USRE36397E - Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity - Google Patents
Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity Download PDFInfo
- Publication number
- USRE36397E USRE36397E US09/219,843 US21984398A USRE36397E US RE36397 E USRE36397 E US RE36397E US 21984398 A US21984398 A US 21984398A US RE36397 E USRE36397 E US RE36397E
- Authority
- US
- United States
- Prior art keywords
- inhibitor
- iaddend
- iadd
- benzamide
- adp
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Expired - Lifetime
Links
- -1 poly(ADP-ribose) Polymers 0.000 title claims abstract description 151
- 239000003112 inhibitor Substances 0.000 title claims abstract description 70
- HOKKHZGPKSLGJE-GSVOUGTGSA-N N-Methyl-D-aspartic acid Chemical compound CN[C@@H](C(O)=O)CC(O)=O HOKKHZGPKSLGJE-GSVOUGTGSA-N 0.000 title claims abstract description 42
- 206010029350 Neurotoxicity Diseases 0.000 title claims abstract description 42
- 206010044221 Toxic encephalopathy Diseases 0.000 title claims abstract description 42
- 230000007135 neurotoxicity Effects 0.000 title claims abstract description 42
- 231100000228 neurotoxicity Toxicity 0.000 title claims abstract description 42
- 102000003960 Ligases Human genes 0.000 title claims abstract description 35
- 108090000364 Ligases Proteins 0.000 title claims abstract description 35
- 108091026813 Poly(ADPribose) Proteins 0.000 title claims abstract description 35
- 230000002792 vascular Effects 0.000 claims abstract description 10
- 208000015122 neurodegenerative disease Diseases 0.000 claims abstract description 9
- 230000001404 mediated effect Effects 0.000 claims abstract description 7
- KXDAEFPNCMNJSK-UHFFFAOYSA-N Benzamide Chemical group NC(=O)C1=CC=CC=C1 KXDAEFPNCMNJSK-UHFFFAOYSA-N 0.000 claims description 80
- 238000000034 method Methods 0.000 claims description 31
- 201000010099 disease Diseases 0.000 claims description 19
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 19
- 239000008194 pharmaceutical composition Substances 0.000 claims description 13
- GSCPDZHWVNUUFI-UHFFFAOYSA-N 3-aminobenzamide Chemical compound NC(=O)C1=CC=CC(N)=C1 GSCPDZHWVNUUFI-UHFFFAOYSA-N 0.000 claims description 10
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 claims description 10
- 229930195712 glutamate Natural products 0.000 claims description 10
- 241000124008 Mammalia Species 0.000 claims description 9
- LFUJIPVWTMGYDG-UHFFFAOYSA-N isoquinoline-1,5-diol Chemical group N1=CC=C2C(O)=CC=CC2=C1O LFUJIPVWTMGYDG-UHFFFAOYSA-N 0.000 claims description 9
- QIKYZXDTTPVVAC-UHFFFAOYSA-N 4-Aminobenzamide Chemical compound NC(=O)C1=CC=C(N)C=C1 QIKYZXDTTPVVAC-UHFFFAOYSA-N 0.000 claims description 8
- 230000004770 neurodegeneration Effects 0.000 claims description 8
- 208000024891 symptom Diseases 0.000 claims description 7
- 239000000203 mixture Substances 0.000 claims description 5
- 230000037396 body weight Effects 0.000 claims description 4
- 238000009472 formulation Methods 0.000 claims description 4
- 230000002401 inhibitory effect Effects 0.000 claims description 4
- 230000003389 potentiating effect Effects 0.000 claims description 4
- 239000002775 capsule Substances 0.000 claims description 3
- 239000000839 emulsion Substances 0.000 claims description 3
- 238000000338 in vitro Methods 0.000 claims description 3
- 239000000725 suspension Substances 0.000 claims description 3
- 102000004868 N-Methyl-D-Aspartate Receptors Human genes 0.000 claims 3
- 108090001041 N-Methyl-D-Aspartate Receptors Proteins 0.000 claims 3
- 239000008174 sterile solution Substances 0.000 claims 2
- 208000024827 Alzheimer disease Diseases 0.000 abstract description 6
- 208000023105 Huntington disease Diseases 0.000 abstract description 6
- 238000011282 treatment Methods 0.000 abstract description 6
- 208000018737 Parkinson disease Diseases 0.000 abstract description 5
- 208000023516 stroke disease Diseases 0.000 abstract 1
- 102100023712 Poly [ADP-ribose] polymerase 1 Human genes 0.000 description 51
- 101001113483 Homo sapiens Poly [ADP-ribose] polymerase 1 Proteins 0.000 description 50
- 101000630284 Homo sapiens Proline-tRNA ligase Proteins 0.000 description 50
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 48
- BAWFJGJZGIEFAR-NNYOXOHSSA-N NAD zwitterion Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 BAWFJGJZGIEFAR-NNYOXOHSSA-N 0.000 description 18
- 230000000694 effects Effects 0.000 description 16
- 229950006238 nadide Drugs 0.000 description 16
- XEYBHCRIKKKOSS-UHFFFAOYSA-N disodium;azanylidyneoxidanium;iron(2+);pentacyanide Chemical compound [Na+].[Na+].[Fe+2].N#[C-].N#[C-].N#[C-].N#[C-].N#[C-].[O+]#N XEYBHCRIKKKOSS-UHFFFAOYSA-N 0.000 description 12
- 229940083618 sodium nitroprusside Drugs 0.000 description 12
- 108020004414 DNA Proteins 0.000 description 11
- 230000004913 activation Effects 0.000 description 11
- 210000004027 cell Anatomy 0.000 description 11
- 230000005764 inhibitory process Effects 0.000 description 11
- 230000036515 potency Effects 0.000 description 9
- SRNWOUGRCWSEMX-KEOHHSTQSA-N ADP-beta-D-ribose Chemical group C([C@H]1O[C@H]([C@@H]([C@@H]1O)O)N1C=2N=CN=C(C=2N=C1)N)OP(O)(=O)OP(O)(=O)OC[C@H]1O[C@@H](O)[C@H](O)[C@@H]1O SRNWOUGRCWSEMX-KEOHHSTQSA-N 0.000 description 8
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 8
- 230000006378 damage Effects 0.000 description 8
- 210000002569 neuron Anatomy 0.000 description 8
- 230000005778 DNA damage Effects 0.000 description 7
- 231100000277 DNA damage Toxicity 0.000 description 7
- IOVCWXUNBOPUCH-UHFFFAOYSA-M Nitrite anion Chemical compound [O-]N=O IOVCWXUNBOPUCH-UHFFFAOYSA-M 0.000 description 7
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 6
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- 210000004556 brain Anatomy 0.000 description 6
- 230000001537 neural effect Effects 0.000 description 6
- PWJFNRJRHXWEPT-UHFFFAOYSA-N ADP ribose Natural products C1=NC=2C(N)=NC=NC=2N1C1OC(COP(O)(=O)OP(O)(=O)OCC(O)C(O)C(O)C=O)C(O)C1O PWJFNRJRHXWEPT-UHFFFAOYSA-N 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- ZIIQCSMRQKCOCT-YFKPBYRVSA-N S-nitroso-N-acetyl-D-penicillamine Chemical compound CC(=O)N[C@@H](C(O)=O)C(C)(C)SN=O ZIIQCSMRQKCOCT-YFKPBYRVSA-N 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 239000005711 Benzoic acid Substances 0.000 description 4
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 4
- 235000010233 benzoic acid Nutrition 0.000 description 4
- 230000030833 cell death Effects 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- 230000002147 killing effect Effects 0.000 description 4
- SSMIFVHARFVINF-UHFFFAOYSA-N 4-amino-1,8-naphthalimide Chemical compound O=C1NC(=O)C2=CC=CC3=C2C1=CC=C3N SSMIFVHARFVINF-UHFFFAOYSA-N 0.000 description 3
- 239000005104 Neeliglow 4-amino-1,8-naphthalimide Substances 0.000 description 3
- YJQPYGGHQPGBLI-UHFFFAOYSA-N Novobiocin Natural products O1C(C)(C)C(OC)C(OC(N)=O)C(O)C1OC1=CC=C(C(O)=C(NC(=O)C=2C=C(CC=C(C)C)C(O)=CC=2)C(=O)O2)C2=C1C YJQPYGGHQPGBLI-UHFFFAOYSA-N 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 238000000692 Student's t-test Methods 0.000 description 3
- 150000003936 benzamides Chemical class 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 229960003966 nicotinamide Drugs 0.000 description 3
- 239000011570 nicotinamide Substances 0.000 description 3
- YJQPYGGHQPGBLI-KGSXXDOSSA-N novobiocin Chemical compound O1C(C)(C)[C@H](OC)[C@@H](OC(N)=O)[C@@H](O)[C@@H]1OC1=CC=C(C(O)=C(NC(=O)C=2C=C(CC=C(C)C)C(O)=CC=2)C(=O)O2)C2=C1C YJQPYGGHQPGBLI-KGSXXDOSSA-N 0.000 description 3
- 229960002950 novobiocin Drugs 0.000 description 3
- 229920000642 polymer Polymers 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000005730 ADP ribosylation Effects 0.000 description 2
- 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 2
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 2
- DAYLJWODMCOQEW-TURQNECASA-O NMN(+) Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP(O)(O)=O)O2)O)=C1 DAYLJWODMCOQEW-TURQNECASA-O 0.000 description 2
- 102000007999 Nuclear Proteins Human genes 0.000 description 2
- 108010089610 Nuclear Proteins Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 210000001218 blood-brain barrier Anatomy 0.000 description 2
- 208000037516 chromosome inversion disease Diseases 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 230000003013 cytotoxicity Effects 0.000 description 2
- 231100000135 cytotoxicity Toxicity 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 239000007789 gas Substances 0.000 description 2
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 238000007914 intraventricular administration Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000002858 neurotransmitter agent Substances 0.000 description 2
- 235000005152 nicotinamide Nutrition 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 150000003254 radicals Chemical class 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 239000012085 test solution Substances 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 230000002476 tumorcidal effect Effects 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- MRXDGVXSWIXTQL-HYHFHBMOSA-N (2s)-2-[[(1s)-1-(2-amino-1,4,5,6-tetrahydropyrimidin-6-yl)-2-[[(2s)-4-methyl-1-oxo-1-[[(2s)-1-oxo-3-phenylpropan-2-yl]amino]pentan-2-yl]amino]-2-oxoethyl]carbamoylamino]-3-phenylpropanoic acid Chemical compound C([C@H](NC(=O)N[C@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C=O)C1NC(N)=NCC1)C(O)=O)C1=CC=CC=C1 MRXDGVXSWIXTQL-HYHFHBMOSA-N 0.000 description 1
- QFNFXYPFSYJCDF-UHFFFAOYSA-N 1-hydroxy-2h-isoquinolin-3-one Chemical compound C1=CC=CC2=C(O)NC(=O)C=C21 QFNFXYPFSYJCDF-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
- PQGCEDQWHSBAJP-TXICZTDVSA-N 5-O-phosphono-alpha-D-ribofuranosyl diphosphate Chemical compound O[C@H]1[C@@H](O)[C@@H](O[P@](O)(=O)OP(O)(O)=O)O[C@@H]1COP(O)(O)=O PQGCEDQWHSBAJP-TXICZTDVSA-N 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- OLVPQBGMUGIKIW-UHFFFAOYSA-N Chymostatin Natural products C=1C=CC=CC=1CC(C=O)NC(=O)C(C(C)CC)NC(=O)C(C1NC(N)=NCC1)NC(=O)NC(C(O)=O)CC1=CC=CC=C1 OLVPQBGMUGIKIW-UHFFFAOYSA-N 0.000 description 1
- 108020004638 Circular DNA Proteins 0.000 description 1
- 231100001074 DNA strand break Toxicity 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 102000006947 Histones Human genes 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- MRAUNPAHJZDYCK-BYPYZUCNSA-N L-nitroarginine Chemical compound OC(=O)[C@@H](N)CCCNC(=N)N[N+]([O-])=O MRAUNPAHJZDYCK-BYPYZUCNSA-N 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 108010047956 Nucleosomes Proteins 0.000 description 1
- 102100032347 Poly(ADP-ribose) glycohydrolase Human genes 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 102000000505 Ribonucleotide Reductases Human genes 0.000 description 1
- 108010041388 Ribonucleotide Reductases Proteins 0.000 description 1
- 230000006295 S-nitrosylation Effects 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 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 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 101710162629 Trypsin inhibitor Proteins 0.000 description 1
- 229940122618 Trypsin inhibitor Drugs 0.000 description 1
- 108010006886 Vitrogen Proteins 0.000 description 1
- DFPAKSUCGFBDDF-ZQBYOMGUSA-N [14c]-nicotinamide Chemical compound N[14C](=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-ZQBYOMGUSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 230000001476 alcoholic effect Effects 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 239000008365 aqueous carrier Substances 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 230000001964 calcium overload Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 230000002490 cerebral effect Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 108010086192 chymostatin Proteins 0.000 description 1
- 230000001054 cortical effect Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000000779 depleting effect Effects 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 230000002964 excitative effect Effects 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 239000011261 inert gas Substances 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 238000005184 irreversible process Methods 0.000 description 1
- 210000004153 islets of langerhan Anatomy 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 230000005787 mitochondrial ATP synthesis coupled electron transport Effects 0.000 description 1
- 230000002438 mitochondrial effect Effects 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 230000000324 neuroprotective effect Effects 0.000 description 1
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 1
- BOPGDPNILDQYTO-NNYOXOHSSA-N nicotinamide-adenine dinucleotide Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 BOPGDPNILDQYTO-NNYOXOHSSA-N 0.000 description 1
- 231100001096 no neurotoxicity Toxicity 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 210000001623 nucleosome Anatomy 0.000 description 1
- 235000015097 nutrients Nutrition 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
- 235000008390 olive oil Nutrition 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 229950000964 pepstatin Drugs 0.000 description 1
- 108010091212 pepstatin Proteins 0.000 description 1
- FAXGPCHRFPCXOO-LXTPJMTPSA-N pepstatin A Chemical compound OC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)CC(C)C FAXGPCHRFPCXOO-LXTPJMTPSA-N 0.000 description 1
- 108010078356 poly ADP-ribose glycohydrolase Proteins 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 208000037821 progressive disease Diseases 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000008263 repair mechanism Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 238000012549 training Methods 0.000 description 1
- 239000002753 trypsin inhibitor Substances 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000005909 tumor killing Effects 0.000 description 1
- 238000002211 ultraviolet spectrum Methods 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/47—Quinolines; Isoquinolines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/16—Amides, e.g. hydroxamic acids
- A61K31/165—Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
Definitions
- the invention relates to the use of inhibitors of poly(ADP-ribose) synthetase to prevent NMDA neurotoxicity.
- Nitric oxide is a messenger molecule that regulates macrophage killing of tumor cells and bacteria (C. F. Nathan and J. B. Hibbs, Jr., Curr. Opin. Immunol. 3, 65 (1991)), blood vessel relaxation (S. Moncada, R. M. J. Palmer, E. A. Higgs, Pharmacol. Rev. 43, 109 (1991); L. J. Ignarro, Ann. Rev. Pharmacol. Toxicol. 30, 535 (1990)) and also is a neurotransmitter (D. S. Bredt and S. H. Snyder, Neuron 8, 3 (1992)).
- Mechanisms proposed for NO neurotoxicity as well as tumoricidal and bactericidal actions include mono-ADP-ribosylation and S-nitrosylation of glyceraldehyde-3-phosphate dehydrogenase (J. Zhang and S. H. Snyder, Proc. Natl. Acad. Sci. U.S.A. 89, 9382 (1992); A. Y. Kots et al., FEBS Lett. 300, 9 (1992); S. Dimmeler, F. Lottspeich, B. Brune, J. Biol. Chem. 267, 16771 (1992); L. Molina y Vedia et al., J. Biol. Chem.
- Another object of the invention is to provide a method of treating a neurodegenerative disease, such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
- a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
- a method of preventing or treating diseases caused by NMDA neurotoxicity comprises administering to a mammal in need thereof a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
- a method of preventing or treating vascular stroke damage in a mammal, in particular a human patient comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
- a method of treating a mammal for a neurodegenerative disease caused by NMDA neurotoxicity in particular preventing or treating a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease in a human patient, is provided, which method comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
- a pharmaceutically acceptable formulation which comprises an inhibitor of poly(ADP-ribose) synthetase in a pharmaceutically acceptable vehicle.
- the present invention provides the art with methods and formulations for treating neurological damage due to neurodegenerative diseases and vascular stroke.
- FIG. 1 Activation of PARS by NO-damaged DNA.
- B Autoradiography of PARS auto-poly(ADP-ribosyl)ation on 7.5% SDS-PAGE. Abbreviations AmNAP, 4amino-1,8-naphthalimide, DHIQ, 1,5-dihydroxyisoquinoline.
- FIG. 2 Inhibition of NMDA and NO mediated Neurotoxicity by PARS Inhibitors. 100 ⁇ M benzamide reduces NMDA, SNP (500 ⁇ M) and SNAP (300 ⁇ M) mediated neurotoxicity.
- Data are the means ⁇ SEM (n>8). Each data point represents 4,000-12,000 neurons counted (21). *p ⁇ 0.01, Student's t-test.
- FIG. 3 A Model of NO-mediated Cytotoxicity.
- NO damaged DNA activates PARS which depletes NAD by poly-ADP-ribosylating neuclear proteins.
- Poly(ADP-ribose) is rapidly degraded by poly(ADP-ribose) glycohydrolase.
- the futile cycle continues during the prolonged PARS activation. It takes an equivalent of four ATP's to resynthesize NAD from nicotinamide (NAm) via nicotinamide mononucleotide (NMN), a reaction that requires phosphoribosyl pyrophosphate (PRPP) and ATP.
- NAm nicotinamide
- NPN nicotinamide mononucleotide
- PRPP phosphoribosyl pyrophosphate
- NO activates PARS in association with damage to DNA.
- PARS activation can kill cells by consuming ⁇ -nicotinamide adenine dinucleotide, the source of ADP-ribose, and ATP.
- PARS inhibitors prevent NMDA neurotoxicity with relative potencies paralleling their enzyme inhibitory actions.
- PARS inhibition The ability of PARS inhibition to provide substantial protection against NMDA neurotoxicity, implicates DNA damage in neuronal killing. PARS activation rapidly leads to energy depletion. For each ADP-ribose unit transferred by PARS, one molecule of NAD is consumed and an equivalent of four molecules of ATP are required to regenerate NAD from nicotinamide, PARS, whose density is up to one enzyme per 10-20 nucleosomes, can be activated 10-20 fold by DNA damage; PARS transfers 50-100 ADP-ribose moieties to each acceptor site of target proteins and its over-activation by substantial DNA damage can markedly deplete cells of NAD and ATP (J. C. Gaal, K. R. Smith, C. K.
- Poly(ADP-ribose) synthetase inhibitors may be used to prevent, treat, arrest, or ameliorate the progression of any disease condition caused by NMDA neurotoxicity.
- diseases include vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states.
- vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states.
- a patient is administered a poly(ADP-ribose) synthetase inhibitor to block damage to the brain.
- Patients with symptoms of Alzheimer's or Huntington's disease are treated with poly(ADP-ribose) synthatase inhibitors to halt the progression of the disease.
- the symptoms of these disease states are known by one skilled in this art.
- Inhibitors of poly(ADP-ribose) synthetase are compounds which compete for the substrate binding site of poly(ADP-ribose) synthetase or other sites on the enzyme, and include both reversible and irreversible inhibitors.
- the present invention contemplates the use of any physiologically acceptable inhibitor which inhibits poly(ADP-ribose) synthetase activity.
- the effectiveness of a compound, and its relative potency as a poly(ADP-ribose) synthetase inhibitor can be tested and routinely determined by measuring inhibition of poly(ADP-ribose) synthetase activity.
- Poly(ADP-ribose) synthetase activity can be assayed according to the method of Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986).
- various benzamide derivatives have been found to have the ability to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. These include: 3-aminobenzamide and 4-aminobenzamide. Benzoic acid was found not to have any activity.
- novobiocin an inhibitor of mono(ADP-ribose) synthetase was found not to have any activity in preventing neurotoxicity.
- the dosage and length of treatment depends on the disease state being treated.
- the duration of treatment may be a day, a week or longer and may, as in the case of a chronic progressive illness, such as Alzheimer's, last over the entire lifetime of the patient.
- the inhibitors are administered in a therapeutically effective amount, a typical human dosage of benzamide ranging from about 0.01 mg/kg of body weight to about 10 mg/kg, in single or divided doses.
- the dosage will vary depending on the poly(ADP-ribose) synthetase inhibitor being used and its relative potency. Dosage and length of treatment are readily determinable by the skilled practitioner based on the condition and stage of disease.
- poly(ADP-ribose) synthetase inhibitors may be administered by any route by which drugs are conventionally administered.
- routes of administration include intraperitoneal, intravenous, intramuscular, subcutaneous, intrathecal, intraventricular, as well as oral.
- Typical pharmaceutically acceptable preparations for administration include sterile aqueous or nonaqueous solutions, suspensions and emulsions.
- nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil and injectable organic esters such as ethyl oleate.
- Aqueous carriers include water alcoholic/aqueous and buffered media.
- Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like.
- Oral preparations, such as in capsule, tablets, and other form include additives such as cellulose, silica gel and stearic acid.
- a poly(ADP-ribose) synthetase inhibitor desirably should be able to penetrate the blood brain barrier when peripherally administrated.
- Poly(ADP-ribose) synthetase inhibitors which are unable to penetrate the blood brain barrier can be effectively administered by, for example, an intraventricular route of delivery.
- SIN-1 3-morpholinosyndnonimine
- SNP sodium nitroprusside
- Each 50 ⁇ l assay mixture contained 10 ⁇ g of the nuclear protein fraction and [adenylate- 32 P] NAD (0.1 mM, 10 Ci/mmole), in the presence or absence of 4-amino-1,8-naphthalimide (20 ⁇ M), 1,5-dihydroxyisoquinoline (20 ⁇ M), benzamide (100 ⁇ M, SNP (1 mM), SIN-1 (1 mM), DNA (0.1 pg, pTrcA, In Vitrogen) and DNA that had been treated with NO gas, SNP or SIN-1. Incubation of DNA with NO gas was according to T.
- benzamide derivatives exist with differing potencies as PARS inhibitors.
- benzamide is the most active, 3-aminobenzamide is about half as potent, 4-aminobenzamide is 50-100 fold weaker than benzamide and benzoic acid is inactive (The IC 50 for benzamide in inhibiting PARS in vitro is 22 ⁇ M [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)].
- IC 50 for benzamide in inhibiting PARS in vitro is 22 ⁇ M [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)].
- benzamide would be competing with millimolar endogenous levels of NAD [R. McNerney et al., Biochim. Biophys. Acta 1009, 185 (1989)].
- Benzamide provided the greatest degree of protection, with 3-aminobenzamide exerting somewhat less protection against NMDA neurotoxicity, while 4-aminobenzamide and benzoic acid were inactive (Table 1 ).
- a structurally unrelated PARS inhibitor, 1,5-dihydroxyisoquinoline (10 ⁇ M) was also neuroprotective against NMDA neurotoxicity (Table 1A).
- the absolute as well as the relative potencies of the benzamide derivatives and 1,5-dihydroxyisoquinoline in blocking NMDA toxicity correspond with their potencies as PARS inhibitors.
- NMDA exposure initiates "delayed neurotoxicity" in which poorly characterized irreversible processes ultimately lead to calcium overload and cell death (R. D. Randall and S. A. Thayer, J. Neurosci. 12, 1882 (1992); D. W. Choi, Neuron 1, 623 (1988); D. W. Choi, J. Neurosci. 10, 2493 (1990)).
- benzamide was added prior to the NMDA and during NMDA exposure (FIG. 2, Table 1A). However, in some experiments we have added benzamide up to 1 hr after NMDA exposure and observed similar protection (Table 1B).
- Table 1A PARS inhibitors were applied 30 min before and during NMDA application.
- Table 1B PARS inhibitors were applied after NMDA administration at the indicated times for 20 to 24 hr. Data are the means ⁇ SEM (n ⁇ 8). Each data point represents 4,000-12,000 neurons counted. *p ⁇ 0.001 Student's t-test.
- Neurotoxicity was determined by exposing the neurons to the various test solutions as previously described (Dawson, supra). NADA, SNP or SNAP were applied to the cells for 5 min, then the cells were washed and replaced with MEM, 21 mM glucose overnight in the incubator. Twenty to 24 hr after exposure to test solutions, the neurons were exposed to 0.4% trypan blue in CSS to stain the residue of non-viable cells and to assess toxicity. Viable and non-viable cells were counted. At least two separate experiments utilizing four separate wells were performed for each data point shown. Significant overall values were obtained by using a one-way, between groups analysis of variance. Specific comparisons on all possible pair combinations were made with the Student's t-test for independent means.
- Nitrite formation in a human kidney 293 cell line stably transfected with the cDNA for brain NOS was measured in response to A23187 (10 ⁇ M) as described (D. S. Bredt, C. D. Ferris and S. H. Snyder, J. Biol. Chem. 267, 10976 (1992).
- A23187 (10 ⁇ M) elicited the formation of 17.1 ⁇ 2.1 ⁇ M nitrite in 2 hr.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Inhibitors of poly(ADP-ribose) synthetase can be used to prevent neurotoxicity mediated through N-methyl-D-aspartate (NMDA) receptors. Poly(ADP-ribose) synthetase inhibitors can be used therapeutically in the treatment of vascular stroke and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and Huntington's disease.
Description
The U.S. Government has a paid-up license in this invention and the right in limited circumstances to require the patent owner to license others on reasonable terms as provided for by the terms of grants awarded by the National Institutes of Health and the U.S. Public Health Service, including USPHS grants DA-00266, Contract DA-271-90-7408, Research Scientist Award DA-00074, USPHS CIDA NS-01578 and an Intramural Research Training Award from the N.I.H.
The invention relates to the use of inhibitors of poly(ADP-ribose) synthetase to prevent NMDA neurotoxicity.
Nitric oxide (NO) is a messenger molecule that regulates macrophage killing of tumor cells and bacteria (C. F. Nathan and J. B. Hibbs, Jr., Curr. Opin. Immunol. 3, 65 (1991)), blood vessel relaxation (S. Moncada, R. M. J. Palmer, E. A. Higgs, Pharmacol. Rev. 43, 109 (1991); L. J. Ignarro, Ann. Rev. Pharmacol. Toxicol. 30, 535 (1990)) and also is a neurotransmitter (D. S. Bredt and S. H. Snyder, Neuron 8, 3 (1992)). When formed in high quantities in response to actions of the excitatory neurotransmitter glutamate acting at N-methyl- D-aspartate (NMDA) receptors. NO mediates neuronal killing (V. L. Dawson et al., Proc. Natl. Acad. Sci. U.S.A. 88, 6368 (1991); V. L. Dawson et al., J. Neurosci. 13, 2651 (1993)). Toxicity due to NMDA accounts for neural damage in vascular stroke, as NO synthase (NOS) inhibitors prevent stroke damage (J. P. Nowicki, D. Duval, H. Poignet, B. Scatton, Eur. J. Pharmac. 204, 339 (1991)). Mechanisms proposed for NO neurotoxicity as well as tumoricidal and bactericidal actions include mono-ADP-ribosylation and S-nitrosylation of glyceraldehyde-3-phosphate dehydrogenase (J. Zhang and S. H. Snyder, Proc. Natl. Acad. Sci. U.S.A. 89, 9382 (1992); A. Y. Kots et al., FEBS Lett. 300, 9 (1992); S. Dimmeler, F. Lottspeich, B. Brune, J. Biol. Chem. 267, 16771 (1992); L. Molina y Vedia et al., J. Biol. Chem. 267, 24929 (1992)), inhibition of mitochondrial enzymes such as cis-aconitase (J. -C. Drapier and J. B. Hibbs, Jr., J. Clin. Invest. 78, 790 (1986)), inhibition of the mitochondrial electron transport chain (Nathan and Hibbs, supra), inhibition of ribonucleotide reductase (Lepoivre, B. Chenais, A. Yapo, G. Lemaire, J. Biol. Chem. 265, 14143 (1990); N. S. Kwon, D. J. Stuehr, C. F. Nathan, J. Exp. Med. 174, 761 (1991)), and DNA damage (D. A. Wink et al., Science 254, 1001 (1991); T. Nguyen et al., Proc. Natl. Acad. Sci. U.S.A. 89, 3030 (1992)). DNA damage activates poly(ADP-ribose) synthetase (PARS, EC 2.4.2.30) (G. de Murcia, J. Menissier-de Murcia, V. Schreiber, BioEssays 13,455 (1991); J. E. Cleaver and W. F. Morgan, Mutation Res. 257, 1 (1991); J. C. Gaal, K. R. Smith, C. K. Pearson, Trends in Biological Sciences 12, 129 (1987); N. A. Berger, Rad. Res. 101, 4 (1985)). PARS is a nuclear enzyme which, upon activation by DNA strand breaks, adds up to 100 ADP-ribose units to nuclear proteins such as histones and PARS itself.
There is a continuing need in the art for effective methods of preventing, treating or ameliorating diseases caused by NMDA neurotoxicity, such as vascular stroke, Alzheimer's disease, Huntington's disease, and Parkinson's disease.
It is an object of the invention to provide a method of treating diseases caused by NMDA neurotoxicity.
It is another object of the invention to provide a method of treating vascular stroke.
Another object of the invention is to provide a method of treating a neurodegenerative disease, such as Huntington's disease, Alzheimer's disease and Parkinson's disease.
It is still another object of the invention to provide pharmaceutical formulations for treating neurodegenerative diseases and vascular stroke damage.
These and other objects of the invention are provided by one or more of the embodiments described below.
In one embodiment of the invention, a method of preventing or treating diseases caused by NMDA neurotoxicity is provided, which method comprises administering to a mammal in need thereof a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In another embodiment of the invention, a method of preventing or treating vascular stroke damage in a mammal, in particular a human patient, is provided, which comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In still another embodiment of the invention, a method of treating a mammal for a neurodegenerative disease caused by NMDA neurotoxicity, in particular preventing or treating a neurodegenerative disease such as Huntington's disease, Alzheimer's disease and Parkinson's disease in a human patient, is provided, which method comprises administering a therapeutically effective amount of a poly(ADP-ribose) synthetase inhibitor.
In yet another embodiment of the invention, a pharmaceutically acceptable formulation is provided which comprises an inhibitor of poly(ADP-ribose) synthetase in a pharmaceutically acceptable vehicle.
Thus the present invention provides the art with methods and formulations for treating neurological damage due to neurodegenerative diseases and vascular stroke.
FIG. 1. Activation of PARS by NO-damaged DNA. (A) PARS activity, (means ±S.E.M., n=3), after different treatments. (B) Autoradiography of PARS auto-poly(ADP-ribosyl)ation on 7.5% SDS-PAGE. Abbreviations AmNAP, 4amino-1,8-naphthalimide, DHIQ, 1,5-dihydroxyisoquinoline.
FIG. 2. Inhibition of NMDA and NO mediated Neurotoxicity by PARS Inhibitors. 100 μM benzamide reduces NMDA, SNP (500 μM) and SNAP (300 μM) mediated neurotoxicity.
Data are the means ±SEM (n>8). Each data point represents 4,000-12,000 neurons counted (21). *p≦0.01, Student's t-test.
FIG. 3. A Model of NO-mediated Cytotoxicity. NO damaged DNA activates PARS which depletes NAD by poly-ADP-ribosylating neuclear proteins. Poly(ADP-ribose) is rapidly degraded by poly(ADP-ribose) glycohydrolase. The futile cycle continues during the prolonged PARS activation. It takes an equivalent of four ATP's to resynthesize NAD from nicotinamide (NAm) via nicotinamide mononucleotide (NMN), a reaction that requires phosphoribosyl pyrophosphate (PRPP) and ATP.
It is a discovery of the present invention that NO activates PARS in association with damage to DNA. PARS activation can kill cells by consuming β-nicotinamide adenine dinucleotide, the source of ADP-ribose, and ATP. Moreover, PARS inhibitors prevent NMDA neurotoxicity with relative potencies paralleling their enzyme inhibitory actions.
Our observation that NO activates PARS fits with other studies indicating that NO damages DNA (D. A. Wink et al., Science 254, 1001 (1991); T. Nguyen et al., Proc. Natl. Acad. Sci. U.S.A. 89, 3030 (1992)). The protection against NMDA neurotoxicity provided by PARS inhibitors appears to reflect PARS inhibition, as the relative potencies of these drugs in blocking neurotoxicity parallel their activities as PARS inhibitors.
The ability of PARS inhibition to provide substantial protection against NMDA neurotoxicity, implicates DNA damage in neuronal killing. PARS activation rapidly leads to energy depletion. For each ADP-ribose unit transferred by PARS, one molecule of NAD is consumed and an equivalent of four molecules of ATP are required to regenerate NAD from nicotinamide, PARS, whose density is up to one enzyme per 10-20 nucleosomes, can be activated 10-20 fold by DNA damage; PARS transfers 50-100 ADP-ribose moieties to each acceptor site of target proteins and its over-activation by substantial DNA damage can markedly deplete cells of NAD and ATP (J. C. Gaal, K. R. Smith, C. K. Pearson, Trends in Biological Sciences 12, 129 (1987); N. A. Berger, Rad. Res. 101, 4 (1985)). Neurotoxicity is similarly associated with a change in the NADH/NAD redox state and energy depletion. (See, for example, O. H. Lowry et al., J. Biol. Chem. 239, 18 (1964); D. Uematsu et al., Brain Research 482, 129 (1989); but also see K. M. Raley and P. Lipton, Neurosci. Lett. 110, 118 (1990)). The protection against NMDA neurotoxicity afforded by PARS inhibition supports a mechanism of cell death in which DNA damage overwhelms repair mechanisms leading to energy depletion by activation of PARS (FIG. 3).
Two major effects of glutamate neurotoxicity suggested to participate in neuronal killing are the formation of free radicals and energy depletion (B. Meldrum and J. Garthwaite, Trends Pharmacol. Sci. 11, 379 (1990)). Our findings indicate that both participate, with NO, a free radical, triggering DNA damage, which in turn activates PARS, ultimately depleting energy sources from the cell. This mechanism of cell death might also account for the tumoricidal effects of NO formed by macrophages, which fits with observations that 3-aminobenzamide protects pancreatic islet cells from macrophage cytotoxicity (B. Kallman et al., Life Sci. 51, 671 (1992)).
Poly(ADP-ribose) synthetase inhibitors may be used to prevent, treat, arrest, or ameliorate the progression of any disease condition caused by NMDA neurotoxicity. Such conditions include vascular strokes and neurodegenerative diseases, such as Alzheimer's, Huntington's and Parkinson's diseases, as well as other disease states. For example, following the symptoms of a stroke, a patient is administered a poly(ADP-ribose) synthetase inhibitor to block damage to the brain. Patients with symptoms of Alzheimer's or Huntington's disease are treated with poly(ADP-ribose) synthatase inhibitors to halt the progression of the disease. The symptoms of these disease states are known by one skilled in this art.
Inhibitors of poly(ADP-ribose) synthetase are compounds which compete for the substrate binding site of poly(ADP-ribose) synthetase or other sites on the enzyme, and include both reversible and irreversible inhibitors. The present invention contemplates the use of any physiologically acceptable inhibitor which inhibits poly(ADP-ribose) synthetase activity. The effectiveness of a compound, and its relative potency as a poly(ADP-ribose) synthetase inhibitor, can be tested and routinely determined by measuring inhibition of poly(ADP-ribose) synthetase activity. Poly(ADP-ribose) synthetase activity can be assayed according to the method of Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986).
Both benzamide and 1,5-dihydroxy-isoquinoline, two poly(ADP-ribose) synthetase inhibitors, have been found to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. In addition, various benzamide derivatives have been found to have the ability to prevent neurotoxicity in proportion to their relative potencies as poly(ADP-ribose) synthetase inhibitors. These include: 3-aminobenzamide and 4-aminobenzamide. Benzoic acid was found not to have any activity. In addition, novobiocin, an inhibitor of mono(ADP-ribose) synthetase was found not to have any activity in preventing neurotoxicity.
The dosage and length of treatment depends on the disease state being treated. The duration of treatment may be a day, a week or longer and may, as in the case of a chronic progressive illness, such as Alzheimer's, last over the entire lifetime of the patient. The inhibitors are administered in a therapeutically effective amount, a typical human dosage of benzamide ranging from about 0.01 mg/kg of body weight to about 10 mg/kg, in single or divided doses. The dosage will vary depending on the poly(ADP-ribose) synthetase inhibitor being used and its relative potency. Dosage and length of treatment are readily determinable by the skilled practitioner based on the condition and stage of disease.
In therapeutic use, poly(ADP-ribose) synthetase inhibitors may be administered by any route by which drugs are conventionally administered. Such routes of administration include intraperitoneal, intravenous, intramuscular, subcutaneous, intrathecal, intraventricular, as well as oral.
Typical pharmaceutically acceptable preparations for administration include sterile aqueous or nonaqueous solutions, suspensions and emulsions. Examples of nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil and injectable organic esters such as ethyl oleate. Aqueous carriers include water alcoholic/aqueous and buffered media. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like. Oral preparations, such as in capsule, tablets, and other form include additives such as cellulose, silica gel and stearic acid.
To be effective therapeutically, a poly(ADP-ribose) synthetase inhibitor desirably should be able to penetrate the blood brain barrier when peripherally administrated. Poly(ADP-ribose) synthetase inhibitors which are unable to penetrate the blood brain barrier can be effectively administered by, for example, an intraventricular route of delivery.
The following examples are provided to exemplify various aspects of the invention and are not intended to limit the scope of the invention.
This example demonstrates the activation of PARS by NO-treated DNA, as well as the inhibition of such activation by PARS inhibitors.
In rat brain nuclear extracts, PARS activity was almost tripled dose-dependently by adding DNA that had been pre-incubated with NO (FIG. 1A). Addition of covalently closed circular DNA by itself had no effect on PARS activity. Both 4amino-1,8-naphthalimide and 1,5-dihydroxyisoquinoline, two potent PARS inhibitors, reduced the activity to <5% of basal levels (FIG. 1A). The major protein ADP-ribosylated in the nuclear extracts was PARS itself (FIG. 1B). Similarly, DNA that had been treated with 3-morpholinosyndnonimine (SIN-1) and sodium nitroprusside (SNP), two NO donors, could stimulate poly (ADP-ribose) synthesis, which was inhibited by benzamide, another PARS inhibitor. SNP and SIN-1 had no effect on PARS by themselves.
Whole brains from 1-day old rats were homogenized in 20% (w/v) buffer A [50 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM DTT, 50 mM NaCl, 0.25M sucrose, 0.2 mM phenylmethylsulfonyl fluoride, 1 μg/ml each of chymostatin, leupeptin, pepstatin and trypsin inhibitor]. The homogenate was centrifuged at 1000 g for 15 min. The pellet was washed with buffer A and centrifuged again. The washed pellet, termed nuclear fraction, was resuspended in buffer A. PARS activity was assayed according to I. U. Schranfstatter, et al., J. Clin. Inves. 77, 1312 (1986). Each 50 μl assay mixture contained 10 μg of the nuclear protein fraction and [adenylate-32 P] NAD (0.1 mM, 10 Ci/mmole), in the presence or absence of 4-amino-1,8-naphthalimide (20 μM), 1,5-dihydroxyisoquinoline (20 μM), benzamide (100 μM, SNP (1 mM), SIN-1 (1 mM), DNA (0.1 pg, pTrcA, In Vitrogen) and DNA that had been treated with NO gas, SNP or SIN-1. Incubation of DNA with NO gas was according to T. Nguyen et al., Proc. Natl. Acad. Sci. USA 89, 3030 (1992). SNP and SIN-1 treated DNA doubled PARS activity, while both compounds had no effects on PARS by themselves. When [14 C]NAD's were used in place of [32 P]NAD in PARS assay, radioactive polymers were only formed from [adenine-14 C] NAD but not from [carbonyl-14 C]NAD.
McDonald and Moss recently demonstrated that NO-enhanced modification of GAPDH by NAD involves the transfer of the entire NAD to a thiol group rather than ADP-ribosylation (J. P. Nowicki, D. Duval, H. Poignet, B. Scatton, Eur. J. Pharmac. 204, 339 (1991)). To ensure that the polymer formed from NAD in our study was poly(ADP-ribose), we used both [14 C-adenine] NAD and [14 C-nicotinamide] NAD and found radioactivity could only be incorporated into the polymers from the former compound.
This example demonstrates that PARS inhibitors inhibit the neurotoxicity elicited by NMDA.
To determine directly if PARS activation participates in NMDA neurotoxicity, we monitored neurotoxicity elicited by NMDA in cerebral cortical cultures in which NO synthase inhibitors provide marked protection (V. L. Dawson et al., Proc. Natl. Acad. Sci. U.S.A. 88, 6368 (1991); V. L. Dawson et al., J. Neurosci. 13, 2651 (1993)) (FIG. 2). Increasing concentrations of NMDA progressively augment neuronal killing. Benzamide (100 μM) provided 40-50% protection at all NMDA concentrations examined. At 50 μM benzamide, no significant protection was detected, while 500 μM benzamide provided about 30% greater protection than at 100 μM (Table 1A). NO did not interact with benzamide.
To test whether NO interacts directly with benzamide, we incubated benzamide (1 mM) with SNP (1 mM) for five days at 25° C., and analyzed it by HPLC on a C18 column. SNP-treated benzamide was 100% recovered at the same elution time point with the same UV spectrum as that of unreated benzamide. When SNP-treated benzamide and benzamide were mixed and analyzed only one peak was observed.
A variety of benzamide derivatives exist with differing potencies as PARS inhibitors. In the family of benzamide and its derivatives, benzamide is the most active, 3-aminobenzamide is about half as potent, 4-aminobenzamide is 50-100 fold weaker than benzamide and benzoic acid is inactive (The IC50 for benzamide in inhibiting PARS in vitro is 22 μM [M. Banasik, H. Komura, M. Shimoyama, K. Ueda, J. Biol. Chem. 267, 1569 (1992)]. In intact cells a higher value would be anticipated as benzamide would be competing with millimolar endogenous levels of NAD [R. McNerney et al., Biochim. Biophys. Acta 1009, 185 (1989)].
Benzamide provided the greatest degree of protection, with 3-aminobenzamide exerting somewhat less protection against NMDA neurotoxicity, while 4-aminobenzamide and benzoic acid were inactive (Table 1 ). A structurally unrelated PARS inhibitor, 1,5-dihydroxyisoquinoline (10 μM), was also neuroprotective against NMDA neurotoxicity (Table 1A). The absolute as well as the relative potencies of the benzamide derivatives and 1,5-dihydroxyisoquinoline in blocking NMDA toxicity correspond with their potencies as PARS inhibitors. It is unlikely that the PARS inhibitors could prevent NMDA neurotoxicity through inhibition of mono(ADP-ribose) synthetase as their IC50 's for mono(ADP-ribose) synthetase inhibition are at least 100 times greater than their IC50 's as PARS inhibitors. Furthermore, novobiocin (1 mM) a relatively selective mono(ADP-ribose) synthetase inhibitor was ineffective against NMDA neurotoxicity (Table 1A).
Brief (5 rain) NMDA exposure initiates "delayed neurotoxicity" in which poorly characterized irreversible processes ultimately lead to calcium overload and cell death (R. D. Randall and S. A. Thayer, J. Neurosci. 12, 1882 (1992); D. W. Choi, Neuron 1, 623 (1988); D. W. Choi, J. Neurosci. 10, 2493 (1990)). In most experiments benzamide was added prior to the NMDA and during NMDA exposure (FIG. 2, Table 1A). However, in some experiments we have added benzamide up to 1 hr after NMDA exposure and observed similar protection (Table 1B).
TABLE 1 ______________________________________ % Cell Death Treatment (+ S.E.M.) ______________________________________ A. PARS Inhibitors Protect Against NMDA Neurotoxicity 500 μM NMDA 57.1 ± 4.7 +50 μM Benzamide 60.1 ± 12.1 +100 μM Benzamide 39.5 ± 5.0* +500 μM Benazmide 21.9 ± 4.1* +100 μM 3-aminobenzamide 45.9 ± 5.1* +100 μM 4-aminobenzamide 55.8 ± 7.6 +1 mM Benzoic Acid 58.9 ± 5.7 +10 μM 1.5 dihydroxyisoquinoline 39.0 ± 4.5* +1 mM Novobiocin 62.4 ± 8.9 B. PARS Inhibitors Rescue Neurons From NMDA Neurotoxicity 500 μM NMDA 74.7 ± 7.8 +100μM Benzamide 5 min 35.6 ± 8.7* 30 min 40.4 ± 5.6* 1 hour 43.4 ______________________________________ ± 3.9*
Table 1A, PARS inhibitors were applied 30 min before and during NMDA application. In Table 1B, PARS inhibitors were applied after NMDA administration at the indicated times for 20 to 24 hr. Data are the means ±SEM (n≧8). Each data point represents 4,000-12,000 neurons counted. *p≦0.001 Student's t-test.
Primary neuronal cultures from cortex were prepared from fetal Sprague-Dawley rats, gestation day 13-14. Mature neurons (grater than 21 days in culture) were used in all experiments.
Neurotoxicity was determined by exposing the neurons to the various test solutions as previously described (Dawson, supra). NADA, SNP or SNAP were applied to the cells for 5 min, then the cells were washed and replaced with MEM, 21 mM glucose overnight in the incubator. Twenty to 24 hr after exposure to test solutions, the neurons were exposed to 0.4% trypan blue in CSS to stain the residue of non-viable cells and to assess toxicity. Viable and non-viable cells were counted. At least two separate experiments utilizing four separate wells were performed for each data point shown. Significant overall values were obtained by using a one-way, between groups analysis of variance. Specific comparisons on all possible pair combinations were made with the Student's t-test for independent means.
This example demonstrates that NO elicits neurotoxicity by activation of PARS.
To ascertain whether NO itself elicits neurotoxicity by activation of PARS, we administered the NO releasers SNP and S-nitroso-N-acetylpenicillamine (SNAP). Benzamide (100 μM) reduced SNP and SNAP neurotoxicity by 45% to 50% (FIG. 2). Benzamide does not inhibit NOS. In human kidney 293 cells stably transfected with the cDNA of brain. NOS, stimulated levels of nitrite, an indirect measure of NO production, were not affected by 100 μM benzamide, but were abolished by nitroarginic. This experiment also confirms that benzamide did not interact with NO.
Nitrite formation in a human kidney 293 cell line stably transfected with the cDNA for brain NOS was measured in response to A23187 (10 μM) as described (D. S. Bredt, C. D. Ferris and S. H. Snyder, J. Biol. Chem. 267, 10976 (1992). A23187 (10 μM) elicited the formation of 17.1±2.1 μM nitrite in 2 hr. Benzamide (100 μM) did not prevent the formation of nitrite (15.9±3.8 μM nitrite) and 100 μM nitroarginine diminished nitrite formation to 3.9±1.6 μM nitrite (n=3).
Claims (17)
1. A method of treating a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity in a mammal comprising:
administering to a mammal which demonstrates symptoms of a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity a therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase.
2. The method of claim 1 wherein said inhibitor is administered intravenously, intraperitoneally, intramuscularly, intraventricularly, or orally.
3. The method of claim 2 wherein said inhibitor is benzamide.
4. The method of claim 1 wherein said inhibitor is benzamide. .[.5. The method of claim 1 wherein said inhibitor is a benzamide derivative selected from the group consisting of 3-aminobenzamide and
4-aminobenzamide..].6. The method of claim .[.5.]. .Iadd.1 .Iaddend.wherein said inhibitor is 3-aminobenzamide. .[.7. The method of
claim 5 wherein said inhibitor is 4-aminobenzamide..].8. The method of
claim 1 wherein said inhibitor is 1,5-dihydroxy-isoquinoline. 9. The
method of claim 1 wherein said disease is vascular stroke. 10. The method
of claim 1 wherein said disease is a neurodegenerative disease. 11. A pharmaceutical formulation comprising .Iadd.a therapeutically effective amount of .Iaddend.an inhibitor of poly(ADP-ribose) synthetase .Iadd.containing a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide .Iaddend.in a
pharmaceutically acceptable vehicle. 12. The pharmaceutical formulation of claim 11 wherein said inhibitor is benzamide. .[.13. The pharmaceutical formulation of claim 11 wherein said inhibitor is a benzamide derivative selected from the group consisting of 3-aminobenzamide and 4-amino
benzamide..].14. The pharmaceutical formulation of claim .[.13.]. .Iadd.11 .Iaddend.wherein said inhibitor is 3-aminobenzamide. .[.15. The pharmaceutical formulation of claim 13 wherein said inhibitor is
4-aminobenzamide..].16. The pharmaceutical formulation of claim 11 wherein
said inhibitor is 1,5-dihydroxy-isoquinoline. 17. The pharmaceutical formulation of claim 11 wherein said inhibitor has an IC50 for inhibiting poly(ADP-ribose) synthetase in vitro of at least .Iadd.as
potent as .Iaddend.22 μM. 18. The pharmaceutical formulation of claim 11 wherein said inhibitor has an IC50 for inhibiting poly(ADP-ribose)
synthetase in vitro of at least .Iadd.as potent as .Iaddend.33 μM. 19. The pharmaceutical formulation of claim 11 wherein said formulation is a capsule or tablet containing a single or divided dose of said inhibitor, wherein said dose is sufficient to treat .[.NMDA.]. .Iadd.glutamate
.Iaddend.neurotoxicity. 20. The pharmaceutical formulation of claim 11 which is a sterile solution, suspension or emulsion, in a single or divided dose, for administration to a mammal which demonstrates symptoms of a disease condition caused by .[.NMDA.]. .Iadd.glutamate .Iaddend.neurotoxicity. .Iadd.21. The pharmaceutical formulation of claim 11 wherein said formulation is a capsule or tablet containing a single or divided dose of said inhibitor, wherein said dose is sufficient to treat glutamate neurotoxicity mediated through NMDA receptors. .Iaddend..Iadd.22. The pharmaceutical formulation of claim 11 which is a sterile solution, suspension or emulsion, in a single or divided dose, for administration to a mammal which demonstrates symptoms of a disease condition caused by glutamate neurotoxicity mediated through NMDA receptors. .Iaddend..Iadd.23. A method of claim 1 wherein said therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase is in the form of a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide. .Iaddend..Iadd.24. A method of treating a disease condition caused by glutamate neurotoxicity in a mammal comprising:
administering to a mammal which demonstrates symptoms of a disease condition caused by glutamate neurotoxicity mediated through NMDA receptors a therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase. .Iaddend..Iadd.25. The method of claim 24 wherein said inhibitor is administered intravenously, intraperitoneally, intramuscularly, intraventricularly, or orally. .Iaddend..Iadd.26. The method of claim 24 wherein said inhibitor is benzamide. .Iaddend..Iadd.27. The method of claim 25 wherein said inhibitor is benzamide.
.Iaddend..Iadd.8. The method of claim 24 wherein said inhibitor is 3-aminobenzamide. .Iaddend..Iadd.29. The method of claim 24 wherein said inhibitor is 1,5-dihydroxy-isoquinoline. .Iaddend..Iadd.30. The method of claim 24 wherein said disease is vascular stroke. .Iaddend..Iadd.31. The method of claim 24 wherein said disease is a neurodegenerative disease. .Iaddend..Iadd.32. A method of claim 24 wherein said therapeutically effective amount of an inhibitor of poly (ADP-ribose) synthetase is in the form of a single or divided dose having a potency equivalent to about 10 mg/kg of body weight or less of benzamide. .Iaddend.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US09/219,843 USRE36397E (en) | 1994-02-04 | 1998-12-23 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US08/191,508 US5587384A (en) | 1994-02-04 | 1994-02-04 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
US09/219,843 USRE36397E (en) | 1994-02-04 | 1998-12-23 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US08/191,508 Reissue US5587384A (en) | 1994-02-04 | 1994-02-04 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
Publications (1)
Publication Number | Publication Date |
---|---|
USRE36397E true USRE36397E (en) | 1999-11-16 |
Family
ID=22705771
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US08/191,508 Ceased US5587384A (en) | 1994-02-04 | 1994-02-04 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
US09/219,843 Expired - Lifetime USRE36397E (en) | 1994-02-04 | 1998-12-23 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US08/191,508 Ceased US5587384A (en) | 1994-02-04 | 1994-02-04 | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity |
Country Status (7)
Country | Link |
---|---|
US (2) | US5587384A (en) |
AU (1) | AU1695495A (en) |
FI (1) | FI963040A0 (en) |
IL (1) | IL112496A0 (en) |
NO (1) | NO963253D0 (en) |
WO (1) | WO1995020952A1 (en) |
ZA (1) | ZA95861B (en) |
Cited By (54)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6306889B1 (en) | 1997-09-03 | 2001-10-23 | Guilford Pharmaceuticals Inc. | Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage |
US6348475B1 (en) | 2000-06-01 | 2002-02-19 | Guilford Pharmaceuticals Inc. | Methods, compounds and compositions for treating gout |
US6380211B1 (en) | 1997-09-03 | 2002-04-30 | Guilford Pharmaceutical Inc. | Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity |
US6387902B1 (en) | 1998-12-31 | 2002-05-14 | Guilford Pharmaceuticals, Inc. | Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP |
US6395749B1 (en) | 1998-05-15 | 2002-05-28 | Guilford Pharmaceuticals Inc. | Carboxamide compounds, methods, and compositions for inhibiting PARP activity |
US6426415B1 (en) | 1997-09-03 | 2002-07-30 | Guilford Pharmaceuticals Inc. | Alkoxy-substituted compounds, methods and compositions for inhibiting parp activity |
US6440455B1 (en) | 1997-09-02 | 2002-08-27 | Children's Medical Center Corporation | Methods for modulating the axonal outgrowth of central nervous system neurons |
US6444676B1 (en) | 1999-12-20 | 2002-09-03 | Iok-Hou Pang | Use of PARP inhibitors in the treatment of glaucoma |
US20020177614A1 (en) * | 2001-03-23 | 2002-11-28 | Merril Carl R. | Methods for treating nuerodegenerative diseases including alzheimer's |
US6514983B1 (en) | 1997-09-03 | 2003-02-04 | Guilford Pharmaceuticals Inc. | Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage |
US6545011B2 (en) | 2000-07-13 | 2003-04-08 | Guilford Pharmaceuticals Inc. | Substituted 4,9-dihydrocyclopenta[imn]phenanthridine-5-ones, derivatives thereof and their uses |
US6635642B1 (en) | 1997-09-03 | 2003-10-21 | Guilford Pharmaceuticals Inc. | PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same |
US6716828B1 (en) | 1999-09-01 | 2004-04-06 | Guilford Pharmaceuticals, Inc. | Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage |
US6723733B2 (en) | 2000-05-19 | 2004-04-20 | Guilford Pharmaceuticals, Inc. | Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses |
US20040138502A1 (en) * | 2001-03-08 | 2004-07-15 | Traynelis Stephen F | Ph-dependent nmda receptor antagonists |
US20050096342A1 (en) * | 1995-10-23 | 2005-05-05 | Medivation, Inc. | Neurodegenerative disorders |
WO2006078711A2 (en) | 2005-01-19 | 2006-07-27 | Mgi Gp, Inc. | Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting parp |
US20060276497A1 (en) * | 2000-05-09 | 2006-12-07 | Cephalon, Inc. | Novel multicyclic compounds and the use thereof |
US20070117835A1 (en) * | 2005-10-04 | 2007-05-24 | David Hung | Methods and compositions for treating Huntington's disease |
US20070179174A1 (en) * | 2003-12-08 | 2007-08-02 | Bachurin Sergei O | Methods and compositions for slowing aging |
US20070248702A1 (en) * | 2004-06-22 | 2007-10-25 | Javier Fernandez-Ruiz | Use of CB2 receptors agonists for the treatment of Huntington's disease |
US20080161255A1 (en) * | 2003-05-29 | 2008-07-03 | Michael Brownlee | Use of Parp Inhibitors for Prevention and Treatment of Diabetic and Insulin Resistance Complications |
US20080234310A1 (en) * | 2003-12-08 | 2008-09-25 | Bachurin Sergei O | Methods and Compositions for Slowing Aging |
US20110071088A1 (en) * | 2003-12-16 | 2011-03-24 | Childrens Medical Center Corporation | Method for treating neurological disorders |
WO2012027224A1 (en) | 2010-08-24 | 2012-03-01 | Dana-Farber Cancer Institute, Inc. | Methods for predicting anti-cancer response |
US8420680B2 (en) | 2007-06-29 | 2013-04-16 | Emory University | NMDA receptor antagonists for neuroprotection |
WO2013055911A1 (en) | 2011-10-14 | 2013-04-18 | Dana-Farber Cancer Institute, Inc. | Znf365/zfp365 biomarker predictive of anti-cancer response |
WO2013130347A1 (en) | 2012-02-23 | 2013-09-06 | The Children's Hospital Corporation | Methods for predicting anti-cancer response |
WO2015109264A1 (en) | 2014-01-17 | 2015-07-23 | Cedars-Sinai Medical Center | Receptor targeting constructs and uses thereof |
WO2015164743A2 (en) | 2014-04-24 | 2015-10-29 | Dana-Farber Cancer Institute, Inc. | Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response |
US9279156B2 (en) | 2011-06-17 | 2016-03-08 | Myriad Genetics, Inc. | Methods and materials for assessing allelic imbalance |
WO2016044707A1 (en) | 2014-09-18 | 2016-03-24 | Cedars-Sinai Medical Center | Compositions and methods for treating fibrosis |
WO2016057367A1 (en) | 2014-10-06 | 2016-04-14 | Dana-Farber Cancer Institute, Inc. | Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response |
WO2016073647A2 (en) | 2014-11-04 | 2016-05-12 | University Of Southern California | COMPOSITIONS AND METHODS FOR TREATING HIF-1alpha OVER-EXPRESSING CANCERS |
WO2016094273A1 (en) | 2014-12-08 | 2016-06-16 | Dana-Farber Cancer Institute, Inc. | Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents |
US9388472B2 (en) | 2011-12-21 | 2016-07-12 | Myriad Genetics, Inc. | Methods and materials for assessing loss of heterozygosity |
WO2016138574A1 (en) | 2015-03-02 | 2016-09-09 | Sinai Health System | Homologous recombination factors |
WO2016144673A1 (en) | 2015-03-06 | 2016-09-15 | Dana-Farber Cancer Institute, Inc. | Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers |
WO2017066561A2 (en) | 2015-10-16 | 2017-04-20 | President And Fellows Of Harvard College | Regulatory t cell pd-1 modulation for regulating t cell effector immune responses |
WO2018057618A1 (en) | 2016-09-20 | 2018-03-29 | Dana-Farber Cancer Institute, Inc. | Compositions and methods for identification, assessment, prevention, and treatment of aml using usp10 biomarkers and modulators |
US10308986B2 (en) | 2013-03-14 | 2019-06-04 | Children's Medical Center Corporation | Cancer diagnosis, treatment selection and treatment |
US10400287B2 (en) | 2014-08-15 | 2019-09-03 | Myriad Genetics, Inc. | Methods and materials for assessing homologous recombination deficiency |
WO2020033791A1 (en) | 2018-08-09 | 2020-02-13 | Verseau Therapeutics, Inc. | Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof |
WO2020223121A1 (en) | 2019-04-30 | 2020-11-05 | Dana-Farber Cancer Institute, Inc. | Methods for treating cancer using combinations of anti-cx3cr1 and immune checkpoint blockade agents |
WO2021150925A1 (en) | 2020-01-24 | 2021-07-29 | Dana-Farber Cancer Institute, Inc. | Uses of biomarkers for improving immunotherapy |
US11091808B2 (en) | 2012-06-07 | 2021-08-17 | Institut Curie | Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors |
US11149316B2 (en) | 2013-12-09 | 2021-10-19 | Institut Curie | Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors |
WO2022086852A2 (en) | 2020-10-19 | 2022-04-28 | Dana-Farber Cancer Institute, Inc. | Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy |
WO2022104104A2 (en) | 2020-11-13 | 2022-05-19 | Dana-Farber Cancer Institute, Inc. | Personalized fusion cell vaccines |
WO2022159793A2 (en) | 2021-01-25 | 2022-07-28 | Dana-Farber Cancer Institute, Inc. | Methods and compositions for identifying neuroendocrine prostate cancer |
WO2022159575A1 (en) | 2021-01-20 | 2022-07-28 | Bioentre Llc | Ctla4-binding proteins and methods of treating cancer |
WO2022261183A2 (en) | 2021-06-08 | 2022-12-15 | Dana-Farber Cancer Institute, Inc. | Compositions and methods for treating and/or identifying an agent for treating intestinal cancers |
WO2023043888A1 (en) | 2021-09-15 | 2023-03-23 | Celloram Inc. | Method of treating cancer |
WO2023240287A1 (en) | 2022-06-10 | 2023-12-14 | Bioentre Llc | Combinations of ctla4 binding proteins and methods of treating cancer |
Families Citing this family (49)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
GB9404485D0 (en) * | 1994-03-09 | 1994-04-20 | Cancer Res Campaign Tech | Benzamide analogues |
US5589483A (en) * | 1994-12-21 | 1996-12-31 | Geron Corporation | Isoquinoline poly (ADP-ribose) polymerase inhibitors to treat skin diseases associated with cellular senescence |
US6358975B1 (en) | 1997-08-15 | 2002-03-19 | Johns Hopkins University | Method of using selective parp inhibitors to prevent or treat neurotoxicity |
WO1999008680A1 (en) * | 1997-08-15 | 1999-02-25 | The Johns Hopkins University | Method of using selective parp inhibitors to prevent or treat neurotoxicity |
US6235748B1 (en) | 1997-09-03 | 2001-05-22 | Guilford Pharmaceuticals Inc. | Oxo-substituted compounds, process of making, and compositions and methods for inhibiting parp activity |
US6333148B1 (en) | 1998-05-01 | 2001-12-25 | University Of Kentucky Research | Genes encoding several poly (ADP-ribose) glycohydrolase (PARG) enzymes, the proteins and fragments thereof, and antibodies immunoreactive therewith |
WO1999059973A1 (en) * | 1998-05-15 | 1999-11-25 | Guilford Pharmaceuticals Inc. | Carboxamide compounds, compositions, and methods for inhibiting parp activity |
US20030078212A1 (en) * | 1998-10-30 | 2003-04-24 | Jia-He Li | Pharmaceutical compositions containing poly(adp-ribose) glycohydrolase inhibitors and methods of using the same |
US6201020B1 (en) | 1998-12-31 | 2001-03-13 | Guilford Pharmaceuticals, Inc. | Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp |
RO121778B1 (en) | 1999-01-11 | 2008-04-30 | Agouron Pharmaceuticals, Inc. | Tricyclic inhibitors of poly(adp-ribose) polymerase |
US6187822B1 (en) * | 1999-06-11 | 2001-02-13 | University Of Medicine & Dentistry Of Nj | Wound treatment through inhibition of adenosine diphosphate ribosyl transferase |
ECSP003637A (en) | 1999-08-31 | 2002-03-25 | Agouron Pharma | TRICYCLE POLY INHIBITORS (ADP-RIBOSA) POLYMERASES |
US6531464B1 (en) * | 1999-12-07 | 2003-03-11 | Inotek Pharmaceutical Corporation | Methods for the treatment of neurodegenerative disorders using substituted phenanthridinone derivatives |
US6476048B1 (en) | 1999-12-07 | 2002-11-05 | Inotek Pharamaceuticals Corporation | Substituted phenanthridinones and methods of use thereof |
US6277990B1 (en) | 1999-12-07 | 2001-08-21 | Inotek Corporation | Substituted phenanthridinones and methods of use thereof |
US7122679B2 (en) * | 2000-05-09 | 2006-10-17 | Cephalon, Inc. | Multicyclic compounds and the use thereof |
US7151102B2 (en) * | 2000-10-30 | 2006-12-19 | Kudos Pharmaceuticals Limited | Phthalazinone derivatives |
US6635786B2 (en) | 2001-01-16 | 2003-10-21 | Guilford Pharmaceuticals, Inc. | Symmetrically disubstituted aromatic compounds and pharmaceutical compositions for inhibiting poly (ADP-ribose) glycohydrolase, and methods for their use |
US7072771B2 (en) * | 2001-06-07 | 2006-07-04 | University Of Kentucky Research Foundation | Selective PARP-1 targeting for designing chemo/radio sensitizing agents |
KR100955015B1 (en) * | 2001-08-15 | 2010-04-28 | 이코스 코포레이션 | 2H-phthalazin-1-one and its use |
DE60335359D1 (en) * | 2002-04-30 | 2011-01-27 | Kudos Pharm Ltd | phthalazinone |
GB0305681D0 (en) | 2003-03-12 | 2003-04-16 | Kudos Pharm Ltd | Phthalazinone derivatives |
US7449464B2 (en) * | 2003-03-12 | 2008-11-11 | Kudos Pharmaceuticals Limited | Phthalazinone derivatives |
MXPA05010563A (en) * | 2003-03-31 | 2005-11-23 | Pfizer | Salts of tricyclic inhibitors of poly(adp-ribose) polymerases. |
DK1684736T3 (en) | 2003-12-01 | 2011-11-21 | Kudos Pharm Ltd | DNA damage repair inhibitors for cancer treatment |
GB0419072D0 (en) | 2004-08-26 | 2004-09-29 | Kudos Pharm Ltd | Phthalazinone derivatives |
GB0428111D0 (en) * | 2004-12-22 | 2005-01-26 | Kudos Pharm Ltd | Pthalazinone derivatives |
GB0521373D0 (en) | 2005-10-20 | 2005-11-30 | Kudos Pharm Ltd | Pthalazinone derivatives |
CN101484436A (en) * | 2006-06-15 | 2009-07-15 | 库多斯药物有限公司 | 2 -oxyheteroarylamide derivatives as parp inhibitors |
US20090181951A1 (en) * | 2006-06-15 | 2009-07-16 | Kudos Pharmaceuticals Limited | Parp inhibitors |
CN101484421A (en) * | 2006-06-15 | 2009-07-15 | 库多斯药物有限公司 | 2 -oxybenzamide derivatives as parp inhibitors |
TWI404716B (en) * | 2006-10-17 | 2013-08-11 | Kudos Pharm Ltd | Phthalazinone derivative |
US20080280910A1 (en) * | 2007-03-22 | 2008-11-13 | Keith Allan Menear | Phthalazinone derivatives |
TW200900396A (en) * | 2007-04-10 | 2009-01-01 | Kudos Pharm Ltd | Phthalazinone derivatives |
US20090023727A1 (en) * | 2007-07-05 | 2009-01-22 | Muhammad Hashim Javaid | Phthalazinone derivatives |
US7981890B2 (en) | 2007-09-14 | 2011-07-19 | Astrazeneca Ab | Phthalazinone derivatives |
AU2008313467B2 (en) * | 2007-10-17 | 2013-08-29 | Kudos Pharmaceuticals Limited | 4- [3- (4-cyclopropanecarbonyl-piperazine-1-carbonyl) -4 -fluoro-benzyl] -2H-phthalazin-1-one |
AR070221A1 (en) | 2008-01-23 | 2010-03-25 | Astrazeneca Ab | DERIVATIVES OF FTALAZINONA POLYMERASE INHIBITORS, PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND USES OF THE SAME TO PREVENT AND / OR TREAT CANCERIGENE TUMORS, ISCHEMICAL INJURIES AND OTHER ASSOCIATED DISEASES. |
UA106878C2 (en) | 2008-10-07 | 2014-10-27 | Астразенека Юк Лімітед | PHARMACEUTICAL COMPOSITION CONTAINING 4- [3- (4-TSYKLOPROPANKARBONILPIPERAZYN-1-carbonyl) -4-FTORBENZYL] -2N FTALAZYN-1-OH or a salt thereof, or solvate thereof, in solid dispersion with a matrix polymer KOPOVIDONOM |
UY32790A (en) * | 2009-07-15 | 2011-02-28 | Astrazeneca Ab | FTALAZINONA COMPOUND |
WO2011058367A2 (en) | 2009-11-13 | 2011-05-19 | Astrazeneca Ab | Diagnostic test for predicting responsiveness to treatment with poly(adp-ribose) polymerase (parp) inhibitor |
DK3325623T6 (en) | 2015-07-23 | 2021-03-15 | Inst Curie | USE OF A COMBINATION OF DBAIT MOLECULE AND PARP INHIBITORS FOR CANCER OF CANCER |
GB201519573D0 (en) | 2015-11-05 | 2015-12-23 | King S College London | Combination |
WO2018162439A1 (en) | 2017-03-08 | 2018-09-13 | Onxeo | New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule |
EP3615026B1 (en) | 2017-04-28 | 2021-03-03 | Akribes Biomedical GmbH | A parp inhibitor in combination with a glucocorticoid and/or ascorbic acid and/or a protein growth factor for the treatment of impaired wound healing |
CN111819282A (en) | 2018-03-13 | 2020-10-23 | 欧恩科斯欧公司 | Dbait molecule against acquired resistance in cancer therapy |
GB201913030D0 (en) | 2019-09-10 | 2019-10-23 | Francis Crick Institute Ltd | Treatment of hr deficient cancer |
WO2021148581A1 (en) | 2020-01-22 | 2021-07-29 | Onxeo | Novel dbait molecule and its use |
WO2024261243A1 (en) | 2023-06-21 | 2024-12-26 | Hemispherian As | Combination comprising a deoxycytidine derivative and a parp inhibitor for use in a method of treating hr proficient cancer |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4594415A (en) * | 1982-03-29 | 1986-06-10 | Robins Roland K | Azole dinucleotide compositions and methods of use |
EP0355750A1 (en) * | 1988-08-19 | 1990-02-28 | Warner-Lambert Company | Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process |
US5177075A (en) * | 1988-08-19 | 1993-01-05 | Warner-Lambert Company | Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process |
US5262564A (en) * | 1992-10-30 | 1993-11-16 | Octamer, Inc. | Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents |
US5338851A (en) * | 1993-03-31 | 1994-08-16 | Eli Lilly And Company | Synthesis of cis-decahydroisoquinoline-3-carboxylic acids |
-
1994
- 1994-02-04 US US08/191,508 patent/US5587384A/en not_active Ceased
-
1995
- 1995-01-31 IL IL11249695A patent/IL112496A0/en unknown
- 1995-02-02 WO PCT/US1995/001189 patent/WO1995020952A1/en active Application Filing
- 1995-02-02 AU AU16954/95A patent/AU1695495A/en not_active Abandoned
- 1995-02-03 ZA ZA95861A patent/ZA95861B/en unknown
-
1996
- 1996-08-01 FI FI963040A patent/FI963040A0/en not_active Application Discontinuation
- 1996-08-02 NO NO963253A patent/NO963253D0/en unknown
-
1998
- 1998-12-23 US US09/219,843 patent/USRE36397E/en not_active Expired - Lifetime
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4594415A (en) * | 1982-03-29 | 1986-06-10 | Robins Roland K | Azole dinucleotide compositions and methods of use |
EP0355750A1 (en) * | 1988-08-19 | 1990-02-28 | Warner-Lambert Company | Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process |
US5177075A (en) * | 1988-08-19 | 1993-01-05 | Warner-Lambert Company | Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process |
US5262564A (en) * | 1992-10-30 | 1993-11-16 | Octamer, Inc. | Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents |
US5338851A (en) * | 1993-03-31 | 1994-08-16 | Eli Lilly And Company | Synthesis of cis-decahydroisoquinoline-3-carboxylic acids |
Non-Patent Citations (24)
Title |
---|
Banasik et al., "Specific Inhibitors of Poly(ADP-Ribose)Synthetase and Mono (ADP-Ribosyl) Transferase", J Biol Chem, 267:1569-1575 (1992). |
Banasik et al., Specific Inhibitors of Poly(ADP Ribose)Synthetase and Mono (ADP Ribosyl) Transferase , J Biol Chem, 267:1569 1575 (1992). * |
Choi, Dennis, "Glutamate Neurotoxicity and Diseases of the Nervous System", Neuron, 1:623-634 (1988). |
Choi, Dennis, "The Role of Glutamate Neurotoxicity in Hypoxic-Ischemic Neuronal Death", Ann Rev. Neurosci., 13:171-82 (1990). |
Choi, Dennis, Glutamate Neurotoxicity and Diseases of the Nervous System , Neuron, 1:623 634 (1988). * |
Choi, Dennis, The Role of Glutamate Neurotoxicity in Hypoxic Ischemic Neuronal Death , Ann Rev. Neurosci., 13:171 82 (1990). * |
Cosi et al., J. Neuroscience Research, 39(1), 38 46, Sep. 1994. * |
Cosi et al., J. Neuroscience Research, 39(1), 38-46, Sep. 1994. |
Dawson et al., "Mechanisms of Nitric Oxide-Mediated Neurotoxicity in Primary Brain Cultures", J Neuroscience, 13:2651-2661 (1993). |
Dawson et al., "Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures", Proc Natl Acad Sci, 88:6568-6371 (1991). |
Dawson et al., Mechanisms of Nitric Oxide Mediated Neurotoxicity in Primary Brain Cultures , J Neuroscience, 13:2651 2661 (1993). * |
Dawson et al., Nitric Oxide Mediates Glutamate Neurotoxicity in Primary Cortical Cultures , Proc Natl Acad Sci, 88:6568 6371 (1991). * |
Meldrum et al., "Excitatory amino acid neurotoxicity and neurodegenerative disease", TIPS, 11:379-387 (1990). |
Meldrum et al., Excitatory amino acid neurotoxicity and neurodegenerative disease , TIPS, 11:379 387 (1990). * |
Nowicki et al., "Nitric Oxide Mediates Neuronal Death After Focal Cerebral Ischemia in the Mouse", European J Pharmacology, 204, 339-340 (1991). |
Nowicki et al., Nitric Oxide Mediates Neuronal Death After Focal Cerebral Ischemia in the Mouse , European J Pharmacology, 204, 339 340 (1991). * |
Purnell et al., "Novel inhibitors of Poly (ADP-Ribose) Synthetase", Biochem J, 185:775-777 (1980). |
Purnell et al., Novel inhibitors of Poly (ADP Ribose) Synthetase , Biochem J, 185:775 777 (1980). * |
Suto et al., "Dihydroisoquinolinones: the design and synthesis of a new series of potent inhibitors of poly (ADP-ribose) polymerase", Anti-cancer Drug Design, 7:107-117 (1991). |
Suto et al., Dihydroisoquinolinones: the design and synthesis of a new series of potent inhibitors of poly (ADP ribose) polymerase , Anti cancer Drug Design, 7:107 117 (1991). * |
Wallis et al., "Neuroprotection Against Nitric Oxide Injury with Inhibitors of ADP-ribosylation", Neuropharmacology, 5(3):245-248 (1993). |
Wallis et al., Neuroprotection Against Nitric Oxide Injury with Inhibitors of ADP ribosylation , Neuropharmacology, 5(3):245 248 (1993). * |
Zhang et al., "Nitric Oxide Activation of Poly(ADP-Ribose) Synthetase in Neurotoxicity", Science, 263:687-689 (1994). |
Zhang et al., Nitric Oxide Activation of Poly(ADP Ribose) Synthetase in Neurotoxicity , Science, 263:687 689 (1994). * |
Cited By (78)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060140866A1 (en) * | 1995-10-23 | 2006-06-29 | Zefirov Nikolai S | Agents for treating neurodegenerative disorders |
US7071206B2 (en) | 1995-10-23 | 2006-07-04 | Medivation, Inc. | Agents for treating neurodegenerative disorders |
US20050096342A1 (en) * | 1995-10-23 | 2005-05-05 | Medivation, Inc. | Neurodegenerative disorders |
US6440455B1 (en) | 1997-09-02 | 2002-08-27 | Children's Medical Center Corporation | Methods for modulating the axonal outgrowth of central nervous system neurons |
US20070213294A1 (en) * | 1997-09-02 | 2007-09-13 | Children's Medical Center Corporation | Methods for modulating the axonal growth of central nervous system neurons |
US6426415B1 (en) | 1997-09-03 | 2002-07-30 | Guilford Pharmaceuticals Inc. | Alkoxy-substituted compounds, methods and compositions for inhibiting parp activity |
US6514983B1 (en) | 1997-09-03 | 2003-02-04 | Guilford Pharmaceuticals Inc. | Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage |
US6380211B1 (en) | 1997-09-03 | 2002-04-30 | Guilford Pharmaceutical Inc. | Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity |
US6635642B1 (en) | 1997-09-03 | 2003-10-21 | Guilford Pharmaceuticals Inc. | PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same |
US6346536B1 (en) | 1997-09-03 | 2002-02-12 | Guilford Pharmaceuticals Inc. | Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same |
US6306889B1 (en) | 1997-09-03 | 2001-10-23 | Guilford Pharmaceuticals Inc. | Compounds, methods and pharmaceutical compositions for treating neural or cardiovascular tissue damage |
US6395749B1 (en) | 1998-05-15 | 2002-05-28 | Guilford Pharmaceuticals Inc. | Carboxamide compounds, methods, and compositions for inhibiting PARP activity |
US6387902B1 (en) | 1998-12-31 | 2002-05-14 | Guilford Pharmaceuticals, Inc. | Phenazine compounds, methods and pharmaceutical compositions for inhibiting PARP |
US7307080B2 (en) | 1999-09-01 | 2007-12-11 | Mgi Gp, Inc. | Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage |
US6716828B1 (en) | 1999-09-01 | 2004-04-06 | Guilford Pharmaceuticals, Inc. | Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage |
US20050074470A1 (en) * | 1999-09-01 | 2005-04-07 | Guilford Pharmaceuticals, Inc. | Compounds, methods and pharmaceutical compositions for treating cellular damage, such as neural or cardiovascular tissue damage |
US6444676B1 (en) | 1999-12-20 | 2002-09-03 | Iok-Hou Pang | Use of PARP inhibitors in the treatment of glaucoma |
US20060276497A1 (en) * | 2000-05-09 | 2006-12-07 | Cephalon, Inc. | Novel multicyclic compounds and the use thereof |
US6723733B2 (en) | 2000-05-19 | 2004-04-20 | Guilford Pharmaceuticals, Inc. | Sulfonamide and carbamide derivatives of 6(5H)phenanthridinones and their uses |
US6348475B1 (en) | 2000-06-01 | 2002-02-19 | Guilford Pharmaceuticals Inc. | Methods, compounds and compositions for treating gout |
US6545011B2 (en) | 2000-07-13 | 2003-04-08 | Guilford Pharmaceuticals Inc. | Substituted 4,9-dihydrocyclopenta[imn]phenanthridine-5-ones, derivatives thereof and their uses |
US20040138502A1 (en) * | 2001-03-08 | 2004-07-15 | Traynelis Stephen F | Ph-dependent nmda receptor antagonists |
US7375136B2 (en) | 2001-03-08 | 2008-05-20 | Emory University | pH-dependent NMDA receptor antagonists |
US20020177614A1 (en) * | 2001-03-23 | 2002-11-28 | Merril Carl R. | Methods for treating nuerodegenerative diseases including alzheimer's |
US20080161255A1 (en) * | 2003-05-29 | 2008-07-03 | Michael Brownlee | Use of Parp Inhibitors for Prevention and Treatment of Diabetic and Insulin Resistance Complications |
US20070179174A1 (en) * | 2003-12-08 | 2007-08-02 | Bachurin Sergei O | Methods and compositions for slowing aging |
US20080234310A1 (en) * | 2003-12-08 | 2008-09-25 | Bachurin Sergei O | Methods and Compositions for Slowing Aging |
US20110071088A1 (en) * | 2003-12-16 | 2011-03-24 | Childrens Medical Center Corporation | Method for treating neurological disorders |
US8912144B2 (en) | 2003-12-16 | 2014-12-16 | Children's Medical Center Corporation | Method for treating stroke via administration of NEP1-40 and inosine |
US20070248702A1 (en) * | 2004-06-22 | 2007-10-25 | Javier Fernandez-Ruiz | Use of CB2 receptors agonists for the treatment of Huntington's disease |
WO2006078711A2 (en) | 2005-01-19 | 2006-07-27 | Mgi Gp, Inc. | Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting parp |
EP2319847A2 (en) | 2005-01-19 | 2011-05-11 | Eisai Inc. | Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP |
US20070117834A1 (en) * | 2005-10-04 | 2007-05-24 | David Hung | Methods and compositions for treating Huntington's disease |
US20070117835A1 (en) * | 2005-10-04 | 2007-05-24 | David Hung | Methods and compositions for treating Huntington's disease |
US9079852B2 (en) | 2007-06-29 | 2015-07-14 | Emory University | NMDA receptor antagonists for neuroprotection |
US8420680B2 (en) | 2007-06-29 | 2013-04-16 | Emory University | NMDA receptor antagonists for neuroprotection |
US9512485B2 (en) | 2010-08-24 | 2016-12-06 | Dana-Farber Cancer Institute. Inc. | Methods for predicting anti-cancer response |
EP3109325A1 (en) | 2010-08-24 | 2016-12-28 | Dana-Farber Cancer Institute, Inc. | Methods for predicting anti-cancer response |
WO2012027224A1 (en) | 2010-08-24 | 2012-03-01 | Dana-Farber Cancer Institute, Inc. | Methods for predicting anti-cancer response |
US10577662B2 (en) | 2010-08-24 | 2020-03-03 | Dana-Farber Cancer Institute, Inc. | Methods for predicting anti-cancer response |
US9574229B2 (en) | 2011-06-17 | 2017-02-21 | Myriad Genetics, Inc. | Methods and materials for assessing allelic imbalance |
US11225685B2 (en) | 2011-06-17 | 2022-01-18 | Myriad Genetics, Inc. | Methods and materials for assessing allelic imbalance |
US9279156B2 (en) | 2011-06-17 | 2016-03-08 | Myriad Genetics, Inc. | Methods and materials for assessing allelic imbalance |
US10626449B2 (en) | 2011-06-17 | 2020-04-21 | Myriad Genetics, Inc. | Methods and materials for assessing allelic imbalance |
WO2013055911A1 (en) | 2011-10-14 | 2013-04-18 | Dana-Farber Cancer Institute, Inc. | Znf365/zfp365 biomarker predictive of anti-cancer response |
US10612098B2 (en) | 2011-12-21 | 2020-04-07 | Myriad Genetics, Inc. | Methods and materials for assessing loss of heterozygosity |
US9388472B2 (en) | 2011-12-21 | 2016-07-12 | Myriad Genetics, Inc. | Methods and materials for assessing loss of heterozygosity |
US11299782B2 (en) | 2012-02-23 | 2022-04-12 | Children's Medical Center Corporation | Methods for predicting anti-cancer response |
EP3415915A1 (en) | 2012-02-23 | 2018-12-19 | Children's Medical Center Corporation | Methods for predicting anti-cancer response |
US10190160B2 (en) | 2012-02-23 | 2019-01-29 | Children's Medical Center Corporation | Methods for predicting anti-cancer response |
WO2013130347A1 (en) | 2012-02-23 | 2013-09-06 | The Children's Hospital Corporation | Methods for predicting anti-cancer response |
US11091808B2 (en) | 2012-06-07 | 2021-08-17 | Institut Curie | Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors |
US10308986B2 (en) | 2013-03-14 | 2019-06-04 | Children's Medical Center Corporation | Cancer diagnosis, treatment selection and treatment |
US11149316B2 (en) | 2013-12-09 | 2021-10-19 | Institut Curie | Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors |
WO2015109264A1 (en) | 2014-01-17 | 2015-07-23 | Cedars-Sinai Medical Center | Receptor targeting constructs and uses thereof |
EP3791889A1 (en) | 2014-01-17 | 2021-03-17 | Cedars-Sinai Medical Center | C-met receptor targeting constructs and uses thereof |
WO2015164743A2 (en) | 2014-04-24 | 2015-10-29 | Dana-Farber Cancer Institute, Inc. | Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response |
US10400287B2 (en) | 2014-08-15 | 2019-09-03 | Myriad Genetics, Inc. | Methods and materials for assessing homologous recombination deficiency |
US12221656B2 (en) | 2014-08-15 | 2025-02-11 | Myriad Genetics, Inc. | Methods and materials for assessing homologous recombination deficiency |
WO2016044707A1 (en) | 2014-09-18 | 2016-03-24 | Cedars-Sinai Medical Center | Compositions and methods for treating fibrosis |
WO2016057367A1 (en) | 2014-10-06 | 2016-04-14 | Dana-Farber Cancer Institute, Inc. | Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response |
WO2016073647A2 (en) | 2014-11-04 | 2016-05-12 | University Of Southern California | COMPOSITIONS AND METHODS FOR TREATING HIF-1alpha OVER-EXPRESSING CANCERS |
WO2016094273A1 (en) | 2014-12-08 | 2016-06-16 | Dana-Farber Cancer Institute, Inc. | Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents |
WO2016138574A1 (en) | 2015-03-02 | 2016-09-09 | Sinai Health System | Homologous recombination factors |
WO2016144673A1 (en) | 2015-03-06 | 2016-09-15 | Dana-Farber Cancer Institute, Inc. | Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers |
WO2017066561A2 (en) | 2015-10-16 | 2017-04-20 | President And Fellows Of Harvard College | Regulatory t cell pd-1 modulation for regulating t cell effector immune responses |
WO2018057618A1 (en) | 2016-09-20 | 2018-03-29 | Dana-Farber Cancer Institute, Inc. | Compositions and methods for identification, assessment, prevention, and treatment of aml using usp10 biomarkers and modulators |
WO2020033791A1 (en) | 2018-08-09 | 2020-02-13 | Verseau Therapeutics, Inc. | Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof |
WO2020223121A1 (en) | 2019-04-30 | 2020-11-05 | Dana-Farber Cancer Institute, Inc. | Methods for treating cancer using combinations of anti-cx3cr1 and immune checkpoint blockade agents |
WO2021150925A1 (en) | 2020-01-24 | 2021-07-29 | Dana-Farber Cancer Institute, Inc. | Uses of biomarkers for improving immunotherapy |
EP4343004A2 (en) | 2020-10-19 | 2024-03-27 | Dana-Farber Cancer Institute, Inc. | Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy |
WO2022086852A2 (en) | 2020-10-19 | 2022-04-28 | Dana-Farber Cancer Institute, Inc. | Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy |
WO2022104104A2 (en) | 2020-11-13 | 2022-05-19 | Dana-Farber Cancer Institute, Inc. | Personalized fusion cell vaccines |
WO2022159575A1 (en) | 2021-01-20 | 2022-07-28 | Bioentre Llc | Ctla4-binding proteins and methods of treating cancer |
WO2022159793A2 (en) | 2021-01-25 | 2022-07-28 | Dana-Farber Cancer Institute, Inc. | Methods and compositions for identifying neuroendocrine prostate cancer |
WO2022261183A2 (en) | 2021-06-08 | 2022-12-15 | Dana-Farber Cancer Institute, Inc. | Compositions and methods for treating and/or identifying an agent for treating intestinal cancers |
WO2023043888A1 (en) | 2021-09-15 | 2023-03-23 | Celloram Inc. | Method of treating cancer |
WO2023240287A1 (en) | 2022-06-10 | 2023-12-14 | Bioentre Llc | Combinations of ctla4 binding proteins and methods of treating cancer |
Also Published As
Publication number | Publication date |
---|---|
IL112496A0 (en) | 1995-03-30 |
WO1995020952A1 (en) | 1995-08-10 |
FI963040A0 (en) | 1996-08-01 |
ZA95861B (en) | 1996-08-02 |
NO963253D0 (en) | 1996-08-02 |
AU1695495A (en) | 1995-08-21 |
US5587384A (en) | 1996-12-24 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
USRE36397E (en) | Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity | |
JP2620413B2 (en) | Methods and compositions for inhibiting diseases associated with oxidative damage | |
Slemmer et al. | Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging | |
Zhang et al. | Nitric oxide activation of poly (ADP-ribose) synthetase in neurotoxicity | |
Weiss et al. | AMPA receptor activation potentiates zinc neurotoxicity | |
EP0581856B1 (en) | Method of preventing nmda receptor complex-mediated neuronal damage | |
CA2149924C (en) | Use of phenserine to treat cognitive disorders | |
Liu et al. | Prostaglandin release by spinal cord injury mediates production of hydroxyl radical, malondialdehyde and cell death: a site of the neuroprotective action of methylprednisolone | |
CZ20021904A3 (en) | Pyrrolidine acetamide derivative per se or in combination used for treating central nervous system disorders | |
US5025032A (en) | Phenyl butyl nitrone compositions and methods for treatment of oxidative tissue damage | |
Scott et al. | Role of poly (ADP-ribose) synthetase activation in the development of experimental allergic encephalomyelitis | |
Gupta et al. | Promising effects of emoxypine and its succinate derivative in the management of various diseases-with insights on recent patent applications | |
Braida et al. | Eptastigmine: ten years of pharmacology, toxicology, pharmacokinetic, and clinical studies | |
JP2022539185A (en) | Methylthioninium as a Cognitive Enhancer | |
Marques et al. | Muscle regeneration in dystrophic mdx mice is enhanced by isosorbide dinitrate | |
Smith et al. | Inhibition of the betaine-GABA transporter (mGAT2/BGT-1) modulates spontaneous electrographic bursting in the medial entorhinal cortex (mEC) | |
Giacobini | Modulation of brain acetylcholine levels with cholinesterase inhibitors as a treatment of Alzheimer disease | |
US6362160B1 (en) | Immunophilin-binding agents prevent glutamate neurotoxicity associated with vascular stroke and neurodegenerative diseases | |
WO2001022960A1 (en) | Treatment of carbon monoxide poisoning | |
Alonso et al. | Nitric-oxide-related and non-related mechanisms in the acetylcholine-evoked relaxations in cat femoral arteries | |
Schwarcz et al. | Quinolinic acid and kynurenic acid: glia-derived modulators of excitotoxic brain injury | |
Andres-Mach et al. | 7-Nitroindazole enhances dose-dependently the anticonvulsant activities of conventional antiepileptic drugs in the mouse maximal electroshock-induced seizure model | |
Díaz-Trelles et al. | Antihistamine terfenadine inhibits calcium influx, cGMP formation, and NMDA receptor-dependent neurotoxicity following activation of L-type voltage sensitive calcium channels | |
RU2314828C1 (en) | Means for treating spasmodic syndrome cases | |
Slemmer et al. | Do antioxidants and free radical scavengers still hold promise for the treatment of stroke, traumatic brain injury and aging? |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
FEPP | Fee payment procedure |
Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY |
|
FPAY | Fee payment |
Year of fee payment: 4 |
|
FPAY | Fee payment |
Year of fee payment: 8 |
|
FEPP | Fee payment procedure |
Free format text: PAT HOLDER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: LTOS); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY |
|
REFU | Refund |
Free format text: REFUND - PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: R1553); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY |
|
FPAY | Fee payment |
Year of fee payment: 12 |