneys and related areas where Endometriosis can be present.
- GnRH agonists are commonly used on women in all stages of the disease and may sometimes have serious side affects.
- GnRH (gonadotropin releasing hormone) analogues are classified into 2 groups: agonists and antagonists.
- Agonists are commonly used in the treatment of Endometriosis by suppressing the manufacture of follicle stimulating hormone (FSH) and luteinizing hormone (LH), common hormones required in ovulation. When they are not secreted, the body will go into "pseudo-menopause,” stalling the growth of more implants. However, these are again only stop-gap measures that can be utilized only for short term intervals. Once the body returns to it's normal state, the Endometriosis will again begin to implant itself.
- FSH follicle stimulating hormone
- LH luteinizing hormone
- Angiogenesis is likely to be involved in the pathogenesis of endometriosis.
- the transplantation theory when the exfoliated endometrium is attached to the peritoneal layer, the establishment of a new blood supply is essential for the survival of the endometrial implant and development of endometriosis (Donnez et al, 1998, Hum. Reprod., 13, 1686- 1690).
- Endometrial growth and repair after menstruation are associated with profound angiogenesis. Abnormalities in these processes result in excessive or unpredictable bleeding patterns and are common in many women. It is therefore important to understand which factors regulate normal endometrial angiogenesis.
- VEGF Vascular endothelial growth factor
- VEGF vascular endothelial growth factor
- Sources of this factor include the eutopic endometrium, ectopic endometriotic tissue and peritoneal fluid macrophages.
- Important to its etiology is the correct peritoneal environment in which the exfoliated endometrium is seeded and implants. Established ectopic tissue is then dependent on the peritoneal environment for its survival, an environment that supports angiogenesis.
- Pavco et al International PCT Publication No. WO 97/15662, describes methods and reagents for treating diseases or conditions related to levels of vascular endothelial growth factor receptor.
- inhibitors including certain ribozyme and antisense nucleic acid molecules, of specific genes, including cathepsin D, AEBP-1, stromelysin-3, cystatin B, protease inhibitor 1, sFRP4, gelsolin, IGFBP-3, dual specificity phosphatase 1, PAEP, Ig gamma chain, ferritin, complement component 3, pro- alpha- 1 type HI collagen, proline 4-hydroxylase, alpha-2 type I collagen, claudin-4, melanoma adhesion protein, procollagen C-endopeptidase enhancer, nascent-polypeptide-associated complex alpha polypeptide, elongation factor 1 alpha (EF-1-alpha).
- vitamin D3 25 hydroxylase CSRP-1, steroidogenic acute regulatory protein, apolipoprotein E, transcobalamin IL prosaposin, early growth response 1 (EGR1), ribosomal protein S6, adenosine deaminase RNA-specific protein, RAD21, guanine nucleotide binding protein beta polypeptide 2-like 1 (RACKl) and podocalyxin genes which are all differentially expressed in tissues within individual patients with endometriosis.
- EGR1 early growth response 1
- ribosomal protein S6, adenosine deaminase RNA-specific protein, RAD21, guanine nucleotide binding protein beta polypeptide 2-like 1 (RACKl) and podocalyxin genes which are all differentially expressed in tissues within individual patients with endometriosis.
- This invention features nucleic acid-based molecules, for example, enzymatic nucleic acid molecules, allozymes, antisense nucleic acids, 2-5A antisense chimeras, triplex forming oligonucleotides, decoy RNA, dsRNA, siRNA, aptamers, and antisense nucleic acids containing nucleic acid cleaving chemical groups, and methods to modulate vascular endothelial growth factor (VEGF) and/or vascular endothelial growth factor receptor (VEGFr) gene expression.
- VEGF vascular endothelial growth factor
- VEGFr vascular endothelial growth factor receptor
- Non-limiting examples of genes that encode vascular endothelial growth factor receptors of the invention include VEGFRl, VEGFR2 or combinations thereof.
- the instant invention features nucleic acid-based molecules and methods that modulate the expression of vascular endothelial growth factor and/or vascular endothelial growth factor receptors, such as VEGFRl and/or VEGFR2, that are useful in preventing, treating, controlling, and/or diagnosing angiogenesis related diseases and conditions, including but not limited to tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and female reproductive disorders and conditions, including but not limited to endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), and menopausal dysfunction.
- angiogenesis related diseases and conditions including but not limited to tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or mela
- the invention features one or more nucleic acid-based molecules and methods that independently or in combination modulate the expression of gene(s) encoding vascular endothelial growth factor receptors.
- the present invention features nucleic acid molecules that modulate the expression of VEGF (for example Genbank Accession No. NM_003376), VEGFRl receptor (for example Genbank Accession No. NM_002019), and VEGFR2 receptor (for example Genbank Accession No.
- NM_002253 that are useful in preventing, treating, controlling, and/or diagnosing tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and female reproductive disorders and conditions, including but not limited to endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), and menopausal dysfunction.
- cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma
- ocular indications such as diabetic retinopathy, or age related macular degeneration
- female reproductive disorders and conditions including but not limited to endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS),
- the present invention features a compound having Formula I: (SEQ ID NO: 5977) 5' g s a s g s u s ugcUGAuGagg ccgaaa ggccGaaAgucugB 3'
- each a is 2'-O-methyl adenosine nucleotide
- each g is a 2'-O-methyl guanosine nucleotide
- each c is a 2'-O-methyl cytidine nucleotide
- each u is a 2'-O-methyl uridine nucleotide
- each A is adenosine
- each G is guanosine
- each s individually represents a phosphorothioate mternucleotide linkage
- U is 2'-deoxy-2'-C-allyl uridine
- B is an inverted deoxyabasic moiety.
- This compound is also referred to as ANGIOZYMETM ribozyme.
- the present invention features a compound having Formula II: (SEQ ID NO: 5978).
- each a is 2'-O-methyl adenosine nucleotide
- each g is a 2'-O-methyl guanosine nucleotide
- each c is a 2'-O-methyl cytidine nucleotide
- each u is a 2'-O-methyl uridine nucleotide
- each A is adenosine
- each G is guanosine
- each s individually represents a phosphorothioate internucleotide linkage
- U is 2'-deoxy-2'-C-allyl uridine
- B is an inverted deoxyabasic moiety.
- the invention features a composition comprising a nucleic acid molecule of the invention in a pharmaceutically acceptable carrier. In another embodiment, the invention features a composition comprising a compound of Formula I and/or Formula II in a pharmaceutically acceptable carrier or diluent.
- the invention features a method of administering to a cell, for example a mammalian cell, including a human cell, a nucleic acid molecule of the invention comprising contacting the cell with the nucleic acid molecule under conditions suitable for administration, for example in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- the invention features a method of administering to a cell, for example a mammalian cell , including a human cell, a compound of Formula I and/or Formula Ilcomprising contacting the cell with the compound under conditions suitable for administration, for example in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- the present invention features a mammalian cell comprising a nucleic acid molecule of the invention, wherein the mammalian cell is, for example, a human cell.
- the present invention also features a mammalian cell comprising the compound of Formula I and/or Formula U, wherein the mammalian cell is, for example, a human cell.
- the invention features a method of inhibiting angiogenesis, for example tumor angiogenesis, or ocular indications such as diabetic retinopathy, or age related macular degeneration, or endometrial neovascularization, in a subject comprising contacting the subject with a nucleic acid molecule of the invention, under conditions suitable for the inhibition.
- the invention features a method of inhibiting angiogenesis, for example tumor angiogenesis, or ocular indications such as diabetic retinopathy, or age related macular degeneration, or endometrial neovascularization, in a subject, comprising contacting the subject with a compound of Formula I and/or Formula II, under conditions suitable for the inhibition.
- the invention features a method of treatment of a subjecthaving an ocular condition associated with the increased level of a VEGF receptor, for example diabetic retinopathy, or age related macular degeneration, comprising contacting cells of the subjectwith a nucleic acid molecule, such as an enzymatic nucleic acid molecule targeted against a VEGF receptor RNA, e.g., molecule according to Formula I and/or H, under conditions suitable for the treatment.
- a nucleic acid molecule such as an enzymatic nucleic acid molecule targeted against a VEGF receptor RNA, e.g., molecule according to Formula I and/or H
- the invention features a method of treatment of a subjecthaving a condition associated with an increased level of VEGR and/or a VEGF receptor, for example tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, ocular diseases or ocular indications such as diabetic retinopathy, or age related macular degeneration, rhuematoid arthritis, psoriasis endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), or menopausal dysfunction, comprising contacting cells of the subject with a nucleic acid molecule of the invention, such as a compound of Formula I and/or Formula H, under conditions suitable for the treatment.
- a nucleic acid molecule of the invention such as a compound of Formula I and/or Formula H
- the inventive method of treatment further comprises the use of one or more drug therapies under conditions suitable for the treatment.
- drug therapies that can be used in combination with nucleic acid molecules of the invention include to 5-fluoro uridine, Leucovorin, Irinotecan (CAMPTOSAR® or CPT-11 or Camptothecin-11 or Campto), Paclitaxel, or Carboplatin, GnRH (gonadotropin releasing hormone) agonists, Lupron Depot (Leuprolide Acetate), Synarel (naferalin acetate), Zolodex (goserelin acetate), Suprefact (buserelin acetate), Danazol, or oral contraceptives including but not limited to Depo-Provera or Provera (medroxyprogesterone acetate), or any other estrogen/progesterone contraceptive.
- the invention features a method of administering to a mammal, for example a human, a nucleic acid molecule of the invention comprising contacting the mammal with the nucleic acid molecule under conditions suitable for the administration, for example, in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- the invention features a method of administering to a mammal, for example a human, a compound of Formula I and/or Formula U comprising contacting the mammal with the compound under conditions suitable for the administration, for example, in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- the invention features a nucleic acid molecule which down regulates expression of a vascular endothelial growth factor (VEGF) and/or vascular endothelial growth factor receptor (VEGFr) gene, for example, wherein the VEGFr gene comprises VEGFRl or VEGFR2 and any combination thereof.
- VEGF vascular endothelial growth factor
- VEGFr vascular endothelial growth factor receptor
- a nucleic acid molecule of the invention such as an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups, is adapted to treat, control and/or diagnose tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, ocular diseases or ocular indications, such as diabetic retinopathy, or age related macular degeneration, rhuematoid arthritis, psoriasis endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), or menopausal dysfunction.
- tumor angiogenesis cancers such as breast cancer
- nucleic acid molecules are also useful for the prevention of the diseases and conditions including diabetic retinopathy, macular degeneration, neovascular glaucoma, myopic degeneration, verruca vulgaris, angiofibroma of tuberous sclerosis, port-wine stains, Sturge Weber syndrome, Kippel-Trenaunay- Weber syndrome, Osier- Weber-Rendu syndrome and other diseases or conditions that are related to the levels of VEGFRl or VEGFR2 in a cell or tissue.
- diseases and conditions including diabetic retinopathy, macular degeneration, neovascular glaucoma, myopic degeneration, verruca vulgaris, angiofibroma of tuberous sclerosis, port-wine stains, Sturge Weber syndrome, Kippel-Trenaunay- Weber syndrome, Osier- Weber-Rendu syndrome and other diseases or conditions that are related to the levels of VEGFRl or VEGFR2 in a cell or tissue.
- the invention features a composition in a pharmaceutically acceptable carrier or diluent, comprising the nucleic acid molecule of the instant invention.
- an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention is adapted for birth control.
- an enzymatic nucleic acid molecule of the invention is in a hammerhead, friozyme, Zinzyme, DNAzyme, Amberzyme, or G-cleaver configuration.
- an enzymatic nucleic acid molecule of the invention comprises between 8 and 100 bases complementary to RNA of VEGFRl and/or VEGFR2 gene. In another embodiment, an enzymatic nucleic acid molecule of the invention comprises between 14 and 24 bases complementary to RNA of VEGFRl and/or VEGFR2 gene.
- a siRNA molecule of the invention comprises a double stranded
- RNA wherein one strand of the RNA is complementary to RNA of a VEGFRl and/or VEGFR2 gene.
- a siRNA molecule of the invention comprises a double stranded RNA wherein one strand of the RNA comprises a portion of a sequence of RNA having a VEGFRl and/or VEGFR2 sequence, hi yet another embodiment, a siRNA molecule of the invention comprises a double stranded RNA wherein both strands of RNA are connected by a non-nucleotide linker.
- a siRNA molecule of the invention comprises a double stranded RNA wherein both strands of RNA are connected by a nucleotide linker, such as a loop or stem loop structure.
- a single strand component of a siRNA molecule of the invention is from about 14 to about 50 nucleotides in length. In another embodiment, a single strand component of a siRNA molecule of the invention is about 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28 nucleotides in length, hi yet another embodiment, a single strand component of a siRNA molecule of the invention is about 23 nucleotides in length. In one embodiment, a siRNA molecule of the invention is from about 28 to about 56 nucleotides in length, hi another embodiment, a siRNA molecule of the invention is about 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, or 52 nucleotides in length.
- a siRNA molecule of the invention is about 46 nucleotides in length.
- an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention is chemically synthesized.
- an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention comprises at least one 2'-sugar modification.
- an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5 A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing nucleic acid cleaving chemical groups of the invention comprises at least one nucleic acid base modification.
- an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention comprises at least one phosphate backbone modification.
- the invention features a mammalian cell, for example a human cell, comprising a nucleic acid molecule of the invention.
- the invention features a method of reducing VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 expression or activity in a cell comprising contacting the cell with a nucleic acid molecule of the invention that modulates the expression and/or activity of VEGF and/or VEGFr, under conditions suitable for the reduction.
- a method of treatment of a subject having a condition associated with the level of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 is featured, wherein the method further comprises the use of one or more drug therapies under conditions suitable for the treatment.
- the invention features a method for treatment of a subject having tumor angiogenesis, tumor angiogenesis, cancers including but not limited to tumor and cancer types shown under Diagnosis in Table III, ocular diseases or ocular indications such as diabetic retinopathy, or age related macular degeneration, rhuematoid arthritis, psoriasis and/or endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), or menopausal dysfunction, comprising administering to the subject a nucleic acid molecule of the invention that modulates the expression and/or activity of VEGF and/or VEGFr under conditions suitable for the treatment.
- ocular diseases or ocular indications such as diabetic retinopathy, or age related macular degeneration, rhuematoid arthritis, psoriasis and/or endometriosis, endometrial carcinoma, gy
- the invention features a method for birth control in a subject comprising administering to the subject a nucleic acid molecule of the invention that modulates the expression and/or activity of VEGF and/or VEGFr under conditions suitable for the treatment.
- the invention features a method of cleaving RNA encoded by a VEGF, VEGFRl and/or VEGFR2 gene comprising contacting an enzymatic nucleic acid molecule of the invention having endonuclease activity with RNA encoded by a VEGFRl and/or VEGFR2 gene under conditions suitable for the cleavage, for example, wherein the cleavage is carried out in the presence of a divalent cation, such as Mg2 + .
- a divalent cation such as Mg2 +
- a nucleic acid molecule of the invention comprises a cap structure, for example a 3 ',3 '-linked or 5 ',5 '-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5'-end, or 3 '-end, or both the 5'-end and the 3'-end of the enzymatic nucleic acid molecule.
- a nucleic acid molecule of the invention comprises a cap structure, for example a 3',3'-linked or 5',5'-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5'-end, or 3'-end, or both the 5'-end and the 3'-end of the antisense nucleic acid molecule.
- the invention features an expression vector comprising a nucleic acid sequence encoding at least one nucleic acid molecule of the invention such that the vector allows expression of the nucleic acid molecule.
- the invention features a mammalian cell, for example, a human cellcomprising an expression vector of the invention.
- an expression vector of the invention further comprises a sequence for a nucleic acid molecule complementary to RNA encoded by a VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 gene.
- an expression vector of the invention comprises a nucleic acid sequence encoding two or more nucleic acid molecules of the invention, which can be the same or different.
- the invention features a method for treatment or control of tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and/or endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), or menopausal dysfunction, comprising administering to a subject a nucleic acid molecule of the invention that modulates the expression and/or activity of VEGF and/or VEGFr, such as an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of
- the method of treatment features a nucleic acid molecule of the invention, such as an enzymatic nucleic acid or antisense nucleic acid molecule, that comprises at least five ribose residues, at least ten 2'-0-methyl modifications, and a 3'- end modification, such as a 3 '-3' inverted abasic moiety.
- a nucleic acid molecule of the invention further comprises phosphorothioate linkages on at least three of the 5' terminal nucleotides.
- the mvention features a method of administering to a mammal, for example a human, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2- 5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention, comprising contacting the mammal with the nucleic acid molecule under conditions suitable for the administration, for example, in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
- the mvention features a method of administering to a mammal an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing nucleic acid cleaving chemical groups of the invention in conjunction with other therapies, comprising contacting the mammal, for example a human, with the nucleic acid molecule and the other therapy under conditions suitable for the administration.
- other therapies contemplated by the instant invention that can be used in conjunction with the nucleic acid molecules of the instant invention include, but are not limited to, 5-fluoro uridine, Leucovorin, Irinotecan (CAMPTOSAR® or CPT-11 or Camptothecin-11 or Campto), Paclitaxel, or Carboplatin, GnRH (gonadotropin releasing hormone) agonists, Lupron Depot (Leuprolide Acetate), Synarel (naferalin acetate), Zolodex (goserelin acetate), Suprefact (buserelin acetate), Danazol, or oral contraceptives including but not limited to Depo-Provera or Provera (medroxyprogesterone acetate), or other estrogen/progesterone contraceptive.
- the invention features the use of an enzymatic nucleic acid molecule, to down-regulate the expression of VEGFRl and/or VEGFR2 genes in the treatment or control of tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and/or endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), or menopausal dysfunction.
- Such enzymatic nucleic acid molecule can be in the hammerhead, NCH, G-cleaver, Amberzyme, Zinzyme, and/or DNAzyme motif.
- the invention features the use of an enzymatic nucleic acid moleculeto down-regulate the expression of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 genes, as a method of birth control.
- an enzymatic nucleic acid molecule can be in the hammerhead, NCH, G-cleaver, Amberzyme, Zinzyme, and/or DNAzyme motif, hi one embodiment, the nucleic acid molecules of the invention have complementarity to the substrate. sequences in Tables V and VI. Examples of enzymatic nucleic acid molecules of the invention are shown in Tables V and VI. Examples of such enzymatic nucleic acid molecules consist essentially of sequences defined in these Tables.
- inhibitor By “inhibit”, “down-regulate”, or “reduce”, it is meant that the expression of the gene, or level of nucleic acids or equivalent nucleic acids encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, such as VEGFRl, VEGFR2 and/or flk-1, is reduced below that observed in the absence of the nucleic acid molecules of the invention.
- inhibition, down-regulation or reduction with enzymatic nucleic acid molecule preferably is below that level observed in the presence of an enzymatically inactive or attenuated molecule that is able to bind to the same site on the target nucleic acid, but is unable to cleave that nucleic acid
- inhibition, down-regulation, or reduction with antisense oligonucleotides is preferably below that level observed in the presence of, for example, an oligonucleotide with scrambled sequence or with mismatches.
- inhibition, down-regulation, or reduction of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 with the nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence.
- up-regulate is meant that the expression of a gene, or level of nucleic acids or equivalent nucleic acids encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, such as VEGFRl and/or VEGFR2, is greater than that observed in the absence of the nucleic acid molecules of the invention.
- the expression of a gene, such as VEGF and or VEGFr, such as VEGFRl and/or VEGFR2 gene can be increased in order to treat, prevent, ameliorate, or modulate a pathological condition caused or exacerbated by an absence or low level of gene expression.
- module is meant that the expression of a gene, or level of nucleic acids or equivalent nucleic acids encoding one or more proteins or protein subunits, or activity of one or more proteins protein subunit(s) is up-regulated or down-regulated, such that the expression, level, or activity is greater than or less than that observed in the absence of the nucleic acid molecules of the invention.
- enzymatic nucleic acid molecule it is meant a nucleic acid molecule which has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity which is active to specifically cleave a target nucleic acid. That is, the enzymatic nucleic acid molecule is able to intermolecularly cleave a nucleic acid and thereby inactivate a target nucleic acid molecule. These complementary regions allow sufficient hybridization of the enzymatic nucleic acid molecule to the target nucleic acid and thus permit cleavage.
- nucleic acids can be modified at the base, sugar, and/or phosphate groups.
- enzymatic nucleic acid is used interchangeably with phrases such as ribozymes, catalytic RNA, enzymatic RNA, catalytic DNA, aptazyme or aptamer-binding ribozyme, regulatable ribozyme, catalytic oligonucleotides, nucleozyme, DNAzyme, RNA enzyme, endoribonuclease, endonuclease, minizyme, leadzyme, oligozyme or DNA enzyme. All of these terminologies describe nucleic acid molecules with enzymatic activity.
- enzymatic nucleic acid molecules described in the instant application are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target nucleic acid regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart a nucleic acid cleaving and/or ligation activity to the molecule (Cech et al, U.S. Patent No. 4,987,071; Cech et al, 1988, 260 JAMA 3030).
- enzymatic nucleic acids act by first binding to a target nucleic acid. Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target nucleic acid.
- the enzymatic nucleic acid first recognizes and then binds a target nucleic acid through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target nucleic acid. Strategic cleavage of such a target nucleic acid will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its nucleic acid target, it is released from that nucleic acid to search for another target and can repeatedly bind and cleave new targets.
- ribozyme is able to cleave many molecules of target nucleic acid, hi addition, the ribozyme is a highly specific inhibitor of gene expression, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target nucleic acid, but also on the mechanism of target nucleic acid cleavage. Single mismatches, or base-substitutions, near the site of cleavage can completely eliminate catalytic activity of a ribozyme.
- an enzymatic nucleic acid molecule of the invention is formed in a hammerhead or hairpin motif, but can also be formed in the motif of a hepatitis delta virus, group I intron, group II intron or RNase P RNA (in association with an RNA guide sequence), Neurospora VS RNA, DNAzymes, NCH cleaving motifs, or G-cleavers.
- Group II introns are described by Griffin et al, 1995, Chem. Biol. 2, 761; Michels and Pyle, 1995, Biochemistry 34, 2965; Pyle et al, International PCT Publication No. WO 96/22689; an example of a Group I intron is described by . Cech et al, U.S. Patent 4,987,071; and examples of DNAzymes are described by Usman et al, International PCT Publication No. WO 95/11304; Chartrand et al, 1995, NAR 23, 4092; Breaker et al, 1995, Chem. Bio.
- nucleic acid molecule as used herein is meant a molecule having nucleotides.
- the nucleic acid can be single, double, or multiple stranded and can comprise modified or unmodified nucleotides or non-nucleotides or various mixtures and combinations thereof.
- enzymatic portion or “catalytic domain” is meant that portion/region of a enzymatic nucleic acid molecule essential for cleavage of a nucleic acid substrate (for example see Figure 6).
- substrate binding arm or “substrate binding domain” is meant that portion/region of a enzymatic nucleic acid which is able to interact, for example via complementarity (i.e., able to base-pair with), with a portion of its substrate.
- complementarity i.e., able to base-pair with
- nucleic acid of the invention can have binding arms that are contiguous or non-contiguous and can be of varying lengths.
- the length of the binding arm(s) are preferably greater than or equal to four nucleotides and of sufficient length to stably interact with the target nucleic acid; preferably 12-100 nucleotides; more preferably 14-24 nucleotides long (see for example Werner and Uhlenbeck, supra; Hamman et al, supra; Hampel et al, EP0360257; Berzal-Herranz et al, 1993, EMBO J., 12, 2567-73) or between 8 and 14 nucleotides long.
- the design is such that the length of the binding arms are symmetrical (i.e., each of the binding arms is of the same length; e.g., four and four, five and five nucleotides, or six and six nucleotides, or seven and seven nucleotides long) or asymmetrical (i.e., the binding arms are of different length; e.g., three and five, six and three nucleotides; three and six nucleotides long; four and five nucleotides long; four and six nucleotides long; four and seven nucleotides long; and the like).
- Inozyme or "NCH” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as NCH Rz in Figure 6 and in Ludwig et al, International PCT Publication No. WO 98/58058 and US Patent Application Serial No. 08/878,640.
- Inozymes possess endonuclease activity to cleave nucleic acid substrates having a cleavage triplet NCH/, where N is a nucleotide, C is cytidine and H is adenosine, uridine or cytidine, and "/" represents the cleavage site.
- H is used interchangeably with X.
- Inozymes can also possess endonuclease activity to cleave nucleic acid substrates having a cleavage triplet NCN/, where N is a nucleotide, C is cytidine, and "/" represents the cleavage site.
- "F in Figure 6 represents an Inosine nucleotide, preferably a ribo-Inosine or xylo-Inosine nucleoside.
- G-cleaver motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as G-cleaver Rz in Figure 6 and in Eckstein et al, US 6,127,173.
- G-cleavers possess endonuclease activity to cleave nucleic acid substrates having a cleavage triplet NYN/, where N is a nucleotide, Y is uridine or cytidine and "/" represents the cleavage site.
- G-cleavers can be chemically modified as is generally shown in Figure 6.
- amberzyme motif or configuration an enzymatic nucleic acid molecule comprising a motif as is generally described in Beigehnan et al, Intemational PCT publication No. WO 99/55857 and US Patent Application Serial No. 09/476,387.
- Amberzymes possess endonuclease activity to cleave nucleic acid substrates having a cleavage triplet NG/N, where N is a nucleotide, G is guanosine, and "/" represents the cleavage site.
- Amberzymes can be chemically modified to increase nuclease stability through substitutions using modified nucleotides.
- nucleoside and/or non- nucleoside linkers can be used to substitute the 5'-gaa-3' loops shown in the figure.
- Amberzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2'-OH) group within its own nucleic acid sequence for activity.
- zinzyme motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described in Figure 7 and in Beigehnan et al, International
- Zinzymes possess endonuclease activity to cleave nucleic acid substrates having a cleavage triplet including but not limited to YG/Y, where Y is uridine or cytidine, and G is guanosine and "/" represents the cleavage site.
- Zinzymes can be chemically modified to increase nuclease stability through substitutions as are generally shown in Figure 7, including substituting 2'-O-methyl guanosine nucleotides for guanosine nucleotides.
- differing nucleotide and/or non-nucleotide linkers can be used to substitute the 5'-gaa-2' loop shown in the figure.
- Zinzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2'-OH) group within its own nucleic acid sequence for activity.
- DNAzyme' an enzymatic nucleic acid molecule that does not require the presence of a 2' -OH group within its own nucleic acid sequence for activity, hi particular embodiments the enzymatic nucleic acid molecule can have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2'-OH groups.
- DNAzymes can be synthesized chemically or expressed endogenously in vivo, by means of a single stranded DNA vector or equivalent thereof.
- DNAzyme An example of a DNAzyme is shown in Figure 8 and is generally reviewed in Usman et al, US patent No., 6,159,714; Chartrand et al, 1995, NAR 23, 4092; Breaker et al, 1995, Chem. Bio. 2, 655; Santoro et al, 1997, PNAS 94, 4262; Breaker, 1999, Nature Biotechnology, 17, 422-423; and Santoro et. al, 2000, J. Am. Chem. Soc, 122, 2433-39.
- the "10-23" DNAzyme motif is one particular type of DNAzyme that was evolved using in vitro selection, see Santoro et al, supra and as generally described in Joyce et al, US 5,807,718. Additional DNAzyme motifs can be selected for using techniques similar to those described in these references, and hence, are within the scope of the present invention.
- a nucleic acid molecule of the invention is long enough to provide the intended function under the expected condition.
- a nucleic acid molecule of the invention needs to be of "sufficient length” to provide stable interaction with a target nucleic acid molecule under the expected binding conditions and environment.
- "sufficient length” means that the binding arm sequence is long enough to provide stable binding to a target site under the expected reaction conditions and environment. The binding arms are not so long as to prevent useful turnover of the nucleic acid molecule.
- stably interact is meant interaction of an oligonucleotides with target nucleic acid (e.g., by forming hydrogen bonds with complementary nucleotides in the target under physiological conditions) that is sufficient to the intended purpose (e.g., cleavage of target nucleic acid by an enzyme).
- RNA to VEGF, VEGFRl and/or VEGFR2 is meant to include nucleic acid molecules having homology (partial or complete) to a nucleic acid encoding VEGF, VEGFRl and/or VEGFR2 proteins or encoding proteins with similar function as VEGF, VEGFRl and/or VEGFR2 proteins in various organisms, including human, rodent, primate, rabbit, pig, protozoans, fungi, plants, and other microorganisms and parasites.
- the equivalent nucleic acid sequence also includes, in addition to the coding region, regions such as 5'- untranslated region, 3 '-untranslated region, introns, intron-exon junction and the like.
- nucleotide sequence of two or more nucleic acid molecules is partially or completely identical.
- antisense nucleic acid a non-enzymatic nucleic acid molecule that binds to target nucleic acid by means of RNA-RNA or RNA-DNA or RNA-PNA (protein nucleic acid; Eghol et al, 1993 Nature 365, 566) interactions and alters the activity of the target nucleic acid (for a review, see Stein and Cheng, 1993 Science 261, 1004 and Woolf et al, US patent No. 5,849,902).
- antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule.
- an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop.
- an antisense molecule can be complementary to two (or even more) noncontiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both.
- antisense DNA can be used to target nucleic acid by means of DNA-RNA interactions, thereby activating RNase H, which digests the target nucleic acid in the duplex.
- the antisense oligonucleotides can comprise one or more RNAse H activating region, which is capable of activating RNAse H cleavage of a target nucleic acid.
- Antisense DNA can be synthesized chemically or expressed via the use of a single stranded DNA expression vector or equivalent thereof.
- RNase H activating region is meant a region (generally greater than or equal to 4- 25 nucleotides in length, preferably from 5-11 nucleotides in length) of a nucleic acid molecule capable of binding to a target nucleic acid to form a non-covalent complex that is recognized by cellular RNase H enzyme (see for example Arrow et al, US 5,849,902; Arrow et al, US 5,989,912).
- the RNase H enzyme binds to a nucleic acid molecule-target nucleic acid complex and cleaves the target nucleic acid sequence.
- the RNase H activating region comprises, for example, phosphodiester, phosphorothioate (preferably at least four of the nucleotides are phosphorothiote substitutions; more specifically, 4-11 of the nucleotides are phosphorothiote substitutions); phosphorodithioate, 5'-thiophosphate, or methylphosphonate backbone chemistry or a combination thereof.
- the RNase H activating region can also comprise a variety of sugar chemistries.
- the RNase H activating region can comprise deoxyribose, arabino, fluoroarabino or a combination thereof, nucleotide sugar chemistry.
- 2-5A antisense chimera an antisense oligonucleotide containing a 5'- phosphorylated 2-5 '-linked adenylate residue. These chimeras bind to target nucleic acid in a sequence-specific manner and activate a cellular 2-5 A-dependent ribonuclease which, in turn, cleaves the target nucleic acid (Torrence et al, 1993 Proc. Natl. Acad. Sci. USA 90, 1300; Silverman et al, 2000, Methods Enzymol, 313, 522-533; Player and Torrence, 1998, Pharmacol. Ther., 78, 55-113).
- triplex forming oligonucleotides an oligonucleotide that can bind to a double-stranded polynucleotide, such as DNA, in a sequence-specific manner to form a triple- strand helix. Formation of such triple helix structure has been shown to inhibit transcription of the targeted gene (Duval- Valentin et al, 1992 Proc. Natl. Acad. Sci. USA 89, 504; Fox, 2000, Curr. Med. Chem., 7, 17-37; Praseuth et. al, 2000, Biochim. Biophys. Ada, 1489, 181- 206).
- RNA RNA sequences including but not limited to structural genes encoding a polypeptide.
- complementarity refers to the ability of a nucleic acid to form hydrogen bond(s) with another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types, hi reference to nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its target or complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., enzymatic nucleic acid cleavage, antisense or triple helix inhibition. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al, 1987, CSH Symp. Quant. Biol.
- a percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule which can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary).
- Perfectly complementary means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence.
- RNA is meant a molecule comprising at least one ribonucleotide residue.
- ribonucleotide or “2'-OH” is meant a nucleotide with a hydroxyl group at the 2' position of a ⁇ -D-ribo-furanose moiety.
- nucleic acid decoy molecule or “decoy” as used herein is meant a nucleic acid molecule that mimics the natural binding domain for a ligand. The decoy therefore competes with the natural binding target for the binding of a specific ligand. For example, it has been shown that over-expression of HTV trans-activation response (TAR) RNA can act as a “decoy” and efficiently binds HIV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger et al., 1990, Cell, 63, 601-608).
- TAR trans-activation response
- aptamer or “nucleic acid aptamer” as used herein is meant a nucleic acid molecule that binds specifically to a target molecule wherein the nucleic acid molecule has sequence that is distinct from sequence recognized by the target molecule in its natural setting.
- an aptamer can be a nucleic acid molecule that binds to a target molecule where the target molecule does not naturally bind to a nucleic acid.
- the target molecule can be any molecule of interest.
- the aptamer can be used to bind to a ligand binding domain of a protein, thereby preventing interaction of the naturally occurring ligand with the protein.
- the nucleic acid molecules of the instant invention can bind to VEGFRl or VEGFR2 receptors to block activity of the receptor.
- VEGFRl or VEGFR2 receptors can be readily generated using techniques generally known in the art, see for example Gold et al, US 5,475,096 and 5,270,163; Gold et al, 1995, Annu. Rev. Biochem., 64, 763; Brody and Gold, 2000, J. Biotechnol, 74, 5; Sun, 2000, Curr. Opin. Mol. Ther., 2, 100; Kusser, 2000, J. Biotechnol, 74, 27; Hermann and Patel, 2000, Science, 287, 820; and Jayasena, 1999, Clinical Chemistry, 45, 1628.
- double stranded RNA or “dsRNA” as used herein refers to a double stranded RNA molecule capable of RNA interference "RNAi", including short interfering RNA “siRNA” see for example Bass, 2001, Nature, 411, 428-429; Elbashir et al., 2001, Nature, 411, 494-498; and Kreutzer et al, International PCT Publication No. WO 00/44895; Zernicka-Goetz et al, International PCT Publication No. WO 01/36646; Fire, International PCT Publication No. WO 99/32619; Plaetinck et al, International PCT Publication No.
- nucleic acid sensor molecule or “allozyme” as used herein is meant a nucleic acid molecule comprising an enzymatic domain and a sensor domain, where the enzymatic nucleic acid domain's ability to catalyze a chemical reaction is dependent on the interaction with a target signaling molecule, such as a nucleic acid, polynucleotide, oligonucleotide, peptide, polypeptide, or protein, for example VEGF, VEGFRl and/or VEGFR2.
- a target signaling molecule such as a nucleic acid, polynucleotide, oligonucleotide, peptide, polypeptide, or protein, for example VEGF, VEGFRl and/or VEGFR2.
- nucleic acid sensor molecule can provide enhanced catalytic activity of the nucleic acid sensor molecule, increased binding affinity of the sensor domain to a target nucleic acid, and/or improved nuclease/chemical stability of the nucleic acid sensor molecule, and are hence within the scope of the present invention (see for example Usman et al, US Patent Application No. 09/877,526, George et al, US Patent Nos. 5,834,186 and 5,741,679, Shih et al, US Patent No. 5,589,332, Nathan et al, US Patent No 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al, International PCT Publication Nos. WO 00/26226 and 98/27104, and Sullenger et al, US Patent Application Serial No. 09/205,520).
- sensor component or “sensor domain” of the nucleic acid sensor molecule as used herein is meant, a nucleic acid sequence (e.g., RNA or DNA or analogs thereof) which interacts with a target signaling molecule, for example a nucleic acid sequence in one or more regions of a target nucleic acid molecule or more than one target nucleic acid molecule, and which interaction causes the enzymatic nucleic acid component of the nucleic acid sensor molecule to either catalyze a reaction or stop catalyzing a reaction.
- a target signaling molecule for example a nucleic acid sequence in one or more regions of a target nucleic acid molecule or more than one target nucleic acid molecule, and which interaction causes the enzymatic nucleic acid component of the nucleic acid sensor molecule to either catalyze a reaction or stop catalyzing a reaction.
- the ability of the sensor component, for example, to modulate the catalytic activity of the nucleic acid sensor molecule, is inhibited or diminished.
- the sensor component can comprise recognition properties relating to chemical or physical signals capable of modulating the nucleic acid sensor molecule via chemical or physical changes to the structure of the nucleic acid sensor molecule.
- the sensor component can be derived from a naturally occurring nucleic acid binding sequence, for example, RNAs that bind to other nucleic acid sequences in vivo.
- the sensor component can be derived from a nucleic acid molecule (aptamer) which is evolved to bind to a nucleic acid sequence within a target nucleic acid molecule (see for example Gold et al, US 5,475,096 and 5,270,163).
- the sensor component can be covalently linked to the nucleic acid sensor molecule, or can be non-covalently associated. A person skilled in the art will recognize that all that is required is that the sensor component is able to selectively inhibit the activity of the nucleic acid sensor molecule to catalyze a reaction.
- target molecule or “target signaling molecule” is meant a molecule capable of interacting with a nucleic acid sensor molecule, specifically a sensor domain of a nucleic acid sensor molecule, in a manner that causes the nucleic acid sensor molecule to be active or inactive.
- the interaction of the signaling agent with a nucleic acid sensor molecule can result in modification of the enzymatic nucleic acid component of the nucleic acid sensor molecule via chemical, physical, topological, or conformational changes to the structure of the molecule, such that the activity of the enzymatic nucleic acid component of the nucleic acid sensor molecule is modulated, for example is activated or deactivated.
- Signaling agents can comprise target signaling molecules such as macromolecules, ligands, small molecules, metals and ions, nucleic acid molecules including but not limited to RNA and DNA or analogs thereof, proteins, peptides, antibodies, polysaccharides, lipids, sugars, microbial or cellular metabolites, pharmaceuticals, and organic and inorganic molecules in a purified or unpurified form, for example VEGF, VEGFRl and or VEGFR2.
- target signaling molecules such as macromolecules, ligands, small molecules, metals and ions, nucleic acid molecules including but not limited to RNA and DNA or analogs thereof, proteins, peptides, antibodies, polysaccharides, lipids, sugars, microbial or cellular metabolites, pharmaceuticals, and organic and inorganic molecules in a purified or unpurified form, for example VEGF, VEGFRl and or VEGFR2.
- triplex forming oligonucleotides refers to an oligonucleotide that can bind to a double-stranded DNA in a sequence-specific manner to form a triple-strand helix. Formation of such a triple helix structure has been shown to inhibit transcription of a targeted gene (Duval-Valentin et al, 1992 Proc. Natl Acad. Sci. USA 89, 504; Fox, 2000, Curr. Med. Chem., 7, 17-37; Praseuth et. al, 2000, Biochim. Biophys. Ada, 1489, 181-206).
- the nucleic acid molecules that modulate the expression of VEGF and or VEGFr represent a novel therapeutic approach to treat or control a variety of angiogenesis related disorders and conditions, including but not limited to tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and/or endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), and/or menopausal dysfunction.
- tumor angiogenesis cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma
- ocular indications such as diabetic retinopathy, or age related macular degeneration
- endometriosis endometrial carcinoma
- gynecologic bleeding disorders irregular menstrual cycles
- nucleic acid molecules that modulate the expression of VEGF and/or VEGFr such as VEGFRl and/or VEGFR2 specific nucleic acids also represent a novel approach to control ovulation or embryonic implantation and therefore provide a novel means of birth control.
- a nucleic acid molecule of the instant invention can be between 12 and 100 nucleotides in length.
- An exemplary enzymatic nucleic acid molecule of the invention is shown as Formula I and/or Formula ⁇ .
- enzymatic nucleic acid molecules of the invention are preferably between 15 and 50 nucleotides in length, more preferably between 25 and 40 nucleotides in length, e.g., 34, 36, or 38 nucleotides in length (for example see Jarvis et al, 1996, J. Biol. Chem., 271, 29107- 29112).
- Exemplary DNAzymes of the invention are preferably between 15 and 40 nucleotides in length, more preferably between 25 and 35 nucleotides in length, e.g., 29, 30, 31, or 32 nucleotides in length (see for example Santoro et al, 1998, Biochemistry, 37, 13330-13342; Chartrand et al, 1995, Nucleic Acids Research, 23, 4092-4096).
- Exemplary antisense molecules of the invention are preferably between 15 and 75 nucleotides in length, more preferably between 20 and 35 nucleotides in length, e.g., 25, 26, 27, or 28 nucleotides in length (see for example Woolf et al, 1992, PNAS, 89, 7305-7309; Milner et al, 1997, Nature Biotechnology, 15, 537-541).
- Exemplary triplex forming oligonucleotide molecules of the invention are preferably between 10 and 40 nucleotides in length, more preferably between 12 and 25 nucleotides in length, e.g., 18, 19, 20, or 21 nucleotides in length (see for example Maher et al, 1990, Biochemistry, 29, 8820-8826; Srrobel and Dervan, 1990, Science, 249, 73-75).
- Those skilled in the art will recognize that all that is required is that the nucleic acid molecule be of length and conformation sufficient and suitable for the nucleic acid molecule to catalyze a reaction contemplated herein.
- the length of the nucleic acid molecules of the instant invention are not limiting within the general limits stated.
- a nucleic acid molecule that modulates, for example, down-regulates, VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 replication or expression comprises between 8 and 100 bases complementary to a nucleic acid molecule of VEGFRl and/or VEGFR2. More preferably, a nucleic acid molecule that modulates VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 replication or expression comprises between 14 and 24 bases complementary to a nucleic acid molecule of VEGFRl and or VEGFR2.
- the invention provides a method for producing a class of nucleic acid-based gene modulating agents which exhibit a high degree of specificity for the nucleic acid of a desired target.
- a nucleic acid molecule of the invention is preferably targeted to a highly conserved sequence region of target nucleic acids encoding VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 (specifically VEGF, VEGFRl and/or VEGFR2 genes) such that specific treatment of a disease or condition can be provided with either one or several nucleic acid molecules of the invention.
- target nucleic acids encoding VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 (specifically VEGF, VEGFRl and/or VEGFR2 genes) such that specific treatment of a disease or condition can be provided with either one or several nucleic acid molecules of the invention.
- Such nucleic acid molecules can be delivered exogenously to specific tissue or cellular targets as required.
- cell is used in its usual biological sense, and does not refer to an entire multicellular organism.
- the cell can, for example, be in vitro, e.g., in cell culture, or present in a multicellular organism, including,, e.g., birds, plants and mammals such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats.
- the cell may be prokaryotic
- eukaryotic e.g., mammalian or plant cell
- VEGFRl and/or VEGFR2 proteins protein receptor or a mutant protein derivative thereof, having vascular endothelial growth factor receptor activity, for example, having the ability to bind vascular endothelial growth factor and/or having tyrosine kinase activity.
- highly conserved sequence region is meant, a nucleotide sequence of one or more regions in a target gene does not vary significantly from one generation to the other or from one biological system to the other.
- Angiogenesis refers to formation of new blood vessels which is an essential process in reproduction, development and wound repair.
- Tumor angiogenesis refers to the induction of the growth of blood vessels from surrounding tissue into a solid tumor. Tumor growth and tumor metastasis are dependent on angiogenesis (for a review see Folkman, 1985 supra; Folkman 1990 J. Natl. Cancer Inst., 82, 4; Folkman and Shing, 1992 J. Biol. Chem. 267, 10931).
- Angiogenesis plays an important role in other diseases such as arthritis wherein new blood vessels have been shown to invade the joints and degrade cartilage (Folkman and Shing, supra).
- Retinopathy refers to inflammation of the retina and or degenerative condition of the retina which may lead to occlusion of the retina and eventual blindness
- h diabetic retinopathy angiogenesis causes the capillaries in the retina to invade the vitreous resulting in bleeding and blindness which is also seen in neonatal retinopathy (for a review see
- Nucleic acid-based inhibitors of VEGF and/or VEGFr such as VEGFRl and or VEGFR2 expression are useful for the prevention, treatment, and/or control of angiogenesis related disorders and conditions, including but not limited to, tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and/or endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), menopausal dysfunction, and other diseases or conditions that are related to or will respond to the levels of VEGF, VEGFRl and/or VEGFR2 in a cell or tissue, alone or in combination with other therapies.
- tumor angiogenesis cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma
- VEGF and/or VEGFr such as VEGFRl and/or VEGFR2 expression (specifically VEGF, VEGFRl and/or VEGFR2 gene RNA levels) and thus reduction in the level of the respective protein relieves, to some degree, the symptoms of the disease or condition.
- Nucleic acid-based inhibitors of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 expression are also useful as birth control agents, for example by inhibition of ovulation or embryonic uterine implantation.
- the nucleic acid molecules of the invention can be added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells or tissues.
- nucleic acid complexes can be locally admimstered to relevant tissues ex vivo, or in vivo through injection or infusion pump, with or without their incorporation in biopolymers.
- nucleic acid inhibitors comprise sequences, which are complementary to polynucleotides, for example DNA and RNA, having VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 sequence.
- Triplex molecules of the invention can be provided targeted to DNA target regions, and containing the DNA equivalent of a target sequence or a sequence complementary to the specified target (substrate) sequence.
- Antisense molecules typically are complementary to a target sequence along a single contiguous sequence of the antisense molecule.
- an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop.
- the antisense molecule can be complementary to two (or even more) non-contiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both.
- the active nucleic acid molecule of the invention for example, an enzymatic nucleic acid molecule, contains an enzymatic center or core equivalent to those in the examples, and binding arms able to bind nucleic acid such that cleavage at the target site occurs.
- a core region can, for example, include one or more loop, stem-loop structure, or linker which does not prevent enzymatic activity.
- a particular region of a nucleic acid molecule of the invention can be such a loop, stem-loop, nucleotide linker, and/or non-nucleotide linker and can be represented generally as sequence "X".
- a core region may, for example, include one or more loop or stem-loop structures which do not prevent enzymatic activity.
- a core sequence for a hammerhead enzymatic nucleic acid can comprise a conserved sequence, such as 5'-CUGAUGAG-3' and 5'-CGAA- 3' connected by "X", where X is 5'-GCCGUUAGGC-3' (SEQ ID NO 5979), or any other Stem II region known in the art, or a nucleotide and/or non-nucleotide linker.
- sequences or non-nucleotide linkers can be present that do not interfere with the function of the nucleic acid molecule.
- Sequence X can be a linker of ⁇ 2 nucleotides in length, preferably 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 26, 30, where the nucleotides can preferably be internally base-paired to form a stem of preferably > 2 base pairs.
- sequence X can be a non-nucleotide linker
- the nucleotide linker X can be a nucleic acid aptamer, such as an ATP aptamer, HTV Rev aptamer (RRE), HIV Tat aptamer (TAR) and others (for a review see Gold et al, 1995, Annu. Rev. Biochem., 64, 763; and Szostak & Ellington, 1993, in The RNA World, ed. Gesteland and Atkins, pp. 511, CSH Laboratory Press).
- a nucleic acid aptamer includes a nucleic acid sequence capable of interacting with a ligand.
- the ligand can be any natural or a synthetic molecule, including but not limited to a resin, metabolites, nucleosides, nucleotides, drugs, toxins, transition state analogs, peptides, lipids, proteins, amino acids, nucleic acid molecules, hormones, carbohydrates, receptors, cells, viruses, bacteria and others.
- non-nucleotide linker X is as defined herein.
- non-nucleotide include either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid, or polyhydrocarbon compounds. Specific examples include those described by Seela and Kaiser, Nucleic Acids Res. 1990, 7 ⁇ °:6353 and Nucleic Acids Res. 1987, 75:3113; Cload and Schepartz, J. Am. Chem. Soc. 1991, 773:6324; Richardson and Schepartz, J Am. Chem. Soc. 1991, 775:5109; Ma et al, Nucleic Acids Res.
- non-nucleotide further means any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity.
- the group or compound can be abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine.
- the invention features an enzymatic nucleic acid molecule having one or more non-nucleotide moieties, and having enzymatic activity to cleave an RNA or DNA molecule.
- nucleic acid molecules that interact with target nucleic acid molecules and down-regulate VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 (specifically VEGF, VEGFRl and/or VEGFR2 gene) activity are expressed from transcription units inserted into DNA or RNA vectors.
- the recombinant vectors are preferably DNA plasmids or viral vectors.
- Enzymatic nucleic acid molecule or antisense expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus.
- the recombinant vectors capable of expressing the enzymatic nucleic acid molecules or antisense are delivered as described above, and persist in target cells.
- viral vectors can be used that provide for transient expression of enzymatic nucleic acid molecules or antisense. Such vectors can be repeatedly administered as necessary.
- the enzymatic nucleic acid molecules or antisense bind to the target nucleic acid and down-regulate its function or expression.
- Delivery of enzymatic nucleic acid molecule or antisense expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex- planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell.
- Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector.
- vectors any nucleic acid- and/or viral-based technique used to deliver a desired nucleic acid.
- subject or “patient” is meant an organism, which is a donor or recipient of explanted cells, or the cells themselves.
- Subject or “Patient” also refers to an organism to which the nucleic acid molecules of the invention can be admimstered.
- a subject or patient is a mammal or mammalian cells. More preferably, a subject or patient is a human or human cells.
- enhanced enzymatic activity is meant to include activity measured in cells and/or in vivo where the activity is a reflection of both the catalytic activity and the stability of the nucleic acid molecules of the invention.
- the product of these properties can be increased in vivo compared to an all RNA enzymatic nucleic acid or all DNA enzyme, i some cases, the activity or stability of the nucleic acid molecule can be decreased (i.e., less than ten-fold), but the overall activity of the nucleic acid molecule is enhanced, in vivo.
- nucleic acid molecules of the instant invention can be used to treat diseases or conditions discussed above.
- the patient can be treated, or other appropriate cells can be treated, as is evident to those skilled in the art, individually or in combination with one or more drugs under conditions suitable for the treatment.
- the described molecules of the invention can be used in combination with other known treatments to treat conditions or diseases discussed above.
- the described molecules can be used in combination with one or more known therapeutic agents to treat angiogenesis related disorders and conditions, including but not limited to tumor angiogenesis, cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma, or ocular indications such as diabetic retinopathy, or age related macular degeneration, and/or endometriosis, birth control, endometrial tumors, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), menopausal dysfunction, endometrial carcinoma, and/or other diseases or conditions which respond to the modulation of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 expression.
- tumor angiogenesis cancers such as breast cancer, lung cancer, colorectal cancer, renal cancer, pancreatic cancer, or melanoma
- ocular indications such as diabetic retinopathy, or age related macular degeneration
- Figure 1 shows a secondary structure model of ANGIOZYMETM ribozyme bound to its RNA target.
- Figure 2 shows a time course of inhibition of primary tumor growth following systemic administration of ANGIOZYMETM in the LLC mouse model.
- Figure 3 shows inhibition of primary tumor growth following systemic administration of ANGIOZYMETM according to a certain dosing regimen in the LLC mouse model.
- Figure 4 shows a dose-dependent inhibition of tumor metastases following systemic administration of ANGIOZYMETM in a mouse colorectal model.
- Figure 5 is a graph showing the plasma concentration profile of ANGIOZYMETM after a single subcutaneous (SC) dose of 10, 30, 100 or 300 mgm 2 .
- Figure 6 shows examples of chemically stabilized ribozyme motifs.
- HH Rz represents hammerhead ribozyme motif (Usman et al, 1996, Curr. Op. Struct. Bio., 1, 527);
- NCH Rz represents the NCH ribozyme motif (Ludwig et al, International PCT Publication No. WO
- G-CIeaver represents G- cleaver ribozyme motif (Kore et al, 1998, Nucleic Acids Research 26, 4116-4120, Eckstein et al, US 6,127,173).
- N or n represent independently a nucleotide which can be same or different and have complementarity to each other;
- rl represents ribo-Inosine nucleotide; arrow indicates the site of cleavage within the target.
- Position 4 of the HH Rz and the NCH Rz is shown as having 2'-C-allyl modification, but those skilled in the art will recognize that this position can be modified with other modifications well known in the art, so long as such modifications do not significantly inhibit the activity of the ribozyme.
- Figure 7 shows an example of a Zinzyme A ribozyme motif that is chemically stabilized (see for example Beigehnan et al, International PCT publication No. WO 99/55857 and US Patent Application Serial No. 09/918,728).
- Figure 8 shows an example of a DNAzyme motif described by Santoro et al, 1997
- Figure 9 shows data demonstrating the inhibition of soluble VEGFRl in a clinical study using ANGIOZYME (SEQ ID NO: 5977) .
- Figure 10 shows an generalized outline for the mouse model of proliferative retinopathy showing the points of ribozyme administration.
- Figure 11 shows a graph demonstrating the efficacy of a VEGF-receptor-targeted enzymatic nucleic acid molecule in a mouse model of proliferative retinopathy.
- Enzymatic Nucleic Acid Several varieties of naturally-occurring enzymatic nucleic acids are presently known. Pn addition, several in vitro selection (evolution) strategies (Orgel, 1979, Proc. R. Soc.
- the enzymatic nature of an enzymatic nucleic acid molecule has significant advantages, one advantage being that the concentration of enzymatic nucleic acid molecule necessary to affect a therapeutic treatment is lower. This advantage reflects the ability of the enzymatic nucleic acid molecule to act enzymatically. Thus, a single enzymatic nucleic acid molecule is able to cleave many molecules of target nucleic acid.
- the enzymatic nucleic acid molecule is a highly specific inhibitor, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target nucleic acid , but also on the mechanism of target nucleic acid cleavage. Single mismatches, or base- substitutions, near the site of cleavage can be chosen to completely eliminate catalytic activity of a enzymatic nucleic acid molecule.
- Nucleic acid molecules having an endonuclease enzymatic activity are able to repeatedly cleave other separate nucleic acid molecules in a nucleotide base sequence-specific manner. With the proper design, such enzymatic nucleic acid molecules can be targeted to RNA transcripts, and achieve efficient cleavage in vitro (Zaug et al, 324, Nature 429 1986; Uhlenbeck, 1987 Nature 328, 596; Kim et al, 84 Proc. Natl. Acad. Sci. USA 8788, 1987; Dreyfus, 1988, Einstein Quart. J. Bio.
- trans-cleaving enzymatic nucleic acid molecules can be used as therapeutic agents for human disease (Usman & McSwiggen, 1995 Ann. Rep. Med. Chem. 30, 285-294; Christoffersen and Marr, 1995 J. Med. Chem. 38, 2023-2037).
- Enzymatic nucleic acid molecules can be designed to cleave specific nucleic acid targets within the background of cellular nucleic acid. Such a cleavage event renders the nucleic acid non-functional and abrogates protein expression from that nucleic acid. In this manner, synthesis of a protein associated with a disease state can be selectively inhibited (Warashina et al, 1999, Chemistry and Biology, 6, 237-250).
- Enzymatic nucleic acid molecules of the invention that are allosterically regulated can be used to down-regulate VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2, expression.
- allosteric enzymatic nucleic acids or allozymes see for example Usman et al, US Patent Application No. 09/877,526, George et al, US Patent Nos. 5,834,186 and 5,741,679, Shih et al, US Patent No. 5,589,332, Nathan et al, US Patent No 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al, International PCT Publication Nos.
- WO 00/26226 and 98/27104 are designed to respond to a signaling agent, for example, mutant VEGFRl and/or VEGFR2 protein, wild-type VEGFRl and/or VEGFR2 protein, mutant VEGFRl and/or VEGFR2 RNA, wild-type VEGFRl and/or VEGFR2 RNA, other proteins and/or RNAs involved in VEGF signal transduction, compounds, metals, polymers, molecules and/or drugs that are targeted to VEGFRl and/or VEGFR2 expression, which in turn modulates the activity of the enzymatic nucleic acid molecule.
- a signaling agent for example, mutant VEGFRl and/or VEGFR2 protein, wild-type VEGFRl and/or VEGFR2 protein, mutant VEGFRl and/or VEGFR2 RNA, wild-type VEGFRl and/or VEGFR2 RNA, other proteins and/or RNAs involved in VEGF signal transduction, compounds, metals, polymers, molecules
- the activity of the allosteric enzymatic nucleic acid is activated or inhibited such that the expression of a particular target is selectively down-regulated.
- the target can comprise wild-type VEGFRl and/or VEGFR2, mutant VEGFRl and/or VEGFR2, and/or a predetermined component of the VEGF signal transduction pathway, hi a specific example, allosteric enzymatic nucleic acid molecules that are activated by interaction with a RNA encoding VEGF protein are used as therapeutic agents in vivo.
- RNA encoding the VEGF protein activates the allosteric enzymatic nucleic acid molecule that subsequently cleaves the RNA encoding a VEGFRl and/or VEGFR2 protein resulting in the inhibition of VEGFRl and/or VEGFR2 protein expression.
- an allozyme can be activated by a VEGF and or VEGFr, such as VEGFRl and/or VEGFR2 protein, peptide, or mutant polypeptide that causes the allozyme to inhibit the expression of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 genes, by, for example, cleaving RNA encoded by VEGF, VEGFRl and/or VEGFR2 gene, hi this non-limiting example, the allozyme acts as a decoy to inhibit the function of VEGF, VEGFRl and/or VEGFR2 and also inhibit the expression of VEGF, VEGFRl and/or VEGFR2 once activated by the VEGF, VEGFRl and/or VEGFR2 protein.
- a VEGF and or VEGFr such as VEGFRl and/or VEGFR2 protein, peptide, or mutant polypeptide that causes the allozyme to inhibit the expression of VEGF and/or VE
- Antisense molecules can be modified or unmodified RNA, DNA, or mixed polymer oligonucleotides and primarily function by specifically binding to matching sequences resulting in inhibition of peptide synthesis (Wu-Pong, Nov 1994, BioPharm, 20- 33).
- the antisense oligonucleotide binds to target RNA by Watson Crick base-pairing and blocks gene expression by preventing ribosomal translation of the bound sequences either by steric blocking or by activating RNase H enzyme.
- Antisense molecules can also alter protein synthesis by interfering with RNA processing or transport from the nucleus into the cytoplasm (Mukhopadhyay & Roth, 1996, Crit. Rev. in Oncogenesis 7, 151-190).
- antisense molecules have been described that utilize novel configurations of chemically modified nucleotides, secondary structure, and/or RNase H substrate domains (Woolf et al, International PCT Publication No. WO 98/13526; Thompson et al, International PCT Publication No. WO 99/54459; Hartmann et al, USSN 60/101,174 which was filed on September 21, 1998) all of these are incorporated by reference herein in their entirety.
- antisense deoxyoligoribonucleotides can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex.
- Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector or equivalents and variations thereof.
- TFO Triplex Forming Oligonucleotides
- RNA degradation found in higher vertebrates (Mitra et al, 1996, Proc Nat Acad Sci USA 93, 6780-6785).
- Two types of enzymes, 2-5A synthetase and RNase L, are required for RNA cleavage.
- the 2-5A synthetases require double stranded RNA to form 2'-5' oligoadenylates (2-5 A).
- 2-5 A then acts as an allosteric effector for utilizing RNase L which has the ability to cleave single stranded RNA.
- RNase L which has the ability to cleave single stranded RNA.
- the ability to form 2-5A structures with double stranded RNA makes this system particularly useful for inhibition of viral replication.
- oligoadenylate structures can be covalently linked to antisense molecules to form chimeric oligonucleotides capable of RNA cleavage (Torrence, supra). These molecules putatively bind and activate a 2-5A dependent RNase, the oligonucleotide/enzyme complex then binds to a target RNA molecule which can then be cleaved by the RNase enzyme.
- RNAi Double-stranded RNAs can suppress expression of homologous genes through an evolutionarily conserved process named RNA interference (RNAi) or post-transcriptional gene silencing (PTGS).
- silencing is the degradation of target mRNAs by an RNP complex, which contains short interfering RNAs (siRNAs) as guides to substrate selection.
- Short interfering RNAs are typically 21 to 23 nucleotides in length.
- a bidentate nuclease called Dicer has been implicated as the protein responsible for siRNA production.
- dsRNA double-stranded RNA
- the dsRNA feeds into a biological pathway and is broken into short pieces of short interfering (si) RNAs.
- the siRNA triggers the degradation of the messenger RNA that matches its sequence (see for example Tuschl et al, International PCT Publication No. WO 01/75164; Bass, 2001, Nature, 411, 428-429; Elbasbir et al., 2001, Nature, 411, 494-498; and Kreutzer et al, International PCT Publication No. WO 00/44895).
- Targets for useful nucleic acid molecules of the invention such as enzymatic nucleic acid molecules, dsRNA, and antisense nucleic acids can be determined as disclosed in Draper et al, WO 93/23569; Sullivan et al, WO 93/23057; Thompson et al, WO 94/02595; Draper et al, WO 95/04818; McSwiggen et al, US Patent No. 5,525,468, and hereby incorporated by reference herein in totality.
- Other examples include the following PCT applications, which concern inactivation of expression of disease-related genes: WO 95/23225, WO 95/13380, WO 94/02595, incorporated by reference herein.
- Enzymatic nucleic acid molecules and antisense to such targets are designed as described in those applications and synthesized to be tested in vitro and in vivo, as also described.
- the sequences of human VEGF, VEGFRl and/or VEGFR2 RNAs are screened for optimal nucleic acid target sites using a computer-folding algorithm. Potential nucleic acid binding cleavage sites are identified.
- mouse targeted enzymatic nucleic acid molecules can be useful to test efficacy of action of the nucleic acid molecule prior to testing in humans.
- Nucleic acid molecule binding/cleavage sites are identified, for example enzymatic nucleic acid, antisense, and dsRNA mediated binding sites are chosen.
- the nucleic acid molecules are individually analyzed by computer folding (Jaeger et al, 1989 Proc. Natl. Acad. Sci. USA, 86, 7706) to assess whether the sequences fold into the appropriate secondary structure.
- Those nucleic acid molecules with unfavorable intramolecular interactions such as between the binding arms and the catalytic core can be eliminated from consideration. Varying binding arm lengths can be chosen to optimize activity.
- Nucleic acids such as antisense, RNAi, and/or enzymatic nucleic acid molecule binding/cleavage sites are identified and are designed to anneal to various sites in the nucleic acid target.
- the binding arms of enzymatic nucleic acid molecules of the invention are complementary to the target site sequences described above.
- Antisense and RNAi sequences are designed to have partial or complete complementarity to the nucleic acid target.
- the nucleic acid molecules can be chemically synthesized. The method of synthesis used follows the procedure for normal DNA/RNA synthesis as described below and in Usman et al, 1987 J. Am. Chem.
- nucleic acid motifs small refers to nucleic acid motifs less than about 100 nucleotides in length, preferably less than about 80 nucleotides in length, and more preferably less than about 50 nucleotides in length; e.g., antisense oligonucleotides, enzymatic nucleic acids, aptamers, allozymes, decoys, siRNA etc.
- small nucleic acid motifs are preferably used for exogenous delivery.
- the simple structure of these molecules increases the ability of the nucleic acid to invade targeted regions of RNA structure.
- Exemplary molecules of the instant invention are chemically synthesized, and others can similarly be synthesized.
- oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'- end.
- small scale syntheses are conducted on a 394 Applied Biosystems, hie. synthesizer using a 0.2 ⁇ mol scale protocol with a 2.5 min coupling step for 2'-O-methylated nucleotides and a 45 sec coupling step for 2'-deoxy nucleotides.
- Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle.
- syntheses at the 0.2 ⁇ mol scale can be performed on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, CA) with minimal modification to the cycle.
- synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methyl imidazole in THF (ABI) and 10% acetic anhydride/ 10% 2,6-lutidine in THF (ABI); and oxidation solution is 16.9 mM I 2 , 49 mM pyridine, 9% water in THF (PERSEPTIVETM). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, hie. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2- Benzodithiol-3-one 1,1-dioxide, 0.05 M in acetonitrile) is used.
- Deprotection of the D ⁇ A polynucleotides is performed as follows: the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65 °C for 10 min. After cooling to -20 °C, the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeC ⁇ :H2O/3:l:l, vortexed and the supernatant is then added to the first supernatant. The combined supematants, containing the oligoribonucleotide, are dried to a white powder.
- RNA oligonucleotides including certain nucleic acid molecules of the invention follows the procedure as described in Usman et al, 1987, J. Am. Chem. Soc, 109, 7845; Scaringe et al, 1990, Nucleic Acids Res., 18, 5433; and Wincott et al, 1995, Nucleic Acids Res. 23, 2677-2684 Wincott et al, 1997, Methods Mol. Bio. , 74, 59, and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
- common nucleic acid protecting and coupling groups such as dimethoxytrityl at the 5'-end, and phosphoramidites at the 3'-end.
- small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 ⁇ mol scale protocol with a 7.5 min coupling step for alkylsilyl protected nucleotides and a 2.5 min coupling step for 2'-O-methylated nucleotides.
- Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle.
- syntheses at the 0.2 ⁇ mol scale can be done on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, CA) with minimal modification to the cycle.
- Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by colorimetric quantitation of the trityl fractions, are typically 97.5-99%.
- synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methyl imidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); oxidation solution is 16.9 mM I 2 , 49 mM pyridine, 9% water in THF (PERSEPTTV ⁇ TM). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American Intemational Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-l,2-Benzodithiol-3-one 1,1- dioxide ⁇ .05 M in acetonitrile) is used.
- Deprotection of the R ⁇ A is performed using either a two-pot or one-pot protocol.
- the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65 °C for 10 min. After cooling to -20 °C, the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeC ⁇ :H2O/3:l:l, vortexed and the supernatant is then added to the first supernatant.
- the combined supematants, containing the oligoribonucleotide, are dried to a white powder.
- the base deprotected oligoribonucleotide is resuspended in anhydrous TEA/HF/NMP solution (300 ⁇ L of a solution of 1.5 mL N- methylpyrrolidinone, 750 ⁇ L TEA and 1 mL TEA » 3HF to provide a 1.4 M HF concentration) and heated to 65 °C. After 1.5 h, the oligomer is quenched with 1.5 M NH4HCO3.
- the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 33% ethanolic methylamine/DMSO: 1/1 (0.8 mL) at 65 °C for 15 min.
- the vial is brought to r.t. TEA»3HF
- RNA is detritylated with 0.5% TFA for 13 min. The cartridge is then washed again with water, salt exchanged with 1 M NaCl and washed with water again.
- the oligonucleotide is then eluted with 30% acetonitrile.
- Inactive hammerhead ribozymes or binding attenuated control (BAC) oligonucleotides are synthesized by substituting a U for G5 and a U for A14 (numbering from Hertel, K. J., et al, 1992, Nucleic Acids Res., 20, 3252). Similarly, one or more nucleotide substitutions can be introduced in otlier enzymatic nucleic acid molecules to inactivate the molecule and such molecules can serve as a negative control.
- nucleic acid molecules of the present invention can be synthesized separately and joined together post-synthetically, for example by ligation (Moore et al, 1992,
- the nucleic acid molecules of the present invention are modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2'-amino, 2'- C-allyl, 2'-flouro, 2'-0-methyl, 2'-H (for a review see Usman and Cedergren, 1992, TIBS 17, 34; Usman et al, 1994, Nucleic Acids Symp. Ser. 31, 163).
- Ribozymes are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al, Supra, the totality of which is hereby incorporated herein by reference) and are re-suspended in water.
- oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-O-methyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al, 1994, Nucleic Acids Symp. Ser. 31, 163; Burgin et al, 1996, Biochemistry, 35, 14090).
- nuclease resistant groups for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-O-methyl, 2'-H, nucleotide base modifications
- Nucleic acid molecules having chemical modifications that maintain or enhance activity are provided. Such nucleic acid is also generally more resistant to nucleases than unmodified nucleic acid. Thus, in a cell and/or in vivo the activity may not be significantly lowered.
- Therapeutic nucleic acid molecules delivered exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state.
- nucleic acid molecules must be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of RNA and DNA (Wincott et al, 1995 Nucleic Acids Res.
- nucleic acid molecules of the invention include one or more G- clamp nucleotides.
- a G-clamp nucleotide is a modified cytosine analog wherein the modifications confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, see for example Lin and Matteucci, 1998, J. Am. Chem. Soc, 120, 8531-8532.
- nucleic acid molecules of the mvention include one or more LNA "locked nucleic acid" nucleotides such as a 2', 4'-C mythylene bicyclo nucleotide (see for example Wengel et al, International PCT Publication No. WO 00/66604 and WO 99/14226).
- the invention features conjugates and/or complexes of nucleic acid molecules targeting VEGF receptors such as VEGFRl and/or VEGFR2.
- VEGF receptors such as VEGFRl and/or VEGFR2.
- conjugates and/or complexes can be used to facilitate delivery of molecules into a biological system, such as cells.
- the conjugates and complexes provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention.
- the present invention encompasses the design and synthesis of novel conjugates and complexes for the delivery of molecules, including but not limited to small molecules, lipids, phospholipids, nucleosides, nucleotides, nucleic acids, antibodies, toxins, negatively charged polymers and other polymers, for example proteins, peptides, hormones, carbohydrates, polyethylene glycols, or polyamines, across cellular membranes.
- the transporters described are designed to be used either individually or as part of a multi- component system, with or without degradable linkers.
- Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules.
- biodegradable nucleic acid linker molecule refers to a nucleic acid molecule that is designed as a biodegradable linker to connect one molecule to another molecule, for example, a biologically active molecule.
- the stability of the biodegradable nucleic acid linker molecule can be modulated by using various combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, for example, 2'-O-methyl, 2'-fluoro, 2'-amino, 2'-O-amino, 2'-C-allyl, 2'-O-allyl, and other 2'-modified or base modified nucleotides.
- the biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphoms based linkage, for example, a phosphoramidate or phosphodiester linkage.
- the biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
- biodegradable refers to degradation in a biological system, for example enzymatic degradation or chemical degradation.
- biologically active molecule refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system.
- biologically active molecules contemplated by the instant invention include therapeutically active molecules such as antibodies, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, 2,5-A chimeras, siRNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
- Biologically active molecules of the invention also include molecules capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules, for example, lipids and polymers such as polyamines, polya ides, polyethylene glycol and other polyethers.
- phospholipid refers to a hydrophobic molecule comprising at least one phosphoms group.
- a phospholipid can comprise a phosphoms containing group and saturated or unsaturated alkyl group, optionally substituted with OH, COOH, oxo, amine, or substituted or unsubstituted aryl groups.
- nucleic acid molecules e.g., enzymatic nucleic acid molecules and antisense nucleic acid molecules
- delivered exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state.
- These nucleic acid molecules should be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of nucleic acid molecules described in the instant invention and in the art have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability as described above.
- nucleic acid catalysts having chemical modifications that maintain or enhance enzymatic activity are provided.
- Such nucleic acids are also generally more resistant to nucleases than unmodified nucleic acid.
- the activity of the nucleic acid may not be significantly lowered.
- enzymatic nucleic acids are useful in a cell and/or in vivo even if activity over all is reduced 10 fold (Burgin et al, 1996, Biochemistry, 35, 14090).
- Such enzymatic nucleic acids herein are said to "maintain" the enzymatic activity of an all RNA ribozyme or all DNA DNAzyme.
- nucleic acid molecules comprise a 5' and/or a 3'- cap structure.
- cap structure is meant chemical modifications, which have been incorporated at either terminus of the oligonucleotide (see for example Wincott et al, WO 97/26270, incorporated by reference herein). These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell.
- the cap can be present at the 5'-terminus (5'-cap) or at the 3'-terminus (3'-cap) or can be present on both terminus, hi non-limiting examples, the 5 '-cap includes inverted abasic residue (moiety), 4', 5 '-methylene nucleotide; l-(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha- nucleotides; modified base nucleotide; phosphorodithioate linkage; tAreo-pentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5- dihydroxypentyl nucleotide, 3 '-3
- the 3 '-cap includes, for example 4', 5 '-methylene nucleotide; 1- (beta-D-erythrofuranosyl) nucleotide; 4'-thio nucleotide, carbocyclic nucleotide; 5'-amino- alkyl phosphate; l,3-diamino-2-propyl phosphate, 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; t ⁇ reo-pentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,
- non-nucleotide any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity.
- the group or compound is abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine.
- alkyl refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
- the alkyl group has 1 to 12 carbons. More preferably it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
- alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups.
- the alkenyl group has 1 to 12 carbons. More preferably it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
- alkyl also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups.
- the alkynyl group has 1 to 12 carbons. More preferably it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons.
- alkyl groups can also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups.
- An "aryl” group refers to an aromatic group which has at least one ring having a conjugated p electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted.
- the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups.
- alkylaryl refers to an alkyl group (as described above) covalently joined to an aryl group (as described above).
- Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
- Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
- Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
- An "amide” refers to an -C(O)-NH-R, where R is either alkyl, aryl, alkylaryl or hydrogen.
- An “ester” refers to an -C(O)-OR', where R is either alkyl, aryl, alkylaryl or hydrogen.
- nucleotide is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a phosphorylated sugar.
- Nucleotides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1' position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar and a phosphate group.
- the nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see for example, Usman and McSwiggen, supra; Eckstein et al, International PCT Publication No. WO 92/07065; Usman et al, International PCT Publication No. WO 93/15187; Uhhnan & Peyman, supra all are hereby incorporated by reference herein).
- modified nucleic acid bases known in the art as summarized by Limbach et al, 1994, Nucleic Acids Res. 22, 2183.
- nucleic acids include, for example, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g.
- 6-methyluridine 6-methyluridine
- propyne quesosine, 2- thiouridine, 4-thiouridine, wybutosine, wybutoxosine, 4-acetylcytidine, 5- (carboxyhydroxymethyl)uridine, 5 '-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluridine, beta-D-galactosylqueosine, 1-methyladenosine, 1- methylinosine, 2,2-dimethylguanosine, 3-methylcytidine, 2-methyladenosine, 2- methylguanosine, N6-methyladenosine, 7-methylguanosine, 5-methoxyaminomethyl-2- thiouridine, 5-methylaminomethyluridine, 5-methylcarbonylmethyluridine, 5- methyloxyuridine, 5-methyl-2-thiouridine, 2-methylthio-N6-isopentenyladenosine, beta-D- mannosylque
- modified bases in this aspect is meant nucleotide bases other than adenine, guanine, cytosine and uracil at 1' position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule.
- nucleoside is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a sugar.
- Nucleosides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1' position of a nucleoside sugar moiety.
- Nucleosides generally comprise a base and sugar group.
- the nucleosides can be unmodified or modified at the sugar, and/or base moiety, (also referred to interchangeably as nucleoside analogs, modified nucleosides, non-natural nucleosides, non- standard nucleosides and other; see for example, Usman and McSwiggen, supra; Eckstein et al, Intemational PCT Publication No.
- nucleic acids Some of the non- limiting examples of chemically modified and other natural nucleic acid bases that can be introduced into nucleic acids include, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g.
- modified bases in this aspect is meant nucleoside bases other than adenine, guanine, cytosine and uracil at 1' position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule.
- the invention features modified enzymatic nucleic acid molecules with phosphate backbone modifications comprising one or more phosphorothioate, phosphorodithioate, methylphosphonate, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl, substitutions.
- abasic sugar moieties lacking a base or having other chemical groups in place of a base at the 1' position, for example a 3',3'-linked or 5',5'-linked deoxyabasic ribose derivative (for more details see Wincott et al, Intemational PCT publication No. WO 97/26270).
- unmodified nucleoside is meant one of the bases adenine, cytosine, guanine, thymine, uracil joined to the 1' carbon of ⁇ -D-ribo-furanose.
- modified nucleoside any nucleotide base which contains a modification in the chemical structure of an unmodified nucleotide base, sugar and/or phosphate.
- amino is meant 2'-NH 2 or 2'-0- NH 2 , which can be modified or unmodified. Such modified groups are described, for example, in Eckstein et al, U.S. Patent 5,672,695 and
- nucleic acid e.g., antisense and ribozyme
- modifications to nucleic acid can be made to enhance the utility of these molecules.
- modifications can enhance shelf-life, half-life in vitro, stability, and ease of introduction of such oligonucleotides to the target site, including, e.g., enhancing penetration of cellular membranes and conferring the ability to recognize and bind to targeted cells.
- nucleic acid-based molecules of the invention can lead to better treatment of the disease progression by affording the possibility of combination therapies (e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs) and or other chemical or biological molecules).
- combination therapies e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs) and or other chemical or biological molecules).
- the treatment of patients with nucleic acid molecules can also include combinations of different types of nucleic acid molecules.
- Therapies can be devised which include a mixture of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs), allozymes, antisense, dsRNA, aptamers, and/or 2-5A chimera molecules to one or more targets to alleviate symptoms of a disease.
- nucleic acid molecules Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; and Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995 which are both incorporated herein by reference.
- Sullivan et al, PCT WO 94/02595 further describes the general methods for delivery of enzymatic RNA molecules. These protocols can be utilized for the delivery of virtually any nucleic acid molecule.
- Nucleic acid molecules can be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
- the nucleic acid/vehicle combination is locally delivered by direct injection or by use of an infusion pump.
- Other routes of delivery include, but are not limited to oral (tablet or pill form) and/or intrathecal delivery (Gold, 1997, Neuroscience, 76, 1153-1158).
- Other approaches include the use of various transport and carrier systems, for example though the use of conjugates and biodegradable polymers.
- the molecules of the instant invention can be used as pharmaceutical agents.
- Pharmaceutical agents prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state in a patient.
- the polynucleotides of the invention can be administered (e.g., RNA, DNA or protein) and introduced into a patient by any standard means, with or without stabilizers, buffers, and the like, to form a pharmaceutical composition.
- RNA, DNA or protein e.g., RNA, DNA or protein
- standard protocols for formation of liposomes can be followed.
- the compositions of the present invention can also be formulated and used as tablets, capsules or elixirs for oral administration; suppositories for rectal administration; sterile solutions; suspensions for injectable administration; and the other compositions known in the art.
- the present invention also includes pharmaceutically acceptable formulations of the compounds described.
- formulations include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, acetic acid, and benzene sulfonic acid.
- a pharmacological composition or formulation refers to a composition or formulation in a form suitable for administration, e.g., systemic administration, into a cell or patient, preferably a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect.
- systemic administration in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body.
- Administration routes which lead to systemic absorption include, without limitations: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular.
- Each of these administration routes expose the desired negatively charged polymers, e.g., nucleic acids, to an accessible diseased tissue.
- the rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size.
- the use of a liposome or other drug carrier comprising the compounds of the instant invention can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES).
- RES reticular endothelial system
- a liposome formulation which can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful.
- This approach can provide enhanced delivery of the drug to target cells by taking advantage of the specificity of macrophage and lymphocyte immune recognition of abnormal cells, such as cells implicated in endometriosis, birth control, endometrial tumors, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), menopausal dysfunction, and endometrial carcinoma.
- compositions or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention in the physical location most suitable for their desired activity.
- agents suitable for formulation with the nucleic acid molecules of the instant invention include: PEG conjugated nucleic acids, phospholipid conjugated nucleic acids, nucleic acids containing lipophilic moieties, phosphorothioates, P-glycoprotein inhibitors (such as Pluronic P85) which can enhance entry of drugs into various tissues, for example the CNS (Jolliet- Riant and Tillement, 1999, Fundam. Clin.
- biodegradable polymers such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery after implantation (Emerich, DF et al, 1999, Cell Transplant, 8, 47-58) Alkermes, hie. Cambridge, MA; and loaded nanoparticles, such as those made of polybutylcyanoacrylate, which can deliver drugs across the blood brain barrier and can alter neuronal uptake mechanisms (Prog Neuropsychopharmacol Biol Psychiatry, 23, 941-949, 1999).
- Other non-limiting examples of delivery strategies, including CNS delivery of the nucleic acid molecules of the instant invention include material described in Boado et al, 1998, J. Pharm.
- the invention also features the use of the composition comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes).
- Nucleic acid molecules of the invention can also comprise covalently attached PEG molecules of various molecular weights. These formulations offer a method for increasing the accumulation of drugs in target tissues. This class of drug carriers resists opsonization and elimination by the mononuclear phagocytic system (MPS or RES), thereby enabling longer blood circulation times and enhanced tissue exposure for the encapsulated drag (Lasic et al. Chem. Rev. 1995, 95, 2601-2627; Ishiwata et al, Chem. Pharm. Bull.
- MPS or RES mononuclear phagocytic system
- liposomes have been shown to accumulate selectively in tumors, presumably by extravasation and capture in the neovascularized target tissues (Lasic et al, Science 1995, 267, 1275-1276; Oku et al, 1995, Biochim. Biophys. Ada, 1238, 86-90).
- the long-circulating liposomes enhance the pharmacokinetics and pharmacodynamics of DNA and RNA, particularly compared to conventional cationic liposomes which are known to accumulate in tissues of the MPS (Liu et al, J. Biol. Chem. 1995, 42, 24864-24870; Choi et al, Intemational PCT Publication No.
- WO 96/10391 Ansell et al, Intemational PCT Publication No. WO 96/10390; Holland et al, International PCT Publication No. WO 96/10392; all of which are incorporated by reference herein).
- Long- circulating liposomes are also likely to protect drugs from nuclease degradation to a greater extent compared to cationic liposomes, based on their ability to avoid accumulation in metabolically aggressive MPS tissues such as the liver and spleen. All of these references are incorporated by reference herein.
- compositions prepared for storage or administration which include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent.
- Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A.R. Gennaro edit. 1985) hereby incorporated by reference herein.
- preservatives, stabilizers, dyes and flavoring agents can be provided. These include sodium benzoate, sorbic acid and esters of p- hydroxybenzoic acid.
- antioxidants and suspending agents can be used.
- a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state.
- the pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer.
- nucleic acid molecules of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
- parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
- a pharmaceutical formulation comprising a nucleic acid molecule of the invention and a pharmaceutically acceptable carrier.
- One or more nucleic acid molecules of the invention can be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants, and if desired other active ingredients.
- compositions containing nucleic acid molecules of the invention can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
- compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
- Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
- excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, com starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
- the tablets can be uncoated or they can be coated by known techniques, hi some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
- a time delay material such as glyceryl monosterate or glyceryl distearate can be employed.
- Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
- an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
- water or an oil medium for example peanut oil, liquid paraffin or olive oil.
- Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
- excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoole
- the aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
- preservatives for example ethyl, or n-propyl p- hydroxybenzoate
- coloring agents for example ethyl, or n-propyl p- hydroxybenzoate
- flavoring agents for example ethyl, or n-propyl p- hydroxybenzoate
- sweetening agents such as sucrose or saccharin.
- Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
- the oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
- Sweetening agents and flavoring agents can be added to provide palatable oral preparations.
- These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
- Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
- a dispersing or wetting agent e.g., glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerin, glycerin, glycerin, glycerin, glycerin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol
- compositions of the invention can also be in the form of oil-in-water emulsions.
- the oily phase can be a vegetable oil or a mineral oil or mixtures of these.
- Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
- the emulsions can also contain sweetening and flavoring agents.
- Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents.
- the pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
- the sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
- the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium.
- any bland fixed oil can be employed including synthetic mono-or diglycerides.
- fatty acids such as oleic acid find use in the preparation of injectables.
- the nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug.
- These compositions can be prepared by mixing the drag with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
- suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
- Such materials include cocoa butter and polyethylene glycols.
- Nucleic acid molecules of the invention can be administered parenterally in a sterile medium.
- the drug depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
- adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.
- Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per patient per day).
- the amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration.
- Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient.
- the specific dose level for any particular patient depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drag combination and the severity of the particular disease undergoing therapy.
- the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
- nucleic acid molecules of the present invention can also be administered to a patient in combination with other therapeutic compounds to increase the overall therapeutic effect.
- the use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
- certain of the nucleic acid molecules of the instant invention can be expressed within cells from eukaryotic promoters (e.g., Izant and Weintraub, 1985, Science, 229, 345; McGarry and Lindquist, 1986, Proc Natl. Acad. Sci, USA 83, 399; Scanlon et al, 1991, Proc. Nat Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al, 1992, Antisense Res.
- eukaryotic promoters e.g., Izant and Weintraub, 1985, Science, 229, 345; McGarry and Lindquist, 1986, Proc Natl. Acad. Sci, USA 83, 399; Scan
- nucleic acid can be expressed in eukaryotic cells from the appropriate DNA/RNA vector.
- the activity of such nucleic acids can be augmented by their release from the primary transcript by a enzymatic nucleic acid (Draper et al, PCT WO 93/23569, and Sullivan et al, PCT WO 94/02595; Ohkawa et al, 1992, Nucleic Acids Symp. Ser., 27, 15-6; Taira et al, 1991, Nucleic Acids Res., 19, 5125-30; Ventura et al, 1993, Nucleic Acids Res., 21, 3249-55; Chowrira et al, 1994, J. Biol.
- RNA molecules of the present invention are preferably expressed from transcription units (see for example Couture et al, 1996, TIG, 12, 510) inserted into DNA or RNA vectors.
- the recombinant vectors are preferably DNA plasmids or viral vectors. Ribozyme expressing viral vectors can be constructed based on, but not hmited to, adeno-associated vims, retrovirus, adenovirus, or alphavirus.
- the recombinant vectors capable of expressing the nucleic acid molecules are delivered as described above, and persist in target cells.
- viral vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary.
- Delivery of nucleic acid molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et al, 1996, TIG, 12, 510).
- the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the nucleic acid molecules of the instant invention.
- the nucleic acid sequence encoding the nucleic acid molecule of the instant invention is operably linked in a manner which allows expression of that nucleic acid molecule.
- the invention features an expression vector comprising: a) a transcription initiation region (e.g., eukaryotic pol I, It or III initiation region); b) a transcription termination region (e.g., eukaryotic pol I, ⁇ or m termination region); c) a nucleic acid sequence encoding at least one of the nucleic acid catalyst of the instant invention; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
- the vector can optionally include an open reading frame (ORF) for a protein operably linked on the 5' side or the 3'-side of the sequence encoding the nucleic acid catalyst of the invention; and/or an intron (intervening sequences).
- ORF open reading frame
- RNA polymerase I RNA polymerase I
- RNA polymerase II RNA polymerase II
- RNA polymerase UI RNA polymerase UI
- Transcripts from pol II or pol IU promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby.
- Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990, Proc. Natl Acad. Sci.
- nucleic acid molecules such as ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al, 1992, Antisense Res. Dev., 2, 3- 15; Ojwang et al, 1992, Proc. Natl. Acad. Sci.
- transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high concentrations of desired RNA molecules such as ribozymes in cells (Thompson et al, supra; Couture and Stinchcomb, 1996, supra; Noonberg et al, 1994, Nucleic Acid Res., 22, 2830; Noonberg et al, US Patent No. 5,624,803; Good et al, 1997, Gene Tlier., 4, 45; Beigehnan et al, Intemational PCT Publication No. WO 96/18736; all of these publications are incorporated by reference herein.
- ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchconib, 1996, supra).
- plasmid DNA vectors such as adenovirus or adeno-associated virus vectors
- viral RNA vectors such as retroviral or alphavirus vectors
- the invention features an expression vector comprising nucleic acid sequence encoding at least one of the nucleic acid molecules of the invention, in a manner which allows expression of that nucleic acid molecule.
- the expression vector comprises in one embodiment; a) a transcription initiation region; b) a transcription termination region; c) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
- the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an open reading frame; d) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3'-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said open reading frame and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
- the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region, said intron and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
- the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) an open reading frame; e) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3 '-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said intron, said open reading frame and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
- Flt-1 (VEGFRl), KDR (VEGFR2) and/or flk-1 are attractive nucleic acid-based therapeutic targets by several criteria.
- the interaction between VEGF and VEGF-R is well- established. Efficacy can be tested in well-defined and predictive animal models. Finally, the disease conditions are serious and current therapies are inadequate.
- nucleic acid-based therapy based on the molecules and methods described herein provides a direct and elegant approach to directly modulate flt-1, KDR and/or flk-1 expression.
- VEGFRl and VEGFR2 mRNAs are highly homologous in certain regions, some nucleic acid target sites are also homologous.
- a single nucleic acid molecule of the invention can target both VEGFRl and VEGFR2 mRNAs.
- a single nucleic acid molecule can sometimes be designed to accommodate a site on both mRNAs by including G/U base pairing.
- the enzymatic nucleic acid can be synthesized with a U at the complementary position and it will bind both to sites.
- the advantage of one enzymatic nucleic acid that targets both VEGFRl and VEGFR2 mRNAs is clear, especially in cases where both VEGF receptors may contribute to the progression of angiogenesis in the disease state. Examples
- Example 1 Enzymatic nucleic acid-mediated inhibition of angiogenesis in vivo
- ribozymes targeted against flt-1 4229 site (SED ID NO: 5977) in the rat cornea model of VEGF induced angiogenesis (see above).
- These ribozymes have either active or inactive catalytic core and either bind and cleave or just bind to VEGF-R mRNA of the flt-1 subtype.
- the active ribozymes that are able to bind and cleave the target RNA, have been shown to inhibit ( 125 I-labeled) VEGF binding in cultured endothelial cells and produce a dose-dependent decrease in VEGF induced endothelial cell proliferation in these cells.
- the catalytically inactive forms of these ribozymes which can only bind to the RNA but cannot catalyze RNA cleavage, failed to inhibit VEGF binding and failed to decrease VEGF induced endothelial cell proliferation.
- the ribozymes and VEGF were co-delivered using the filter disk method: Nitrocellulose filter disks (Millipore®) of 0.057 diameter were immersed in appropriate solutions and were surgically implanted in rat cornea as described by Pandey et al, supra. This delivery method has been shown to deliver rhodamine-labeled free ribozyme to scleral cells and, in all likelihood cells of the pericorneal vascular plexus. Since the active ribozymes show cell culture efficacy and can be delivered to the target site using the disk method, it is essential that these ribozymes be assessed for in vivo anti-angiogenic activity.
- the stimulus for angiogenesis in this study was the treatment of the filter disk with 30 ⁇ M VEGF which is implanted within the cornea's stroma. This dose yields reproducible neovascularization stemming from the pericorneal vascular plexus growing toward the disk in a dose-response study 5 days following implant. Filter disks treated only with the vehicle for VEGF show no angiogenic response.
- the ribozymes were co-adminstered with VEGF on a disk in two different ribozyme concentrations. One concern with the simultaneous administration is that the ribozymes will not be able to inhibit angiogenesis since VEGF receptors can be stimulated.
- VEGF and RIBOZYMES were prepared as a 2X solution for 1 : 1 mixing for final concentrations above, with the exception of solution 1 in which VEGF was 2X and diluted with ribozyme diluent (sterile water).
- the 2X VEGF solution (60 ⁇ M) was prepared from a stock of 0.82 ⁇ g/ ⁇ L in 50 mM Tris base. 200 ⁇ L of VEGF stock was concentrated by speed vac to a final volume of 60.8 ⁇ L, for a final concentration of 2.7 ⁇ g/ ⁇ L or 60 ⁇ M. Six 10 ⁇ L aliquots was prepared for daily mixing. 2X solutions for VEGF and Ribozyme was stored at 4°C until the day of the surgery. Solutions were mixed for each day of surgery. Original 2X solutions was prepared on the day before the first day of the surgery.
- mice were treated with the treatment groups as described above. Animals were allowed to recover for 5 days after treatment with daily observation (scoring 0 - 3). On the fifth day animals were euthanized and digital images of each eye was obtained for quantitaion using Image Pro Plus. Quantitated neovascular surface area were analyzed by ANOVA followed by two post-hoc tests including Dunnets and Tukey-Kramer tests for significance at the 95% confidence level. Dunnets provide information on the significance between the differences within the means of treatments vs. controls while Tukey-Kramer provide information on the significance of differences within the means of each group.
- the flt-1 4229 (SEQ ID NO: 5977) active hammerhead ribozyme at both concentrations was effective at inhibiting angiogenesis while the inactive ribozyme did not show any significant reduction in angiogenesis.
- a statistically signifying reduction in neovascular surface area was observed only with active ribozymes. This result clearly shows that the ribozymes are capable of significantly inhibiting angiogenesis in vivo. Specifically, given ribozyme mechanism of action, the observed inhibition is by the binding and cleavage of target RNA by ribozymes.
- Ribozymes Hammerhead ribozymes and controls designed to have attenuated activity (attenuated controls) were synthesized and purified as previously described above.
- the attenuated ribozyme controls maintain the binding arm sequence of the parent ribozyme and thus are still capable of binding to the mRNA target. However, they have two nucleotide changes in the core sequence that substantially reduce their ability to carry out the cleavage reaction.
- Ribozymes were designed to target Flt-1 or KDR mRNA sites conserved in human, mouse, and rat. In general, ribozymes with binding arms of seven nucleotides were designed and tested.
- In vitro ribozyme cleavage assays In vitro RNA cleavage rates on a 15 nucleotide synthetic RNA substrate were measured as previously described above.
- HMVEC-d Human dermal microvascular endothelial cells
- FBS fetal bovine serum
- VEGF 165 (165 amino acids) was selected for cell culture and animal studies because it is the predominant form of the four native forms of VEGF generated by alternative mRNA splicing. Cell culture assays were carried out in triplicate.
- Ribozymes or attenuated controls were formulated for cell culture studies and used immediately. Formulations were carried out with LlPOFECTAMINETM (Gibco BRL) at a 3:1 lipid to phosphate charge ratio in serum-free medium (OPTI-MEMTM, Gibco BRL) by mixing for 20 minutes at room temperature. For example, a 3:1 lipid to phosphate charge ratio was established by complexing 200 nM ribozyme with 10.8 ⁇ g/ ⁇ L LlPOFECTAMINETM (13.5 ⁇ M DOSPA).
- HMVEC-d were seeded (5 x 10 3 cells/well) in 48-well plates
- RNAse protection assay HMVEC-d were seeded (2 x 10 5 cells/well) in 6-well plates (Costar) and allowed to grow 32-36 hours in Growth medium at 37°C. Cells were treated with LlPOFECTAMINETM complexes containing 200 nM ribozyme or attenuated control for 2 h as described under "Proliferation Assay” and then incubated in Growth medium containing 20 ng/mL VEGF ⁇ 65 for 24 hours. Cells were harvested and an RNAse protection assay was carried out using the Ambion Direct Protect kit and protocol with the exception that 50 mM EDTA was added to the lysis buffer to eliminate the possibility of ribozyme cleavage during sample preparation.
- RNA probes targeting portions of Flt-1 and KDR were prepared by transcription in the presence of [ 32 P]-UTP. Samples were analyzed on polyacrylamide gels and the level of protected RNA fragments was quantified using a Molecular Dynamics Phosphorhnager. The levels of Flt-1 and KDR were normalized to the level of cyclophilin (human cyclophilin probe template, Ambion) in each sample. The coefficient of variation for cyclophilin levels was 11% [265940 cpm ⁇ 29386 (SD)] for all conditions tested here (ie. in the presence of either active ribozymes or attenuated controls). Thus, cyclophilin is useful as an internal standard in these studies.
- Rat corneal pocket assay of VEGF-induced angiogenesis Rat corneal pocket assay of VEGF-induced angiogenesis:
- VEGF soaked disk For comeal implantation, 0.57 mm diameter nitrocellulose disks, prepared from 0.45 ⁇ m pore diameter nitrocellulose filter membranes (Millipore Corporation), were soaked for 30 min in 1 ⁇ L of 30 ⁇ M VEGF 165 in 82 mM Tris ⁇ Cl (pH 6.9) in covered petri dishes on ice.
- Corneal surgery The rat comeal model used in this study was a modified from Koch et al Supra and Pandey et al, supra. Briefly, corneas were irrigated with 0.5% povidone iodine solution followed by normal saline and two drops of 2% lidocaine. Under a dissecting microscope (Leica MZ-6), a stromal pocket was created and a presoaked filter disk (see above) was inserted into the pocket such that its edge was 1 mm from the comeal linibus.
- test solution ribozyme, attenuated control or sterile water vehicle
- test solution ribozyme, attenuated control or sterile water vehicle
- NSA neovascular surface area
- Ribozyme-mediated reduction of VEGF-induced cell proliferation Ribozyme cleavage of Flt-1 or KDR mRNA should result in a decrease in the density of cell surface VEGF receptors. This decrease should limit VEGF binding and consequently interfere with the mitogenic signaling induced by VEGF.
- proliferation assays using cultured human microvascular cells were carried out. Ribozymes included in the proliferation assays were initially chosen by their ability to decrease the level of VEGF binding to treated cells, hi these initial studies, ribozymes targeting 20 sites in the coding region of each mRNA were screened.
- the active ribozymes tested decreased the relative proliferation of HMVEC-d after VEGF stimulation, an effect that increased with ribozyme concentration. This concentration dependency was not observed following treatment with the attenuated controls designed for these sites, hi fact, little or no change in cell growth was noted following treatment with the attenuated controls, even though these controls can still bind to the specific target sequences. At 200 nM, there was a distinct "window" between the anti-proliferative effects of each ribozyme and its attenuated control; a trend also observed at lower doses.
- the first order rate constants were 0.434 ⁇ 0.024 min l and 0.002 ⁇ 1 x 10 "4 min "1 , respectively.
- the attenuated controls retain a very slight level of cleavage activity under these optimized conditions, the decrease in in vitro cleavage activity between each active ribozyme and its paired attenuated control is about two orders of magnitude.
- an active core is essential for cleavage activity in vitro and is also necessary for ribozyme activity in cell culture.
- Ribozyme-mediated reduction of VEGF-induced angiogenesis in vivo To assess whether ribozymes targeting VEGF receptor mRNA could impact the complex process of angiogenesis, prototypic mti-Flt-1 and KDR ribozymes that were identified in cell culture studies were screened in a rat comeal pocket assay of VEGF-induced angiogenesis. In this assay, corneas implanted with VEGF-containing filter disks exhibited a robust neovascular response in the comeal region between the disk and the comeal limbus (from which the new vessels emerge). Disks containing a vehicle solution elicited no angiogenic response. In separate studies, intraconjunctival injections of sterile water vehicle did not affect the magnitude of the VEGF-induced angiogenic response, hi addition, ribozyme injections alone did not induce angiogenesis.
- the dose-related effects of anti-Flt-1 or KDR ribozymes on the VEGF-induced angiogenic response were then examined.
- the antiangiogenic effect of the ar ⁇ i-Flt-1 (site 4229) and KDR (site 726) ribozymes and their attenuated controls over a dose range from 1 to 100 ⁇ g, respectively was determined.
- the maximal antiangiogenic response 48 and 36% for axAi-Flt-1 and KDR ribozymes, respectively) was observed at a dose of 10 ⁇ g.
- the dxxXi-Flt-1 ribozyme produced a significantly greater antiangiogenic response than its attenuated control at 3 and 10 ⁇ g (p ⁇ 0.05). Its attenuated control exhibited a small but significant antiangiogenic response at doses above 10 ⁇ g compared to vehicle treated VEGF controls (p ⁇ 0.05). At its maximum, this response was not significantly greater than that observed with the lowest dose of active anti- t-i ribozyme.
- the mti-KDR ribozyme significantly inhibited angiogenesis from 3 to 30 ⁇ g (p ⁇ 0.05). The a ti-KDR attenuated control had no significant effect at any dose tested.
- Example 3 In vivo inhibition of tumor growth and metastases by VEGF-R ribozymes.
- the tumors in this study were derived from a cell line (LLC-HM) which gives rise to reproducible numbers of spontaneous lung metastases when propagated in vivo.
- LLC-HM line was obtained from Dr. Michael O'Reilly, Harvard University. Tumor neovascularization in Lewis lung carcinoma has been shown to be VEGF-dependent. Tumors from mice bearing LLC-HM (selected for the highly metastatic phenotype by serial propagation) were harvested 20 days post-inoculation. A tumor brei suspension was prepared from these tumors according to standard protocols. On day 0 of the study, 0.5 x 10 6 viable LLC-HM tumor cells were injected subcutaneously (sc) into the dorsum or flank of previously untreated mice (100 ⁇ L injectate).
- Ribozyme solutions were prepared to deliver to another set of animals 100, 10, 3, or 1 mg/kg/day of ANGIOZYMETM via Alzet mini-pumps.
- a total of 10 animals per dose or saline control group were surgically implanted on the left flank with osmotic mini-pumps pre- filled with the respective test solution three days following tumor inoculation. Pumps were attached to indwelling jugular vein catheters.
- Figure 2 shows the antitumor effects of ANGIOZYMETM.
- ANGIOZYMETM significantly reduced (p ⁇ 0.05) the number of lung metastatic foci in animals inoculated either in the flank regions.
- Figure 3 illustrates the dose-dependent anti-metastatic effect of ANGIOZYMETM compared to saline control.
- KM12L4a-16 is a human colorectal cancer cell line.
- KM12L4a-16 cells were implanted into the spleen of nude mice.
- Alzet minipumps were implanted and continuous subcutaneous delivery of either saline or 12, 36 or 100 mg/kg/ day of ANGIOZYMETM was initiated.
- the spleens containing the primary tumors were removed.
- the Alzet minipumps were replaced with fresh pumps so that delivery of saline or ANGIOZYMETM was continuous over a 28 day period from day 3 to day 32. Animals were euthanized on day 41 and the liver tumor burden was evaluated.
- Example 4 Effect of ANGIOZYMETM alone or in combination with chemotherapeutic agents in the mouse Lewis Lung Carcinoma Model.
- RPI.13141 an attenuated version of RPI.4610 in which four nucleotides in the catalytic core are changed so that the cleavage activity is dramatically decreased.
- RPI.13141 maintains the base composition and binding arms of RPI.4610 and so is still capable of binding to the target site.
- the second control also has changes to the catalytic core (three) to inhibit cleavage activity, but in addition the sequence of the binding arms has been scrambled so that it can no longer bind to the target sequence.
- One nucleotide in the arm of RPI.13030 is also changed to maintain the same base composition as RPI.4610.
- Ribozyme administrations Ribozymes and controls were resuspended in normal saline. Administration was initiated seven days following tumor inoculation. Animals either received a daily subcutaneous injection (30 mg/kg test substance) from day 7 to day 20 or were instrumented with an Alzet osmotic minipump (12 ⁇ L/day flow rate) containing a solution of ribozyme or control. Subcutaneous infusion pumps delivered the test substances (30 mg kg/day) from day 7 to 20 (14-day pumps, 420 mg/kg total test substance) or days 7-34 (28-day pumps, 840 mg/kg total test substance). Where indicated, chemotherapeutic agents were given in combination with ribozyme treatment.
- Cyclophosphamide was given by intraperitoneal administration on days 7, 9 and 11 (125 mg/kg).
- Gemcitabine was given by intraperitoneal administration on days 8, 11 and 14 (125 mg/kg). Untreated, uninstrumented animals were used as comparison. Five animals were included in each group.
- ANGIOZYMETM The antiangiogenic ribozyme, ANGIOZYMETM, was tested in a model of Lewis lung carcinoma alone and in combination with two chemotherapeutic agents. Previously (see above), 30 mg/kg/day ANGIOZYMETM alone was determined to inhibit both primary tumor growth and lung metastases in a highly metastatic variant of Lewis lung (continuous 14-day iv deliveryvt ⁇ Alzet minipump, manuscript in preparation). In this study, 30 mg/kg/day ANGIOZYMETM delivered either as a daily subcutaneous bolus injection or as a continuous infusion from an Alzet minipump resulted in a delay in tumor growth.
- tumor growth to 500 mm was delayed by ⁇ 7 days in animals being treated with ANGIOZYMETM compared to an unfreated group. Growth of tumors in animals being treated with either of two attenuated controls was delayed by only ⁇ 2 days.
- ANGIOZYMETM delivered by subcutaneous bolus was also tested in combination with either Gemcytabine or cyclophosphamide.
- Tumor growth delay increased by about 3 days in the presence of combination therapy with ANGIOZYMETM and Gemcytabine over the effects of either treatment alone.
- the combination of ANGIOZYMETM and cyclophosphamide did not increase tumor growth delay over that of cyclophosphamide alone, however, suboptimal doses of cyclophosphamide were not included in this study. Neither of the attenuated controls increased the effect of the chemotherapeutic agents.
- ANGIOZYMETM The effect of ANGIOZYMETM on metastases to the lung was also determined in the presence and absence of additional chemotherapeutic freatment. Macrometastases to the lungs were counted in two animals in each treatment group on day 20. In the presence of ANGIOZYMETM, with or without a chemotherapeutic agent, the lung metastases were reduced to zero. Treatment with either Gemcytabine or cyclophosphamide alone (mean number of metastases 4.5 and 4, respectively) were not as effective as ANGIOZYMETM alone or when used in combination with ANGIOZYMETM. Neither of the attenuated controls increased the effect of the chemotherapeutic agents.
- ANGIOZYMETM delivered by a daily subcutaneous bolus resulted in zero metastases (Fig.4) in the two animals counted, it is possible that the additional burden of being instrumented with the minipump contributes to a slightly decreased response to ANGIOZYMETM.
- the sequence of human VEGFRl and/or VEGFR2 genes are screened for accessible sites using a computer-folding algorithm. Regions of the RNA that do not form secondary folding structures and contain potential enzymatic nucleic acid molecule and/or antisense binding/cleavage sites are identified.
- An exemplary sequence of an enzymatic nucleic acid molecule of the invention is shown in Formula I and/or Formula JJ (SEQ ID Nos: 5977 and 5978, respectively).
- Other nucleic acid molecules and targets contemplated by the invention are described in Pavco et al, US Patent Application No. 09/870,161, incorporated by reference herein in its entirety.
- nucleic acid molecules of the invention including antisense, aptamers, dsRNA, siRNA, and/or 2,5-A chimeras, can be designed to modulate the expression of the nucleic acid targets described in Pavco et al, US Patent Application No. 09/870,161.
- Enzymatic nucleic acid molecule target sites are chosen by analyzing sequences of human VEGFRl receptor (for example Genbank Accession No. NM_002019), and VEGFR2 receptor (for example Genbank Accession No. NM_002253) genes and prioritizing the sites on the basis of folding. Enzymatic nucleic acid molecules are designed that can bind each target and are individually analyzed by computer folding (Christoffersen et al, 1994 J. Mol. Struc Theochem, 311, 273; Jaeger et al, 1989, Proc. Natl. Acad. Sci. USA, 86, 7706) to assess whether the enzymatic nucleic acid molecule sequences fold into the appropriate secondary structure.
- binding arm lengths can be chosen to optimize activity. Generally, at least 4 bases on each arm are able to bind to, or otherwise interact with, the target RNA.
- Example 7 Chemical Synthesis and Purification of Ribozymes and Antisense for Efficient Cleavage and/or blocking of VEGFRl and/or VEGFR2 RNA
- Enzymatic nucleic acid molecules and antisense constructs are designed to anneal to various sites in the RNA message.
- the binding arms of the enzymatic nucleic acid molecules are complementary to the target site sequences described above, while the antisense constructs are fully complementary to the target site sequences described above.
- RNAi molecules likewise have one strand of RNA or a portion of RNA complementarity to the target site sequence or a portion of the target site sequence.
- complementarity within the double-strand RNAi structure is formed from two separate individual RNA strands or from self-complementary areas of a topologically closed, individual RNA sfrand which can be optionally circular.
- the nucleic acid molecules were chemically synthesized. The method of synthesis used followed the procedure for normal RNA synthesis as described above and in Usman et al, (1987 J. Am. Chem.
- Nucleic acid molecules are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbeck, 1989, Methods Enzymol. 180, 51). Nucleic acid molecules of the invention are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al, supra; the totality of which is hereby incorporated herein by reference) and are resuspended in water. Examples of sequences of chemically synthesized enzymatic nucleic acid molecules are shown in Formula I (SEQ ID NO: 5977), Formula II (SEQ ID NO: 5978) and in Pavco et al, US Patent Application No. 09/870, 161.
- Example 8 Enzymatic Nucleic Acid Molecule Cleavage of VEGFRl and/or VEGFR2 RNA Target in vitro
- Enzymatic nucleic acid molecules targeted to the human VEGFRl and/or VEGFR2 are targeted to the human VEGFRl and/or VEGFR2
- RNA are designed and synthesized as described above. These enzymatic nucleic acid molecules can be tested for cleavage activity in vitro, for example, using the following procedure.
- the target sequences and the nucleotide location within the VEGFRl and/or VEGFR2 RNA are described in Pavco et al, US Patent Application No. 09/870,161.
- Full-length or partially full-length, internally-labeled target RNA for enzymatic nucleic acid molecule cleavage assay is prepared by in vitro transcription in the presence of [a- 3 p] CTP, passed over a G 50 Sephadex column by spin chromatography and used as substrate RNA without further purification.
- substrates are 5'-32p-end labeled using T4 polynucleotide kinase enzyme.
- Assays are performed by pre-warming a 2X concentration of purified enzymatic nucleic acid molecule in enzymatic nucleic acid molecule cleavage buffer (50 mM Tris-HCl, pH 7.5 at 37°C, 10 mM M Ci2) and the cleavage reaction was initiated by adding the 2X enzymatic nucleic acid molecule mix to an equal volume of substrate RNA (maximum of 1-5 nM) that was also pre-warmed in cleavage buffer. As an initial screen, assays are carried out for 1 hour at 37 C using a final concentration of either 40 nM or 1 mM enzymatic nucleic acid molecule, ie., enzymatic nucleic acid molecule excess.
- enzymatic nucleic acid molecule excess 50 mM Tris-HCl, pH 7.5 at 37°C, 10 mM M Ci2
- the reaction is quenched by the addition of an equal volume of 95% formamide, 20 mM EDTA, 0.05%) bromophenol blue and 0.05% xylene cyanol after which the sample is heated to 95 C for 2 minutes, quick chilled and loaded onto a denaturing polyacrylamide gel.
- Substrate RNA and the specific RNA cleavage products generated by enzymatic nucleic acid molecule cleavage are visualized on an autoradiograph of the gel. The percentage of cleavage is determined by Phosphor Imager® quantitation of bands representing the intact subsfrate and the cleavage products.
- Example 9 Phase I/H Study of Repetitive Dosing of ANGIOZYMETM Targeting the VEGFRl (FLT-1) Receptor of VEGF
- a ribozyme therapeutic agent ANGIOZYMETM (SEQ ID NO: 5977), was assessed by daily subcutaneous administration in a phase I/JJ trial for 31 patients with refractory solid tumors. Demographic information relating to patients enrolled in the study are shown in Table III. The primary study endpoint was to determine the safety and maximum tolerated dose of ANGIOZYMETM. Secondary endpoints assessed ANGIOZYMETM pharmacokinetics and clinical response. Patients were treated at the following doses: 3 patients received doses of 10 mg/m ⁇ /day, 4 patients received 30 mg/m.2/day, 20 patients received 100 mg ⁇ /day, and 4 patients received 300 mg/m ⁇ /day. All but one patient were dosed for a minimum of 29 consecutive days with 24-hour pharmacokinetic analyses on Day 1 and 29. Clinical response was assessed monthly. Results The data from 20 patients indicated that
- ANGIOZYMETM was well tolerated, with no systemic adverse events.
- Figure 5 shows the plasma concentration profile of ANGIOZYMETM after a single subcutaneous dose of 10, 30, 100, or 300 mg/m 2 .
- the pharmacokinetic parameters of ANGIOZYMETM after subcutaneous bolus administration are outlined in Table IV.
- An MTD maximum tolerated dose
- One patient in the 300 mg/m 2 /d group experienced a grade 3 injection site reaction.
- Patients in the other groups experienced intermittent grade 1 and grade 2 injection site reactions with erythema and induration. No systemic or laboratory toxicities were observed.
- Example 10 Down-regulation of VEGFRl gene expression to treat gynecologic neovascularization dependent conditions
- ANGIOZYMETM One patient in the Phase I/ ⁇ trial described in Example 19 was menstruating prior to enrollment in the ANGIOZYMETM monotherapy trial. After 1-2 months on trial, the patient's menstmal cycles ceased. The patient remained on trial for approximately 11 months and did not menstruate. The patient then went off the trial for about 4 months and the menstmal cycles resumed. Re-enrollment in the ANGIOZYMETM trial resulted in the patient's menstmal cycle stopping again. This clinical observation suggests that ANGIOZYMETM is interfering with the patient's menstrual cycle, perhaps by inhibiting neovascularization of uterine tissue.
- ANGIOZYMETM has a direct effect on the endometrial tissue or an effect on LH FSH stimulation.
- ANGIOZYMETM SEQ ID NO: 5977
- other nucleic acid molecules of the instant invention of various clinical targets and/or processes associated with female reproduction and gynecologic neovascularization, such as endometriosis, birth control, gynecologic bleeding disorders, irregular menstrual cycles, ovulation, premenstrual syndrome (PMS), menopausal dysfunction, endometrial carcinoma or other condition associated with the expression of VEGFRl and/or VEGFR2 VEGF receptors.
- Example 22 In vivo inhibition of neovascularization in an ocular animal model by VEGF-R ribozymes. Summary of the Mouse Model: A mouse model of proliferative retinopathy (Aiello et al., 1995, Proc. Natl. Acad. Sci. USA 92: 10457-10461; Robinson et al, 1996, Proc. Natl. Acad. Sci. USA 93: 4851-4856; Pierce et al, 1996, Archives of Ophthalmology 114: 1219- 1228) in which neovascularization of the mouse retina is induced by exposure of 7-day old mice to 75% oxygen followed by a return to normal room air.
- the initial period in high oxygen causes an obliteration of developing blood vessels in the retina. Exposure to room air five days later is perceived as hypoxia by the now underperfused retina. The result is an immediate upregulation of VEGF mRNA and VEGF protein (between 6-12 hours) followed by an extensive retinal neovascularization that peaks in ⁇ 5 days.
- this model is more representative of retinopathy of prematurity than diabetic retinopathy, it is an accepted small animal model in which to study neovascular pathophysiology of the retina.
- Tumor angiogenesis Angiogenesis has been shown to be necessary for tumors to grow into pathological size (Folkman, 1971, PNAS 76, 5217-5221; Wellstein & Czubayko, 1996, Breast Cancer Res and Treatment 38, 109-119). In addition, it allows tumor cells to travel through the circulatory system during metastasis. Increased levels of gene expression of a number of angiogenic factors such as vascular endothelial growth factor (VEGF) have been reported in vascularized and edema-associated brain tumors (Berkman et al, 1993 J. Clini. Invest. 91, 153).
- VEGF vascular endothelial growth factor
- Neovascularization has been shown to cause or exacerbate ocular diseases including but not limited to, macular degeneration, neovascular glaucoma, diabetic retinopathy, myopic degeneration, and trachoma (Norrby, 1997, APMIS 105, 417-437).
- Aiello et al, 1994 New Engl. J. Med. 331, 1480 showed that the ocular fluid, of a majority of patients suffering from diabetic retinopathy and other retinal disorders, contains a high concentration of VEGF.
- Miller et al, 1994 Am. J. Paihol 145, 574 reported elevated levels of VEGF mRNA in patients suffering from retinal ischemia. These observations support a direct role for VEGF in ocular diseases. Other factors including those that stimulate VEGF synthesis may also contribute to these indications.
- Rheumatoid arthritis Immunohistochemistry and in situ hybridization studies on tissues from the joints of patients suffering from rheumatoid arthritis show an increased level of VEGF and its receptors (Fava et al, 1994 J. Exp. Med. 180, 341). Additionally, Koch et al, 1994 J. Immunol. 152, 4149, found that VEGF-specific antibodies were able to significantly reduce the mitogenic activity of synovial tissues from patients suffering from rheumatoid arthritis. These observations support a direct role for VEGF in rheumatoid arthritis. Other angiogenic factors including those of the present invention may also be involved in arthritis.
- Endometriosis Various studies indicate that VEGF is directly implicated in endometriosis. hi one study, VEGF concentrations measured by ELISA in peritoneal fluid were found to be significantly higher in women with endometriosis than in women without endometriosis (24.1 ⁇ 15 ng/ml vs 13.3 ⁇ 7.2 ng/ml in normals), hi patients with endometriosis, higher concentrations of VEGF were detected in the proliferative phase of the menstmal cycle (33 + 13 ng/ml) compared to the secretory phase (10.7 + 5 ng/ml). The cyclic variation was not noted in fluid from normal patients (McLaren et al, 1996, Human Reprod.
- VEGF-positive staining of human ectopic endometrium was shown to be localized to macrophages (double immunofluorescent staining with CD 14 marker).
- Peritoneal fluid macrophages demonstrated VEGF staining in women with and without endometriosis.
- increased activation of macrophages was demonstrated in fluid from women with endometriosis compared with controls.
- Peritoneal fluid macrophage conditioned media from patients with endometriosis resulted in significantly increased cell proliferation ([ 3 H] thymidine incorporation) in HUVEC cells compared to controls.
- peritoneal fluid macrophages with VEGFR2 mRNA The percentage of peritoneal fluid macrophages with VEGFR2 mRNA was higher during the secretory phase, and significantly higher in fluid from women with endometriosis (80 ⁇ 15%) compared with controls (32 ⁇ 20%).
- Flt-rnRNA was detected in peritoneal fluid macrophages from women with and without endometriosis, but there was no difference between the groups or any evidence of cyclic dependence (McLaren et al, 1996, J. Clin. Invest. 98, 482-489).
- VEGF has been found to be expressed in secretory columnar epithelium (estrogen-responsive) lining both the oviducts and the uterus in female mice. During the secretory phase, VEGF expression was shown to have shifted to the underlying stroma composing the functional endometrium. In addition to examining the endometrium, neovascularization of ovarian follicles and the corpus luteum, as well as angiogenesis in embryonic implantation sites have been analyzed. For these processes, VEGF was expressed in spatial and temporal proximity to forming vasculature (Shweiki et al, 1993, J. Clin. Invest. 91, 2235-2243).
- the nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of VEGF, VEGFRl and/or VEGFR2 levels, hi addition, the nucleic acid molecules can be used to treat disease state related to VEGF and/or VEGFr, such as VEGFRl and or VEGFR2 levels.
- VEGFRl and/or VEGFR2 levels include, but are not limited to, gynecologic neovascularization, such as endometriosis, endometrial carcinoma, gynecologic bleeding disorders, irregular menstmal cycles, ovulation, premenstrual syndrome (PMS), menopausal dysfunction, other diseases and conditions discussed herein, and other diseases or conditions that are related to or respond to the levels of VEGF and/or VEGFr, such as VEGFRl and or VEGFR2, in a cell or tissue, alone or in combination with other therapies
- GnRH gonadotropin releasing hormone
- Lupron Depot Leuprolide Acetate
- Synarel noferalin acetate
- Zolodex goserelin acetate
- Suprefact buserelin acetate
- Danazol or oral contraceptives including, but not limited to, Depo-Provera or Provera (medroxyprogesterone acetate), or any other estrogen/progesterone contraceptive
- Various chemotherapies can be readily combined with nucleic acid molecules of the invention for the treatment of endometrial carcinoma.
- chemotherapies that can be combined with nucleic acid molecules of the instant invention include various combinations of cytotoxic drags to kill the cancer cells.
- These drugs include but are not limited to paclitaxel (Taxol), docetaxel, cisplatin, methotrexate, cyclophosphamide, doxorubin, fluorouracil carboplatin, edatrexate, gemcitabine, vinorelbine etc.
- paclitaxel Texol
- docetaxel cisplatin
- methotrexate cyclophosphamide
- doxorubin fluorouracil carboplatin
- edatrexate gemcitabine
- vinorelbine vinorelbine
- VEGF-R mRNAs there are several animal models in which the anti-angiogenesis effect of nucleic acids of the present invention, such as ribozymes, directed against VEGF-R mRNAs can be tested.
- a comeal model has been used to study angiogenesis in rat and rabbit since recraitment of vessels can easily be followed in this normally avascular tissue (Pandey et al, 1995 Science 268: 567-569).
- a small Teflon or Hydron disk prefreated with an angiogenesis factor e.g. bFGF or VEGF
- Angiogenesis is monitored 3 to 5 days later.
- Ribozymes directed against VEGF-R mRNAs would be delivered in the disk as well, or dropwise to the eye over the time course of the experiment.
- hypoxia has been shown to cause both increased expression of VEGF and neovascularization in the retina (Pierce et al, 1995 Proc. Natl. Acad. Sci. USA. 92: 905-909; Shweiki et al, 1992 J. Clin. Invest. 91: 2235-2243).
- VEGF is at least partially responsible for tumor angiogenesis (Plate et al, 1992 Nature 359, 845).
- Animal models have been developed in which glioblastoma cells are implanted subcutaneously into nude mice and the progress of tumor growth and angiogenesism is studied (Kim et al, 1993 supra; Millauer et al, 1994 supra).
- Matrigel an extract of basement membrane that becomes a solid gel when injected subcutaneously (Passaniti et al, 1992 Lab. Invest. 67: 519-528).
- angiogenesis factors such as VEGF
- vessels grow into the Matrigel over a period of 3 to 5 days and angiogenesis can be assessed.
- Ribozymes directed against VEGF-R mRNAs can be delivered in the Matrigel to assess anti-angiogesis effect.
- the cornea model is the most common and well characterized anti-angiogenic agent efficacy screening model.
- This model involves an avascular tissue into which vessels are recruited by a stimulating agent (growth factor, thermal or alkalai bum, endotoxin).
- the comeal model utilizes the intrastromal comeal implantation of a Teflon pellet soaked in a VEGF-Hydron solution to recruit blood vessels toward the pellet which can be quantitated using standard microscopic and image analysis techniques.
- ribozymes are applied topically to the eye or bound within Hydron on the Teflon pellet itself.
- This avascular cornea as well as the Matrigel provide for low background assays. While the comeal model has been performed extensively in the rabbit, studies in the rat have also been conducted.
- the mouse model (Passaniti et al., supra) is a non-tissue model which utilizes Matrigel, an extract of basement membrane (Kleinman et al., 1986) or Millipore® filter disk, which can be impregnated with growth factors and anti-angiogenic agents in a liquid form prior to injection. Upon subcutaneous administration at body temperature, the Matrigel or Millipore® filter disk forms a solid implant.
- VEGF embedded in the Matrigel or Millipore® filter disk would be used to recruit vessels within the matrix of the Matrigel or Millipore® filter disk which can be processed histologically for endothelial cell specific vWF (factor Vm antigen) immunohistochemistry, Trichrome-Masson stain, or hemoglobin content.
- vWF factor Vm antigen
- the Matrigel or Millipore® filter disk are avascular; however, it is not tissue.
- ribozymes are administered within the matrix of the Matrigel or Millipore® filter disk to test their anti-angiogenic efficacy.
- delivery issues in this model as with delivery of ribozymes by Hydron- coated Teflon pellets in the rat comea model, are minimized due to the homogeneous presence of the ribozyme within the respective matrix.
- Vessel recruitment using FGF should not be affected in either model by anti- VEGFr mRNA ribozymes.
- Other models of angiogenesis including vessel formation in the female reproductive system using hormonal manipulation (Shweiki et al, 1993 supra); a variety of vascular solid tumor models which involve indirect correlations with angiogenesis (O'Reilly et al, 1994 supra; Senger et al, 1993 supra; Takahasi et al, 1994 supra; Kim et al, 1993 supra); and retinal neovascularization following transient hypoxia (Pierce et al, 1995 supra), were not selected for efficacy screening due to their non-specific nature, although they can be useful models due to a demonstrated correlation between VEGF and angiogenesis.
- mice For a typical systemic study involving 10 mice (20 g each) per dose group, 5 doses (1,
- Identifying a common animal model for systemic efficacy testing of ribozymes is an efficient way of screening ribozymes for systemic efficacy.
- the Lewis lung carcinoma and B-16 murine melanoma models are well accepted models of primary and metastatic cancer and are used for initial screening of anti-cancer agents. These murine models are not dependent upon the use of immunodeficient mice, are relatively inexpensive, and minimize housing concerns. Both the Lewis lung and B-16 melanoma models involve subcutaneous implantation of approximately 10 6 tumor cells from metastatically aggressive tumor cell lines (Lewis lung lines 3LL or D122, LLc-LN7; B-16- BL6 melanoma) in C57BL/6J mice. Alternatively, the Lewis lung model can be produced by the surgical implantation of tumor spheres (approximately 0.8 mm in diameter). Metastasis also can be modeled by injecting the tumor cells directly intraveneously.
- systemic pharmacotherapy with a wide variety of agents usually begins 1-7 days following tumor implantation inoculation with either continuous or multiple administration regimens.
- Concurrent pharmacokinetic studies can be performed to determine whether sufficient tissue levels of ribozymes can be achieved for pharmacodynamic effect to be expected.
- primary tumors and secondary lung metastases can be removed and subjected to a variety of in vitro studies (i.e. target RNA reduction).
- Flt-1, KDR and/or flk-1 protein levels can be measured clinically or experimentally by FACS analysis.
- Flt-1, KDR and or flk-1 encoded mRNA levels can be assessed by Northern analysis, RNase-protection, primer extension analysis and/or quantitative RT-PCR.
- Ribozymes that block flt-1, KDR and/or flk-1 protein encoding mRNAs and therefore result in decreased levels of flt-1, KDR and or flk-1 activity by more than 20% in vitro can be identified.
- Ribozymes and/or genes encoding them are delivered by either free' delivery, liposome delivery, cationic lipid delivery, adeno-associated vims vector delivery, adenovirus vector delivery, retrovirus vector delivery or plasmid vector delivery in these animal model experiments (see above).
- Subjects can be treated by locally administering nucleic acids targeted against VEGF-R by direct injection.
- Routes of administration include, but are not limited to, intravascular, intramuscular, subcutaneous, intraarticular, aerosol inhalation, oral (tablet, capsule or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery.
- Surgically induced models of endometriosis have been developed in rats, mice, and rabbits. Non-human primates demonstrate spontaneous endometriosis, but surgical induction can also be used, hi addition to the surgical technique, cycle monitoring can be performed by daily vaginal cytology in primates. For all of the surgically induced models of endometriosis, the following general procedure is used.
- An initial laparotomy is performed to implant tissue from a donor animal. A portion of one uterine hom (or one complete hom in the case of mice) is removed. The endometrium of this piece of uterus is separated from the myometrium and cut into small segments (4-10 mm2). Segments (approximately 3) are sutured to various locations within the abdominal cavity (peritoneum, intestinal mesentery vessels, uterus, broad ligament). Cummings and Metcalf (1996) attached whole segments of mouse uterus without separating the endometrium from the myometrium. Implants are allowed to grow for 3-6 weeks. A second laparotomy is sometimes performed to verify development of endometriosis-like foci (vascularization and cysts filled with clear fluid).
- Endpoints include, but are not limited to, changes in the surface area of the implants and tissue mass of the ectopic endometrial implants (see for example Brogniez et al, 1995, Human Reprod. 10, 927-931; Cummings et al, 1996, Tox. Appl Pharm. 138, 131-139; Cummings and Metcalf, 1996, Proc. Soc. Exp. Biol.
- Gemcytabine and cyclophosphamide are non-limiting examples of chemotherapeutic agents that can be combined with or used in conjunction with the nucleic acid molecules (e.g. ribozymes and antisense molecules) of the instant invention.
- chemotherapeutic agents that can be combined with or used in conjunction with the nucleic acid molecules (e.g. ribozymes and antisense molecules) of the instant invention.
- ribozymes and antisense molecules e.g. ribozymes and antisense molecules
- Lippincott Company Philadelphia, USA; incorporated herein by reference) and include, without limitations, folates, antifolates, pyrimidine analogs, fluoropyrimidines, purine analogs, adenosine analogs, topoisomerase I inhibitors, anthrapyrazoles, retinoids, antibiotics, anthacyclins, platinum analogs, alkylating agents, nifrosoureas, plant derived compounds such as vinca alkaloids, epipodophyllotoxins, tyrosine kinase inhibitors, taxols, radiation therapy, surgery, nutritional supplements, gene therapy, radiotherapy, for example 3D-CRT, immunotoxin therapy, for example ricin, and monoclonal antibodies.
- chemotherapeutic compounds include but are not limited to Paclitaxel; Docetaxel; Methotrexate; Doxorubin; Edatrexate; Vinorelbine; Tomaxifen; Leucovorin; 5- fluoro uridine (5-FU); notecan (CAMPTOSAR® or CPT-11 or Camptothecin-11 or Campto); Cisplatin; Carboplatin; Amsacrine; Cytarabine; Bleomycin; Mitomycin C; Dactinomycin; Mithramycin; Hexamethylmelamine; dacarbazine; L-asperginase; Nitrogen mustard; Melphalan, Chlorambucil; Busulfan; Ifosfamide; 4-hydroperoxycyclophosphamide, Thiotepa; Tamoxifen, Herceptin; IMC C225; ABX-EGF: and combinations thereof.
- the nucleic acid molecules of this invention can be used as diagnostic tools to examine genetic drift and mutations within diseased cells or to detect the presence of VEGF and/or VEGFr, such as VEGFRl and/or VEGFR2 RNA in a cell.
- VEGF and/or VEGFr such as VEGFRl and/or VEGFR2 RNA in a cell.
- the close relationship between enzymatic nucleic acid molecule activity and the structure of the target RNA allows the detection of mutations in any region of the molecule which alters the base-pairing and three-dimensional structure of the target RNA.
- By using multiple enzymatic nucleic acid molecules described in this invention one can map nucleotide changes which are important to RNA structure and function in vitro, as well as in cells and tissues.
- Cleavage of target RNAs with enzymatic nucleic acid molecules can be used to inhibit gene expression and define the role (essentially) of specified gene products in the progression of disease. In this manner, other genetic targets can be defined as important mediators of the disease.
- combinational therapies e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules and/or other chemical or biological molecules.
- enzymatic nucleic acid molecules of this invention include detection of the presence of mRNAs associated with VEGF, VEGFRl and/or VEGFR2-related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with an enzymatic nucleic acid molecule using standard methodology.
- enzymatic nucleic acid molecules which cleave only wild-type or mutant forms of the target RNA are used for the assay. The first enzymatic nucleic acid molecule is used to identify wild-type RNA present in the sample and the second enzymatic nucleic acid molecule is used to identify mutant RNA in the sample.
- synthetic substrates of both wild-type and mutant RNA are cleaved by both enzymatic nucleic acid molecules to demonstrate the relative enzymatic nucleic acid molecule efficiencies in the reactions and the absence of cleavage of the "non-targeted" RNA species.
- the cleavage products from the synthetic substrates also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population.
- each analysis requires two enzymatic nucleic acid molecules, two substrates and one unknown sample which is combined into six reactions.
- the presence of cleavage products is determined using an RNAse protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel.
- sequence-specific enzymatic nucleic acid molecules of the instant invention can have many of the same applications for the study of RNA that DNA restriction endonucleases have for the study of DNA (Nathans et al, 1975 Ann. Rev. Biochem. 44:273).
- the pattern of restriction fragments can be used to establish sequence relationships between two related RNAs, and large RNAs can be specifically cleaved to fragments of a size more useful for study.
- the ability to engineer sequence specificity of the enzymatic nucleic acid molecule is ideal for cleavage of RNAs of unknown sequence.
- Applicant has described the use of nucleic acid molecules to down-regulate gene expression of target genes in bacterial, microbial, fungal, viral, and eukaryotic systems including plant, or mammalian cells.
- RNAseP RNA M1 RNA
- Size -290 to 400 nucleotides.
- RNA portion of a ribonucleoprotein enzyme Cleaves tRNA precursors to form mature tRNA.
- RNA RNAs of the tobacco ringspot virus, arabis mosaic virus and chicory yellow mottle virus which uses RNA as the infectious agent ( Figure 3).
- HDV Hepatitis Delta Virus
- Neurospora VS RNA Ribozyme Size -144 nucleotides (at present)
- Phosphoramidites 15 31 ⁇ L 45 sec 233 sec 465 sec S-Ethyl Tetrazole 38.7 31 ⁇ L 45 sec 233 min 465 sec Acetic Anhydride 655 124 ⁇ L 5 sec 5 sec 5 sec A/-Methyl Imidazole 1245 124 ⁇ L 5 sec 5 sec 5 sec TCA 700 732 ⁇ L 10 sec 10 sec 10 sec Iodine 20.6 244 ⁇ L 15 sec 15 sec 15 sec 15 sec
- Wait time does not include contact time during delivery.
- AAACCAAU A CAAUCAUA 216 TATGATTG GGCTAGCTACAACGA ATTGGTTT 1918
- GCUUUUAU A UCACAGAU 448 ATCTGTGA GGCTAGCTACAACGA ATAAAAGC 2150
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- General Chemical & Material Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Endocrinology (AREA)
- Reproductive Health (AREA)
- Genetics & Genomics (AREA)
- Diabetes (AREA)
- Biomedical Technology (AREA)
- General Engineering & Computer Science (AREA)
- Heart & Thoracic Surgery (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Cardiology (AREA)
- Rheumatology (AREA)
- Biotechnology (AREA)
- Molecular Biology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Physical Education & Sports Medicine (AREA)
- Gynecology & Obstetrics (AREA)
- Dermatology (AREA)
- Microbiology (AREA)
- Pregnancy & Childbirth (AREA)
- Urology & Nephrology (AREA)
- Emergency Medicine (AREA)
- Neurology (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Ophthalmology & Optometry (AREA)
Abstract
Description
Claims
Priority Applications (26)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP02752028A EP1390385A4 (en) | 2001-05-29 | 2002-05-29 | NUCLEIC ACID MODULATION OF DISEASES AND DISORDERS OF THE REPRODUCTIVE APPARATUS IN WOMEN |
CA002448320A CA2448320A1 (en) | 2001-05-29 | 2002-05-29 | Ribozyme based treatment of female reproductive diseases |
JP2003500106A JP2005500025A (en) | 2001-05-29 | 2002-05-29 | Nucleic acid-based regulation of female reproductive diseases and conditions |
AU2002344237A AU2002344237B8 (en) | 2001-05-29 | 2002-05-29 | Nucleic Acid Based Modulation of Female Reproductive Diseases and Conditions |
US10/287,949 US20040102389A1 (en) | 1995-10-26 | 2002-11-04 | Nucleic acid-mediated treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) |
US10/306,747 US20030216335A1 (en) | 2001-11-30 | 2002-11-27 | Method and reagent for the modulation of female reproductive diseases and conditions |
EP03742833A EP1521768A4 (en) | 2002-02-20 | 2003-02-20 | INHIBITION INDUCED BY INTERFERENCE OF VASCULAR ENDOTHELIAL GROWTH FACTOR RNA AND GENE EXPRESSION OF VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR USING SHORT INTERFERENT NUCLEIC ACIDS (SINA) |
GB0404898A GB2396864B (en) | 2002-02-20 | 2003-02-20 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression |
AU2003216323A AU2003216323B2 (en) | 2002-02-20 | 2003-02-20 | Inhibition of vascular endothelial growth factor (vegf) and vegf receptor gene expression using short interfereing nucleic acid (sina) |
CA002456444A CA2456444A1 (en) | 2002-02-20 | 2003-02-20 | Rna interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (sina) |
GB0427955A GB2406569B (en) | 2002-02-20 | 2003-02-20 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfe |
PCT/US2003/005022 WO2003070910A2 (en) | 2002-02-20 | 2003-02-20 | INHIBITION OF VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) AND VEGF RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) |
JP2003569803A JP2005517436A (en) | 2002-02-20 | 2003-02-20 | RNA interference-mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acids (siNA) |
US10/665,951 US20040138163A1 (en) | 2002-05-29 | 2003-09-18 | RNA interference mediated inhibition of vascular edothelial growth factor and vascular edothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/664,668 US20070203333A1 (en) | 2001-11-30 | 2003-09-18 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/670,011 US20040209832A1 (en) | 2001-11-30 | 2003-09-23 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/683,990 US20040198682A1 (en) | 2001-11-30 | 2003-10-10 | RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (siNA) |
US10/712,633 US20040220128A1 (en) | 1995-10-26 | 2003-11-13 | Nucleic acid based modulation of female reproductive diseases and conditions |
US10/726,236 US20040142895A1 (en) | 1995-10-26 | 2003-12-02 | Nucleic acid-based modulation of gene expression in the vascular endothelial growth factor pathway |
US10/758,155 US20050075304A1 (en) | 2001-11-30 | 2004-01-12 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/764,957 US20050054596A1 (en) | 2001-11-30 | 2004-01-26 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
GBGB0404461.6A GB0404461D0 (en) | 2001-05-29 | 2004-02-27 | Ribozyme based treatment of female reproductive diseases |
US10/831,620 US20050148530A1 (en) | 2002-02-20 | 2004-04-23 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/844,076 US7176304B2 (en) | 2002-02-20 | 2004-05-11 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/922,761 US20050267058A1 (en) | 2001-05-18 | 2004-08-20 | RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (sINA) |
JP2008148548A JP2009000105A (en) | 2002-02-20 | 2008-06-05 | RNA interference-mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acids (siNA) |
Applications Claiming Priority (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US87016101A | 2001-05-29 | 2001-05-29 | |
US09/870,161 | 2001-05-29 | ||
US33446101P | 2001-11-30 | 2001-11-30 | |
US60/334,461 | 2001-11-30 | ||
US10/138,674 US7034009B2 (en) | 1995-10-26 | 2002-05-03 | Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) |
US10/138,674 | 2002-05-03 |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/138,674 Continuation-In-Part US7034009B2 (en) | 1995-10-26 | 2002-05-03 | Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) |
Related Child Applications (6)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/287,949 Continuation-In-Part US20040102389A1 (en) | 1995-10-26 | 2002-11-04 | Nucleic acid-mediated treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) |
US10/664,668 Continuation-In-Part US20070203333A1 (en) | 2001-11-30 | 2003-09-18 | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US10/683,990 Continuation-In-Part US20040198682A1 (en) | 2001-05-18 | 2003-10-10 | RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (siNA) |
US10/712,633 Continuation-In-Part US20040220128A1 (en) | 1995-10-26 | 2003-11-13 | Nucleic acid based modulation of female reproductive diseases and conditions |
US10/712,633 Continuation US20040220128A1 (en) | 1995-10-26 | 2003-11-13 | Nucleic acid based modulation of female reproductive diseases and conditions |
US10/726,236 Continuation-In-Part US20040142895A1 (en) | 1995-10-26 | 2003-12-02 | Nucleic acid-based modulation of gene expression in the vascular endothelial growth factor pathway |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2002096927A2 true WO2002096927A2 (en) | 2002-12-05 |
WO2002096927A3 WO2002096927A3 (en) | 2003-02-20 |
Family
ID=27385212
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2002/017674 WO2002096927A2 (en) | 1995-10-26 | 2002-05-29 | Ribozyme based treatment of female reproductive diseases |
Country Status (6)
Country | Link |
---|---|
EP (1) | EP1390385A4 (en) |
JP (2) | JP2005500025A (en) |
AU (1) | AU2002344237B8 (en) |
CA (1) | CA2448320A1 (en) |
GB (1) | GB0404461D0 (en) |
WO (1) | WO2002096927A2 (en) |
Cited By (49)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
GB2396864A (en) * | 2002-02-20 | 2004-07-07 | Sirna Therapeutics Inc | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression |
WO2005021749A1 (en) * | 2003-08-28 | 2005-03-10 | Novartis Ag | Interfering rna duplex having blunt-ends and 3’-modifications |
GB2396616B (en) * | 2002-02-20 | 2005-09-21 | Sirna Therapeutics Inc | RNA interference mediated inhibition of gene expression using double stranded short interfering nucleic acid (SINA) |
EP1578933A2 (en) * | 2002-07-24 | 2005-09-28 | The Trustees of The University of Pennsylvania | Compositions and methods for si-rna inhibition of angiogenesis |
EP1603535A2 (en) * | 2003-03-18 | 2005-12-14 | Ethicon, Inc. | Aromatase inhibitor diagnosis and therapy |
US7022828B2 (en) | 2001-04-05 | 2006-04-04 | Sirna Theraputics, Inc. | siRNA treatment of diseases or conditions related to levels of IKK-gamma |
WO2006047394A1 (en) * | 2004-10-22 | 2006-05-04 | Benitec, Inc. | Therapeutic rnai agents for treating psoriasis |
JP2006519610A (en) * | 2003-03-12 | 2006-08-31 | バスジーン セラピューティクス, インコーポレイテッド | Nucleic acid compounds for inhibiting angiogenesis and tumor growth |
EP1711510A2 (en) * | 2004-02-05 | 2006-10-18 | Intradigm Corporation | Rnai therapeutics for treatment of eye neovascularization diseases |
JP2006526989A (en) * | 2003-06-06 | 2006-11-30 | セネスコ テクノロジーズ,インコーポレイティド | Inhibition of apoptosis-specific eIF-5A ("eIF-5A") using antisense oligonucleotides and siRNA as anti-inflammatory therapy |
WO2006064519A3 (en) * | 2004-12-14 | 2006-12-21 | Nat Inst Immunology | Dnazymes for inhibition of japanese encephalitis virus replication |
US7176304B2 (en) * | 2002-02-20 | 2007-02-13 | Mcswiggen James | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
EP1939291A2 (en) * | 2001-11-09 | 2008-07-02 | Centre National de la Recherche Scientifique (CNRS) | Inhibitor oligonucleotides and their use for specific repression of a gene |
US7534878B2 (en) | 2005-04-12 | 2009-05-19 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US7659390B2 (en) | 2002-02-20 | 2010-02-09 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA) |
US7833992B2 (en) * | 2001-05-18 | 2010-11-16 | Merck Sharpe & Dohme | Conjugates and compositions for cellular delivery |
US7858769B2 (en) | 2004-02-10 | 2010-12-28 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using multifunctional short interfering nucleic acid (multifunctional siNA) |
US7872118B2 (en) | 2006-09-08 | 2011-01-18 | Opko Ophthalmics, Llc | siRNA and methods of manufacture |
WO2011019423A2 (en) | 2009-05-20 | 2011-02-17 | Schering Corporation | Modulation of pilr receptors to treat microbial infections |
US7893244B2 (en) | 2005-04-12 | 2011-02-22 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
WO2011031600A1 (en) | 2009-09-10 | 2011-03-17 | Schering Corporation | Use of il-33 antagonists to treat fibrotic disease |
EP2316441A1 (en) * | 2005-02-17 | 2011-05-04 | Hadasit Medical Research Services And Development | Bisphosphonates for treating endometriosis |
JP2011116755A (en) * | 2003-02-28 | 2011-06-16 | Johns Hopkins Univ | T cell regulation |
US7964578B2 (en) * | 2001-05-18 | 2011-06-21 | Sirna Therapeutics, Inc. | Conjugates and compositions for cellular delivery |
WO2011084357A1 (en) | 2009-12-17 | 2011-07-14 | Schering Corporation | Modulation of pilr to treat immune disorders |
US8084600B2 (en) | 2006-05-04 | 2011-12-27 | Novartis Ag | Short interfering ribonucleic acid (siRNA) with improved pharmacological properties |
US8084436B2 (en) | 2003-11-03 | 2011-12-27 | Isis Pharmaceuticals, Inc. | Modulation of SGLT2 expression |
EP2314691A3 (en) * | 2002-11-14 | 2012-01-18 | Dharmacon, Inc. | Fuctional and hyperfunctional siRNA |
EP2446890A1 (en) | 2007-04-30 | 2012-05-02 | Allergan, Inc. | High viscosity macromolecular compositions for treating ocular conditions |
US8202845B2 (en) | 2002-04-18 | 2012-06-19 | Acuity Pharmaceuticals, Inc. | Means and methods for the specific modulation of target genes in the CNS and the eye and methods for their identification |
US8258105B2 (en) | 2003-10-07 | 2012-09-04 | Isis Pharmaceuticals, Inc. | Antisense oligonucleotides optimized for kidney targeting |
US20130101657A1 (en) * | 2004-06-25 | 2013-04-25 | The Johns Hopkins University | Angiogenesis inhibitors |
US8470792B2 (en) | 2008-12-04 | 2013-06-25 | Opko Pharmaceuticals, Llc. | Compositions and methods for selective inhibition of VEGF |
US8546349B2 (en) | 2010-07-28 | 2013-10-01 | Thermo Fisher Scientific Biosciences Inc. | siRNA targeting VEGFA and methods for treatment in vivo |
US8575329B2 (en) | 2002-11-14 | 2013-11-05 | Thermo Fisher Scientific Biosciences Inc. | siRNA targeting kinase insert domain receptor (KDR) |
CN103627709A (en) * | 2013-12-06 | 2014-03-12 | 孙仑泉 | Deoxyribozyme capable of suppressing genetic expression of vascular endothelial growth factor |
US8871737B2 (en) | 2010-09-22 | 2014-10-28 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US8916538B2 (en) | 2012-03-21 | 2014-12-23 | Vertex Pharmaceuticals Incorporated | Solid forms of a thiophosphoramidate nucleotide prodrug |
US8980865B2 (en) | 2011-12-22 | 2015-03-17 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US9012427B2 (en) | 2012-03-22 | 2015-04-21 | Alios Biopharma, Inc. | Pharmaceutical combinations comprising a thionucleotide analog |
US9181551B2 (en) | 2002-02-20 | 2015-11-10 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US9243246B2 (en) | 2010-08-24 | 2016-01-26 | Sirna Therapeutics, Inc. | Single-stranded RNAi agents containing an internal, non-nucleic acid spacer |
US9260471B2 (en) | 2010-10-29 | 2016-02-16 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA) |
US9657294B2 (en) | 2002-02-20 | 2017-05-23 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US9987292B2 (en) | 2009-06-22 | 2018-06-05 | Ampio Pharmaceuticals, Inc. | Method for treatment of diseases |
US9993427B2 (en) | 2013-03-14 | 2018-06-12 | Biorest Ltd. | Liposome formulation and manufacture |
US9994853B2 (en) | 2001-05-18 | 2018-06-12 | Sirna Therapeutics, Inc. | Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference |
US10058562B2 (en) | 2012-12-19 | 2018-08-28 | Ampio Pharmaceuticals, Inc. | Methods of treatment of diseases |
US10508277B2 (en) | 2004-05-24 | 2019-12-17 | Sirna Therapeutics, Inc. | Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference |
Families Citing this family (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7517864B2 (en) | 2001-05-18 | 2009-04-14 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
AU2006203062B2 (en) * | 2002-02-20 | 2009-03-12 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
US8093259B2 (en) * | 2006-05-25 | 2012-01-10 | Novartis Ag | 4-methyl-3-[[4-(3-pyridinyl)-2-pyrimidinyl]amino]-N-[5-(4-methyl-1H-imidazol-1-yl)-3-(trifluoromethyl)phenyl]-benzamide for treatment of melanoma |
WO2013053076A1 (en) | 2011-10-10 | 2013-04-18 | Zensun (Shanghai)Science & Technology Limited | Compositions and methods for treating heart failure |
AU2014270882B2 (en) | 2013-05-22 | 2019-03-14 | Zensun (Shanghai) Science & Technology, Co. Ltd. | Extended release of neuregulin for treating heart failure |
CN110177544A (en) | 2016-11-29 | 2019-08-27 | 普尔泰克健康有限公司 | For delivering the excretion body of therapeutic agent |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1999004819A1 (en) * | 1997-07-24 | 1999-02-04 | Inex Pharmaceuticals Corporation | Liposomal compositions for the delivery of nucleic acid catalysts |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6346398B1 (en) * | 1995-10-26 | 2002-02-12 | Ribozyme Pharmaceuticals, Inc. | Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor |
WO2003070910A2 (en) * | 2002-02-20 | 2003-08-28 | Ribozyme Pharmaceuticals, Incorporated | INHIBITION OF VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) AND VEGF RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) |
-
2002
- 2002-05-29 WO PCT/US2002/017674 patent/WO2002096927A2/en not_active Application Discontinuation
- 2002-05-29 AU AU2002344237A patent/AU2002344237B8/en not_active Ceased
- 2002-05-29 JP JP2003500106A patent/JP2005500025A/en active Pending
- 2002-05-29 CA CA002448320A patent/CA2448320A1/en not_active Abandoned
- 2002-05-29 EP EP02752028A patent/EP1390385A4/en not_active Withdrawn
-
2004
- 2004-02-27 GB GBGB0404461.6A patent/GB0404461D0/en active Pending
-
2008
- 2008-06-06 JP JP2008149684A patent/JP2009000106A/en active Pending
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1999004819A1 (en) * | 1997-07-24 | 1999-02-04 | Inex Pharmaceuticals Corporation | Liposomal compositions for the delivery of nucleic acid catalysts |
Non-Patent Citations (2)
Title |
---|
PARRY T.J. ET AL.: 'Bioactivity of anti-angiogenic ribozymes targeting Flt-1 and KDR mRNA' NUCLEIC ACIDS RESEARCH vol. 27, no. 13, 1999, pages 2569 - 2577, XP002956534 * |
See also references of EP1390385A2 * |
Cited By (108)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7022828B2 (en) | 2001-04-05 | 2006-04-04 | Sirna Theraputics, Inc. | siRNA treatment of diseases or conditions related to levels of IKK-gamma |
US7964578B2 (en) * | 2001-05-18 | 2011-06-21 | Sirna Therapeutics, Inc. | Conjugates and compositions for cellular delivery |
US7833992B2 (en) * | 2001-05-18 | 2010-11-16 | Merck Sharpe & Dohme | Conjugates and compositions for cellular delivery |
US9994853B2 (en) | 2001-05-18 | 2018-06-12 | Sirna Therapeutics, Inc. | Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference |
EP1939291A3 (en) * | 2001-11-09 | 2012-02-22 | Centre National de la Recherche Scientifique (CNRS) | Inhibitor oligonucleotides and their use for specific repression of a gene |
US8318689B2 (en) | 2001-11-09 | 2012-11-27 | Centre National De La Recherche Scientifique | SiRNA-based cancer treatment |
EP1939291A2 (en) * | 2001-11-09 | 2008-07-02 | Centre National de la Recherche Scientifique (CNRS) | Inhibitor oligonucleotides and their use for specific repression of a gene |
US9771588B2 (en) | 2002-02-20 | 2017-09-26 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
GB2396864A (en) * | 2002-02-20 | 2004-07-07 | Sirna Therapeutics Inc | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression |
GB2396864B (en) * | 2002-02-20 | 2005-03-02 | Sirna Therapeutics Inc | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression |
GB2396616B (en) * | 2002-02-20 | 2005-09-21 | Sirna Therapeutics Inc | RNA interference mediated inhibition of gene expression using double stranded short interfering nucleic acid (SINA) |
US10662428B2 (en) | 2002-02-20 | 2020-05-26 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US8076472B2 (en) | 2002-02-20 | 2011-12-13 | Merck Sharp & Dohme Corp. | RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA) |
US10351852B2 (en) | 2002-02-20 | 2019-07-16 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US10889815B2 (en) | 2002-02-20 | 2021-01-12 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
GB2406569A (en) * | 2002-02-20 | 2005-04-06 | Sirna Therapeutics Inc | Double-stranded, short interfering nucleic aicd (siNA) molecules |
US10000754B2 (en) | 2002-02-20 | 2018-06-19 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US7176304B2 (en) * | 2002-02-20 | 2007-02-13 | Mcswiggen James | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) |
GB2406569B (en) * | 2002-02-20 | 2005-07-20 | Sirna Therapeutics Inc | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfe |
US9181551B2 (en) | 2002-02-20 | 2015-11-10 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US7858771B2 (en) | 2002-02-20 | 2010-12-28 | Merck Sharp & Dohme Corp. | RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA) |
EP1521768A4 (en) * | 2002-02-20 | 2005-07-06 | Sirna Therapeutics Inc | INHIBITION INDUCED BY INTERFERENCE OF VASCULAR ENDOTHELIAL GROWTH FACTOR RNA AND GENE EXPRESSION OF VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR USING SHORT INTERFERENT NUCLEIC ACIDS (SINA) |
US9657294B2 (en) | 2002-02-20 | 2017-05-23 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US9957517B2 (en) | 2002-02-20 | 2018-05-01 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
EP1521768A2 (en) * | 2002-02-20 | 2005-04-13 | Sirna Therapeutics, Inc. | RNA INTERFERENCE MEDIATED INHIBITION OF VASCULAR ENDOTHELIAL GROWTH FACTOR AND VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) |
US7659390B2 (en) | 2002-02-20 | 2010-02-09 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA) |
US9732344B2 (en) | 2002-02-20 | 2017-08-15 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US9738899B2 (en) | 2002-02-20 | 2017-08-22 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA) |
US8946180B2 (en) | 2002-04-18 | 2015-02-03 | Opko Pharmaceuticals, Llc | Means and methods for the specific modulation of target genes in the CNS and the eye and methods for their identification |
US8202845B2 (en) | 2002-04-18 | 2012-06-19 | Acuity Pharmaceuticals, Inc. | Means and methods for the specific modulation of target genes in the CNS and the eye and methods for their identification |
US8541384B2 (en) | 2002-07-24 | 2013-09-24 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
EP1578933A2 (en) * | 2002-07-24 | 2005-09-28 | The Trustees of The University of Pennsylvania | Compositions and methods for si-rna inhibition of angiogenesis |
US7750143B2 (en) | 2002-07-24 | 2010-07-06 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
EP2345718A3 (en) * | 2002-07-24 | 2011-11-16 | The Trustees of The University of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
US7345027B2 (en) | 2002-07-24 | 2008-03-18 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
EP1578933A4 (en) * | 2002-07-24 | 2007-09-19 | Univ Pennsylvania | COMPOSITIONS AND METHOD FOR INHIBITING ANGIOGENESIS BY RNA-IS |
US9150863B2 (en) | 2002-07-24 | 2015-10-06 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
EP2192187A3 (en) * | 2002-07-24 | 2010-08-25 | The Trustees of The University of Pennsylvania | Compositions and methods for si-RNA inhibition of angiogenesis |
US8946403B2 (en) | 2002-07-24 | 2015-02-03 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
US7674895B2 (en) | 2002-07-24 | 2010-03-09 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
US8546345B2 (en) | 2002-07-24 | 2013-10-01 | The Trustees Of The University Of Pennsylvania | Compositions and methods for siRNA inhibition of angiogenesis |
US7148342B2 (en) | 2002-07-24 | 2006-12-12 | The Trustees Of The University Of Pennyslvania | Compositions and methods for sirna inhibition of angiogenesis |
US8575329B2 (en) | 2002-11-14 | 2013-11-05 | Thermo Fisher Scientific Biosciences Inc. | siRNA targeting kinase insert domain receptor (KDR) |
EP2314691A3 (en) * | 2002-11-14 | 2012-01-18 | Dharmacon, Inc. | Fuctional and hyperfunctional siRNA |
JP2011116755A (en) * | 2003-02-28 | 2011-06-16 | Johns Hopkins Univ | T cell regulation |
JP4762889B2 (en) * | 2003-03-12 | 2011-08-31 | バスジーン セラピューティクス,インコーポレイテッド | Nucleic acid compounds for inhibiting angiogenesis and tumor growth |
JP2006519610A (en) * | 2003-03-12 | 2006-08-31 | バスジーン セラピューティクス, インコーポレイテッド | Nucleic acid compounds for inhibiting angiogenesis and tumor growth |
EP1603535A4 (en) * | 2003-03-18 | 2008-10-15 | Ethicon Inc | Aromatase inhibitor diagnosis and therapy |
EP1603535A2 (en) * | 2003-03-18 | 2005-12-14 | Ethicon, Inc. | Aromatase inhibitor diagnosis and therapy |
JP2006526989A (en) * | 2003-06-06 | 2006-11-30 | セネスコ テクノロジーズ,インコーポレイティド | Inhibition of apoptosis-specific eIF-5A ("eIF-5A") using antisense oligonucleotides and siRNA as anti-inflammatory therapy |
CN1845993B (en) * | 2003-08-28 | 2010-06-23 | 诺瓦提斯公司 | Interfering RNA duplexes with blunt ends and 3' modifications |
WO2005021749A1 (en) * | 2003-08-28 | 2005-03-10 | Novartis Ag | Interfering rna duplex having blunt-ends and 3’-modifications |
EP2338995A3 (en) * | 2003-08-28 | 2012-01-11 | Novartis AG | Interfering RNA duplex having blunt-ends and 3'-modifications |
US8097716B2 (en) | 2003-08-28 | 2012-01-17 | Novartis Ag | Interfering RNA duplex having blunt-ends and 3′-modifications |
JP2007503803A (en) * | 2003-08-28 | 2007-03-01 | ノバルティス アクチエンゲゼルシャフト | Interfering RNA duplexes with blunt ends and 3 'modifications |
JP2011160796A (en) * | 2003-08-28 | 2011-08-25 | Novartis Ag | Interfering rna duplex having blunt-end and 3'-modification |
EP2489737A1 (en) * | 2003-08-28 | 2012-08-22 | Novartis AG | Interfering RNA duplex having blunt-ends and 3'-modifications |
US8258105B2 (en) | 2003-10-07 | 2012-09-04 | Isis Pharmaceuticals, Inc. | Antisense oligonucleotides optimized for kidney targeting |
US8084436B2 (en) | 2003-11-03 | 2011-12-27 | Isis Pharmaceuticals, Inc. | Modulation of SGLT2 expression |
EP1711510A4 (en) * | 2004-02-05 | 2008-11-26 | Intradigm Corp | THERAPY WITH RNAI AGENTS APPLIED IN THE TREATMENT OF OCULAR DISEASES RESULTING FROM NEOVASCULARIZATION |
EP1711510A2 (en) * | 2004-02-05 | 2006-10-18 | Intradigm Corporation | Rnai therapeutics for treatment of eye neovascularization diseases |
US7858769B2 (en) | 2004-02-10 | 2010-12-28 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using multifunctional short interfering nucleic acid (multifunctional siNA) |
US10508277B2 (en) | 2004-05-24 | 2019-12-17 | Sirna Therapeutics, Inc. | Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference |
US20130101657A1 (en) * | 2004-06-25 | 2013-04-25 | The Johns Hopkins University | Angiogenesis inhibitors |
US8980930B2 (en) | 2004-06-25 | 2015-03-17 | The Johns Hopkins University | Angiogenesis inhibitors |
WO2006047394A1 (en) * | 2004-10-22 | 2006-05-04 | Benitec, Inc. | Therapeutic rnai agents for treating psoriasis |
WO2006064519A3 (en) * | 2004-12-14 | 2006-12-21 | Nat Inst Immunology | Dnazymes for inhibition of japanese encephalitis virus replication |
EP2316441A1 (en) * | 2005-02-17 | 2011-05-04 | Hadasit Medical Research Services And Development | Bisphosphonates for treating endometriosis |
US7893243B2 (en) | 2005-04-12 | 2011-02-22 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US7723316B2 (en) | 2005-04-12 | 2010-05-25 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US7786092B2 (en) | 2005-04-12 | 2010-08-31 | Intradigm Corporation | Composition and method of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US7534878B2 (en) | 2005-04-12 | 2009-05-19 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US7893244B2 (en) | 2005-04-12 | 2011-02-22 | Intradigm Corporation | Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases |
US8404832B2 (en) | 2006-05-04 | 2013-03-26 | Novartis Ag | Short interfering ribonucleic acid (siRNA) for oral administration |
US9493771B2 (en) | 2006-05-04 | 2016-11-15 | Novartis Ag | Short interfering ribonucleic acid (siRNA) for oral administration |
US8957041B2 (en) | 2006-05-04 | 2015-02-17 | Novartis Ag | Short interfering ribonucleic acid (siRNA) for oral administration |
US8404831B2 (en) | 2006-05-04 | 2013-03-26 | Novartis Ag | Short interfering ribonucleic acid (siRNA) for oral administration |
US8344128B2 (en) | 2006-05-04 | 2013-01-01 | Novartis Ag | Short interfering ribonucleic acid (siRNA) for oral administration |
US8084600B2 (en) | 2006-05-04 | 2011-12-27 | Novartis Ag | Short interfering ribonucleic acid (siRNA) with improved pharmacological properties |
US7872118B2 (en) | 2006-09-08 | 2011-01-18 | Opko Ophthalmics, Llc | siRNA and methods of manufacture |
EP2446890A1 (en) | 2007-04-30 | 2012-05-02 | Allergan, Inc. | High viscosity macromolecular compositions for treating ocular conditions |
US11078262B2 (en) | 2007-04-30 | 2021-08-03 | Allergan, Inc. | High viscosity macromolecular compositions for treating ocular conditions |
US8470792B2 (en) | 2008-12-04 | 2013-06-25 | Opko Pharmaceuticals, Llc. | Compositions and methods for selective inhibition of VEGF |
WO2011019423A2 (en) | 2009-05-20 | 2011-02-17 | Schering Corporation | Modulation of pilr receptors to treat microbial infections |
US9987292B2 (en) | 2009-06-22 | 2018-06-05 | Ampio Pharmaceuticals, Inc. | Method for treatment of diseases |
WO2011031600A1 (en) | 2009-09-10 | 2011-03-17 | Schering Corporation | Use of il-33 antagonists to treat fibrotic disease |
WO2011084357A1 (en) | 2009-12-17 | 2011-07-14 | Schering Corporation | Modulation of pilr to treat immune disorders |
US9150862B2 (en) | 2010-07-28 | 2015-10-06 | Thermo Fisher Scientific Inc. | siRNA targeting VEGFA and methods for treatment in vivo |
US8546349B2 (en) | 2010-07-28 | 2013-10-01 | Thermo Fisher Scientific Biosciences Inc. | siRNA targeting VEGFA and methods for treatment in vivo |
US9845466B2 (en) | 2010-08-24 | 2017-12-19 | Sirna Therapeutics, Inc. | Single-stranded RNAi agents containing an internal, non-nucleic acid spacer |
US9243246B2 (en) | 2010-08-24 | 2016-01-26 | Sirna Therapeutics, Inc. | Single-stranded RNAi agents containing an internal, non-nucleic acid spacer |
US10584335B2 (en) | 2010-08-24 | 2020-03-10 | Sirna Therapeutics, Inc. | Single-stranded RNAi agents containing an internal, non-nucleic acid spacer |
US9278990B2 (en) | 2010-09-22 | 2016-03-08 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US8871737B2 (en) | 2010-09-22 | 2014-10-28 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US11193126B2 (en) | 2010-10-29 | 2021-12-07 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA) |
US9970005B2 (en) | 2010-10-29 | 2018-05-15 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA) |
US9260471B2 (en) | 2010-10-29 | 2016-02-16 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA) |
US11932854B2 (en) | 2010-10-29 | 2024-03-19 | Sirna Therapeutics, Inc. | RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA) |
US9605018B2 (en) | 2011-12-22 | 2017-03-28 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US8980865B2 (en) | 2011-12-22 | 2015-03-17 | Alios Biopharma, Inc. | Substituted nucleotide analogs |
US8916538B2 (en) | 2012-03-21 | 2014-12-23 | Vertex Pharmaceuticals Incorporated | Solid forms of a thiophosphoramidate nucleotide prodrug |
US9856284B2 (en) | 2012-03-21 | 2018-01-02 | Alios Biopharma, Inc. | Solid forms of a thiophosphoramidate nucleotide prodrug |
US9012427B2 (en) | 2012-03-22 | 2015-04-21 | Alios Biopharma, Inc. | Pharmaceutical combinations comprising a thionucleotide analog |
US10058562B2 (en) | 2012-12-19 | 2018-08-28 | Ampio Pharmaceuticals, Inc. | Methods of treatment of diseases |
US10265269B2 (en) | 2013-03-14 | 2019-04-23 | Biorest Ltd. | Liposome formulation and manufacture |
US11633357B2 (en) | 2013-03-14 | 2023-04-25 | Zuli Holdings, Ltd. | Liposome formulation and manufacture |
US9993427B2 (en) | 2013-03-14 | 2018-06-12 | Biorest Ltd. | Liposome formulation and manufacture |
CN103627709A (en) * | 2013-12-06 | 2014-03-12 | 孙仑泉 | Deoxyribozyme capable of suppressing genetic expression of vascular endothelial growth factor |
Also Published As
Publication number | Publication date |
---|---|
EP1390385A2 (en) | 2004-02-25 |
JP2005500025A (en) | 2005-01-06 |
GB0404461D0 (en) | 2004-03-31 |
EP1390385A4 (en) | 2004-11-24 |
WO2002096927A3 (en) | 2003-02-20 |
AU2002344237B8 (en) | 2008-11-06 |
CA2448320A1 (en) | 2002-12-05 |
AU2002344237B2 (en) | 2008-10-23 |
JP2009000106A (en) | 2009-01-08 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2002344237B2 (en) | Nucleic Acid Based Modulation of Female Reproductive Diseases and Conditions | |
AU2002344237A1 (en) | Nucleic Acid Based Modulation of Female Reproductive Diseases and Conditions | |
US20030216335A1 (en) | Method and reagent for the modulation of female reproductive diseases and conditions | |
US6566127B1 (en) | Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor | |
US20040142895A1 (en) | Nucleic acid-based modulation of gene expression in the vascular endothelial growth factor pathway | |
US7034009B2 (en) | Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) | |
US20030064945A1 (en) | Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors | |
US7022828B2 (en) | siRNA treatment of diseases or conditions related to levels of IKK-gamma | |
US7176304B2 (en) | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) | |
US20040220128A1 (en) | Nucleic acid based modulation of female reproductive diseases and conditions | |
US20070203333A1 (en) | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) | |
US20030203870A1 (en) | Method and reagent for the inhibition of NOGO and NOGO receptor genes | |
US20050075304A1 (en) | RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA) | |
US20020177568A1 (en) | Enzymatic nucleic acid treatment of diseases or conditions related to levels of NF-kappa B | |
WO2001088124A2 (en) | Method and reagent for the inhibition of erg | |
US20040102389A1 (en) | Nucleic acid-mediated treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R) | |
EP1767632A2 (en) | A method for local administration of synthetic double-stranded oligonucleotides targeting a VEGF receptor | |
JP2002509721A (en) | Methods and reagents for the treatment of diseases or conditions related to molecules involved in the angiogenic response | |
US20030073207A1 (en) | Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors | |
US20030186909A1 (en) | Nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors | |
US20030113891A1 (en) | Method and reagent for the inhibition of NOGO and NOGO receptor genes | |
WO2001062911A2 (en) | Antisense and catalytically acting nucleic acid molecules targeted to grb2- related with insert domain (grid) proteins and their uses | |
US20030060611A1 (en) | Method and reagent for the inhibition of NOGO gene | |
JP2007505605A6 (en) | RNA interference-mediated suppression of vascular endothelial growth factor gene expression and vascular endothelial growth factor gene receptor gene expression mediated by RNA interference using small interfering nucleic acids (siNA) | |
JP2007505605A (en) | RNA interference-mediated suppression of vascular endothelial growth factor gene expression and vascular endothelial growth factor gene receptor gene expression mediated by RNA interference using small interfering nucleic acids (siNA) |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 10287949 Country of ref document: US |
|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
AK | Designated states |
Kind code of ref document: A3 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A3 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
WWE | Wipo information: entry into national phase |
Ref document number: 10664668 Country of ref document: US |
|
WWE | Wipo information: entry into national phase |
Ref document number: 10712633 Country of ref document: US |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2002344237 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2002752028 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2448320 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2003500106 Country of ref document: JP |
|
WWP | Wipo information: published in national office |
Ref document number: 2002752028 Country of ref document: EP |
|
REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 2002752028 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 10664668 Country of ref document: US |