NZ625066B2 - Metalloporphyrin neurological treatments - Google Patents
Metalloporphyrin neurological treatments Download PDFInfo
- Publication number
- NZ625066B2 NZ625066B2 NZ625066A NZ62506612A NZ625066B2 NZ 625066 B2 NZ625066 B2 NZ 625066B2 NZ 625066 A NZ625066 A NZ 625066A NZ 62506612 A NZ62506612 A NZ 62506612A NZ 625066 B2 NZ625066 B2 NZ 625066B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- agent
- unsubstituted
- use according
- substituted
- compound
- Prior art date
Links
- 238000011282 treatment Methods 0.000 title description 38
- 230000000926 neurological effect Effects 0.000 title description 2
- 150000001875 compounds Chemical class 0.000 claims abstract description 160
- 239000000126 substance Substances 0.000 claims abstract description 91
- 239000003795 chemical substances by application Substances 0.000 claims description 210
- 125000000217 alkyl group Chemical group 0.000 claims description 86
- 230000000694 effects Effects 0.000 claims description 69
- -1 2-chloroethyl ethyl Chemical group 0.000 claims description 56
- 206010010904 Convulsion Diseases 0.000 claims description 55
- 229910052751 metal Inorganic materials 0.000 claims description 48
- 239000002184 metal Substances 0.000 claims description 48
- 125000004404 heteroalkyl group Chemical group 0.000 claims description 44
- 230000000573 anti-seizure effect Effects 0.000 claims description 43
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 42
- 125000001072 heteroaryl group Chemical group 0.000 claims description 41
- 229940022698 acetylcholinesterase Drugs 0.000 claims description 39
- 108010022752 Acetylcholinesterase Proteins 0.000 claims description 38
- 210000005036 nerve Anatomy 0.000 claims description 37
- 239000000812 cholinergic antagonist Substances 0.000 claims description 33
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 claims description 32
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 claims description 32
- 229940121948 Muscarinic receptor antagonist Drugs 0.000 claims description 30
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 30
- 229910052739 hydrogen Inorganic materials 0.000 claims description 29
- 239000001257 hydrogen Substances 0.000 claims description 29
- 208000029028 brain injury Diseases 0.000 claims description 27
- 229910052736 halogen Inorganic materials 0.000 claims description 24
- 150000002367 halogens Chemical class 0.000 claims description 22
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 19
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 claims description 16
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 claims description 16
- 229910052802 copper Inorganic materials 0.000 claims description 16
- 239000010949 copper Substances 0.000 claims description 16
- 229910052742 iron Inorganic materials 0.000 claims description 16
- 229910052759 nickel Inorganic materials 0.000 claims description 16
- 229910052725 zinc Inorganic materials 0.000 claims description 16
- 239000011701 zinc Substances 0.000 claims description 16
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 15
- 239000010941 cobalt Substances 0.000 claims description 15
- 229910017052 cobalt Inorganic materials 0.000 claims description 15
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 claims description 15
- JBKPUQTUERUYQE-UHFFFAOYSA-O pralidoxime Chemical group C[N+]1=CC=CC=C1\C=N\O JBKPUQTUERUYQE-UHFFFAOYSA-O 0.000 claims description 15
- 239000011572 manganese Substances 0.000 claims description 14
- 239000000544 cholinesterase inhibitor Substances 0.000 claims description 13
- WPBNNNQJVZRUHP-UHFFFAOYSA-L manganese(2+);methyl n-[[2-(methoxycarbonylcarbamothioylamino)phenyl]carbamothioyl]carbamate;n-[2-(sulfidocarbothioylamino)ethyl]carbamodithioate Chemical compound [Mn+2].[S-]C(=S)NCCNC([S-])=S.COC(=O)NC(=S)NC1=CC=CC=C1NC(=S)NC(=O)OC WPBNNNQJVZRUHP-UHFFFAOYSA-L 0.000 claims description 13
- PWHULOQIROXLJO-UHFFFAOYSA-N Manganese Chemical compound [Mn] PWHULOQIROXLJO-UHFFFAOYSA-N 0.000 claims description 12
- 239000003814 drug Substances 0.000 claims description 12
- 229910052748 manganese Inorganic materials 0.000 claims description 12
- 125000006575 electron-withdrawing group Chemical group 0.000 claims description 11
- 150000002431 hydrogen Chemical class 0.000 claims description 10
- 229960003370 pralidoxime Drugs 0.000 claims description 10
- 230000006378 damage Effects 0.000 claims description 8
- QMGVPVSNSZLJIA-FVWCLLPLSA-N strychnine Chemical compound O([C@H]1CC(N([C@H]2[C@H]1[C@H]1C3)C=4C5=CC=CC=4)=O)CC=C1CN1[C@@H]3[C@]25CC1 QMGVPVSNSZLJIA-FVWCLLPLSA-N 0.000 claims description 8
- 125000000547 substituted alkyl group Chemical group 0.000 claims description 8
- 125000003107 substituted aryl group Chemical group 0.000 claims description 8
- 125000005346 substituted cycloalkyl group Chemical group 0.000 claims description 8
- AGGKEGLBGGJEBZ-UHFFFAOYSA-N tetramethylenedisulfotetramine Chemical compound C1N(S2(=O)=O)CN3S(=O)(=O)N1CN2C3 AGGKEGLBGGJEBZ-UHFFFAOYSA-N 0.000 claims description 8
- 208000027418 Wounds and injury Diseases 0.000 claims description 7
- 239000008280 blood Substances 0.000 claims description 7
- 210000004369 blood Anatomy 0.000 claims description 7
- 208000014674 injury Diseases 0.000 claims description 7
- 238000004519 manufacturing process Methods 0.000 claims description 7
- DYAHQFWOVKZOOW-UHFFFAOYSA-N Sarin Chemical group CC(C)OP(C)(F)=O DYAHQFWOVKZOOW-UHFFFAOYSA-N 0.000 claims description 6
- 230000007171 neuropathology Effects 0.000 claims description 6
- HJTAZXHBEBIQQX-UHFFFAOYSA-N 1,5-bis(chloromethyl)naphthalene Chemical compound C1=CC=C2C(CCl)=CC=CC2=C1CCl HJTAZXHBEBIQQX-UHFFFAOYSA-N 0.000 claims description 5
- XFXPMWWXUTWYJX-UHFFFAOYSA-N Cyanide Chemical compound N#[C-] XFXPMWWXUTWYJX-UHFFFAOYSA-N 0.000 claims description 5
- QMGVPVSNSZLJIA-UHFFFAOYSA-N Nux Vomica Natural products C1C2C3C4N(C=5C6=CC=CC=5)C(=O)CC3OCC=C2CN2C1C46CC2 QMGVPVSNSZLJIA-UHFFFAOYSA-N 0.000 claims description 5
- GOLCXWYRSKYTSP-UHFFFAOYSA-N arsenic trioxide Inorganic materials O1[As]2O[As]1O2 GOLCXWYRSKYTSP-UHFFFAOYSA-N 0.000 claims description 5
- 229940049706 benzodiazepine Drugs 0.000 claims description 5
- QKSKPIVNLNLAAV-UHFFFAOYSA-N bis(2-chloroethyl) sulfide Chemical compound ClCCSCCCl QKSKPIVNLNLAAV-UHFFFAOYSA-N 0.000 claims description 5
- 239000003445 gaba agent Substances 0.000 claims description 5
- 230000002401 inhibitory effect Effects 0.000 claims description 5
- JGFYQVQAXANWJU-UHFFFAOYSA-M sodium fluoroacetate Chemical compound [Na+].[O-]C(=O)CF JGFYQVQAXANWJU-UHFFFAOYSA-M 0.000 claims description 5
- KZBUYRJDOAKODT-UHFFFAOYSA-N Chlorine Chemical compound ClCl KZBUYRJDOAKODT-UHFFFAOYSA-N 0.000 claims description 4
- 241001279009 Strychnos toxifera Species 0.000 claims description 4
- LCCNCVORNKJIRZ-UHFFFAOYSA-N parathion Chemical compound CCOP(=S)(OCC)OC1=CC=C([N+]([O-])=O)C=C1 LCCNCVORNKJIRZ-UHFFFAOYSA-N 0.000 claims description 4
- 229960005453 strychnine Drugs 0.000 claims description 4
- YGYAWVDWMABLBF-UHFFFAOYSA-N Phosgene Chemical compound ClC(Cl)=O YGYAWVDWMABLBF-UHFFFAOYSA-N 0.000 claims description 3
- QGLZXHRNAYXIBU-WEVVVXLNSA-N aldicarb Chemical compound CNC(=O)O\N=C\C(C)(C)SC QGLZXHRNAYXIBU-WEVVVXLNSA-N 0.000 claims description 2
- 125000004400 (C1-C12) alkyl group Chemical group 0.000 claims 6
- 102000012440 Acetylcholinesterase Human genes 0.000 claims 3
- SVUOLADPCWQTTE-UHFFFAOYSA-N 1h-1,2-benzodiazepine Chemical compound N1N=CC=CC2=CC=CC=C12 SVUOLADPCWQTTE-UHFFFAOYSA-N 0.000 claims 1
- 125000001340 2-chloroethyl group Chemical group [H]C([H])(Cl)C([H])([H])* 0.000 claims 1
- 102100026459 POU domain, class 3, transcription factor 2 Human genes 0.000 claims 1
- 101710133394 POU domain, class 3, transcription factor 2 Proteins 0.000 claims 1
- 230000001149 cognitive effect Effects 0.000 claims 1
- LJSQFQKUNVCTIA-UHFFFAOYSA-N diethyl sulfide Chemical compound CCSCC LJSQFQKUNVCTIA-UHFFFAOYSA-N 0.000 claims 1
- 230000004064 dysfunction Effects 0.000 claims 1
- 229960001416 pilocarpine Drugs 0.000 description 139
- QCHFTSOMWOSFHM-UHFFFAOYSA-N SJ000285536 Natural products C1OC(=O)C(CC)C1CC1=CN=CN1C QCHFTSOMWOSFHM-UHFFFAOYSA-N 0.000 description 127
- XMYHQLIAYLLEKT-UHFFFAOYSA-I manganese(3+);5,10,15,20-tetrakis(1,3-diethylimidazol-1-ium-2-yl)porphyrin-22,24-diide;pentachloride Chemical compound [Cl-].[Cl-].[Cl-].[Cl-].[Cl-].[Mn+3].CCN1C=C[N+](CC)=C1C(C1=CC=C([N-]1)C(C1=[N+](C=CN1CC)CC)=C1C=CC(=N1)C(C1=[N+](C=CN1CC)CC)=C1C=CC([N-]1)=C1C2=[N+](C=CN2CC)CC)=C2N=C1C=C2 XMYHQLIAYLLEKT-UHFFFAOYSA-I 0.000 description 125
- QCHFTSOMWOSFHM-WPRPVWTQSA-N (+)-Pilocarpine Chemical compound C1OC(=O)[C@@H](CC)[C@H]1CC1=CN=CN1C QCHFTSOMWOSFHM-WPRPVWTQSA-N 0.000 description 124
- 238000000034 method Methods 0.000 description 92
- 241000700159 Rattus Species 0.000 description 63
- 125000001424 substituent group Chemical group 0.000 description 56
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 46
- 125000003118 aryl group Chemical group 0.000 description 40
- 125000000753 cycloalkyl group Chemical group 0.000 description 36
- 102100033639 Acetylcholinesterase Human genes 0.000 description 35
- 229930003347 Atropine Natural products 0.000 description 33
- RKUNBYITZUJHSG-UHFFFAOYSA-N Hyosciamin-hydrochlorid Natural products CN1C(C2)CCC1CC2OC(=O)C(CO)C1=CC=CC=C1 RKUNBYITZUJHSG-UHFFFAOYSA-N 0.000 description 33
- 229960000396 atropine Drugs 0.000 description 33
- RKUNBYITZUJHSG-SPUOUPEWSA-N atropine Chemical compound O([C@H]1C[C@H]2CC[C@@H](C1)N2C)C(=O)C(CO)C1=CC=CC=C1 RKUNBYITZUJHSG-SPUOUPEWSA-N 0.000 description 33
- 230000036542 oxidative stress Effects 0.000 description 33
- 229960003529 diazepam Drugs 0.000 description 26
- AAOVKJBEBIDNHE-UHFFFAOYSA-N diazepam Chemical compound N=1CC(=O)N(C)C2=CC=C(Cl)C=C2C=1C1=CC=CC=C1 AAOVKJBEBIDNHE-UHFFFAOYSA-N 0.000 description 26
- 238000007920 subcutaneous administration Methods 0.000 description 26
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 23
- 230000008901 benefit Effects 0.000 description 23
- 108010053070 Glutathione Disulfide Proteins 0.000 description 22
- 229960003180 glutathione Drugs 0.000 description 22
- YPZRWBKMTBYPTK-BJDJZHNGSA-N glutathione disulfide Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@H](C(=O)NCC(O)=O)CSSC[C@@H](C(=O)NCC(O)=O)NC(=O)CC[C@H](N)C(O)=O YPZRWBKMTBYPTK-BJDJZHNGSA-N 0.000 description 22
- 210000001320 hippocampus Anatomy 0.000 description 22
- 239000011780 sodium chloride Substances 0.000 description 22
- 230000002195 synergetic effect Effects 0.000 description 22
- 210000004556 brain Anatomy 0.000 description 21
- 239000000203 mixture Substances 0.000 description 21
- 125000004093 cyano group Chemical group *C#N 0.000 description 19
- 230000004112 neuroprotection Effects 0.000 description 19
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 18
- 230000001225 therapeutic effect Effects 0.000 description 18
- 230000008499 blood brain barrier function Effects 0.000 description 17
- 210000001218 blood-brain barrier Anatomy 0.000 description 17
- 230000003197 catalytic effect Effects 0.000 description 16
- 230000000971 hippocampal effect Effects 0.000 description 16
- 238000002347 injection Methods 0.000 description 16
- 239000007924 injection Substances 0.000 description 16
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 15
- 230000003078 antioxidant effect Effects 0.000 description 15
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 15
- VLSMHEGGTFMBBZ-OOZYFLPDSA-M Kainate Chemical compound CC(=C)[C@H]1C[NH2+][C@H](C([O-])=O)[C@H]1CC([O-])=O VLSMHEGGTFMBBZ-OOZYFLPDSA-M 0.000 description 14
- 230000000324 neuroprotective effect Effects 0.000 description 14
- 238000004458 analytical method Methods 0.000 description 13
- 239000003963 antioxidant agent Substances 0.000 description 13
- 208000005809 status epilepticus Diseases 0.000 description 13
- ZETXHVRPKUXQIT-UHFFFAOYSA-N 1-methyl-4-phenylpiperidine Chemical compound C1CN(C)CCC1C1=CC=CC=C1 ZETXHVRPKUXQIT-UHFFFAOYSA-N 0.000 description 12
- JVJFIQYAHPMBBX-UHFFFAOYSA-N 4-hydroxynonenal Chemical compound CCCCCC(O)C=CC=O JVJFIQYAHPMBBX-UHFFFAOYSA-N 0.000 description 12
- 235000006708 antioxidants Nutrition 0.000 description 12
- 201000010099 disease Diseases 0.000 description 12
- 238000007912 intraperitoneal administration Methods 0.000 description 12
- 150000003254 radicals Chemical class 0.000 description 12
- 239000003642 reactive oxygen metabolite Substances 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 11
- 210000004027 cell Anatomy 0.000 description 11
- FBTSQILOGYXGMD-LURJTMIESA-N 3-nitro-L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C([N+]([O-])=O)=C1 FBTSQILOGYXGMD-LURJTMIESA-N 0.000 description 10
- 230000001078 anti-cholinergic effect Effects 0.000 description 10
- 150000002500 ions Chemical class 0.000 description 10
- DDLIGBOFAVUZHB-UHFFFAOYSA-N midazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NC=C2CN=C1C1=CC=CC=C1F DDLIGBOFAVUZHB-UHFFFAOYSA-N 0.000 description 10
- 229960003793 midazolam Drugs 0.000 description 10
- 238000001543 one-way ANOVA Methods 0.000 description 10
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 10
- 206010018341 Gliosis Diseases 0.000 description 9
- 230000003247 decreasing effect Effects 0.000 description 9
- 230000007387 gliosis Effects 0.000 description 9
- 125000005842 heteroatom Chemical group 0.000 description 9
- 230000002438 mitochondrial effect Effects 0.000 description 9
- 229910052760 oxygen Inorganic materials 0.000 description 9
- 239000008194 pharmaceutical composition Substances 0.000 description 9
- 239000013543 active substance Substances 0.000 description 8
- 125000004429 atom Chemical group 0.000 description 8
- 229940079593 drug Drugs 0.000 description 8
- 230000016273 neuron death Effects 0.000 description 8
- 229910052757 nitrogen Inorganic materials 0.000 description 8
- 230000008599 nitrosative stress Effects 0.000 description 8
- 208000024891 symptom Diseases 0.000 description 8
- 210000003568 synaptosome Anatomy 0.000 description 8
- 230000002503 metabolic effect Effects 0.000 description 7
- 230000001537 neural effect Effects 0.000 description 7
- 210000001519 tissue Anatomy 0.000 description 7
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 6
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 6
- 102000019197 Superoxide Dismutase Human genes 0.000 description 6
- 108010012715 Superoxide dismutase Proteins 0.000 description 6
- OIPILFWXSMYKGL-UHFFFAOYSA-N acetylcholine Chemical compound CC(=O)OCC[N+](C)(C)C OIPILFWXSMYKGL-UHFFFAOYSA-N 0.000 description 6
- 229960004373 acetylcholine Drugs 0.000 description 6
- 125000002947 alkylene group Chemical group 0.000 description 6
- 125000004432 carbon atom Chemical group C* 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 230000018109 developmental process Effects 0.000 description 6
- 238000007918 intramuscular administration Methods 0.000 description 6
- 210000002569 neuron Anatomy 0.000 description 6
- 239000001301 oxygen Substances 0.000 description 6
- 230000036284 oxygen consumption Effects 0.000 description 6
- 239000002574 poison Substances 0.000 description 6
- 231100000614 poison Toxicity 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 230000035882 stress Effects 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 5
- 229910052799 carbon Inorganic materials 0.000 description 5
- 230000030833 cell death Effects 0.000 description 5
- 210000003169 central nervous system Anatomy 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 230000004770 neurodegeneration Effects 0.000 description 5
- 239000000575 pesticide Substances 0.000 description 5
- 230000003389 potentiating effect Effects 0.000 description 5
- 230000000069 prophylactic effect Effects 0.000 description 5
- 230000001681 protective effect Effects 0.000 description 5
- 238000004445 quantitative analysis Methods 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 108010024636 Glutathione Proteins 0.000 description 4
- LEVWYRKDKASIDU-IMJSIDKUSA-N L-cystine Chemical compound [O-]C(=O)[C@@H]([NH3+])CSSC[C@H]([NH3+])C([O-])=O LEVWYRKDKASIDU-IMJSIDKUSA-N 0.000 description 4
- 208000021966 Motor seizure Diseases 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 102000014415 Muscarinic acetylcholine receptor Human genes 0.000 description 4
- 108050003473 Muscarinic acetylcholine receptor Proteins 0.000 description 4
- 206010029350 Neurotoxicity Diseases 0.000 description 4
- OUUQCZGPVNCOIJ-UHFFFAOYSA-M Superoxide Chemical compound [O-][O] OUUQCZGPVNCOIJ-UHFFFAOYSA-M 0.000 description 4
- 206010044221 Toxic encephalopathy Diseases 0.000 description 4
- 230000006727 cell loss Effects 0.000 description 4
- 239000003179 convulsant agent Substances 0.000 description 4
- 125000004122 cyclic group Chemical group 0.000 description 4
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 4
- 235000018417 cysteine Nutrition 0.000 description 4
- 229960003067 cystine Drugs 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 125000004474 heteroalkylene group Chemical group 0.000 description 4
- 125000005549 heteroarylene group Chemical group 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 230000003818 metabolic dysfunction Effects 0.000 description 4
- 230000004065 mitochondrial dysfunction Effects 0.000 description 4
- 230000004898 mitochondrial function Effects 0.000 description 4
- 230000007135 neurotoxicity Effects 0.000 description 4
- 231100000228 neurotoxicity Toxicity 0.000 description 4
- 230000004792 oxidative damage Effects 0.000 description 4
- CMFNMSMUKZHDEY-UHFFFAOYSA-M peroxynitrite Chemical compound [O-]ON=O CMFNMSMUKZHDEY-UHFFFAOYSA-M 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 229910052698 phosphorus Inorganic materials 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 238000011301 standard therapy Methods 0.000 description 4
- 229910052717 sulfur Inorganic materials 0.000 description 4
- 238000007910 systemic administration Methods 0.000 description 4
- 125000003837 (C1-C20) alkyl group Chemical group 0.000 description 3
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 description 3
- 125000004209 (C1-C8) alkyl group Chemical group 0.000 description 3
- 102100040958 Aconitate hydratase, mitochondrial Human genes 0.000 description 3
- 238000011740 C57BL/6 mouse Methods 0.000 description 3
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 241000347881 Kadua laxiflora Species 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 239000000729 antidote Substances 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000003542 behavioural effect Effects 0.000 description 3
- 230000005779 cell damage Effects 0.000 description 3
- 208000037887 cell injury Diseases 0.000 description 3
- 231100000481 chemical toxicant Toxicity 0.000 description 3
- 239000000460 chlorine Substances 0.000 description 3
- 229910052801 chlorine Inorganic materials 0.000 description 3
- 238000011260 co-administration Methods 0.000 description 3
- 208000010877 cognitive disease Diseases 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- 125000001153 fluoro group Chemical group F* 0.000 description 3
- 230000004907 flux Effects 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 210000005153 frontal cortex Anatomy 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 230000002414 glycolytic effect Effects 0.000 description 3
- 230000004054 inflammatory process Effects 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 239000010977 jade Substances 0.000 description 3
- 125000005647 linker group Chemical group 0.000 description 3
- 230000003859 lipid peroxidation Effects 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 125000004433 nitrogen atom Chemical group N* 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000007911 parenteral administration Methods 0.000 description 3
- 230000035699 permeability Effects 0.000 description 3
- 238000002203 pretreatment Methods 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000000241 respiratory effect Effects 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 229910052710 silicon Inorganic materials 0.000 description 3
- 238000013222 sprague-dawley male rat Methods 0.000 description 3
- 238000010254 subcutaneous injection Methods 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 125000000008 (C1-C10) alkyl group Chemical group 0.000 description 2
- 125000006552 (C3-C8) cycloalkyl group Chemical group 0.000 description 2
- GVJHHUAWPYXKBD-UHFFFAOYSA-N (±)-α-Tocopherol Chemical compound OC1=C(C)C(C)=C2OC(CCCC(C)CCCC(C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-UHFFFAOYSA-N 0.000 description 2
- GBNVXYXIRHSYEG-UHFFFAOYSA-N 1-chloro-2-ethylsulfanylethane Chemical compound CCSCCCl GBNVXYXIRHSYEG-UHFFFAOYSA-N 0.000 description 2
- 229930000680 A04AD01 - Scopolamine Natural products 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 2
- 108010009924 Aconitate hydratase Proteins 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 2
- 102000016938 Catalase Human genes 0.000 description 2
- 108010053835 Catalase Proteins 0.000 description 2
- 206010009346 Clonus Diseases 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108010036781 Fumarate Hydratase Proteins 0.000 description 2
- 102100036160 Fumarate hydratase, mitochondrial Human genes 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- 241001559542 Hippocampus hippocampus Species 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- STECJAGHUSJQJN-GAUPFVANSA-N Hyoscine Natural products C1([C@H](CO)C(=O)OC2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-GAUPFVANSA-N 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- STECJAGHUSJQJN-UHFFFAOYSA-N N-Methyl-scopolamin Natural products C1C(C2C3O2)N(C)C3CC1OC(=O)C(CO)C1=CC=CC=C1 STECJAGHUSJQJN-UHFFFAOYSA-N 0.000 description 2
- 229910019142 PO4 Inorganic materials 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 2
- NBBJYMSMWIIQGU-UHFFFAOYSA-N Propionic aldehyde Chemical compound CCC=O NBBJYMSMWIIQGU-UHFFFAOYSA-N 0.000 description 2
- UCKMPCXJQFINFW-UHFFFAOYSA-N Sulphide Chemical compound [S-2] UCKMPCXJQFINFW-UHFFFAOYSA-N 0.000 description 2
- 102220470415 Thymosin beta-10_R27A_mutation Human genes 0.000 description 2
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 2
- 230000009056 active transport Effects 0.000 description 2
- 230000001154 acute effect Effects 0.000 description 2
- 125000002252 acyl group Chemical group 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 150000001298 alcohols Chemical class 0.000 description 2
- 125000003545 alkoxy group Chemical group 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 150000001449 anionic compounds Chemical class 0.000 description 2
- 229940065524 anticholinergics inhalants for obstructive airway diseases Drugs 0.000 description 2
- 229940055075 anticholinesterase parasympathomimetics Drugs 0.000 description 2
- 210000001130 astrocyte Anatomy 0.000 description 2
- 150000001557 benzodiazepines Chemical class 0.000 description 2
- 125000003310 benzodiazepinyl group Chemical group N1N=C(C=CC2=C1C=CC=C2)* 0.000 description 2
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Chemical compound BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 2
- 229910052794 bromium Inorganic materials 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 150000001721 carbon Chemical group 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 239000002575 chemical warfare agent Substances 0.000 description 2
- 210000000038 chest Anatomy 0.000 description 2
- 230000001713 cholinergic effect Effects 0.000 description 2
- 239000002779 cholinesterase reactivator Substances 0.000 description 2
- 230000002301 combined effect Effects 0.000 description 2
- 230000003111 delayed effect Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 235000014113 dietary fatty acids Nutrition 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 238000007876 drug discovery Methods 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- 230000008029 eradication Effects 0.000 description 2
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 2
- 239000000194 fatty acid Substances 0.000 description 2
- 229930195729 fatty acid Natural products 0.000 description 2
- 229910052731 fluorine Inorganic materials 0.000 description 2
- 210000003194 forelimb Anatomy 0.000 description 2
- 125000001188 haloalkyl group Chemical group 0.000 description 2
- 125000005843 halogen group Chemical group 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 230000001744 histochemical effect Effects 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- ZCYVEMRRCGMTRW-YPZZEJLDSA-N iodine-125 Chemical compound [125I] ZCYVEMRRCGMTRW-YPZZEJLDSA-N 0.000 description 2
- 150000002611 lead compounds Chemical class 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- RLSSMJSEOOYNOY-UHFFFAOYSA-N m-cresol Chemical compound CC1=CC=CC(O)=C1 RLSSMJSEOOYNOY-UHFFFAOYSA-N 0.000 description 2
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 2
- LZCOQTDXKCNBEE-IKIFYQGPSA-N methscopolamine Chemical compound C1([C@@H](CO)C(=O)O[C@H]2C[C@@H]3[N+]([C@H](C2)[C@@H]2[C@H]3O2)(C)C)=CC=CC=C1 LZCOQTDXKCNBEE-IKIFYQGPSA-N 0.000 description 2
- 229960001383 methylscopolamine Drugs 0.000 description 2
- 238000012544 monitoring process Methods 0.000 description 2
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 239000002858 neurotransmitter agent Substances 0.000 description 2
- YPZRWBKMTBYPTK-UHFFFAOYSA-N oxidized gamma-L-glutamyl-L-cysteinylglycine Natural products OC(=O)C(N)CCC(=O)NC(C(=O)NCC(O)=O)CSSCC(C(=O)NCC(O)=O)NC(=O)CCC(N)C(O)=O YPZRWBKMTBYPTK-UHFFFAOYSA-N 0.000 description 2
- 150000002978 peroxides Chemical class 0.000 description 2
- 229940097156 peroxyl Drugs 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 2
- 239000010452 phosphate Substances 0.000 description 2
- 239000011574 phosphorus Substances 0.000 description 2
- 230000004962 physiological condition Effects 0.000 description 2
- 231100000572 poisoning Toxicity 0.000 description 2
- 230000000607 poisoning effect Effects 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 230000004224 protection Effects 0.000 description 2
- 230000002685 pulmonary effect Effects 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 238000011552 rat model Methods 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000029058 respiratory gaseous exchange Effects 0.000 description 2
- 230000008458 response to injury Effects 0.000 description 2
- STECJAGHUSJQJN-FWXGHANASA-N scopolamine Chemical compound C1([C@@H](CO)C(=O)O[C@H]2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-FWXGHANASA-N 0.000 description 2
- 229960002646 scopolamine Drugs 0.000 description 2
- 238000001228 spectrum Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000007929 subcutaneous injection Substances 0.000 description 2
- 125000004434 sulfur atom Chemical group 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 229910052722 tritium Inorganic materials 0.000 description 2
- 230000007306 turnover Effects 0.000 description 2
- 238000007492 two-way ANOVA Methods 0.000 description 2
- 125000004417 unsaturated alkyl group Chemical group 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- MNULEGDCPYONBU-WMBHJXFZSA-N (1r,4s,5e,5'r,6'r,7e,10s,11r,12s,14r,15s,16s,18r,19s,20r,21e,25s,26r,27s,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trio Polymers O([C@@H]1CC[C@@H](/C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)[C@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)O[C@H]([C@H]2C)[C@H]1C)CC)[C@]12CC[C@@H](C)[C@@H](C[C@H](C)O)O1 MNULEGDCPYONBU-WMBHJXFZSA-N 0.000 description 1
- MNULEGDCPYONBU-DJRUDOHVSA-N (1s,4r,5z,5'r,6'r,7e,10s,11r,12s,14r,15s,18r,19r,20s,21e,26r,27s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers O([C@H]1CC[C@H](\C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)C(C)C(=O)[C@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)OC([C@H]2C)C1C)CC)[C@]12CC[C@@H](C)[C@@H](CC(C)O)O1 MNULEGDCPYONBU-DJRUDOHVSA-N 0.000 description 1
- DIWRORZWFLOCLC-HNNXBMFYSA-N (3s)-7-chloro-5-(2-chlorophenyl)-3-hydroxy-1,3-dihydro-1,4-benzodiazepin-2-one Chemical compound N([C@H](C(NC1=CC=C(Cl)C=C11)=O)O)=C1C1=CC=CC=C1Cl DIWRORZWFLOCLC-HNNXBMFYSA-N 0.000 description 1
- MNULEGDCPYONBU-YNZHUHFTSA-N (4Z,18Z,20Z)-22-ethyl-7,11,14,15-tetrahydroxy-6'-(2-hydroxypropyl)-5',6,8,10,12,14,16,28,29-nonamethylspiro[2,26-dioxabicyclo[23.3.1]nonacosa-4,18,20-triene-27,2'-oxane]-3,9,13-trione Polymers CC1C(C2C)OC(=O)\C=C/C(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)C\C=C/C=C\C(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-YNZHUHFTSA-N 0.000 description 1
- MNULEGDCPYONBU-VVXVDZGXSA-N (5e,5'r,7e,10s,11r,12s,14s,15r,16r,18r,19s,20r,21e,26r,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers C([C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)OC([C@H]1C)[C@H]2C)\C=C\C=C\C(CC)CCC2OC21CC[C@@H](C)C(C[C@H](C)O)O2 MNULEGDCPYONBU-VVXVDZGXSA-N 0.000 description 1
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 description 1
- 125000004769 (C1-C4) alkylsulfonyl group Chemical group 0.000 description 1
- 125000000229 (C1-C4)alkoxy group Chemical group 0.000 description 1
- 125000006527 (C1-C5) alkyl group Chemical group 0.000 description 1
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 description 1
- UCTWMZQNUQWSLP-VIFPVBQESA-N (R)-adrenaline Chemical compound CNC[C@H](O)C1=CC=C(O)C(O)=C1 UCTWMZQNUQWSLP-VIFPVBQESA-N 0.000 description 1
- 229930182837 (R)-adrenaline Natural products 0.000 description 1
- QELSIJXWEROXOE-IGUOPLJTSA-N 1-[[(2e)-2-(nitrosomethylidene)pyridin-1-yl]methoxymethyl]pyridin-1-ium-4-carboximidate;dihydrochloride Chemical compound [Cl-].[Cl-].C1=CC(C(=O)N)=CC=[N+]1COCN1C(=C/[NH+]=O)/C=CC=C1 QELSIJXWEROXOE-IGUOPLJTSA-N 0.000 description 1
- 125000004206 2,2,2-trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- 125000004174 2-benzimidazolyl group Chemical group [H]N1C(*)=NC2=C([H])C([H])=C([H])C([H])=C12 0.000 description 1
- 125000002941 2-furyl group Chemical group O1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 1
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 125000000389 2-pyrrolyl group Chemical group [H]N1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- 125000000175 2-thienyl group Chemical group S1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- NCAJWYASAWUEBY-UHFFFAOYSA-N 3-[20-(2-carboxyethyl)-9,14-diethyl-5,10,15,19-tetramethyl-21,22,23,24-tetraazapentacyclo[16.2.1.1^{3,6}.1^{8,11}.1^{13,16}]tetracosa-1(21),2,4,6(24),7,9,11,13,15,17,19-undecaen-4-yl]propanoic acid Chemical compound N1C2=C(C)C(CC)=C1C=C(N1)C(C)=C(CC)C1=CC(C(C)=C1CCC(O)=O)=NC1=CC(C(CCC(O)=O)=C1C)=NC1=C2 NCAJWYASAWUEBY-UHFFFAOYSA-N 0.000 description 1
- 125000000474 3-butynyl group Chemical group [H]C#CC([H])([H])C([H])([H])* 0.000 description 1
- 125000003682 3-furyl group Chemical group O1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 description 1
- 125000001397 3-pyrrolyl group Chemical group [H]N1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- MNULEGDCPYONBU-UHFFFAOYSA-N 4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers CC1C(C2C)OC(=O)C=CC(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)CC=CC=CC(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-UHFFFAOYSA-N 0.000 description 1
- 125000000339 4-pyridyl group Chemical group N1=C([H])C([H])=C([*])C([H])=C1[H] 0.000 description 1
- KDDQRKBRJSGMQE-UHFFFAOYSA-N 4-thiazolyl Chemical group [C]1=CSC=N1 KDDQRKBRJSGMQE-UHFFFAOYSA-N 0.000 description 1
- CWDWFSXUQODZGW-UHFFFAOYSA-N 5-thiazolyl Chemical group [C]1=CN=CS1 CWDWFSXUQODZGW-UHFFFAOYSA-N 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical group [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- 229940100578 Acetylcholinesterase inhibitor Drugs 0.000 description 1
- ORILYTVJVMAKLC-UHFFFAOYSA-N Adamantane Natural products C1C(C2)CC3CC1CC2C3 ORILYTVJVMAKLC-UHFFFAOYSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 229920000856 Amylose Polymers 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 1
- 206010006482 Bronchospasm Diseases 0.000 description 1
- APPSHIJNEBEURU-UHFFFAOYSA-O C12=CC=C(N1)C=C1C=CC(=N1)C=C1C=CC(N1)=CC=1C=CC(N1)=C2.C(C)C=2[NH+](C=CN2)CC Chemical compound C12=CC=C(N1)C=C1C=CC(=N1)C=C1C=CC(N1)=CC=1C=CC(N1)=C2.C(C)C=2[NH+](C=CN2)CC APPSHIJNEBEURU-UHFFFAOYSA-O 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- BMZRVOVNUMQTIN-UHFFFAOYSA-N Carbonyl Cyanide para-Trifluoromethoxyphenylhydrazone Chemical compound FC(F)(F)OC1=CC=C(NN=C(C#N)C#N)C=C1 BMZRVOVNUMQTIN-UHFFFAOYSA-N 0.000 description 1
- 108010062745 Chloride Channels Proteins 0.000 description 1
- 102000011045 Chloride Channels Human genes 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- DBAKFASWICGISY-BTJKTKAUSA-N Chlorpheniramine maleate Chemical compound OC(=O)\C=C/C(O)=O.C=1C=CC=NC=1C(CCN(C)C)C1=CC=C(Cl)C=C1 DBAKFASWICGISY-BTJKTKAUSA-N 0.000 description 1
- 102000003914 Cholinesterases Human genes 0.000 description 1
- 108090000322 Cholinesterases Proteins 0.000 description 1
- 206010009192 Circulatory collapse Diseases 0.000 description 1
- 206010010071 Coma Diseases 0.000 description 1
- 229910017518 Cu Zn Inorganic materials 0.000 description 1
- 229910017752 Cu-Zn Inorganic materials 0.000 description 1
- 229910017943 Cu—Zn Inorganic materials 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- XIQVNETUBQGFHX-UHFFFAOYSA-N Ditropan Chemical compound C=1C=CC=CC=1C(O)(C(=O)OCC#CCN(CC)CC)C1CCCCC1 XIQVNETUBQGFHX-UHFFFAOYSA-N 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 1
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 102100039289 Glial fibrillary acidic protein Human genes 0.000 description 1
- 101710193519 Glial fibrillary acidic protein Proteins 0.000 description 1
- VPNYRYCIDCJBOM-UHFFFAOYSA-M Glycopyrronium bromide Chemical compound [Br-].C1[N+](C)(C)CCC1OC(=O)C(O)(C=1C=CC=CC=1)C1CCCC1 VPNYRYCIDCJBOM-UHFFFAOYSA-M 0.000 description 1
- 238000010268 HPLC based assay Methods 0.000 description 1
- 101000965314 Homo sapiens Aconitate hydratase, mitochondrial Proteins 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-L L-tartrate(2-) Chemical compound [O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O FEWJPZIEWOKRBE-JCYAYHJZSA-L 0.000 description 1
- MKXZASYAUGDDCJ-SZMVWBNQSA-N LSM-2525 Chemical compound C1CCC[C@H]2[C@@]3([H])N(C)CC[C@]21C1=CC(OC)=CC=C1C3 MKXZASYAUGDDCJ-SZMVWBNQSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- 102000006404 Mitochondrial Proteins Human genes 0.000 description 1
- 108010058682 Mitochondrial Proteins Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 102000019315 Nicotinic acetylcholine receptors Human genes 0.000 description 1
- 108050006807 Nicotinic acetylcholine receptors Proteins 0.000 description 1
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 1
- 206010033799 Paralysis Diseases 0.000 description 1
- 208000027089 Parkinsonian disease Diseases 0.000 description 1
- 206010034010 Parkinsonism Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 229910007161 Si(CH3)3 Inorganic materials 0.000 description 1
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 208000003443 Unconsciousness Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 229930003427 Vitamin E Natural products 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 210000001642 activated microglia Anatomy 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 125000003342 alkenyl group Chemical group 0.000 description 1
- 125000004390 alkyl sulfonyl group Chemical group 0.000 description 1
- 125000005237 alkyleneamino group Chemical group 0.000 description 1
- 125000005530 alkylenedioxy group Chemical group 0.000 description 1
- 125000005529 alkyleneoxy group Chemical group 0.000 description 1
- 125000000304 alkynyl group Chemical group 0.000 description 1
- VLSMHEGGTFMBBZ-UHFFFAOYSA-N alpha-Kainic acid Natural products CC(=C)C1CNC(C(O)=O)C1CC(O)=O VLSMHEGGTFMBBZ-UHFFFAOYSA-N 0.000 description 1
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 230000001152 anti-nicotinic effect Effects 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 125000003710 aryl alkyl group Chemical group 0.000 description 1
- 125000000732 arylene group Chemical group 0.000 description 1
- 229940072107 ascorbate Drugs 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- QELSIJXWEROXOE-UHFFFAOYSA-N asoxime chloride Chemical compound [Cl-].[Cl-].C1=CC(C(=O)N)=CC=[N+]1COC[N+]1=CC=CC=C1\C=N\O QELSIJXWEROXOE-UHFFFAOYSA-N 0.000 description 1
- 230000003140 astrocytic effect Effects 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 210000003403 autonomic nervous system Anatomy 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- 210000002469 basement membrane Anatomy 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 229960001081 benzatropine Drugs 0.000 description 1
- GIJXKZJWITVLHI-PMOLBWCYSA-N benzatropine Chemical compound O([C@H]1C[C@H]2CC[C@@H](C1)N2C)C(C=1C=CC=CC=1)C1=CC=CC=C1 GIJXKZJWITVLHI-PMOLBWCYSA-N 0.000 description 1
- 230000002715 bioenergetic effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 125000000319 biphenyl-4-yl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1C1=C([H])C([H])=C([*])C([H])=C1[H] 0.000 description 1
- 230000036471 bradycardia Effects 0.000 description 1
- 208000006218 bradycardia Diseases 0.000 description 1
- 230000007885 bronchoconstriction Effects 0.000 description 1
- SNPPWIUOZRMYNY-UHFFFAOYSA-N bupropion Chemical compound CC(C)(C)NC(C)C(=O)C1=CC=CC(Cl)=C1 SNPPWIUOZRMYNY-UHFFFAOYSA-N 0.000 description 1
- 229960001058 bupropion Drugs 0.000 description 1
- 239000006227 byproduct Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 230000002802 cardiorespiratory effect Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000003518 caustics Substances 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 230000004098 cellular respiration Effects 0.000 description 1
- 210000001638 cerebellum Anatomy 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000013043 chemical agent Substances 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 229960003291 chlorphenamine Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 229940048961 cholinesterase Drugs 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 229960001403 clobazam Drugs 0.000 description 1
- CXOXHMZGEKVPMT-UHFFFAOYSA-N clobazam Chemical compound O=C1CC(=O)N(C)C2=CC=C(Cl)C=C2N1C1=CC=CC=C1 CXOXHMZGEKVPMT-UHFFFAOYSA-N 0.000 description 1
- DGBIGWXXNGSACT-UHFFFAOYSA-N clonazepam Chemical compound C12=CC([N+](=O)[O-])=CC=C2NC(=O)CN=C1C1=CC=CC=C1Cl DGBIGWXXNGSACT-UHFFFAOYSA-N 0.000 description 1
- 229960003120 clonazepam Drugs 0.000 description 1
- 229960004362 clorazepate Drugs 0.000 description 1
- XDDJGVMJFWAHJX-UHFFFAOYSA-M clorazepic acid anion Chemical compound C12=CC(Cl)=CC=C2NC(=O)C(C(=O)[O-])N=C1C1=CC=CC=C1 XDDJGVMJFWAHJX-UHFFFAOYSA-M 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- TVZPLCNGKSPOJA-UHFFFAOYSA-N copper zinc Chemical compound [Cu].[Zn] TVZPLCNGKSPOJA-UHFFFAOYSA-N 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 125000000392 cycloalkenyl group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 229960002433 cysteine Drugs 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 238000001784 detoxification Methods 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 229960001985 dextromethorphan Drugs 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- MZDOIJOUFRQXHC-UHFFFAOYSA-N dimenhydrinate Chemical compound O=C1N(C)C(=O)N(C)C2=NC(Cl)=N[C]21.C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 MZDOIJOUFRQXHC-UHFFFAOYSA-N 0.000 description 1
- 229960004993 dimenhydrinate Drugs 0.000 description 1
- PCHPORCSPXIHLZ-UHFFFAOYSA-N diphenhydramine hydrochloride Chemical compound [Cl-].C=1C=CC=CC=1C(OCC[NH+](C)C)C1=CC=CC=C1 PCHPORCSPXIHLZ-UHFFFAOYSA-N 0.000 description 1
- 238000007323 disproportionation reaction Methods 0.000 description 1
- 230000004771 dopaminergic neurodegeneration Effects 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229960004428 doxacurium Drugs 0.000 description 1
- 230000000792 effect on seizure Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 206010015037 epilepsy Diseases 0.000 description 1
- 230000008579 epileptogenesis Effects 0.000 description 1
- 229960005139 epinephrine Drugs 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 238000010304 firing Methods 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- 239000011737 fluorine Substances 0.000 description 1
- 125000004216 fluoromethyl group Chemical group [H]C([H])(F)* 0.000 description 1
- 239000002778 food additive Substances 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 229960003692 gamma aminobutyric acid Drugs 0.000 description 1
- BTCSSZJGUNDROE-UHFFFAOYSA-N gamma-aminobutyric acid Chemical compound NCCCC(O)=O BTCSSZJGUNDROE-UHFFFAOYSA-N 0.000 description 1
- WIGCFUFOHFEKBI-UHFFFAOYSA-N gamma-tocopherol Natural products CC(C)CCCC(C)CCCC(C)CCCC1CCC2C(C)C(O)C(C)C(C)C2O1 WIGCFUFOHFEKBI-UHFFFAOYSA-N 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 210000005046 glial fibrillary acidic protein Anatomy 0.000 description 1
- 230000009688 glial response Effects 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 230000006545 glycolytic metabolism Effects 0.000 description 1
- 229940015042 glycopyrrolate Drugs 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 125000004366 heterocycloalkenyl group Chemical group 0.000 description 1
- 125000006588 heterocycloalkylene group Chemical group 0.000 description 1
- 229950002932 hexamethonium Drugs 0.000 description 1
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001183 hydrocarbyl group Chemical group 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- 229940031574 hydroxymethyl cellulose Drugs 0.000 description 1
- 229920003063 hydroxymethyl cellulose Polymers 0.000 description 1
- 230000035873 hypermotility Effects 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 230000003933 intellectual function Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000000185 intracerebroventricular administration Methods 0.000 description 1
- 230000004410 intraocular pressure Effects 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- 229940044173 iodine-125 Drugs 0.000 description 1
- OEXHQOGQTVQTAT-JRNQLAHRSA-N ipratropium Chemical compound O([C@H]1C[C@H]2CC[C@@H](C1)[N@@+]2(C)C(C)C)C(=O)C(CO)C1=CC=CC=C1 OEXHQOGQTVQTAT-JRNQLAHRSA-N 0.000 description 1
- 229960001888 ipratropium Drugs 0.000 description 1
- RGXCTRIQQODGIZ-UHFFFAOYSA-O isodesmosine Chemical compound OC(=O)C(N)CCCC[N+]1=CC(CCC(N)C(O)=O)=CC(CCC(N)C(O)=O)=C1CCCC(N)C(O)=O RGXCTRIQQODGIZ-UHFFFAOYSA-O 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- VLSMHEGGTFMBBZ-OOZYFLPDSA-N kainic acid Chemical compound CC(=C)[C@H]1CN[C@H](C(O)=O)[C@H]1CC(O)=O VLSMHEGGTFMBBZ-OOZYFLPDSA-N 0.000 description 1
- 229950006874 kainic acid Drugs 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 231100001231 less toxic Toxicity 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 229960004391 lorazepam Drugs 0.000 description 1
- 230000001045 lordotic effect Effects 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000003712 lysosome Anatomy 0.000 description 1
- 230000001868 lysosomic effect Effects 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- OAWZTKNCHQQRKF-UHFFFAOYSA-L manganese(3+);4-[10,15,20-tris(4-carboxyphenyl)porphyrin-22,24-diid-5-yl]benzoic acid Chemical compound [Mn+3].C1=CC(C(=O)O)=CC=C1C(C1=CC=C([N-]1)C(C=1C=CC(=CC=1)C(O)=O)=C1C=CC(=N1)C(C=1C=CC(=CC=1)C(O)=O)=C1C=CC([N-]1)=C1C=2C=CC(=CC=2)C(O)=O)=C2N=C1C=C2 OAWZTKNCHQQRKF-UHFFFAOYSA-L 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- IMYZQPCYWPFTAG-IQJOONFLSA-N mecamylamine Chemical compound C1C[C@@H]2C(C)(C)[C@@](NC)(C)[C@H]1C2 IMYZQPCYWPFTAG-IQJOONFLSA-N 0.000 description 1
- 229960002525 mecamylamine Drugs 0.000 description 1
- 230000003340 mental effect Effects 0.000 description 1
- GBLRQXKSCRCLBZ-IYQFLEDGSA-N meso-doxacurium Chemical compound COC1=C(OC)C(OC)=CC(C[C@@H]2[N@@+](CCC3=C2C(=C(OC)C(OC)=C3)OC)(C)CCCOC(=O)CCC(=O)OCCC[N@@+]2(C)[C@@H](C3=C(OC)C(OC)=C(OC)C=C3CC2)CC=2C=C(OC)C(OC)=C(OC)C=2)=C1 GBLRQXKSCRCLBZ-IYQFLEDGSA-N 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- 210000000274 microglia Anatomy 0.000 description 1
- 230000006724 microglial activation Effects 0.000 description 1
- 230000002025 microglial effect Effects 0.000 description 1
- 239000004005 microsphere Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 230000006686 mitochondrial oxygen consumption Effects 0.000 description 1
- 230000025608 mitochondrion localization Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 125000006682 monohaloalkyl group Chemical group 0.000 description 1
- 125000004572 morpholin-3-yl group Chemical group N1C(COCC1)* 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 239000000472 muscarinic agonist Substances 0.000 description 1
- 239000003149 muscarinic antagonist Substances 0.000 description 1
- XKTFQMCPGMTBMD-FYHMSGCOSA-N myxothiazol Chemical compound NC(=O)\C=C(\OC)[C@H](C)[C@@H](OC)\C=C\C1=CSC(C=2N=C(SC=2)[C@@H](C)\C=C\C=C\C(C)C)=N1 XKTFQMCPGMTBMD-FYHMSGCOSA-N 0.000 description 1
- 229930187386 myxothiazol Natural products 0.000 description 1
- XKTFQMCPGMTBMD-UHFFFAOYSA-N myxothiazol A Natural products NC(=O)C=C(OC)C(C)C(OC)C=CC1=CSC(C=2N=C(SC=2)C(C)C=CC=CC(C)C)=N1 XKTFQMCPGMTBMD-UHFFFAOYSA-N 0.000 description 1
- GDVKPNORZICDJU-UHFFFAOYSA-N n-[2-[ethoxy(methyl)phosphinothioyl]oxyethyl]-n-propan-2-ylpropan-2-amine Chemical compound CCOP(C)(=S)OCCN(C(C)C)C(C)C GDVKPNORZICDJU-UHFFFAOYSA-N 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- 238000002663 nebulization Methods 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 210000000715 neuromuscular junction Anatomy 0.000 description 1
- 230000003955 neuronal function Effects 0.000 description 1
- 230000003961 neuronal insult Effects 0.000 description 1
- 231100000421 neurotoxicant Toxicity 0.000 description 1
- 239000012244 neurotoxicant Substances 0.000 description 1
- 230000001682 neurotoxicant effect Effects 0.000 description 1
- 239000002581 neurotoxin Substances 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 238000006396 nitration reaction Methods 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 229960004429 obidoxime Drugs 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 229930191479 oligomycin Natural products 0.000 description 1
- MNULEGDCPYONBU-AWJDAWNUSA-N oligomycin A Polymers O([C@H]1CC[C@H](/C=C/C=C/C[C@@H](C)[C@H](O)[C@@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)O[C@@H]([C@@H]2C)[C@@H]1C)CC)[C@@]12CC[C@H](C)[C@H](C[C@@H](C)O)O1 MNULEGDCPYONBU-AWJDAWNUSA-N 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 150000002891 organic anions Chemical class 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 230000008789 oxidative DNA damage Effects 0.000 description 1
- NVOYVOBDTVTBDX-PMEUIYRNSA-N oxitropium Chemical compound CC[N+]1(C)[C@H]2C[C@@H](C[C@@H]1[C@H]1O[C@@H]21)OC(=O)[C@H](CO)C1=CC=CC=C1 NVOYVOBDTVTBDX-PMEUIYRNSA-N 0.000 description 1
- 229960000797 oxitropium Drugs 0.000 description 1
- ZIFJVJZWVSPZLE-UHFFFAOYSA-N oxo-[[1-[[4-(oxoazaniumylmethylidene)pyridin-1-yl]methoxymethyl]pyridin-4-ylidene]methyl]azanium;dichloride Chemical compound [Cl-].[Cl-].C1=CC(=C[NH+]=O)C=CN1COCN1C=CC(=C[NH+]=O)C=C1 ZIFJVJZWVSPZLE-UHFFFAOYSA-N 0.000 description 1
- 125000000963 oxybis(methylene) group Chemical group [H]C([H])(*)OC([H])([H])* 0.000 description 1
- NFHFRUOZVGFOOS-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000001499 parasympathomimetic effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- IMACFCSSMIZSPP-UHFFFAOYSA-N phenacyl chloride Chemical compound ClCC(=O)C1=CC=CC=C1 IMACFCSSMIZSPP-UHFFFAOYSA-N 0.000 description 1
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 1
- 229960002139 pilocarpine hydrochloride Drugs 0.000 description 1
- RNAICSBVACLLGM-GNAZCLTHSA-N pilocarpine hydrochloride Chemical compound Cl.C1OC(=O)[C@@H](CC)[C@H]1CC1=CN=CN1C RNAICSBVACLLGM-GNAZCLTHSA-N 0.000 description 1
- 125000004483 piperidin-3-yl group Chemical group N1CC(CCC1)* 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 125000006684 polyhaloalkyl group Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 150000004032 porphyrins Chemical class 0.000 description 1
- 238000010149 post-hoc-test Methods 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 235000018102 proteins Nutrition 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 230000000541 pulsatile effect Effects 0.000 description 1
- 238000006338 pulse radiolysis reaction Methods 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- JUJWROOIHBZHMG-UHFFFAOYSA-O pyridinium Chemical compound C1=CC=[NH+]C=C1 JUJWROOIHBZHMG-UHFFFAOYSA-O 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 238000012827 research and development Methods 0.000 description 1
- 230000027756 respiratory electron transport chain Effects 0.000 description 1
- 230000036387 respiratory rate Effects 0.000 description 1
- 239000002710 riot control agent Substances 0.000 description 1
- 229940080817 rotenone Drugs 0.000 description 1
- JUVIOZPCNVVQFO-UHFFFAOYSA-N rotenone Natural products O1C2=C3CC(C(C)=C)OC3=CC=C2C(=O)C2C1COC1=C2C=C(OC)C(OC)=C1 JUVIOZPCNVVQFO-UHFFFAOYSA-N 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 229930195734 saturated hydrocarbon Natural products 0.000 description 1
- 230000002000 scavenging effect Effects 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 239000010703 silicon Substances 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 230000009295 sperm incapacitation Effects 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010245 stereological analysis Methods 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 239000003491 tear gas Substances 0.000 description 1
- 210000003478 temporal lobe Anatomy 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000005207 tetraalkylammonium group Chemical group 0.000 description 1
- 125000005958 tetrahydrothienyl group Chemical group 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 125000005309 thioalkoxy group Chemical group 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- LERNTVKEWCAPOY-DZZGSBJMSA-N tiotropium Chemical compound O([C@H]1C[C@@H]2[N+]([C@H](C1)[C@@H]1[C@H]2O1)(C)C)C(=O)C(O)(C=1SC=CC=1)C1=CC=CS1 LERNTVKEWCAPOY-DZZGSBJMSA-N 0.000 description 1
- 229940110309 tiotropium Drugs 0.000 description 1
- OOGJQPCLVADCPB-HXUWFJFHSA-N tolterodine Chemical compound C1([C@@H](CCN(C(C)C)C(C)C)C=2C(=CC=C(C)C=2)O)=CC=CC=C1 OOGJQPCLVADCPB-HXUWFJFHSA-N 0.000 description 1
- 229960004045 tolterodine Drugs 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 231100000816 toxic dose Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000027 toxicology Toxicity 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- PYIHTIJNCRKDBV-UHFFFAOYSA-L trimethyl-[6-(trimethylazaniumyl)hexyl]azanium;dichloride Chemical compound [Cl-].[Cl-].C[N+](C)(C)CCCCCC[N+](C)(C)C PYIHTIJNCRKDBV-UHFFFAOYSA-L 0.000 description 1
- JFJZZMVDLULRGK-URLMMPGGSA-O tubocurarine Chemical compound C([C@H]1[N+](C)(C)CCC=2C=C(C(=C(OC3=CC=C(C=C3)C[C@H]3C=4C=C(C(=CC=4CCN3C)OC)O3)C=21)O)OC)C1=CC=C(O)C3=C1 JFJZZMVDLULRGK-URLMMPGGSA-O 0.000 description 1
- 229960001844 tubocurarine Drugs 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- BDIAUFOIMFAIPU-UHFFFAOYSA-N valepotriate Natural products CC(C)CC(=O)OC1C=C(C(=COC2OC(=O)CC(C)C)COC(C)=O)C2C11CO1 BDIAUFOIMFAIPU-UHFFFAOYSA-N 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 1
- 229920002554 vinyl polymer Polymers 0.000 description 1
- 235000019165 vitamin E Nutrition 0.000 description 1
- 229940046009 vitamin E Drugs 0.000 description 1
- 239000011709 vitamin E Substances 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000002676 xenobiotic agent Substances 0.000 description 1
- 230000002034 xenobiotic effect Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61B—DIAGNOSIS; SURGERY; IDENTIFICATION
- A61B17/00—Surgical instruments, devices or methods
- A61B17/16—Instruments for performing osteoclasis; Drills or chisels for bones; Trepans
- A61B17/17—Guides or aligning means for drills, mills, pins or wires
- A61B17/1739—Guides or aligning means for drills, mills, pins or wires specially adapted for particular parts of the body
- A61B17/1742—Guides or aligning means for drills, mills, pins or wires specially adapted for particular parts of the body for the hip
- A61B17/1746—Guides or aligning means for drills, mills, pins or wires specially adapted for particular parts of the body for the hip for the acetabulum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61B—DIAGNOSIS; SURGERY; IDENTIFICATION
- A61B90/00—Instruments, implements or accessories specially adapted for surgery or diagnosis and not covered by any of the groups A61B1/00 - A61B50/00, e.g. for luxation treatment or for protecting wound edges
- A61B90/10—Instruments, implements or accessories specially adapted for surgery or diagnosis and not covered by any of the groups A61B1/00 - A61B50/00, e.g. for luxation treatment or for protecting wound edges for stereotaxic surgery, e.g. frame-based stereotaxis
- A61B90/11—Instruments, implements or accessories specially adapted for surgery or diagnosis and not covered by any of the groups A61B1/00 - A61B50/00, e.g. for luxation treatment or for protecting wound edges for stereotaxic surgery, e.g. frame-based stereotaxis with guides for needles or instruments, e.g. arcuate slides or ball joints
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- 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
- A61K31/40—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
- A61K31/409—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having four such rings, e.g. porphine derivatives, bilirubin, biliverdine
-
- 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
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
-
- 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
- A61K31/555—Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/08—Antiepileptics; Anticonvulsants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/14—Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P39/00—General protective or antinoxious agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P39/00—General protective or antinoxious agents
- A61P39/02—Antidotes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P39/00—General protective or antinoxious agents
- A61P39/04—Chelating agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- F—MECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
- F16—ENGINEERING ELEMENTS AND UNITS; GENERAL MEASURES FOR PRODUCING AND MAINTAINING EFFECTIVE FUNCTIONING OF MACHINES OR INSTALLATIONS; THERMAL INSULATION IN GENERAL
- F16C—SHAFTS; FLEXIBLE SHAFTS; ELEMENTS OR CRANKSHAFT MECHANISMS; ROTARY BODIES OTHER THAN GEARING ELEMENTS; BEARINGS
- F16C11/00—Pivots; Pivotal connections
- F16C11/04—Pivotal connections
- F16C11/10—Arrangements for locking
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y10—TECHNICAL SUBJECTS COVERED BY FORMER USPC
- Y10T—TECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
- Y10T403/00—Joints and connections
- Y10T403/32—Articulated members
- Y10T403/32254—Lockable at fixed position
- Y10T403/32426—Plural distinct positions
Abstract
Disclosed herein is the use of metalloporphyrin compounds of formulae I-XVII for treating a subject suffering from exposure to a chemical threat agent.
Description
oporphyrin Neurological Treatments
CROSS—REFERENCES TO RELATED ATIONS
[0001] This application claims the benefit of US. Provisional ation No. 61/566,530,
filed December 2, 2011, the entire ts of which is hereby incorporated herein and for all
purposes.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER
FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] This invention was made with Government support under Grant Numbers
R21NS072099 and R0 1NS03 9487 by the National Institutes of Health, and by the Counter ACT
program. The Government may have certain rights in this invention.
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER
PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK
[0003] NOT APPLICABLE
OUND OF THE INVENTION
Chemical warfare agents (e.g., chemical threat agents) are an immense threat to military
nel and civilians. The central nervous system (CTN S) is a sensitive target for chemical
toxicants that interact with receptors and signaling, eg. nerve agents or organophosphate
pesticides. Studies in the literature have established that controlling seizure activity and
downstream consequences is critical for neuroprotection and survival after nerve agent exposure.
Accordingly, there is a need to develop novel and efficacious neuroprotective countermeasures
against chemical threat agents. Provided herein are compositions and methods directed to these
and other problems in the art.
BRIEF Y OF THE IN VENTION
There is provided, inter alia, a novel method for treating a subject suffering from
exposure to a chemical threat agent, the method including administering to the subject an
effective amount of a nd selected from:
a) a compound having the structure of Formula (I) or Formula (11),
R4 R2
R4 R2
Rs R3
(1) (11),
wherein R1, R2, R3, and R4 are each independently —CF3, -C02Rg, ,
R5_N)+\N—R6% O—R7 R10\N \s
\_/ OH N\:/
7 7
W W
R12 E@ \N/ N/R13 /—S +
/ J )\
N R17—N \N
R1/1+ R15 R14 7R16 \__/——
7 7 7
W mm
W WW
7 7 7 701-
R24\N/ S
\:/+ ; R5, R6, R7, R8, R8’, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20,
R21, R22, R23, and R24 are each independently hydrogen, halogen, —CN, —CF3, —OH, —NH2,
—COOH, —COOR25, —CH2COOR25, —CH2COOH, an tituted or substituted alkyl,
unsubstituted or substituted heteroalkyl, unsubstituted or substituted cycloalkyl, unsubstituted or
substituted heterocycloalkyl, unsubstituted or substituted aryl, or an unsubstituted or substituted
heteroaryl; R25 is an unsubstituted alkyl; and M is a metal;
b) a compound having the structure of one of ae (X)—(XV),
[11+
wherein R13, R23, R33, and R4a are independently -(CH2)mCHZOX1 or -(CH2CHZO)nX1; m is 1-6; n
is 3—50; X1 is tuted or unsubstituted €1-12 alkyl; M is a metal; and each A is, independently,
en or an electron withdrawing group; and
c) a compound having the structure of one of Formulae (XVI)—(XVII),
wherein
at least one of R”, or R16, R21, or R26, R3b or R36, and R4b or R40 is, independently, (CH2)pCHzOX2
or -(CH2CHZO)qX2; the other one of Ru, or Rlc, R21, or R26, R3b or R36, and R4b or R40 is,
independently, a C142 alkyl (straight chain or ed); p is 1-6; q is 3—50; X2 is substituted or
unsubstituted C142 alkyl; M is a metal; and each A is, independently, hydrogen or an electron
withdrawing group; wherein said chemical threat agent is an anti-cholinesterase agent, a GABA-
agent or a metabolic poison.
In another aspect, there is provided a method for reducing brain injury in a subject in
need thereof. The method includes administering to the subject an effective amount of a
compound selected from any of Formulae (I)—(XVII), as disclosed herein.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Fig. 1A: AEOL10150 [Formula (VII)] levels in plasma (right axis) and brain
(left axis) of C57Bl6 mice are ed with time after a single s.c. dose of AEOL10150
(lSmg/kg). Points represent mean + S.E.M. (n= 3—4). Legend (Fig. 1A): Brain d circle);
plasma (open circle). Fig. 1B: Histogram depicts TH—positive neurons in the SNpc of C57BL/6
mice counted by stereological analysis after injection of MPTP (lSmg/kg X3, s.c., 24h als)
alone or in the presence of AEOL10150 (lSmg/kg X3 s.c., daily for 3 days beginning lh prior to
MPTP). Fig. 1C: Histogram depicts 4-HNE (4-hydroxynonenal) levels in the SN of the
C57BL/6 mice after injection of MPTP (lSmg/kg X3, s.c., 24h als) alone or in the presence
of AEOL10150 (lSmg/kg X3 s.c., daily for 3 days) *p<0.01 vs. control, # p<0.01 vs. MPTP;
one—way, ANOVA, n=6. Legend (Figs. lB—lC): Control (open); MPTP (closed);
EOL10150 (diagonal lines).
Figure 2. Histograms depict tration of GSH (Fig. 2A) and GSSG (Fig. 2B), and
GSH/GSSG (Fig. 2C) ratio in the ampus of the rat after either pilocarpine alone or in the
presence of AEOL10150 post—treatment. Bars represent mean + S.E.M, **p<0.01 vs. control
rats, #p<0.05 vs. pilocarpine alone treated rats; one—way ANOVA, n=3 rats per group. Legend:
Control (open); pilocarpine (closed); pilocarpine+ AEOL10150 nal lines).
Figure 3. rams depict concentrations of Cysteine (Fig. 3A) and Cystine (Fig.
3B), and Cysteine/Cystine ratio (Fig. 3C) in the hippocampus of the rat after either pilocarpine
alone or in the presence of AEOL10150 post-treatment. Bars ent mean + S.E.M, **p<0.01
vs. control rats, #p<0.05 vs. pilocarpine alone treated rats; one—way ANOVA, n=3 rats per group.
Legend: Control (open); pilocarpine (closed); pilocarpine+AEOL10150 (diagonal lines).
Figure 4. Histograms depict 3-NT/tyrosine ratio (Fig. 4A) and AA (Fig. 4B) in the
hippocampus of the rat after either pilocarpine alone or in the presence of AEOL10150 post—
treatment. Bars represent mean + S.E.M, **p<0.01 vs. control rats, #p<0.05 vs. rpine alone
d rats; one—way ANOVA, n=3 rats per group. Legend: Control (open); pilocarpine
(closed); pilocarpine+AEOL10150 (diagonal lines).
2012/067633
Figure 5. Histograms depict quantitative analysis of Fluoro-jade B fluorescence in the
hilus (Fig. 5A) and CA3 (Fig. 5B) of the rat after either pilocarpine alone or in the ce of
AEOL10150 post-treatment. Bars represent mean + SEM, *p<0.05, **p<0.01 vs. pilocarpine
alone treated rats; one-way ANOVA, n=3 rats per group. Histogram ordering (left to right):
pilocarpine; pilocarpine+AEOL10150; AEOL10150; control.
Figure 6. Photomicrographs depict representative Fluoro-jade B stained images in the
CA3 (Figs. 6A-6C) and hilus (Figs. 6D-6F) of rats after either kainate alone or in the presence of
AEOL 10150 post-treatment. (Figs. 6A, 6D) control; (Figs. 6B, 6E), kainate alone; (Figs. 6C,
6F) kainate in presence of AEOL10150 ent.
[0013] Figure 7. Histograms depicts quantitative analysis of Fluoro-jade B fluorescence in the
hilus (Fig. 7A) and CA3 (Fig. 7B) of the rat after either kainate alone or in the ce of
AEOL 10150 post-treatment. Bars represent mean + SEM, **p<0.01 vs. control rats, #p<0.05
vs. kainate alone treated rats; one—way ANOVA, n=4 rats per group. : Control (open);
rpine (closed); pilocarpine+AEOL1015 0 (diagonal lines).
[0014] Figure 8. Figs. 8A—8B depict representative oxygen consumption rates (OCR) in
isolated hippocampal synaptosomes 48h after injection pilocarpine (Fig. 8A) or kainate (Fig. 8B)
in rats.
Figure 9. Histograms depict 150 effect on 3—NT/Tyrosine (Fig. 9A) and
GSH/GSSG (Fig. 9B) 24 hours ing Pilocarpine (pilo)—induced SE. AEOL 10150 was
injected at a dose of 5 mg/kg every 4 hours s.c. beginning 90 min after SE onset. **p<0.01,
*p<0.05, ***p<0.001 n=3 per group. Legend: Control e) (open); pilogram (closed);
rpine+AEOL10150 (diagonal .
Figure 10. Fig. 10 depicts oxygen ption rates (OCR) in isolated hippocampal
synaptosomes 16h after injection of saline and pilocarpine. Points represent average values from
2 rats per group.
Figure 11. Fig. 11 depicts a time line for studies described in Example 2.
Figure 12. Fig. 12 depicts histograms of the concentration AEOL10150 (pmol/g
tissue) in the hippocampus and piriform cortex after s.c. injection of AEOL10150 in the rat.
Legend: (open): single injection of AEOL10150 (5 mg/kg, s.c.) at 4-hrs; (closed): multiple
injections of AEOL10150 (5 mg/kg, s.c., every 4-hrs, 6 injections total) at 24-hrs. Error bars:
SEM (standard error of the mean). Groups: n=4. Histogram ordering (left to right): hippocampus
WO 30150
single dose; hippocampus multiple dose; piriforrn cortex single dose; piriforrn cortext mulitple
dose. Legend: Single dose of AEOL10150 (open); multiple dose (closed).
Figure 13. Figs. l3A-l3B depict histograms of quantitative analysis of Fluoro-Jade B
histofluorescence staining in the CA3 (Fig. 13A) and Hilus (Fig. 13B) of the rats at 24 h after
ing either pilocarpine (340mg/kg, i.p.) alone or in presence of AEOL10150 (5mg/kg, s.c.,
start at 60 or 90 min post pilocarpine treatment and every 4h therefore until sacrificed). The
Fluoro-Jade B positive signal in a given area of hippocampal subregions from three slides of
each animal was estimated with Image J. Bars represent mean + S.E.M, *p<0.01 vs. saline;
vs. pilocarpine; one way ANOVA, n=6 rats per group. : l (saline) (open);
pilocarpine alone (closed); pilocarpine+AEOL10150 90min post pilocarpine administration
(diagonal lines upper left to lower right); pilocarpine+AEOL10150 60min post pilocarpine
administration (diagonal lines lower left to upper right).
Figure 14. Figs. B depict histograms of GSH and GSSG concentrations, and
GSH/GSSG ratio, and Fig. 14C depicts histogram of 3-nitrotyrosine/tryrosine (3NT/tyr) ratios in
the hippocampus of the rat 24-hr (Fig. 14A and Fig. 14C left panel) or 48-h (Fig. 14B and Fig.
14C right panel) after either pilocarpine alone or in combination with AEOL10150 beginning 90
min after, 60 min after or 30 before and every 4 h thereafter until ice (24h or 48h). Bars
represent mean + S.E.M., 5 vs. saline treatment, # p<0.05 vs. pilo alone treatment, one—
way ANOVA, n=3—6 per group. Legend of ram ng: Fig. 14A: Control (A);
pilocarpine (B); pilocarpine+AEOL10150 (90min post-pilocarpine) (C);
pilocarpine--AEOL10150 (60min post-pilocarpine) (D); Fig. 14B: Control (E); pilocarpine (F);
pilocarpine--AEOL10150 (90min post-pilocarpine) (G); pilocarpine+AEOL10150 (60min post-
pilocarpine) (H); pilocarpine+AEOL10150 (3 0min pre-pilocarpine) (I); Fig. 14C (left panel):
Control (saline) (J); pilocarpine (K); pilocarpine+AEOL10150 (90min post-pilocarpine) (L);
pilocarpine--AEOL10150 (60min post-pilocarpine) (M); Fig. 14C (right panel): Control (saline)
(N); pilocarpine (O); pilocarpine+AEOL10150 (90min post-pilocarpine) (P);
pilocarpine--AEOL10150 (60min post-pilocarpine) (Q); rpine+AEOL10150 (30min pre-
pilocarpine) (R).
Figure 15. Fig. 15A is histogram depicting maximal respiratory capacity of rats treated
with control, pilocarpine and , or pilocarpine and AEOL10150. n=5—8 rat/group.
Histogram ordering (left to right); Control; pilocarpine+saline; pilocarpine+AEOL10150. Fig.
15B depicts time course of oxygen consumption rate (OCR, pmol/min).
2012/067633
Figure 16. Figs. D are photomicrographs of images of microglia (iba 1)
immunofluorescence in rat brain at 24 h after receiving saline (control (Fig. 16A), pilocarpine
alone (Fig. 16B) or with AEOL10150 (5mg/kg, s.c.) 60-min after pilocarpine (Fig. 16C) or 90—
min after rpine (Fig. 16D) every 4-hrs until sacrifice. Fig. 16E and Fig. 16F are histograms
of quantitative analysis of the data provided in Figs. 16A-16D. Bars=mean + S.E.M, *p<0.01 vs.
saline; #p<0.05 vs. pilocarpine; one way ANOVA, n=6 rats per group. Legend (Figs. 16E-16F);
Control (saline) (open); pilocarpine (closed); pilocarpine + AEOL10150 90-min post pilocarpine
(diagonal lines upper left to lower right); rpine (closed); pilocarpine + AEOL10150 60-min
post pilocarpine (diagonal lines lower left to upper right).
[0023] Figure 17. Figs. 17A-16D are histograms depicting concentrations of GSH (Fig. 17A),
GSSG (Fig. 17B), and ratios of GSH/GSSG (Fig. 17C) and 3-nitrotyrosine/tyrosine (Fig. 17D) in
the hippocampus of the rat at 24h after either pilocarpine (340mg/kg) alone with or without
atropine and diazepam or in the ce ofAEOL 10150 treatment (5mg/kg, s.c., every 4h).
Bars represent mean + S.E.M; a p<0.01 vs. l rats; p<0.05 vs. pilocarpine treated rats; c
p<0.05 vs. rpine+atropine+diazepam d rats. one—way ANOVA, n=2—6 rats per group.
saline n=6; pilocarpine, n=2 (3 of 5 dead); pilocarpine+atropine+diazepam, n=6;
pilocarpine+atropine+AEOL10150+diazepam, n=6). Histogram ordering (left to right): Control
(saline) (open); pilocarpine d); pilocarpine + atropine + diazepam (diagonal lines);
pilocarpine + atropine + diazepam + AEOL10150 (cross checkered).
DETAILED DESCRIPTION OF THE INVENTION
1. Definitions
The abbreviations used herein have their conventional meaning within the chemical and
biological arts. The chemical structures and ae set forth herein are constructed according
to the standard rules of chemical valency known in the chemical arts.
[0025] Where substituent groups are specified by their conventional chemical ae,
written from left to right, they equally encompass the chemically identical substituents that
would result from writing the structure from right to left, e.g., —CH20- is equivalent to —OCH2—.
The term “alkyl,” by itself or as part of another substituent, means, unless otherwise
stated, a straight (i.e., unbranched) or branched chain, or combination thereof, which may be
fully saturated, mono- or polyunsaturated and can e di- and multivalent radicals, having
the number of carbon atoms designated (i.e., C1—C10 means one to ten carbons). Examples of
saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-
propyl, isopropyl, n—butyl, t—butyl, yl, sec—butyl, hexyl)methyl, homologs and
s of, for example, yl, n-hexyl, yl, n-octyl, and the like. An unsaturated alkyl
group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl
groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, adienyl),
2,4-pentadienyl, 3-(l,4-pentadienyl), ethynyl, l- and 3-propynyl, 3-butynyl, and the higher
homologs and isomers. An alkoxy is an alkyl attached to the remainder of the molecule via an
oxygen linker (—O—).
The term “alkylene,” by itself or as part of another substituent, means, unless otherwise
stated, a divalent radical derived from an alkyl, as exemplified, but not limited by,
-CH2CH2CH2CH2-. lly, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms,
with those groups having 10 or fewer carbon atoms being preferred in the present invention. A
“lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having
eight or fewer carbon atoms.
[0028] The term “heteroalkyl,” by itself or in combination with another term, means, unless
otherwise stated, a stable straight or ed chain, or combinations thereof, consisting of at
least one carbon atom and at least one heteroatom ed from the group consisting of O, N, P,
Si, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the
nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N, P, S, and Si may
be placed at any or position of the heteroalkyl group or at the position at which the alkyl
group is attached to the remainder of the molecule. Examples include, but are not limited to:
H2—O—CH3, —CH2—CH2—NH—CH3, —CH2—CH2—N(CH3)—CH3, —CH2—S—CH2—CH3, —CH2—CH2,
—S(O)—CH3, —CH2—CH2—S(O)2—CH3, —CH=CH—O—CH3, —Si(CH3)3, —CH2—CH=N—OCH3,
—N(CH3)—CH3, —O—CH3, —O—CH2—CH3, and —CN. Up to two heteroatoms may be
consecutive, such as, for example, —CH2—NH—OCH3.
Similarly, the term “heteroalkylene,” by itself or as part of another substituent, means,
unless otherwise stated, a divalent radical derived from heteroalkyl, as exemplified, but not
limited by, —CH2—CH2—S—CH2—CH2- and —CH2—S-CH2—CH2—NH—CH2—. For heteroalkylene ,
heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy,
alkylenedioxy, alkyleneamino, nediamino, and the like). Still further, for alkylene and
heteroalkylene linking groups, no orientation of the linking group is implied by the direction in
which the formula of the linking group is written. For example, the formula -C(O)2R'- represents
2012/067633
both —C(O)2R'— and —R'C(O)2—. As described above, heteroalkyl groups, as used , include
those groups that are attached to the remainder of the molecule through a heteroatom, such as
—C(O)R', —C(O)NR', —NR'R", —OR', —SR', and/or —SOzR'. Where oalkyl” is recited, followed
by recitations of specific heteroalkyl groups, such as -NR'R" or the like, it will be tood that
the terms heteroalkyl and -NR'R" are not redundant or mutually exclusive. Rather, the specific
heteroalkyl groups are recited to add y. Thus, the term “heteroalkyl” should not be
interpreted herein as excluding specific heteroalkyl groups, such as -NR'R" or the like.
The terms “cycloalkyl” and “heterocycloalkyl,” by themselves or in combination with
other terms, mean, unless otherwise stated, cyclic versions of ” and “heteroalkyl,”
respectively. Additionally, for heterocycloalkyl, a atom can occupy the position at which
the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include,
but are not limited to, ropyl, cyclobutyl, cyclopentyl, cyclohexyl, l-cyclohexenyl,
3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include, but are not
limited to, l-(l,2,5,6-tetrahydropyridyl), l-piperidinyl, 2-piperidinyl, 3-piperidinyl,
4-morpholinyl, 3-morpholinyl, ydrofuran-2—yl, tetrahydrofuran-3 -yl, tetrahydrothienyl,
tetrahydrothien-3 -yl, l-piperazinyl, 2-piperazinyl, and the like. A alkylene” and a
“heterocycloalkylene,” alone or as part of another substituent, means a divalent radical derived
from a cycloalkyl and cycloalkyl, tively.
The terms “halo” or “halogen,” by themselves or as part of another substituent, mean,
unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such
as “haloalkyl” are meant to include monohaloalkyl and polyhaloalkyl. For example, the term
“halo(C1-C4)alkyl” includes, but is not limited to, fluoromethyl, romethyl, trifluoromethyl,
2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
The term “acyl” means, unless otherwise stated, —C(O)R where R is a substituted or
unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted
heteroalkyl, tuted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or
substituted or unsubstituted heteroaryl.
The term “aryl” means, unless otherwise stated, a polyunsaturated, aromatic,
hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3
rings) that are fused together (i.e., a fused ring aryl) or linked covalently. A fused ring aryl refers
to multiple rings fused together wherein at least one of the fused rings is an aryl ring. The term
“heteroaryl” refers to aryl groups (or rings) that contain at least one atom selected from N,
2012/067633
O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s)
are optionally ized. Thus, the term oaryl” includes fused ring heteroaryl groups
(i.e., multiple rings fused together wherein at least one of the fused rings is a heteroaromatic
ring). A 5,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 5
members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
Likewise, a 6,6—fused ring heteroarylene refers to two rings fused together, wherein one ring has
6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
And a 6,5—fused ring heteroarylene refers to two rings fused together, wherein one ring has 6
members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring. A
heteroaryl group can be attached to the der of the molecule through a carbon or
heteroatom. Non-limiting examples of aryl and heteroaryl groups e , l-naphthyl, 2-
yl, 4-biphenyl, l-pyrrolyl, 2—pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2—imidazolyl, 4-imidazolyl,
pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2—phenyloxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-
isoxazolyl, 2—thiazolyl, 4-thiazolyl, 5-thiazolyl, 2—furyl, 3-furyl, 2—thienyl, nyl, 2-pyridyl, 3-
pyridyl, 4-pyridyl, 2—pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2—benzimidazolyl, 5-
indolyl, l-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl.
Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the
group of able substituents described below. An “arylene” and a “heteroarylene,” alone or
as part of another substituent, mean a divalent radical d from an aryl and heteroaryl,
respectively.
The term “oxo,” as used , means an oxygen that is double bonded to a carbon
atom.
The term “alkylsulfonyl,” as used herein, means a moiety having the formula -S(Oz)-R',
where R' is an alkyl group as defined above. R' may have a specified number of carbons (e. g.,
“C1—C4 alkylsulfonyl”).
Each of the above terms (e. g., “alkyl,” “heteroalkyl,” “aryl,” and “heteroaryl”) includes
both tuted and unsubstituted forms of the indicated radical. Preferred substituents for each
type of radical are provided below.
Substituents for the alkyl and heteroalkyl radicals (including those groups often referred
to as ne, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl,
cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of groups ed from,
but not limited to, -OR', :0, =NR', =N—OR', -NR'R", -SR', -halogen, -SiR'R"R"', -OC(O)R',
—C(O)R', —COzR', —CONR'R", —OC(O)NR'R", —NR"C(O)R', —NR'—C(O)NR"R'", —NR”C(O)2R',
—NR—C(NR'R"R'")=NR"", —NR—C(NR'R")=NR"', —S(O)R', —S(O)2R', —S(O)2NR'R", —NRSOzR',
-CN, and -NOZ in a number ranging from zero to (2m'+l), where m' is the total number of carbon
atoms in such radical. R', R", R'", and R"" each preferably independently refer to en,
substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or
unsubstituted heterocycloalkyl, substituted or tituted aryl (e.g., aryl substituted with 1—3
halogens), substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups.
When a compound of the invention es more than one R group, for example, each of the R
groups is ndently selected as are each R', R", R'", and R"" group when more than one of
these groups is present. When R' and R" are attached to the same nitrogen atom, they can be
combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring. For example, -NR'R"
includes, but is not d to, l-pyrrolidinyl and 4-morpholinyl. From the above discussion of
tuents, one of skill in the art will understand that the term “alkyl” is meant to e
groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl
(e.g., —CF3 and —CH2CF3) and acyl (e.g., —C(O)CH3, —C(O)CF3, —C(O)CHZOCH3, and the like).
Similar to the substituents described for the alkyl radical, substituents for the aryl and
heteroaryl groups are varied and are selected from, for example: -OR', -NR'R", -SR', -halogen,
—SiR'R"R"', —OC(O)R', ', , —CONR'R”, NR'R", —NR"C(O)R',
—NR'—C(O)NR"R"', —NR"C(O)2R', —NR—C(NR'R"R"')=NR"", —NR—C(NR'R")=NR'", —S(O)R',
—S(O)2R', —S(O)2NR'R”, —NRSOzR', —CN, —NOz, —R', —N3, —CH(Ph)2, fluoro(C1—C4)alkoxy, and
fluoro(C1-C4)alkyl, in a number ranging from zero to the total number of open valences on the
aromatic ring system; and where R', R", R'", and R'"' are preferably independently selected from
hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, tuted
or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or
unsubstituted aryl, and substituted or unsubstituted heteroaryl. When a compound of the
invention includes more than one R group, for example, each of the R groups is independently
selected as are each R', R", R'", and R"" groups when more than one of these groups is present.
Two or more substituents may optionally be joined to form aryl, heteroaryl, cycloalkyl,
or heterocycloalkyl groups. Such so—called ring-forming substituents are typically, though not
necessarily, found attached to a cyclic base structure. In one embodiment, the ring—forrning
substituents are attached to adjacent s of the base structure. For example, two ring-
g substituents ed to adjacent members of a cyclic base ure create a fused ring
structure. In another embodiment, the ring-forming substituents are attached to a single member
2012/067633
of the base structure. For example, two ring-forming substituents attached to a single member of
a cyclic base structure create a spirocyclic structure. In yet another embodiment, the ring—
forming substituents are attached to non—adj acent members of the base ure.
Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally
form a ring of the formula -T-C(O)-(CRR')q-U-, wherein T and U are independently —NR—, —O—,
-CRR'-, or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents
on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of
the formula -A-(CH2)r-B-, wherein A and B are independently —CRR'—, —O—, —NR—, —S—, —S(O) —,
—S(O)2—, —S(O)2NR'—, or a single bond, and r is an integer of from 1 to 4. One of the single bonds
of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of
the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with
a tuent of the formula -(CRR')S-X'- (C"R'")d-, where s and d are independently integers of
from 0 to 3, and X' is —O—, —NR'—, —S—, —S(O)—, —, or —S(O)2NR'—. The substituents R, R', R",
and R'” are preferably independently ed from hydrogen, substituted or unsubstituted alkyl,
tuted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted
or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
As used herein, the terms “heteroatom” or “ring heteroatom” are meant to include
oxygen (0), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
A ituent group,” as used herein, means a group selected from the ing
moieties:
(A) —OH, —NH2, —SH, —CN, —CF3, -N02, oxo, halogen, unsubstituted alkyl, unsubstituted
heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl,
unsubstituted heteroaryl, and
(B) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, and aryl, substituted with
at least one substituent selected from:
(i) oxo, —OH, —NH2, —SH, —CN, —CF3, —NOz, halogen, tituted alkyl,
unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl,
unsubstituted aryl, unsubstituted heteroaryl, and
(ii) alkyl, heteroalkyl, cycloalkyl, cycloalkyl, aryl, and heteroaryl, substituted
with at least one substituent selected from:
(a) oxo, —OH, —NH2, —SH, —CN, —CF3, -N02, halogen, unsubstituted alkyl,
unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted
heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, and
(b) alkyl, heteroalkyl, lkyl, heterocycloalkyl, aryl, or heteroaryl,
substituted with at least one substituent selected from: oxo, —OH, —NH2, —SH, —CN,
—CF3, —NOz, halogen, unsubstituted alkyl, unsubstituted alkyl, unsubstituted
cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, and unsubstituted
heteroaryl.
A “size—limited substituent” or “ size-limited substituent group,” as used herein, means
a group selected from all of the substituents described above for a “substituent group,” wherein
each substituted or unsubstituted alkyl is a substituted or tituted C1—C20 alkyl, each
substituted or unsubstituted alkyl is a substituted or unsubstituted 2 to 20 membered
heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C4—C8
cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or
unsubstituted 4 to 8 membered heterocycloalkyl.
A “lower substituent” or “ lower substituent group,” as used herein, means a group
selected from all of the substituents described above for a “substituent group,” wherein each
substituted or unsubstituted alkyl is a substituted or unsubstituted C1—C8 alkyl, each substituted or
unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each
substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C5—C7 cycloalkyl, and
each substituted or unsubstituted heterocycloalkyl is a substituted or tituted 5 to 7
membered heterocycloalkyl.
Unless otherwise stated, structures depicted herein are also meant to include all
chemical forms of the ure; i.e., the R and S configurations for each asymmetric
. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric
mixtures of the present compounds are within the scope of the ion.
Unless otherwise stated, structures depicted herein are also meant to include
nds which differ only in the presence of one or more isotopically enriched atoms. For
example, compounds having the present ures except for the replacement of a hydrogen by a
deuterium or tritium, or the replacement of a carbon by 13C— or 14C—enriched carbon are within
the scope of this invention.
The nds of the present invention may also contain unnatural proportions of
atomic isotopes at one or more of atoms that tute such compounds. For example, the
compounds may be radiolabeled with radioactive isotopes, such as for e tritium (3H),
iodine-125 (1251) or carbon-14 (14C). All isotopic variations of the compounds of the present
invention, r radioactive or not, are encompassed within the scope of the t invention.
The terms “a,” “an,” or “a(n)”, when used in reference to a group of substituents herein,
mean at least one. For example, where a compound is substituted with “an” alkyl or aryl, the
compound is optionally substituted with at least one alkyl and/or at least one aryl. Moreover,
where a moiety is substituted with an R substituent, the group may be referred to as “R-
substituted.” Where a moiety is R—substituted, the moiety is substituted with at least one R
substituent, and each R substituent is optionally different. Where a particular R group is present
in the description of a chemical genus (such as Formula (1)), a Roman alphabetic symbol may be
used to distinguish each appearance of that particular R group. For example, where multiple R27
substituents are present, each R27 substituent may be distinguished as R27A, R273, R270, R271), etc.,
wherein each of R27A, R273, R270, R271), etc. is defined within the scope of the tion of R27
and optionally ently. The term “about” in the context of a numeric value refers, absent
express description otherwise, to the c value :: 10% thereof
Description of compounds of the present ion are limited by principles of
chemical bonding known to those skilled in the art. ingly, where a group may be
substituted by one or more of a number of substituents, such substitutions are selected so as to
comply with ples of chemical bonding and to give compounds which are not inherently
le and/or would be known to one of ordinary skill in the art as likely to be unstable under
t conditions, such as aqueous, neutral, and several known physiological conditions. For
example, a heterocycloalkyl or heteroaryl is attached to the remainder of the molecule via a ring
heteroatom in compliance with principles of chemical bonding known to those skilled in the art
thereby avoiding inherently unstable nds.
The terms “effective amount,” “therapeutically effective amount” and the like refer to
the amount of an active agent sufficient to induce a desired biological result. That result may be
alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a
biological system. The term “therapeutically effective amount” is used herein to denote any
amount of a compound disclosed herein or formulation thereof which causes an improvement in
a disease condition (e. g., exposure to a chemical threat agent) upon administration. The amount
will vary with the condition being d, the stage of advancement of the condition, and the
type and concentration of formulation applied. Appropriate amounts in any given instance will
be readily apparent to those skilled in the art or capable of determination by routine
mentation.
The terms “chemical agent, 3’ CCchemical threat agent” and the like refer in the ary
sense to compounds which elicit a pathological condition, e. g., incapacitation, sions, and
the like, in a subject. ary chemical threat agents include ins, r agents, blood
agents, caustics (e. g., acids, bases), choking agents, lung agents, pulmonary agents,
incapacitating agents, long-acting anticoagulants, metals, nerve agents, organic solvents, riot
control agents, tear gas, toxic alcohols, and ng agents. The chemical threat agent may be a
chemical weapon. The chemical threat agent may also be a nerve agent that disrupts the
mechanism by which nerves transfer messages to organs. The disruption may be caused by
inhibiting (i.e. ng the activity of) acetylcholinesterase; e. g., sarin (isopropyl
methylphosphonofluoridate), parathion (0,0-diethyl trophenyl phosphorothioate),
rb ((E)-2—methyl(methylthio)propanal O-methylcarbamoyl , and VX (S
(diisopropylamino)ethyl O-ethyl methylphosphonothioate). The chemical threat agent (e.g.,
nerve agent) may be a phosphorus-containing organic chemical (organophosphate). Some
chemical threat agents (i.e., so-called agents”) interfere with GABA neuronal function
and/or chloride channels, e.g., tetramethylene disulfotetramine, also known as “tetramine
(TETS)” (2,6—dithia— l,3,5,7- tetraaza— tricyclo [3.3.1.13,7] decane 2,2,6,6—tetraoxide). Some
chemical threat agents are so—called “metabolic poisons” or compounds which target the blood,
as known in the art, e. g., cyanide, sodium fluoroacetate, arsenic trioxide, and nine. In one
embodiment, chemical threat agents contemplated herein do not include agents as disclosed in
International Publication No. including a sulfur mustard, chlorine gas,
phosgene, or 2-chloroethyl ethyl sulfide (CEES).
As used herein, “treatment” or “treating,” or “palliating” or “ameliorating” are used
interchangeably herein. These terms refer to an approach for obtaining beneficial or desired
results including but not d to therapeutic benefit and/or a prophylactic benefit. By
therapeutic benefit is meant eradication or amelioration of the underlying disorder being d.
Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the
physiological symptoms associated with the underlying disorder such that an improvement is
observed in the patient, notwithstanding that the patient may still be ed with the underlying
disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of
WO 30150
re to a chemical threat agent, or to a t ing one or more of the physiological
symptoms of a e, even though a diagnosis of this disease may not have been made.
Treatment includes preventing the disease, that is, causing the clinical symptoms of the disease
not to develop by administration of a protective composition prior to the induction of the disease;
suppressing the disease, that is, causing the clinical symptoms of the disease not to develop by
administration of a protective composition after the inductive event but prior to the clinical
appearance or reappearance of the disease; inhibiting the disease, that is, arresting the
development of clinical symptoms by administration of a protective ition after their
initial appearance; ting re-occurring of the disease and/or relieving the disease, that is,
causing the regression of clinical symptoms by administration of a protective composition after
their initial appearance.
The term “pharmaceutically acceptable salt” refers to salts derived from a variety of
organic and inorganic counter ions well known in the art and include, by way of example only,
sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and
when the le contains a basic functionality, salts of organic or inorganic acids, such as
hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
A ct,” “individual,” or “patient,” is used interchangeably herein, which refers to a
vertebrate, preferably a mammal, more ably a human. Mammals e, but are not
limited to, murines, simians, , farm animals, sport animals, and pets. Tissues, cells and
their progeny of a biological entity obtained in vitro or cultured in vitro are also encompassed.
In one embodiment, the subject or patient is a child. In some embodiments, the subject or patient
is a young child. In some embodiments, the subject or t is an infant. In one embodiment,
the subject or patient is an adult.
As defined herein, the term "child" or "children" as used herein means persons over the
age of 3 years and prior to adolescence. As used herein, the term "young child" or "young
children" means persons from the age of more than 12 months up to the age of three years. As
used herein, the term "infant" means a person not more than 12 months of age. The term “adult”
means persons past the age of adolescence.
11. Methods
[0056] There is ed a method for treating a subject suffering from exposure to a chemical
threat agent. The chemical threat agent may be a nerve agent. The chemical threat agent may
2012/067633
function as an anti-cholinesterase agent, a GABA-agent or a lic poison. The method
includes administering to the subject an effective amount of a compound selected from the group
having the structure of any of ae (I)—(XVH) disclosed herein.
In one embodiment, the compound has the structure of Formula (I) or Formula (11),
Rs (1) R3 (11).
In Formula (I), the substituted porphyrin may be bound to a metal, e.g., Formula (II).
The metal may be manganese, iron, cobalt, copper, nickel, or zinc, including ions thereof. For
example, in Formula (II), or in any formula set forth , M is manganese, iron, cobalt,
copper, nickel, or zinc, including ions thereof: Thus, in a specific embodiment, the metal is
manganese and the compound has the structure of Formula (111):
R3 (111).
WO 30150
In any of Formulae (I)-(IH), R1, R2, R3, and R4 are each independently -CF3, -C02R8,
R5—N/Jrk g
N—R6
_COR8,7 \=/ OH va
JVVV‘ J'VVV"
_ RRN/XN/Rw /
E s
\_/— Rfi R15 R14
7 7 7R16 7
JV'V'U" JWV‘
JVVV‘ W
A \
F“— \\
R17—N \N \:N\ \
\=/7 + N—N\
R197 \:N R22
7 7
JVVV‘
\4 \Q.
In one ment for Formulae (I)-(III), R1, R2, R3, and R4 may be
MAW é \ /
\ / / +
or R11
In one embodiment, R1 and R3 are independently -CO2R3 or . R2 and R4 may
independently be —CF3 or JWV‘
. In one embodiment, R1 and R3 are ndently
-CO2R3, and R2 and R4 are —CF3. In one embodiment, R1 and R3 are independently -CO2R3 and
R2 and R4 are independently JVVV‘
Where R1, R2, R3, and R4 contain a positive , one of skill will immediately
recognize that an anionic compound or molecule will be present Where the compound is in
solution. Any applicable anionic compound are molecule may be used as a counterion to the
positively charges substituents, including for example chloride, fluoride, sulfide, a e, a
carbonate, or a phosphate.
Further to this embodiment, R5, R6, R7, R3, R3,, R9, R10, R11, R12, R13, R14, R15, R16, R17,
R13, R19, R20, R21, R22, R23, and R24 are each independently hydrogen, halogen, —CN, —CF3, —OH,
—NH2, —COOH, —COOR25, —CH2COOR25, —CH2COOH, an unsubstituted or substituted alkyl,
unsubstituted or substituted alkyl, unsubstituted or substituted cycloalkyl, unsubstituted or
substituted heterocycloalkyl, unsubstituted or substituted aryl, or an unsubstituted or substituted
heteroaryl. R25 is an unsubstituted alkyl. In one embodiment, R25 is an unsubstituted alkyl such
as C140 alkyl (e.g., —CH3 or a C1_5 alkyl). M is a metal (e. g. is manganese, iron, cobalt, copper,
nickel, or zinc).
In one embodiment, R5, R6, R7, R3, R3,, R9, R10, R11, R12, R13, R14, R15, R16, R17, R13,
R19, R20, R21, R22, R23, and R24 may each independently be hydrogen, halogen, —CN, —CF3, —OH,
—NH2, —COOH, —COOR25, —CH2COOR25, —CH2COOH, substituted or unsubstituted C1—C10 (e.g.,
C1—C6) alkyl, substituted or unsubstituted 2 to 10 membered (e.g., 2 to 6 ed) heteroalkyl,
substituted or unsubstituted C3—C3 (e.g., C5—C7) cycloalkyl, substituted or unsubstituted 3 to 8
membered (e.g., 3 to 6 membered) cycloalkyl, tuted or tituted C5—C3 (e.g., C5—
C6) aryl, or substituted or unsubstituted 5 to 8 ed (e.g., 5 to 6 membered) heteroaryl. In
one embodiment, one or more of R5, R6, R7, R3, R3,, R9, R10, R11, R12, R13, R14, R15, R16, R17, R13,
R19, R20, R21, R22, R23, and R24 is unsubstituted. In one embodiment, R5, R6, R7, R3, R3,, R9, R10,
R11, R12, R13, R14, R15, R16, R17, R13, R19, R20, R21, R22, R23, and R24 are independently hydrogen
or a substituted or unsubstituted C1—C10 (e.g., C1—C6 or C1—C3) alkyl.
In one embodiment, R5, R6, R7, R3, R3,, R9, R10, R11, R12, R13, R14, R15, R16, R17, R13,
R19, R20, R21, R22, R23, and R24, may independently be hydrogen, halogen, —CN, —CF3, —OH, —NH2,
—COOH, —COOR25, —CH2COOR25, —CH2COOH, bstituted or unsubstituted alkyl,
bstituted or unsubstituted heteroalkyl, R26—substituted or unsubstituted cycloalkyl,
R26—substituted or unsubstituted heterocycloalkyl, R26—substituted or unsubstituted aryl, or
R26—substituted or unsubstituted heteroaryl. R26 is halogen, —CN, —CF3, —OH, —NH2, —COOH,
—COOR25, —CH2COOR25, —CH2COOH, R27—substituted or unsubstituted alkyl, R27—substituted or
unsubstituted heteroalkyl, bstituted or unsubstituted cycloalkyl, R27—substituted or
unsubstituted heterocycloalkyl, R27—substituted or unsubstituted aryl, or R27—substituted or
unsubstituted heteroaryl. In one embodiment, R26 is n, —CN, —CF3, —OH, —NH2, —COOH,
R27—substituted or tituted C1—C10 (e.g., C1—C6) alkyl, R27—substituted or unsubstituted 2 to
10 membered (e.g., 2 to 6 membered) heteroalkyl, R27—substituted or unsubstituted C3—C3 (e.g.,
C5—C7) cycloalkyl, R27—substituted or unsubstituted 3 to 8 membered (e.g., 3 to 6 membered)
heterocycloalkyl, R27—substituted or unsubstituted C5—C3 (e.g., C5—C6) aryl, or R27—substituted or
unsubstituted 5 to 8 membered (e.g., 5 to 6 membered) heteroaryl. R27 is halogen, -CN, —CF3,
—OH, —NH2, —COOH, —COOR25, —CH2COOR25, —CH2COOH, R23—substituted or tituted
alkyl, R23—substituted or unsubstituted heteroalkyl, R23—substituted or unsubstituted cycloalkyl,
bstituted or unsubstituted heterocycloalkyl, R2g—substituted or unsubstituted aryl, or
R23—substituted or unsubstituted heteroaryl. In one embodiment, R27 is halogen, —CN, —CF3, —OH,
—NH2, —COOH, R23—substituted or unsubstituted C1—C10 (e.g., C1—C6) alkyl, R23—substituted or
WO 30150
unsubstituted 2 to 10 membered (e. g., 2 to 6 ed) alkyl, R2g—substituted or
unsubstituted C3—C8 (e. g., C5—C7) cycloalkyl, R2g—substituted or tituted 3 to 8 membered
(e. g., 3 to 6 membered) heterocycloalkyl, R2g—substituted or unsubstituted C5—C8 (e. g., C5—C6)
aryl, or R2g—substituted or tituted 5 to 8 membered (e. g., 5 to 6 membered) heteroaryl. R28
is halogen, —CN, —CF3, —OH, —NH2, —COOH, —COOR25, —CH2COOR25, —CH2COOH, unsubstituted
alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl,
unsubstituted aryl, or unsubstituted heteroaryl.
In one ment, R26 and/or R27 are substituted with a substituent group, a size—
limited substituent group or a lower substituent group. In another embodiment, R27 and R28 are
ndently halogen, —CN, —CF3, —OH, —NH2, —COOH, —COOR25, —CH2COOR25, —CH2COOH,
tituted C1—C10 (e.g., C1—C6) alkyl, unsubstituted 2 to 10 membered (e. g., 2 to 6 membered)
heteroalkyl, unsubstituted C3—C8 (e.g., C5—C7) cycloalkyl, unsubstituted 3 to 8 membered (e. g., 3
to 6 membered) heterocycloalkyl, unsubstituted C5—C8 (e. g., C5—C6) aryl, or unsubstituted 5 to 8
ed (e.g., 5 to 6 membered) heteroaryl.
[0067] In one embodiment, each R5, R6, R7, R8, R83 R9, R10, R11, R12, R13, R14, R15, R16, R17,
R18, R19, R20, R21, R22, R23, R24, and R25 may be the same or different and may each
independently be an alkyl, and particularly a C1_20 alkyl, more particularly a C140 alkyl, and even
more particularly a C1_4 alkyl, and even more particularly, a methyl, an ethyl, or a propyl.
In one embodiments R8 and R8, are independently hydrogen or an unsubstituted alkyl
(e. g. an unsubstituted C140 alkyl). R8, may also be hydrogen. R8 may be methyl.
In one embodiment, R9 is —COOH, —COOR25, —CH2COOR25, or —CH2COOH. R9 may
also be —COOR25 or —CH2COOR25. In certain embodiments, R9 is —COOR25. In one related
embodiment, R25 is an unsubstituted C1-C10 alkyl, such as methyl.
2012/067633
In one embodiment, R1 and R3 may each independently be —COz-CH3, or H
E COZCH3
and R2 and R4 may each independently be —CF3, or
g CH2C02CH3
In one embodiment, the metalloporphyrin compound may have the formula:
COZCH3
F30 CFa
COZCHa (IV),
(V), or
(VI).
In another specific ment, R1, R2, R3, and R4 may each independently be
§‘<\ +\N—/ é \—/
k k +T
or (CH2)5CH3_
In one embodiment, the metalloporphyrin compound of the invention may have the
formula:
\_N/T/\N_/
I (
\ \J
J K
(VIII), or
H3C(Hzc)5
HSC(HZC)5—N
(IX).
In one embodiment, each substituted group described in the nds above (e.g.,
Formulae (I)-(IX)) is substituted with at least one substituent group. More specifically, in one
embodiment, each substituted alkyl, tuted heteroalkyl, substituted cycloalkyl, substituted
heterocycloalkyl, substituted aryl, substituted heteroaryl, described in the compounds above
(e. g., Formulae (I)-( IX)) are substituted with at least one substituent group. In one embodiment,
at least one or all of these groups are tuted with at least one size—limited substituent group.
Alternatively, at least one or all of these groups are substituted with at least one lower substituent
group. .
In one embodiment of the compounds described above (e. g., ae (I)—( IX)) each
substituted or unsubstituted alkyl is a substituted or unsubstituted C1—C20 alkyl, each substituted
or tituted heteroalkyl is a substituted or tituted 2 to 20 membered heteroalkyl, each
substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3—C8 cycloalkyl, and
each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8
membered heterocycloalkyl.
In one embodiment, each substituted or unsubstituted alkyl is a substituted or
unsubstituted C1—C8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or
unsubstituted 2 to 8 membered alkyl, each substituted or unsubstituted lkyl is a
substituted or unsubstituted C5—C7 cycloalkyl, and each substituted or unsubstituted
heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered cycloalkyl.
In one embodiment, the compound has the structure of one of Formulae (X)—(XV),
Further to this embodiment, R13, R23, R33, and R4a are independently -(CH2)mCHZOX1
or -(CH2CHZO)nX1; m is 1-6, preferably 1-4, more preferably 1 or 2; n is 3-50, preferably 3-10,
more preferably 3, 4 or 5; X1 is substituted or unsubstituted C142 alkyl, preferably unsubstituted
C142 alkyl (straight chain or branched), more preferably C1_g alkyl, even more preferably C1_4
alkyl; M is a metal (is manganese, iron, , copper, nickel, or zinc); and each A is,
independently, hydrogen or an electron withdrawing group. Each R13, R23, R33, and R4a can be
the same. The terms “electron withdrawing group,” “EWG” and the like refer, in the usual and
customary sense, to an atom or functional group that removes electron density from a system
(e.g., a pi-system) thus making the system more electrophilic.
WO 30150 2012/067633
In one embodiment, the compound has the structure of one of Formulae (XVI)—(XVII),
Further to this embodiment, at least one of Ru, or R16, R2], or R26, R3b or R36, and R4], or
R40 is, independently, -(CH2)pCHZOX2 or -(CH2CHZO)qX2; the other one of Ru, or R16, R2], or
R26, R3b or R36, and R4], or R40 is, independently, a C142 alkyl (straight chain or branched),
preferably a C1_g alkyl, more preferably a C1, C2, C3 or C4 alkyl; p is l-6, ably 104, more
ably 1 or 2; q is 3—50, preferably 3—10, more preferably 3, r or 5; X2 is substituted or
unsubstituted C142 alkyl, ably is C142 alkyl (straight chain or branched), more preferably
C1_g alkyl, even more preferably C1_4 alkyl; M is a metal (e.g. is manganese, iron, cobalt, copper,
nickel, or zinc); and each A is, independently, hydrogen or an electron withdrawing group.
Advantageously, each Rlb, R16, R21), R26, R3b, R3c, R4], and R4c can be the same and is
—(CH2CHZO)qX2.
When the nd is of Formulae (X)—(XVII), each A is, independently, hydrogen or
an electron withdrawing group, for example, a halogen (e. g., Cl, Br or F), N02, or CHO,
preferably each A is hydrogen or halogen, more preferably at least one A is halogen and the
remaining A's are hydrogen, still more preferably l-4 A's are, independently, C1 or Br and the
remaining A's are hydrogen. M is metal ed from the group consisting of manganese, iron,
copper, cobalt, nickel and zinc (preferably manganese).
In one embodiment, each substituted group described in the compounds with structure
of Formulae (X)—(XVII) is substituted with at least one substituent group. More specifically, in
one ment, each substituted alkyl, substituted heteroalkyl, substituted cycloalkyl,
substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, described in the compounds
with structure of Formulae VII) are substituted with at least one substituent group. In one
embodiment, at least one or all of these groups are substituted with at least one size-limited
substituent group. Alternatively, at least one or all of these groups are tuted with at least
one lower substituent group.
In one embodiment of the compounds with structure of Formulae (X)—(XVH), each
substituted or unsubstituted alkyl is a substituted or unsubstituted C1—C20 alkyl, each substituted
or unsubstituted heteroalkyl is a substituted or tituted 2 to 20 membered heteroalkyl, each
substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3—C8 cycloalkyl, and
each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8
ed heterocycloalkyl. In one ment, each substituted or unsubstituted alkyl is a
substituted or unsubstituted C1—C8 alkyl, each substituted or tituted heteroalkyl is a
substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted
cycloalkyl is a tuted or unsubstituted C5—C7 cycloalkyl, and each substituted or
unsubstituted heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered
heterocycloalkyl.
[0084] Further to any embodiment disclosed herein, the compound can be formed with a
rion Z, exemplified but not limited as follows for compounds of Formulae (X)-(XVII):
R13 R13
I I
N“r Z‘ N’r
\ \
In one embodiment, a compound of Formula (I) can be formed with a counterion Z. In
one embodiment, a compound of a (II) can be formed with a counterion Z. In one
embodiment, a compound of Formula (III) can be formed with a counterion Z. In one
embodiment, a compound of Formula (IV) can be formed with a counterion Z. In one
WO 30150
embodiment, a compound of Formula (V) can be formed with a counterion Z. In one
embodiment, a compound of Formula (VI) can be formed with a counterion Z. In one
embodiment, a compound of Formula (VII) can be formed with a counterion Z. In one
embodiment, a compound of Formula (VIII) can be formed with a counterion Z. In one
embodiment, a compound of Formula (IX) can be formed with a rion Z. In one
embodiment, a compound of Formula (X) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XI) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XII) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XIII) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XIV) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XV) can be formed with a counterion Z. In one
embodiment, a compound of a (XVI) can be formed with a counterion Z. In one
embodiment, a compound of Formula (XVII) can be formed with a counterion Z.
The counterion Z is an anion, e. g., halogen ide, bromide, iodide), an organic
anion base (e. g., acetate, and the like), an inorganic base (e.g., sulfide, sulfate, carbonate,
phosphate), or the like.
In one embodiment, the compound has the ure of Formula (I). In one
embodiment, the compound has the structure of Formula (II). Where the compound has the
structure of Formula (II), in one embodiment the metal is ese, iron, cobalt, copper,
nickel, or zinc. In one embodiment, the metal is manganese.
Further to embodiments Where the compound has the structure of a (I) or
R5—N)+\N—R6
Formula (II), in one embodiment R1, R2, R3, and R4 are each \=/ ; and
R5 and R6 are independently unsubstituted alkyl (e. g., tituted C1—C10 alkyl). R5 and R6
may independently be unsubstituted C1—C6 alkyl. R5 and R6 may independently be unsubstituted
C1-C5 alkyl. R5 and R6 may independently be unsubstituted C1-C4 alkyl. R5 and R6 may
independently be unsubstituted C1—C3 alkyl. R5 and R6 may independently be tituted C1—
C2 alkyl. In one ment, the compound has the structure of a (VII) following:
/— \_J _\N /+
(VII).
Further to any embodiment sed herein, in one embodiment the chemical threat
agent causes seizures, neuropathology, or both seizures and neuropathology. In one
ment, the chemical threat agent causes seizures. In one embodiment, the chemical threat
agent causes neuropathology.
In one embodiment, the chemical threat agent is a nerve agent. The terms “nerve
agent” and the like refer, in the usual and customary sense, to compounds that t the
mechanism by which nerves transfer messages. In one embodiment, the nerve agent disrupts
3’ “
nerve signals by inhibiting acetylcholinesterase. The terms “anti-acetylcholinesterase, anti-
cholinesterase” and the like refer, in the usual and ary meaning, to an agent which can
inhibit the activity of acetylcholinesterase (e. g., upon exposure to a al threat agent).
Acetylcholinesterase, as well known in the art, hydrolyzes the neurotransmitter acetylcholine to
afford an acetyl group and choline. In one embodiment, inhibition of acetylcholinesterase results
in sed levels and on of acetylcholine in a subject. The effects of anti-cholinesterases
on the autonomic nervous system can include bradycardia, hypotension, hypersecretion,
bronchoconstriction, GI tract hypermotility, and decreases intraocular pressure. Action at the
neuromuscular junction can include prolonged muscle ction. The effect of anti—
cholinesterases can include seizure and/or neuropathology.
In one embodiment, the chemical threat agent is sarin, parathion, aldicarb or tetramine
(TETS). In one embodiment, the chemical threat agent targets the blood. In one embodiment,
the chemical threat agent is cyanide, sodium fluoroacetate, arsenic trioxide or strychnine.
In one embodiment, the effect of treating a subject suffering from exposure to a
chemical threat agent lasts for a period of time (i.e., tive period”) following administration
of a compound disclosed . In one embodiment, the effect lasts for an effective period of at
least 10, 20, 30, 40, 50, 60, 90, 120, 150, 180, or 240 minutes, or even longer. In one
embodiment, the effect lasts for an effective period of at least 1, 2, 3, 4, 5, 6, 12, or even 24
hours. In one embodiment, the effect lasts for at least 90 minutes.
In one embodiment, a compound with ure of Formulae (I)—(XVII), e. g., a
(VII), is administered (e. g., erally or topically) at a dosage in the range of about 0.01 to
50 mg/kg/day, preferably, 0.1 to 10 mg/kg/day, more preferably 0.1 to 6 mg/kg/day. In one
embodiment, dosage is about 1, 3, 5, 7, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900,
1000 mg/day, or even more, in an adult.
In one embodiment, a nd with structure of Formulae (I)—(XVII) is administered
in ation with any antidote or prophylactic to a chemical threat agent (e.g., nerve agent
such as anti-cholinesterase agent, a GABA-agent or a metabolic poison).
The antidote or prophylactic can be an anticholinergic, an anti-seizure agent, or an
acetylcholinesterase reactivating agent, or a combination of one, two or three of an
anticholinergic, an anti-seizure agent, or an acetylcholinesterase reactivating agent. The
anticholinergic can be an anticholinergic disclosed herein or known in the art. The anti-seizure
agent can be an anti-seizure agent disclosed herein or known in the art. The cholinesterase
reactivating agent can be an acetylcholinesterase reactivating agent disclosed herein or known in
the art. In one embodiment, the antidote or prophylactic agent restores acetylcholinesterase
activity that is inhibited by the chemical threat agent.
[0096] In another aspect, there is provided a method for reducing brain injury in a subject in
need thereof. The method es administering to the subject an effective amount of a
compound ed from any of ae (I)—(XVII), as disclosed above. In one embodiment,
the compound has the structure of Formula (VII).
In one embodiment, the brain injury results from seizure. The seizure can result from
exposure to a chemical threat agent. In one embodiment, the brain injury is cognitive
dysfunction. The terms “cognitive dysfunction” and the like refer, in the usual and customary
sense, to a loss of intellectual functions such as thinking, remembering, reasoning, and the like.
In one embodiment, the brain injury results from seizure, the seizure s from
exposure to a chemical threat agent, and the chemical threat agent is an anti-cholinesterase agent.
[0099] In one embodiment, a compound with ure of Formulae (I)—(XVII), e. g., Formula
(VII), is administered (e. g., erally or topically) at a dosage in the range of about 0.01 to
50 mg/kg/day, preferably, 0.1 to 10 day, more preferably 0.1 to 6 mg/kg/day. In one
embodiment, dosage is about 1, 3, 5, 7, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900,
1000 mg/day, or even more, in an adult.
Further to the method for treating a subject suffering from exposure to a chemical threat
agent, or the method for ng brain injury in a subject in need f, in one embodiment
the method further includes administering to the subject an anticholinergic agent. The terms
“anticholinergic (noun), 3’ 6‘anticholinergic agent” and the like refer, in the usual and customary
sense, to compounds that block the action of the neurotransmitter acetylcholine in the l and
peripheral nervous system. Exemplary anticholinergics at the muscarinic receptor, i.e.,
“antimuscarinic agents” as known in the art, include atropine, benztropine, ipratropium,
oxitropium, tiotropium, glycopyrrolate, oxybutinin, tolterodine, chlorphenamine,
hydramine, dimenhydrinate, and the like. Exemplary anticholinergics at the nicotinic
receptor, i.e., “antinicotinic agents” as known in the art, include bupropion, hexamethonium,
tubocurarine, dextromethorphan, mecamylamine, doxacurium, and the like.
[0101] In one embodiment, the anticholinergic agent is atropine. As well known in the art,
atropine competitively blocks acetylcholine (ACh) at muscarinic receptor sites by competing for
the muscarinic receptors. Thus, blockade of muscarinic receptor ameliorates increased levels of
acetylcholine. In one embodiment, the preferred anticholinergic is less toxic than atropine in a
human subject. Toxicity can be assessed by a variety of methods known in the art including
calculation of LD50, the toxic dose for 50% of a population.
In one ment, an anticholinergic agent is administered at a dosage of about 0.1,
0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 4.0, 5.0, 6.0 mg or even greater in an adult.
Administration can be by bolus or sequential administration of smaller ts of anticholinergic
agent, e. g., administration in rapid succession, e. g., 2, 4, 6, 8, 10, 15, 20 minutes part.
Administration of olinergic agent can be repeated as needed to prevent or treat ms
of parasympathomimetic activity, coma, and/or cardiovascular collapse, as known in the art. In
one embodiment, atropine is administered at a dosage of about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5,
2.0, 2.5, 3.0, 4.0, 5.0, 6.0 mg or even greater in an adult. Dosages for children can be adjusted as
ary. For example, dosages for infants generally less than six months of age can be about
0.10, 0.15, 0.20, 0.25, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75 mg, or more.
Dosages can be administered by sequential administration of smaller aliquots of anticholinergic
agent, e. g., atropine. s for infants and children weighting 15 to 40 pounds may be 0.10,
0.20, 0.30, 0.40, 0.50, 0.60, 0.70, 0.80, 0.90, 1.00, 1.10, 1.20, 1.30, 1.40, 1.50 mg, or even
greater. Dosages for children weighting 40 to 90 pounds may be about 0.20, 0.40, 0.60, 0.80,
1.00, 1.20, 1.40, 1.60, 1.80, 2.00, 2.20, 2.40, 2.60, 2.80, 3.00 mg, or even greater.
In one embodiment, the compound of Formula (VII) is administered in combination
with an anticholinergic. In one embodiment, the anticholinergic is atropine.
Further to the method for treating a t suffering from re to a chemical threat
agent, or the method for reducing brain injury in a subject in need thereof, in one embodiment
the method further includes administering to the subject an anti-seizure agent. The term “anti-
seizure agent” and the like refer, in the usual and customary sense, to compounds useful to
suppress the rapid and excessive firing of neurons as a preliminary to, or duration, a seizure.
Exemplary anti-seizures agents include the benzodiazepines: clobazam, clonazepam, clorazepate,
diazepam, midazolam, lorazepam, and the like. In one embodiment, the anti-seizure agent is a
benzodiazepine. In one embodiment, the anti-seizure agent is diazepam. In one embodiment,
the eizure agent is midazolam.
[0105] In one ment, an anti-seizure agent is administered at a dosage of about 1, 2, 3, 4,
, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 mg, or even greater, in an adult. In one
embodiment, an anti-seizure agent is administered at a dosage of about 10, 50, 100, 200, 300
400, 500 mg/kg, or even greater, in a child. In one embodiment, administration is intravenous
(iv) or intramuscular (i.m.) In one embodiment, administration is intramuscular (i.m.) In one
embodiment, the anti-seizure agent is diazepam which is administered at a dosage of about 1, 2,
3, 4, 5, 6, 7, 8, 9, 10, l2, l4, l6, 18, 20 mg, or even greater, in an adult. In one embodiment, the
anti-seizure agent is midazolam which is administered at a dosage of about 1, 2, 3, 4, 5, 6, 7, 8, 9,
, l2, l4, l6, 18, 20 mg, or even greater, in an adult.
In one embodiment, the compound of a (VII) is stered in combination
with an eizure agent. In one embodiment, the anti-seizure agent is am. In one
embodiment, the anti-seizure agent is midazolam.
Further to the method for treating a subject suffering from re to a chemical threat
agent, or the method for reducing brain injury in a subject in need thereof, in one embodiment
the method further includes administering to the subject in combination an olinergic agent
and an anti-seizure agent, as disclosed herein.
In one ment, the administered nd has the structural of Formula (VII),
and the method further includes administering to the subject in combination an anticholinergic
agent and an anti-seizure agent, as disclosed herein. In one embodiment, the anticholinergic is
atropine. In one embodiment, the anti-seizure agent is diazepam. In one embodiment, the anti-
seizure agent is lam.
In one embodiment, administration of a compound with structure of Formulae (I)-
(XVII), e. g., Formula (VII), in combination with an anticholinergic agent and an anti-seizure
agent results in a synergistic benefit to the subject. The terms “synergistic, 3’ 6‘synergistic
benefit,” “synergistic effect, 3’ CCsynergistic therapeutic effect, 3’ 33synergistically effective amount”
and the like in the context of co—administration of compounds described herein refer to a more
than additive (e.g., supra—additive) se (e.g., biological se) when two or more
compounds are administered with respect to the summed effects upon administration of each
compound in the e of the other compound or nds. For example, if two nds
provide a synergistic therapeutic effect, then the therapeutic effect observed upon co—
administration of both compounds is greater than the summed observed therapeutic effects when
either compound is administered in the absence of the other compound. Likewise, a first amount
of a first nd and a second amount of a second nd together provide a
synergistically effective amount where the therapeutic effect observed upon co-administration of
both nds is greater than the summed observed therapeutic effects when either compound
is administered in the absence of the other compound. Where a synergistic benefit is ed,
the pharmaceutically active agents are provided in a combined synergistic amount.
In one embodiment, administration of a compound with structure of Formulae (I)-
(XVII), e. g., Formula (VII), in combination with an anticholinergic agent and an anti-seizure
agent results is r effectiveness (i.e., synergistic benef1t wherein the compound and agents
are provided in a combined synergistic amount) relative to the summed effects of 1) treatment by
administration of a structure of Formulae (I)-(XVII) alone, and 2) treatment with an
anticholinergic agent in combination with an anti-seizure agent alone. In one ment, the
synergistic benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%,
65%, 70%, 75%, 80%, 85%, 90%, or 95%.
[0111] In one embodiment, a compound with structure of Formulae (I)-(XVII), e. g., Formula
(VII), is administered (e. g., parenterally or lly) at a dosage in the range of about 0.01 to
50 mg/kg, preferably 0.1 to 10 mg/kg, more preferably 1.0 to 6 mg/kg. The effect of this
WO 30150
administration alone can be compared with the combined effect upon administration of an
anticholinergic agent administered at a dosage of about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0, 2.5,
3.0, 4.0, 5.0, or 6.0 mg, preferably in the range of about 2-6 mg, and an anti-seizure agent
administered at a dosage of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 mg,
ably in the range of about 10-30 mg. The effect of the ternary combination of a nd
with structure of Formulae (I)—(XVII), the anticholinergic agent and the anti-seizure agent can be
compared with the summed s of the administration of a compound with structure of
Formulae (I)-(XVII) alone, and the effect of the administration of the binary combination of
anticholinergic agent and anti-seizure agent alone to quantitate a synergistic benefit. In one
ment, the synergistic benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%,
50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%.
In one embodiment, a compound with structure of Formulae (I)-(XVII), e. g., Formula
(VII), is administered (e. g., erally or topically) at a dosage in the range of about 0.01 to
50 mg/kg, preferably 0.1 to 10 mg/kg, more preferably 1.0 to 6 mg/kg. An anticholinergic agent,
preferably atropine, may be administered at a dosage of about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0,
2.5, 3.0, 4.0, 5.0, or 6.0 mg, preferably in the range of about 2-6 mg. An anti-seizure agent,
preferably diazepam or midazolam, may be administered at a dosage of about 1, 2, 3, 4, 5, 6, 7,
8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 mg, preferably in the range of about 10—30 mg. The effect
of the ternary combination of a compound with ure of Formulae (I)-(XVII), e.g., Formula
(VII), the anticholinergic agent and the anti-seizure agent can be ed with the summed
effects of the administration of a compound with structure of Formulae (I)—(XVII), e.g., Formula
(VII), alone, and the effect of the administration of the binary combination of anticholinergic
agent and eizure agent alone to quantitate a synergistic benefit. In one embodiment, the
istic benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%,
65%, 70%, 75%, 80%, 85%, 90%, or 95%.
In one embodiment, a compound with structure of Formula (VII) is administered (e.g.,
parenterally or topically) at a dosage in the range of about 0.01 to 50 mg/kg, preferably 0.1 to
mg/kg, more preferably 1.0 to 6 mg/kg. Atropine may be stered at a dosage of about
0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 4.0, 5.0, or 6.0 mg, preferably in the range of about
2-6 mg. Diazepam may be administered at a dosage of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20,
, 30, 35, 40, 45, 50 mg, preferably in the range of about 10—30 mg. The effect of the ternary
combination of a compound with structure of Formula (VII), atropine and diazepam can be
compared with the summed effects of the administration of a compound with structure of
Formula (VII) alone, and the effect of the administration of the binary combination of ne
and diazepam alone to quantitate a istic benefit. In one embodiment, the synergistic
benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%,
75%, 80%, 85%, 90%, or 95%.
In one embodiment, a compound with structure of Formula (VII) is administered (e.g.,
parenterally or topically) at a dosage in the range of about 0.01 to 50 mg/kg, preferably 0.1 to
mg/kg, more preferably 1.0 to 6 mg/kg. ne may be administered at a dosage of about
0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 4.0, 5.0, or 6.0 mg, preferably in the range of about
2-6 mg. Midazolam may be administered at a dosage of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20,
25, 30, 35, 40, 45, 50 mg, preferably in the range of about 10—30 mg. The effect of the ternary
combination of a compound with structure of Formula (VII), atropine and midazolam can be
ed with the summed effects of the administration of a compound with structure of
a (VII) alone, and the effect of the administration of the binary combination of atropine
and midazolam alone to quantitate a synergistic benefit. In one embodiment, the synergistic
benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%,
75%, 80%, 85%, 90%, or 95%.
Further to the method for treating a subject suffering from exposure to a chemical threat
agent, or the method for reducing brain injury in a subject in need thereof, in one embodiment
the method further includes administering to the subject an acetylcholinesterase reactivating
agent. The terms “acetylcholinesterase vating agent” the like refer, in the usual and
customary sense, to compounds useful to regenerate catalytic activity at an acetylcholinesterase
site which has become deactivated due to al reaction, e. g., with a al threat agent
such as an acetylcholinesterase inhibitor. See e. g., Luo, C., et al., 2007, Biochemistry 46: 1 1771—
11779. Useful acetylcholinesterase reactivating agents are known in the art and include HI—6
([(E)-[1-[(4-carbamoylpyridiniumyl)methoxymethyl]pyridinylidene]methyl]-
oxoazaniumdichloride), pralidoxime (2-pyridine me methyl de (2-PAM), and
obidoxime (1,1'-[oxybis(methylene)]bis{4-[(E)- (hydroxyimino)methyl] pyridinium}), and the
like. See e.g., Dawson, R.M., 1994, J. Appl. Toxicol. 14:317—33 1; Koplovitz, I. & Stewart, J.R.,
1994, Toxicol. Lett. 70:269—279; Marrs, TC, 1993, Pharmac. Ther. 58:51—66; Rousseaux, C.G. &
Dua, A.K., 1989, Can. J. Physiol. Pharmacol, 67:1183—1189. In one embodiment, the methods
provided herein e administeting a nd of Formulae (I)-(XVII) and 2-PAM (e.g,. in a
combined synergistic amount and achieving a synergistic benefit).
2012/067633
In one embodiment, acetylcholinesterase reactivating agent is administered at a dosage
of about 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100 mg/kg, or even greater, in an adult,
preferably about 30 mg/kg. In one embodiment, acetylcholinesterase reactivating agent is
administered at a dosage of about 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, or 100 mg/kg,
or even greater, in a child, preferably 20-50 mg/kg. In one embodiment, initial administration of
acetylcholinesterase reactivating agent is followed by a maintenance infusion of 1-20 mg/kg/hr,
preferably 5-10 hr. Initial administration of acetylcholinesterase reactivating agent can be
by any means, e. g., intravenous, intramuscular, or subcutaneous. In one embodiment, initial
stration is intravenous as a continuous infusion at about 100, 200, 300, 400, 500, 600, 700
mg/hr, or even r. In one embodiment, administration of acetylcholinesterase reactivating
agent is sequential, wherein administration of aliquots of acetylcholinesterase reactivating agent
takes place over a time span of, e.g., 5, 10, 15, 20, 25, 30, 40, 50, 60 minutes, or even longer. In
a preferred embodiment, initial administration of acetylcholinesterase reactivating agent is
intramuscular. In one embodiment, the acetylcholinesterase reactivating agent is pralidoxime.
In one embodiment, pralidoxime is administered at a dosage of about 5, 10, 15, 20, 25, 30, 35,
40, 50 mg/kg, or even r, in an adult. In a preferred embodiment, administration of
pralidoxime is intramuscular, and the dosage of pralidoxime is 1-2g.
In one embodiment, stration of a compound with structure of ae (I)-
(XVII), e.g., Formula (VII), in combination with an anticholinergic agent, an anti-seizure agent,
and an acetylcholinesterase reactivating agent results in a synergistic benefit relative to the
summed effects of 1) treatment by administration of a structure of Formulae (I)-(XVII) alone,
and 2) treatment alone with an anticholinergic agent in ation with an anti-seizure agent in
further combination with an acetylcholinesterase vating agent. In one embodiment, the
synergistic benefit is at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%,
65%, 70%, 75%, 80%, 85%, 90%, or even 95%.
In one embodiment, a compound with structure of Formulae (I)-(XVII), e. g., Formula
(VII), is administered (e. g., erally or topically) at a dosage in the range of about 0.01 to
50 mg/kg, preferably 0.1 to 10 mg/kg, more preferably 1.0 to 6 mg/kg. The effect of this
administration alone can be compared with the combined effect upon administration of an
anticholinergic agent administered at a dosage of about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 1.5, 2.0, 2.5,
3.0, 4.0, 5.0, or 6.0 mg, preferably in the range of about 2—6 mg, an eizure agent
administered at a dosage of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 mg,
preferably in the range of about 10-30 mg, and an acetylcholinesterase reactivating agent at a
dosage of about 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100 mg/kg. The effect of the
quarnary combination of a compound with structure of Formulae (I)-(XVII), e. g., Formula (VII),
the anticholinergic agent, the anti-seizure agent and the acetylcholinesterase reactivating agent
can be compared with the summed effects of the administration of a compound with structure of
Formulae (I)-(XVII) alone, and the effect of the administration of the ternary combination of
anticholinergic agent, anti-seizure agent, and acetylcholinesterase reactivating agent alone to
quantitate a synergistic benefit. In one embodiment, the synergistic benefit is at least 5%, 10%,
%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or
95%. In one embodiment, the compound has the structure of Formula (VII), the anticholinergic
agent is atropine, the anti-seizure agent is am, and the acetylcholinesterase reactivating
agent is pralidoxime. In one ment, the compound has the structure of Formula (VII), the
anticholinergic agent is atropine, the anti-seizure agent is midazolam, and the
acetylcholinesterase reactivating agent is pralidoxime.
In one embodiment, r to the method for treating a subject suffering from exposure
to a chemical threat agent, or the method for reducing brain injury in a subject in need thereof,
the method further includes administering to the subject a compound of Formulae (I)-(XVII),
e. g., Formula (VII), and an cholinesterase reactivating agent in combination with an
anticholinergic agent and an anti-seizure agent. In one ment, the administered nd
has the structural of a (VII), and the method further includes administering to the subject
in ation an acetylcholinesterase reactivating agent, an anticholinergic agent and an anti-
seizure agent, as sed herein. In one embodiment, the anticholinergic is atropine. In one
embodiment, the anti-seizure agent is diazepam. In one embodiment, the anti-seizure agent is
midazolam. In one embodiment, the acetylcholinesterase reactivating agent is pralidoxime.
Further to the method for treating a subject suffering from exposure to a chemical threat
agent, or the method for reducing brain injury in a subject in need thereof wherein the brain
injury results from seizure and the seizure results from re to a chemical threat agent, in
one embodiment administration of a compound haVing the structure of any one of Formulae (I)-
(XVII), e.g., Formula (VII), occurs prior to the exposure to the chemical threat agent. In one
embodiment, stration of a compound of any one of Formulae VII) occurs at least 10,
20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120 min, or even longer, prior to exposure to the
chemical threat agent. In one embodiment, administration of a compound of any one of
Formulae (I)—(XVII) occurs at least 30 min prior to exposure to the chemical threat agent. In one
ment, administration of a compound of any one of Formulae (I)-(XVII) occurs at least 60
WO 30150
min prior to exposure to the chemical threat agent. In one embodiment, administration of a
compound of any one of Formulae (I)-(XVII) occurs at least 90 min prior to exposure to the
al threat agent. In one embodiment, the compound has the structure of a (VII).
Further to the method for reducing brain injury in a subject in need thereof, the effect of
reducing the brain injury lasts for at least 30, 60, 90,120, 180, 240, 300 min, or even ,
ing administration of a compound of Formulae (I)-(XVII). In one embodiment, the effect
of reducing the brain injury lasts at least 30 min. In one embodiment, the effect of reducing the
brain injury lasts at least 60 min. In one embodiment, the effect of reducing the brain injury lasts
at least 90 min. In one embodiment, the effect of reducing the brain injury lasts at least 120 min.
In one ment, the compound has the structure of Formula (VII).
Further to the method for reducing brain injury in a subject in need thereof, in one
embodiment the compound has the structure of Formula (I) or Formula (II). In one embodiment,
the compound has the structure of Formula (II). In one ment, the metal is manganese,
iron, cobalt, copper, nickel, or zinc. In one embodiment, the metal is manganese. In one
embodiment, for the compound with structure of Formula (I) or Formula (II), R1, R2, R3, and R4
JUVV‘
R5—N)+\N—R6
are each \=/
; and R5 and R6 are independently unsubstituted alkyl. In one
embodiment, the compound has the structure of Formula (VII).
III. Pharmaceutical Compositions
The compounds described above, metal bound and metal free forms, can be formulated
into pharmaceutical itions suitable for use in the t methods. Such compositions
e the active agent (metalloporphyrin compounds) together with a pharmaceutically
able carrier, excipient or diluent. The composition can be present in dosage unit form for
example, tablets, capsules or suppositories. The composition can also be in the form of a sterile
solution, e.g., a solution suitable for injection (e. g., subcutaneous, i.p. or iv.) or nebulization.
Compositions can also be in a form suitable for ophthalmic use. The invention also includes
compositions formulated for topical administration, such compositions taking the form, for
example, of a lotion, cream, gel or ointment. The concentration of active agent to be included in
the ition can be selected based on the nature of the agent, the dosage regimen and the
result sought. The compounds can also be encapsulated in lysosomes and thereby targeted to
enhance delivery.
In one embodiment, the metalloporphyrin compound may form part of a
pharmaceutical ition. The pharmaceutical composition may e a metallophorphyrin
compound, as disclosed , and a ceutically acceptable excipient. A
"pharmaceutically acceptable excipien " includes pharmaceutically and physiologically
acceptable, organic or inorganic carrier substances suitable for enteral or parenteral
administration that do not deleteriously react with the active agent. Suitable pharmaceutically
able carriers include water, salt ons (such as Ringer's solution), alcohols, oils,
gelatins, and carbohydrates such as lactose, amylose or , fatty acid esters,
hydroxymethylcellulose, and polyvinyl pyrrolidone. Such preparations can be sterilized and, if
desired, mixed with auxiliary agents such as lubricants, preservatives, izers, wetting agents,
emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances
and the like that do not deleteriously react with the active agent.
[0125] In one embodiment, the treatment compound (e. g., metalloporphyrin compounds or
metalloporphyrin catalytic antioxidant compositions as set forth herein) forms part of a
pharmaceutical composition, wherein said pharmaceutical composition ses the treatment
compound and a pharmaceutical acceptable excipient. In one embodiment, the pharmaceutical
composition includes a permeabilizer (e. g., a late, a fatty acid, or a metal chelator).
[0126] The pharmaceutical composition can be formulated for any route of administration,
including enteral, oral, sublingual, buccal, parenteral, , asal, pulmonary, ,
intravaginal, transdermal, and topical routes. Parenteral administration includes, but is not
limited to, intravenous, intramuscular, subcutaneous, intradermal, intraperitoneal, intrastemal,
intraarterial injection and infusion.
[0127] The pharmaceutical composition can be formulated for immediate release or ed
release, e. g., modif1ed, sustained, extended, delayed, or pulsatile release, using known methods
and excipients.
In one ment, the pharmaceutical composition is formulated as a topical
composition, an injectable composition, an inhalant, a sustained release composition, or an oral
composition. The treatment compound is preferably formulated for eral administration,
e. g., by subcutaneous injection. If subcutaneous or an alternative type of administration is used,
the compounds may be derivatized or formulated such that they have a protracted profile of
action.
In another embodiment, the pharmaceutical composition is formulated a targeted
micelle, a degradable ric dosage form, a porous microsphere, a polymer scaffold, a
liposome, or a hydrogel.
The treatment compound may be formulated according to known methods to prepare
pharmaceutically useful compositions. An exemplary formulation would be one that is stable
and reconstituted with an appropriate diluent or an aqueous solution of high purity with optional
pharmaceutically acceptable carriers, preservatives, ents or stabilizer. See e.g.,
Remington, 1980, PHARMACEUTICAL SCIENCES, 16th n. The pharmaceutical ition
may include a ceutically acceptable buffer to achieve a le pH for stability and for
administration.
For parenteral administration, the treatment compound can be formulated in a unit
dosage injectable form (solution, suspension, or emulsion) with a pharmaceutically acceptable
carrier. Preferably, one or more pharmaceutically acceptable anti-microbial agents may be
added, such as phenol, m—cresol, benzyl alcohol, and the like as known in the art.
In one embodiment, one or more ceutically acceptable salts (e. g., sodium
chloride), sugars (e. g., mannitol), or other ents (e. g., glycerin) may be added to adjust the
ionic strength or tonicity.
[0133] The dosage of the composition to be stered can be determined without undue
mentation and will be dependent upon various s including the nature of the active
agent (including whether metal bound or metal free), the route of administration, the patient, and
the result sought to be achieved. A suitable dosage of mimetic to be administered (e. g., i.v. or
topically) can be expected to be in the range of about 0.01 to 50 mg/kg/day, preferably, 0.1 to
10 mg/kg/day, more preferably 0.1 to 6 mg/kg/day. For aerosol administration, it is expected
that doses will be in the range of 0.001 to 5.0 mg/kg/day, preferably, 0.01 to 1 mg/kg/day.
Suitable doses will vary, for example, with the compound and with the result sought.
The concentration of compound tation in a solution used to treat
cells/tissues/organs in accordance with the methods disclosed herein can be readily ined
and will vary with the active agent, the cell/tissue/organ and the effect sought.
Certain aspects sed herein can be described in greater detail in the non-limiting
examples that follows.
IV. Examples
The ing examples illustrate n specific embodiments of the invention and are
not meant to limit the scope of the invention.
Embodiments herein are further illustrated by the following examples and detailed
protocols. However, the examples are merely intended to illustrate embodiments and are not to
be construed to limit the scope herein. The contents of all references and published patents and
patent ations cited throughout this application are hereby incorporated by reference.
Example 1. Neuroprotection by a catalytic antioxidant following pilocarpine- and kainate-
induced status ticus
Abstract
Rationale: t wishing to be bound by any theory, it is believed that status
ticus (SE) results in profound oxidative stress and mitochondrial dysfunction. Reactive
oxygen species are mediators of mitochondrial dysfunction that may be active in ing
neuronal death associated with the development of temporal lobe sy (TLE). A goal of this
study was to determine if mitochondrial oxidative stress contributes to hippocampal neuronal
death following SE and r a synthetic catalytic antioxidant administered post-SE would
provide neuroprotection in two chemoconvulsant models.
[0139] Methods: Adult Sprague—Dawley rats were injected with vehicle, kainate (11 mg/kg) or
pilocarpine (340 mg/kg) to initiate SE followed by treatment with vehicle or a synthetic
metalloporphyrin catalytic antioxidant, AEOL 10150 (5 mg/kg, s.c.), beginning 60—90 min post—
SE onset and every 4-6 hr until sacrif1ce at 48 h. Evidence for neuroprotection in the
ampus of chemoconvulsant/AEOL 10150-treated rats was measured at 48 h post-SE using
Fluoro-Jade B staining, a marker of degenerating neurons, and Image J analysis. Oxidative
damage was assessed 24 h post—SE by ement of 3-nitrotyrosine/trysoine (3NT/tyr) and
reduced/oxidized glutathione (GSH/GSSG) ratios, respectively by HPLC methods. The
concentrations ofAEOL 10150 in the rat brain were also determined.
Results: -Jade B staining indicative of cell injury was prevalent hout the
hippocampus of pilocarpine and kainate-treated rats at 48 h post-SE. In pilocarpine-treated rats
receiving AEOL 10150, cell injury decreased by imately 40% in CA1, and 60% in CA3
and hilus. In kainate—treated rats receiving AEOL 10150, cell injury decreased by approximately
40% in CA3 and hilus. AEOL 10150 significantly decreased oxidative stress indices (3—NT/tyr
and GSH/GSSG ) in the hippocampus of pilocarpine-treated rats. Measurement ofAEOL
10150 levels in the brain revealed its ability to achieve neuroprotective concentrations in the
hippocampus and cortex following systemic administration.
Conclusions: These data demonstrate the ability of a catalytic metalloporphyrin
antioxidant to inhibit oxidative damage and provide neuroprotection in the hippocampus when
administered 60-90 minutes following SE onset. The results suggest that oxidative stress may be
a ial target for neuroprotection following SE.
Introduction
The l nervous system is a sensitive target for chemical toxicants that interact with
receptors and signaling e. g. nerve agents or organophosphate pesticides. Studies in the ture
have established that controlling seizure activity and downstream consequences is critical for
neuroprotection and survival after nerve agent exposure. Recent efforts by the NIH CounterAct
program to develop medical countermeasures have identified AEOL10150 as a lead compound
with broad efficacy against multiple chemical threats. AEOL10150 is a catalytic idant with
a wide spectrum of activity against superoxide radicals (02:), hydrogen peroxide (H202),
peroxynitrite (ONOO_), and lipid peroxyl ls. Work by our laboratories over the past decade
demonstrates the efficacy of metalloporphyrins in numerous cell and animal models of neuronal
injury. We have also established that ive stress is a al consequence of ged
seizures and contributes to seizure—induced neuronal death. Since elicitation of seizure ty is
an ant mechanism of several chemical threat agents e. g. nerve agents and organophosphate
ides, it is important to determine whether AEOL10150 exerts neuroprotection against such
agents. A goal of this study was to determine if AEOL10150 exerted neuroprotection against
pilocarpine and kainate-induced seizures when administered 90 minutes after ion of
convulsants.
A present goal is to determine if mitochondrial oxidative stress contributes to
hippocampal neuronal death following SE and whether a synthetic catalytic antioxidant
administered post—SE would provide neuroprotection in two chemoconvulsant models.
Methods
Animal. Male Sprague-Dawley rats were treated with pilocarpine hydrochloride (340
mg/kg) i.p. after pre-treatment with methyl-scopolamine (1 mg/kg) i.p. or kainate (l lmg/kg, s.c.)
to induce status epilepticus (SE). The animals were treated by saline or AEOL 10150 (5mg/kg)
s.c. at 90 minutes post—SE and every 4 hours thereafter until being sacrificed. Oxidative stress
was measured at 24 h post—SE and neurons death was assessed by Fluoro—Jade B staining at 48 h
post—SE.
Monitoring behavioral seizures. Behavioral seizure severity during SE was evaluated
by direct observation for 6 h after the initial treatment and scored based on a d Racine
scale with only motor seizures being considered (Class I and 11 seizures were not scored). See
Racine R. J Clin. Neurophysiol. 32:269—279.
., 1972, Electroencephalogr. Briefly, motor seizure
severity was terized as follows: class 111, animals displayed forelimb clonus with a lordotic
posture; class IV, animals reared with concomitant forelimb clonus; and class V animals had a
Class IV seizure and fell over. Only rats having, at least, class III convulsive seizure were
ed in the study.
Histochemical analyses. The brain of the rats paraffin sections (10 um) were cut
coronally and stained with Fluoro-Jade B -Chem Inc., son, AR) follows the method
described in the literature. See e.g., Hopkins, KJ, et al., 2000, Brain Res 864:69—80; Liang LP, et
al., 2008, JNeurosci 28:11550—11556. The Fluoro—Jade B positive signal in a given area was
estimated with Image J (National Institutes of Health, Bethesda, MD).
HPLC assay. Ascorbate, cysteine, cystine, glutathione (GSH), glutathione ide
(GSSG), tyrosine, 3—nitrotyrosine (3 —NT) were med with BSA (Chelmsford, MA) 5600
ray® HPLC equipped with eight ochemical detector cells as previously described in
the literature. See e.g., Beal MF, et al., 1990, JNeurochem 55:1327—1339; Liang LP, et al.,
2007, JNeurosci 6—4333.
Statistical analyses. al analysis was performed using the Kaplan-Meier method.
For all biochemical analyses, two—way ANOVA was used. P values less than 0.05 were
considered significant.
Results
[0149] The structure of AEOL 10150 follows. The antioxidant effect of AEOL10150
compared with Cu—Zn SOD (superoxide dismutase) is provided in Table 1. A unit of SOD
WO 30150
activity is defined as the amount of compound that inhibits one-half the reduction of epinephrine
by superoxide at pH 10.2. CAT: catalase activity measured by Clarke electrode. 4HNE assay by
HPLC.
Ali-KM. 1035‘.)
Table 1. Antioxidant effects of AEOL10150.
Compounds
Figs. lA-lC demonstrate that AEOL10150 penetrates the BBB (blood brain barrier)
following systemic administration in mice and protects against MPTP (l-methylphenyl-
l,2,3,6-tetrahydropyridine) neurotoxicity, as known in the art.
[0151] Figs. 2A—2C demonstrate that AEOL10150 ates pilocarpine—induced GSH/GSSG
changes.
Fig. 3A-3C demonstrate that AEOL10150 attenuates rpine-induced
Cysteine/Cystine changes.
Figs. 4A-4B demonstrate that AEOL10150 ates pilocarpine-induced increase in
3—Nitrotyrosine/Tyrosine ratio.
Figs. 5A-5B demonstrate that AEOL10150 attenuates pilocarpine-induced
ampal cell death.
Figs. 6A-6F and Figs. 7A-7B demonstrate that AEOL10150 attenuates Kainate-induced
hippocampal cell death.
WO 30150
Figs. 8A—8B trate that oxygen consumption rates (OCR) in ed
hippocampal synaptosomes are decreased after injection of pilocarpine or kainate.
Summary. Figures 1-8 demonstrate that AEOL10150 penetrates the BBB following
systemic administration in mice; AEOL10150 inhibits oxidative stress indices 90 minutes post—
pilocarpine or kainate treatment; AEOL10150 ts hippocampal cell loss 90 minutes post—
pilocarpine or e treatment; and pilocarpine— or e-induced seizures result in decreased
oxygen consumption rates.
Example 2. Neuroprotective efficacy of AEOL10150 against neurotoxic agents.
Introduction. al warfare agents (e.g., chemical threat agents) are an immense
threat to military personnel and civilians. The central nervous system (CNS) is a sensitive target
for chemical toxicants that interact with receptors and signaling e. g. nerve agents or
organophospliate pesticides. See e.g., Jett, D.A. & D.T. Yeung, Proc Am Thorac Soc. 7(4): 254—
6. Studies in the literature have established that controlling seizure activity and downstream
consequences is critical for neuroprotection and survival after nerve agent. See e. g, Shih, T.M.,
et al., 2003, Toxicol Appl Pharmacol 188(2):69—80. Accordingly, the goal of this research
project is to develop a novel and efficacious neuroprotective countermeasure t nerve
agents. Recent efforts by the Nllj". CounterAet program to develop medical countermeasures
have identified AEOLlOlSO as a lead compound with broad efficacy against multiple chemical
threats. AEOLl Ol 50 is a catalytic antioxidant with a wide spectrum of activity t
superoxide radicals (01"), hydrogen peroxide (llgOz), peroxynitrite (ONOO), and lipid peroxyl
radicals. See e..,g Day, B.J., 2004, Drug ery Today 9(13):557—66. Work by the Pl and
colleagues over the past decade trates the efficacy of rnetallopor‘phyrins in us cell
and animal models of neuronal injury. See e.g., Patel, M., 2003, Aging Cell 2(4): 2; Patel,
M., 1998, Neurochem 71:1068—1074; Patel, M. & B.J. Day, 1999, Trends Pharmacol Sci 20:359—
364; Patel, M., et al., 1996, Neuron 16:345—355; Sheng, H., et al., 2002, Free Radical Biology &
Medicine 33(7):947—61; Li, Q.Y., et al., 2001, JNeurochem 78(4):746—55; Liang, L.P., et al.,
2000, Neuroscience :563—70. Tl’ie Pl’s laboratory has also established that oxidative stress
is a critical consequence of prolonged seizures and contributes to seizure—induced neuronal
death. See e.g., Liang, L.P., Y, 2000, Id.; Liang, L.P., et al., 2008, sci 28(45):11550—6;
Waldbaum, S., et al., 2010, Journal ofNeurochemistry 115(5): 1 172—1 182. Since elicitation of
seizure activity is an important mechanism of l chemical threat agents e.g nerve agents
and organophospha‘te pesticides, it is important to determine whetl'ier AEOLlOlSO exerts
neuroprotection against such agents.
Several important attributes of AEOL10150 support its rapid development as a lead
medical countermeasure agent. 1) It has completed Phase 1 human clinical trials for safety with a
low incidence of adverse events which can expedite its development. 2) It is efficacious against
several threats including ion, chlorine and mustard gas. See e. g., l, H.C., et al., 2010,
Free Radic Biol Med 48(9): 1 188—96; Gould, N.S., et al., 2009, JPharmacol Exp Ther
328(3):732—9. 3) It has ble cokinetic properties following subcutaneous injection
which is ideal for its use as a medical countermeasure. 4) It is efficacious in experimental models
when administered post-exposure i.e. after the chemical threat agent which allows its self-
administration after chemical exposure.
Thus, the goal of this project is to determine if AEOL10150 is a neuroprotective
l countermeasure against nerve agents using pilocarpine as a surrogate agent. Nerve
agents such as sarin and VX are known to rapidly elicit seizures in animals and exposed
individuals as evidenced by the Tokyo subway attack and use in the Iran-Iraq war. See e.g., Jett,
D.A. & D.T. Yeung, Proc Am Thorac Soc. 7(4):254—6; Jett, D.A., Sci Trans] Med. 2(23):23ps12.
We have shown that pilocarpine—induced es result in profound oxidative stress. See e.g.,
Waldbaum, S., et al., 2010, Id. Therefore, catalytic removal of reactive oxygen species (ROS)
by AEOL10150 is predicted to blunt oxidative stress and prevent downstream changes such as
metabolic dysfunction, gliosis and neuronal loss.
Compelling in vivo preliminary data demonstrate that 1) pilocarpine produces oxidative
stress and mitochondrial ction, 2) AEOL10150 is permeable to the rodent brain, and 3)
ts pilocarpine-induced ive stress and neuronal death. The specific goals of each
specific aims below include ishing the dose, mechanism and therapeutic window of
neuroprotection. The following parameters can be measured: 1) blood brain barrier (BBB)
permeability and pharmacokinetic parameters for zation of dosing, 2) oxidative stress
indices (reduced and oxidized glutathione, otyrosine levels, xy—2’—deoxyguanosine
(8—OHdG), 4-hydroxynonenal (4-HNE) levels), 3) mitochondrial oxygen consumption rates and
glycolytic metabolism 4) seizure activity using 24/7 video EEG monitoring and 5) neuronal loss
(Fluoro-Jade B is) and 5) gliosis (astrocyte and microglial markers).
Specific Aims. A first specific aim is to determine BBB permeability of AEOL10150
in rats. This es a) determine of plasma and brain concentrations, and b) establishment of
optimal dose and dosing n. A second specific aim is to evaluate the neuroprotective
efficacy of AEOL10150 against pilocarpine exposure in rats. This includes a) determining
efficacy and therapeutic window of AEOL10150 on pilocarpine—induced seizures, oxidative
stress, mitochondrial dysfunction, neuronal loss and gliosis, and b) determining the
neuroprotective efficacy of AEOL10150 following administration with standard therapy
(diazepam and atropine).
Background and Significance
Seizures are a critical consequence of nerve agent. Exposure to nerve agents,
metabolic poisons, or high levels of sulfur mustard can trigger seizures and loss of
consciousness. The elicitation of seizures is a common manifestation of nerve agents that target
the CNS. See e.g., Jett, D.A. & D.T. Yeung, 161.; Jett, D.A., Id.. Therefore it is important for
medical countermeasures to intervene at two . The first level of intervention is usually to
rate the symptoms arising due to the specific interaction of the agent and ar targets.
Nerve agents and organophosphate pesticides bind and inhibit acetylcholinesterase (AChE)
leading to a persistent increase cholinergic tone. This produces acute effects of nerve agent
poisoning including muscle paralysis, cardiorespiratory depression, massive secretion from
mucous membranes, eye tion, and blurry or dim vision which can be controlled by ne
and other cholinergic antagonists. A second level of intervention is targeting the delayed injury
response to the threat agents. Seizure activity is the most critical injury response common to
nerve agents and organophosphate exposures. See e.g., Shih, T.M., et al., 2003, l Appl
Pharmacol 188(2):69—80; Shih, T., et al., 1999, JBiomed Sci 6(2):86—96.
Oxidative stress is a consequence of chemical convulsants: An ant by—product
of ondrial metabolism, xenobiotic detoxification and other enzymatic chain reactions is
the production of ROS. Excessive production of ROS can overwhelm idant es
resulting in oxidation of vulnerable cellular targets. Work from this laboratory has demonstrated
that seizures resulting from chemical convulsants such as rpine and kainic acid oxidatively
damage ondrial DNA, susceptible mitochondrial proteins and cellular lipids. See e. g.,
Patel, M., 2004, Free Radic Biol Med 37(12): 195 1-62. In addition to being an acute
consequence of SE, mitochondrial ROS production rges immediately prior to
development of chronic epilepsy assessed by behavioral analysis, suggesting that ROS ion
could contribute to epileptogenesis. See, e. g., Jarrett, S.G., et al., 2008, NeurobiolDis,.30:130—
13 8.
AEOLlGlSG, a catalytic antioxidant is a medical countermeasure with broad
efficacy against multiple agents: Catalytic antioxidants, which are small, lar mimics of
superoxide ase (SOD) and/or catalase, potent inhibitors of lipid peroxides and ONOO'
hold particular promise. See e.g., Day, B.J., 2004, Drug Discov Today 9(13):557—66. Because
they are catalytic, and not merely free l scavengers, these compounds are much more
potent idants than dietary additives such as vitamin E that act stoichiometrically. The
manganese meso-porphyrin catalytic antioxidants (e.g., AEOL10150, Table 2) combine the
broad spectrum of reactivity like the stoichiometric antioxidants with the catalytic efficiency of
the endogenous antioxidant enzymes. Table 2 discloses the effect ofAEOL 10150 to y
superoxide (as measured by pulse radiolysis), hydrogen peroxide (Clark oxygen electrode [Day,
B.J., 2004, Drug Discovery Today 9(13): 57—66]), peroxynitrite (stop-flow) and inhibit lipid
peroxidation (F2-isoprostanes [Kachadourian, R., et al., 2004, Biochemical Pharmacology,
67(1): 77—85]).
Table 2. Structure and antioxidant activities of AEOL10150
Reactive Superoxide Hydrogen Lipid Peroxynitrite
Oxygen peroxide Peroxides
S n ecies
kcal(02)T k(ONOO):
ty 6.78x10 Mlsl 7M s
These synthetic compounds can be chemically modified to increase their ability to cross
the BBB and various lular compartments. Metalloporphyrins have plasma half lives that
range from 4 to 48 hours. Patel et al. first demonstrated the neuroprotective effects of MnTBAP,
a prototypical first generation metalloporphyrin. See e. g., Patel, M., et al., 1996, Id. Since then
their properties have been optimized resulting in the development of AEOL101 13, 150
and the orally bioavailable, AEOL11207. The efficacy of these compounds have been
demonstrated in multiple models of neuronal injury. See e. g, Trova, M.P., et al., 2003,
Bioorganic & Medicinal Chemistry :695—707. For e.g. AEOL11207, a lipophilic
metalloporphyrin, protected against 1-methyl 4-phenyl tetrahydropyridine (MPTP) neurotoxicity
in vivo following oral administration. See e. g., Liang, L.P., et al., 2007, Id. Most
metalloporphyrins are not extensively metabolized by the body and are y excreted
unchanged in the urine. AEOL10150 is a ypical water soluble metalloporphyrin that
possesses extremely high SOD activity. On a weight basis, its SOD activity surpasses that of
CuZnSOD. It also catalyzes the dismutation of H202 and inhibits lipid peroxidation with potent
1C50s and scavenges ONOO' efficiently. See e. g., Day, B.J., Drug Discovery Today, 2004.
9(13): 557—66; Day, B.J., et al., 1999, Id.; Day, B.J. & J.D. Crapo, 1996, Toxicology and Applied
Pharmacology :4—100; Day, B.J., et al., 1997, Archives hemistry and sics
:256—262.
Study 1: 150 penetrates the BBB following systemic administration in
mice and ts against MPTP neurotoxicity.
MPTP is a prototypical neurotoxicant that is widely used to induce parkinsonism in
mice. MPTP neurotoxicity is thought to arise primarily via inhibition of the mitochondrial
on transport chain at the level of complex I and consequent metabolic inhibition and ROS
production. Figs 1A-1C (Example 1) show the ability ofAEOL 10150 to penetrate the mouse
BBB and inhibit MPTP—induced dopaminergic neuronal loss and ive stress.
Results and interpretation: As shown in Fig. 1A, AEOL10150 achieved
concentrations of 150-200 pmoles/g in the mouse brain following single injection. Estimated
AEOL10150 concentrations based on its molecular weight is 100—200nM . Based on
AEOL10150’s potent antioxidant activity profile, these concentrations are expected to exert
neuroprotection.
Study 2. AEOL10150 inhibits oxidative stress indices 90 minutes post-pilocarpine
treatment.
[0171] Previous work in our tory demonstrates marked oxidative stress and
mitochondrial dysfunction in the hippocampus of rats injected with pilocarpine. See e. g.,
Waldbaum, S., et al., 2008, Soc Neurosci Abstr, 511.6. Here we conducted a study to determine
the effects of AEOL10150 on pilocarpine—induced oxidative stress. As shown in Figs. 3A—3B,
ion of AEOL10150 90 minutes after pilocarpine resulted in a statistically significant
tion of oxidative stress indices (3 —nitrotyrosine/tyrosine; 3—NT/tyr and GSH/GSSG )
in the hippocampus 24 hours later.
Study 3. AEOL10150 inhibits hippocampal cell loss 90 minutes post-pilocarpine
treatment.
A study was conducted to determine the effect of AEOL10150 on pilocarpine—induced
hippocampal cell loss.
Methods and results: Rats were injected AEOL10150 90 min s.c .after receiving
saline (CON) or pilocarpine (Pilo) and ced 24h thereafter. Frozen sections (15um) were
cut coronally and stained with Fluoro-Jade B (Histo-Chem lnc., Jefferson, AR) with
modifications of a method described in the literature. See e. g., Hopkins, K.Jet al., 2000, Brain
Res., 864(1): 69—80. Images were captured using a Nikon Optiphot—2 80i microscope equipped
with epifluorescence optics (Nikon Inc., le, NY). The Fluoro—Jade B positive signal of a
given area was measured with Image J software. The e of relative fluorescence density
was expressed as percentage of the control. As shown in Figs. 5A—5B, this study demonstrates
the ability of AEOL10150 to significantly decrease hippocampal cell loss in the hilar and CA3
regions.
Study 4. Pilocarpine decreases oxygen consumption rates in the hippocampus.
A key end point of Specific Aim 2 is to assess metabolic flux n consumption
rates; OCR) in real time using the Seahorse Biosciences extracellular flux analyzer. To support
the feasibility of these studies, we provide pilot data y trating decreased OCR in
hippocampal synaptosomes from pilocarpine- vs saline-treated rats 16 hours after injection
(average values from n=2 rats per groups, Fig. 10). This study suggests that rpine seizures
result in decreased ated OCR in hippocampal synaptosomes and underscores the feasibility
of testing the effects on AEOL10150 on pilocarpine—induced changes in OCR in vivo.
Summary. The data of Studies 1-4 trate the ability of AEOL10150 to penetrate
the mouse brain at therapeutic concentrations and to protect against rpine-induced
oxidative stress and neuronal loss. Moreover, pilocarpine decreases OCR, a key index of
ondrial function.
Research Strategy: Specific Aim 1: To determine BBB permeability of
AEOEJGE 50 in rats.
[0179] Rationale: The pharmacokinetic profile of AEOL10150 can be determined to guide
studies that its efficacy. Measurements of plasma and brain concentrations of the compound in
rats is necessary for determining an optimal dosing n and correlate its biological effects
with in vivo cy in the pilocarpine rat model. In addition, data using AEOL10150 in the
mouse indicate that it crosses the mouse BBB.
[0180] Experimental Approach: Groups of 6-10 rats can be administered 2.5, 5 or 10 mg/kg
of AEOL10150 by the s.c. route (single or multiple i.e. every 4, 8 and 24h) and sacrificed at
various times (1, 3, 6, 12, 24 and 48 hr) following the last injection. Once blood samples are
obtained, rats can be perfused free of blood and the brains (hippocampus, piriform cortex and
frontal cortex) can be collected for analysis. Compounds can be measured using methods known
in the art. See e. g., Kachadourian, R., et al., 2004, Id.; Kachadourian, R., et al., 2003, Journal of
Inorganic Biochemistry, 95(4): 240—8.
AEOL10150 measurement: 150 can be measured in plasma and brain
samples by HPLC—UV methods as previously described for AEOL11207. See e.g., Liang, L.P.,
et al., 2007, JNeurosci 27(16): 4326—33
Analysis: cokinetic is of drug levels can be ed by PKAnalyst®
(MicroMath® software). The following parameters can be obtained: 1) Elimination T1/2 in
plasma and brain; 2) Distribution T1/2 in plasma and brain regions; 3) Volume of distribution of
AEOL10150 (Vd); 4) Time to peak plasma concentrations; 5) Time to peak tissue concentration;
6) AUC (area under the plasma level—time curve), which gives a measure of the extent of drug
bioavailability; and 7) Peak plasma and brain levels of AEOL101050.
[0183] Research gy: Specific Aim 2: To evaluate the neuroprotective y of
AEOL10150 against pilocarpine exposure in rats.
Rationale: The goal of this project is to determine if AEOL10150 is a neuroprotective
medical countermeasure against chemical threat agents that mediate ive stress via
ation of seizures. Work by the PI has demonstrated that various chemical convulsants
including pilocarpine produce profound oxidative stress in vulnerable brain areas. See e.g.,
Liang, L.P., et al., 2000, Neuroscience 101(3):563—570; Waldbaum, S., et al., 2010, Journal of
Neurochemistry : 1 172—1 182; Liang, L.P. & M. Patel, 2006, Free Radic Biol Med
40(2):316—22. Therefore, tic removal of ROS by AEOL10150 is predicted to blunt
ive stress and prevent downstream changes such as metabolic dysfunction, gliosis and
neuronal loss thereby aiding recovery of the brain from the chemical attack. Video-EEG
analysis, oxidative stress indices, mitochondrial functions, glycolytic rates and neuronal
death/gliosis markers and their time points of ment are all based on pilot studies and prior
work in the laboratory.
Aim 2a: The rationale for testing AEOL10150 alone is to ine its therapeutic
window and whether it is sufficient to exert neuroprotection in a pre-treatment and post-
treatment paradigm. In Specific Aim 2a, we can determine a rotective dose of
AEOL10150 and its therapeutic window (by treatment 30 min before, 60 min, 90 min, 3h and 6h
after pilocarpine) in the presence or absence of scopolamine, an olinergic agent which does
not penetrate the BBB. Video EEG analysis can determine if AEOLlOlSO has any effect on
pilocarpine—induced seizure activity. Using an optimal dose, two separate time points (30 min
before and selected time after pilocarpine) can determine AEOLlOlSO’s influence on seizure
activity over a 24h period.
Aim 2b: It is important to determine the neuroprotective efficacy of AEOLlOlSO in the
e and presence of standard therapy for nerve agent exposure i.e. anticholinergic agents and
benzodiazepines. Although pralidoximine (2-PAM) and/or diazepam are first line therapies for
nerve agent poisoning, it is important to determine whether a new therapy can work on its own
and in combination with standard therapies. Aim 2b determines the neuroprotective ability of
AEOLlOlSO in combination with rd ents (anticholinergic agent and diazepam). The
rationale for using atropine vs 2—PAM or carbamates is model—dependent i.e. because pilocarpine
is a muscarinic agonist and therefore 2—PAM, which works via cholinesterase is believed to be
ineffective. Treatment with atropine (0.5-2mg/kg, i.m.) 5 min ilocarpine and diazepam
(10-20 mg/kg, i.p.) 30 min after first motor seizure is based on our experience, literature gs
and standard use of these countermeasures sed in the NIH Strategic Plan for Medical
Countermeasures. See e.g., Shih, T.M., et al., 3004, Toxicol Appl Pharmacol 188(2):69—80;
Shih, T., et al., 1999, JBiomed Sci 6(2):86—96; Shih, T.M., et al., 2011, Toxicol Mech Methods
21(1):53-62. Further studies and Aims l and 2a can determine optimal doses, order and timing
of each agent. The ability of the combination to influence pilocarpine-induced seizures, oxidative
stress and injury can be assessed using video—EEG analysis, oxidative stress indices,
mitochondrial functions and neuronal death/gliosis markers.
mental Approach
Experimental Timing. A schematic diagram of a time line for experimental
conditions for studies for Aims 2a and 2b is disclosed in Fig. 11.
Aim 2a. The goal of this study is to answer the following questions. 1) What is the
optimal dose and dosing regimen for lSO? Pharmacokinetic analysis (Specific Aim 1)
and ement of oxidative stress indices and cell death allow us to determine the dose and
dosing frequency of AEOLlOlSO to optimally inhibit pilocarpine-induced ive stress and
cell death. 2) What is the therapeutic window of lSO neuroprotection? Once a dose and
dosing frequency are established, we can ine the window of opportunity after pilocarpine
treatment that yields tically significant neuroprotection (see end points below). This can be
WO 30150
addressed by g the timing of AEOL10150 treatment before or after pilocarpine (30 min
before, 60 min, 90 min, 3h and 6h after pilocarpine). To determine whether peripheral
cholinergic systems need to be blocked to achieve optimal CNS neuroprotection with AEOL
10150, we plan to include a group of animals injected with scopolamine (30 min prior to
pilocarpine). Finally, 3) does AEOL10150 have any effect on seizure activity? Using an optimal
dose of AEOL10150, its ability to influence seizure activity can be assessed by continuous
video~EEG over a 24 h period. AEOL10150 can be stered 30 min prior to pilocarpine to
determine if pre—treatrnent has an effect and at one selected time point after rpine to
determine any nce on ongoing seizure activity.
[0190] Aim 2b. To determine the neuroprotective cy of AEOL10150 in the absence and
presence of standard therapy for nerve agent exposure, the following treatment groups can be
conducted: 1) control, 2) pilocarpine, 3) pilocarpine+atropine, 4) pilocarpine + diazepam 5)
pilocarpine +atropine+AEOL10150+diazepam. Drug alone control groups (for determining
effects on nts): 1) diazepam, 2) AEOL10150 and 3) ne. Optimal dose and timing of
atropine (0.5-2mg/kg, dose range i.m., 5 min post-pilocarpine) and diazepam (10-20mg/kg dose
range i.p. and 30 min post first motor seizure) can be determined from studies. AEOL 10150 can
be given via s.c. route (dose and timing to be determined from Specific Aim 2a). End points can
be the same as discussed in Specific Aim 2a.
End points. End points for rotection include mitochondrial functions (basal
respiration, ATP turnover, proton leak, and maximal respiratory capacity) and glycolytic rates
which can be measured using an ellular flux analyzer (Seahorse Biosciences) 16 hr after
pilocarpine hippocampal synaptosomes. We have optimized these assays in synaptosomes from
pilocarpine and e injected rats. Mitochondrial aconitase activity can be measured because
of its known sensitivity to ROS and its mitochondrial localization and fumarase activity because
it serves as a control enzyme that is insensitive to ive damage. See e.g., Patel, M., 1996,
Molecular Psychiatry 1:362—3 63. ATP levels (as well as ADP and AMP) can be measured to
monitor bioenergetic status (16hr).
Additional end points for neuroprotection include oxidative stress indices :
Several s of oxidative stress can be measured including GSH and GSSG which assess the
ar redox status, 4-HNE, which is an electrophilic lipid peroxidation end product,
8OHdG/2dG, which is an index of oxidative DNA damage and 3-NT, which is an indicator of
protein nitration. The choice of oxidative stress indices (GSH/GSSG, 4—NHE and 8—OHdg/2dG)
has been aligned with the standard markers used to assess the protective effects of AEOL10150
in other studies. Additionally, we have included two additional markers, 3-NT/tyr and
aconitase/fumarase to obtain ation regarding nitrosative stress (3-NT) and mitochondrial
oxidative stress (aconitase inactivation).
Additional end points for rotection include cell death and gliosis: A principal
end point of this study is evaluation of neuronal viability and glial response to injury. Neuronal
ity can be assessed by Fluoro Jade B analysis which detects injured neurons by
stereological methods. Gliosis can be assessed by analysis of GFAP, a marker of astrocytes and
Ibal, a marker of activated microglia at the 2 and 7d time points.
[0194] Tissue and brain regions: AEOL10150 levels can be measured in plasma and
hippocampus, piriform cortex and frontal cortex. Oxidative stress end points can be measured in
hippocampus, piriform cortex and llum (control region). Neuronal viability and gliosis can
be assessed in the hippocampus, piriform cortex, frontal cortex and cerebellum (control region).
Mitochondrial and ytic function assays can be measured in synaptosomes from
hippocampus and piriform .
Methods: Male Sprague—Dawley rats (200—250 g) can be treated with 340 mg/kg
rpine hydrochloride i.p. alone or after pre-treatment with methyl-scopolamine (1 mg/kg)
i.p. (Aim 2a) or 5 min prior to atropine (0.5-2 mg/kg, i.m.) followed by AEOL10150 at varying
time points following pilocarpine and then am (10-20 mg/kg, i.p.) 30 min after the first
motor seizure (Aim 2b). All rats can be directly observed and those having a minimum of 5 P3
seizures based on a ed Racine scale [Racine, R.J., 1972, Electroencephalogr Clin
Neurophysiol. 32:281—94.] can be treated with either saline or AEOL10150 s.c. beginning 30 min
prior to or 90 min, 3h or 6h post-pilocarpine and every 4, 8, 24 hours thereafter until sacrifice
(Aim 2a). ements of GSH, GSSG, 80HDdG/2dG, 3NT and tyrosine can be performed
with a HPLC ed electrochemical and UV detectors by previous methods. See e. g., Liang,
L.P., et al., 2007, 161.; Day, B.J., et al., 1999, Free Radical Biology and Medicine 26(5—6):730—6;
Hensley, K., et al., 1998, JNeurosci 18(20):8126—32. 4—HNE can be measured by HPLC—EC
methods and GC mass spectrometry for verification. Mitochondrial functions can be analyzed in
isolated osomes (subregions in Aim 2) 16h after pilocarpine using the XF analyzer. The
effect on cellular respiration rates [OCR] can be measured after e, 1.2 uM oligomycin
itor of ATP synthase), 4 uM FCCP (to short-circuit proton circuit and get maximal
ation), luM myxothiazol and 2uM rotenone (to inhibit electron transfer). AMP, ADP and
WO 30150
ATP can be quantified by HPLC—UV at 258 nm. See, e.g., Sellevold, O.F., et al., 1986, JMol
Cell Cardiol 18(5):517—27; Botker, H.E., et al., 1994, JMol Cell Cardiol, 26(1):4l—8.
Statistical analysis: Two—way ANOVA can be used to determine the ences
between ent and drug. Group measures can be expressed as mean :: SEM. The statistical
cance of differences can be assessed with the Neuman—Keul post hoc test. The level of
significance can be set at p < 0.05.
Example 3. Further Investigation of BBB Penetration of AEOL10150.
Because the tetrakis diethylimidazolium porphyrin AEOL10150 has a net charge of at
least +5 under physiological conditions of pH, this compound would not be expected a priori to
transit the blood brain barrier (BBB) solely due to passive ion, e. g., absent an active
transport system in the cells forming the BBB including endothelial, basement membrane and/or
astrocytic cells. Moreover, we find no evidence for an active transport system for AEOL10150.
Thus, it has been surprisingly discovered that AEOL10150 indeed transits the BBB.
In order to determine the extent and distribution of AEOL10150 upon administration
via modes which do not include intracerebral implantation, intracerebroventricular or convection
enhanced diffusion, as known in the art, rats were administered either a single injection
(5 mg/kg, s.c.), or multiple injections (5 mg/kg, s.c., every 4—hr for 24—hrs) of AEOL10150. As
depicted in Fig. 12, the concentration (pmol/g tissue) of AEOL10150 was quantified in the
ampus and piriform cortex. Based on the potent antioxidant activity profile of
AEOL10150, the concentrations reported in Fig. 12 are expected to exert rotection when
administered every 4-hrs for at least 24-hrs, e. g., 24-48 hrs.
Example 4. Therapeutic Window of Neuroprotective Effects of AEOL10150.
Studies were ted to determine the therapeutic effects including timing (i.e., the
so—called “therapeutic window”) of AEOL10150 on pilocarpine—induced oxidative stress and
neuronal injury. Rats received pilocarpine (340 mg/kg, i.p.) alone or in combination with
AEOL10150 (5 mg/kg, s.c.).
As depicted in the rams of Figs. B, quantitative analysis of Fluoro-Jade B
histofluorescence ng in the CA3 (Fig. 13A) and Hilus (Fig. 13B) of the rats at 24 h after
receiving either rpine alone or in the presence of AEOL10150 at 60 or 90 min post
pilocarpine treatment and every 4h therefore until sacrifice. The Fluoro-Jade B ve signals
in a given area of hippocampal subregions from three slides of each animal were estimated with
Image J (available at the rsb.info.nih. gov website). For Figs. l3A—l3B, bars represent mean +
S.E.M, *p<0.01 vs. ; 5 vs. rpine; one way ANOVA, n=6 rats per group.
As depicted in Figs. l4A-l4B, injection of AEOL10150 at 60 or 90 minutes after
pilocarpine administration resulted in inhibition of oxidative stress indices (i.e., GSH, GSSG,
and GSH/GSSG ratios, left, center and right panels, respectively) in the hippocampus.
As depicted in Fig. 14B, 30 min atment of AEOL10150 resulted in maximum
protection of oxidative stress indices followed by 60 min which afforded better protection than
90 min post-pilocarpine treatment.
As depicted in Fig. 14C, 3-nitrotyrosine/tyrosine ratio in the hippocampus of the rat at
24-hrs (left panel) and 48-hrs (right panel) after either rpine alone, or onally with
AEOL10150 administration at 90 min after, 60 min after, or 30-min before (right panel only) and
continued every 4—hrs until sacrifice.
Example 5. Effects of AEOL10150 on Pilocarpine—Induced Mitochondrial Respiratory
[0204] In order to test the hypothesis that mitochondrial ROS production mediates metabolic
dysfunction which occurs following pilocarpine exposure, we ined oxygen consumption
rates (OCR) in osomes ed from , pilocarpine alone and pilocarpine +
AEOL10150 (latter injected 60 min after and every 4h (q4h) thereafter for 24h). Methods for
determining OCR as well known in the art. As depicted in Fig. 15A, maximal respiractory
capacity is rescued by administration of AEOL10150 and pilocarpine. Indeed, maximal
respiratory rates (Fig. 15B) as well as ATP turnover, ne respiration and glycolytic rates
(data not shown) decreased by pilocarpine were largely prevented by AEOL10150. This
es the first ce that metabolic dysfunction following pilocarpine is inhibited by a
catalytic antioxidant, e. g., AEOL10150.
Example 6. AEOL10150 Inhibits Pilocarpine—Induced Microglial Activation.
Oxidative stress and neuronal damage can activate inflammation. Accordingly, we
investigated whether inflammatory cell activation by pilocarpine-induced seizures was inhibited
by AEOL10150. See Figs. l6A-l6D. Hippocampal lial activation in pilocarpine treated
rats analyzed by Ibal antibody straining histochemical procedures was significantly attenuated
by AEOL10150. See Figs. l6E—l6F. This provides evidence that inflammation in this model
likely occurs as a result of oxidative damage, and that scavenging ROS can inhibit inflammation.
Example 7. 150 in Combination Therapy.
In order to determine the effect of combination of AEOL10150 in the treatment of
subjects administered pilocarpine, a rat model was employed. Male Sprague—Dawley rats (3 00—
350g) were divided into four different groups: saline, pilocarpine alone, pilocarpine + atropine+
diazepam, and pilocarpine + atropine+ AEOL10150+diazepam.
Pilocarpine (340 mg/kg, s.c.) or saline was stered to the subjects. Atropine
(lmg/kg, i.m.) or saline was administered at 10min after pilocarpine. AEOL10150 (5mg/kg, s.c.)
or saline was administered at 60min after pilocarpine and every 4 hours thereafter until sacrifice.
am (lOmg/kg, i.p.) or saline was administered at 90 min after pilocarpine.
[0208] As shown in Figs. l7A-l7D, which are histograms depicting concentrations of GSH
(Fig. 17A), GSSG (Fig. 17B), and ratios of GSH/GSSG (Fig. 167) and the ratio 3-nitrotyrosine/
tyrosine (Fig. 17D) in the hippocampus of the rat at 24h after the administration regimen,
addition of AEOL10150 in the treatment protocol is observed to enhance the effects of atropine
and diazepam as judged by increased GSH concentration and SG ratio, and decreased
GSSG and 3—nitrotyrosine/tyrosine ratio.
V. ments
ments contemplated herein include the following.
Embodiment l. A method for treating a subject suffering from exposure to a
chemical threat agent, said method comprising administering to said subject an effective amount
of a compound selected from: a) a compound having the structure of Formula (I) or Formula
(11),
R3 (1) (11),
wherein R1, R2, R3, and R4 are each independently —CF3, -C02R8, —CORg’,
\=/ OH W \=/
mm W
\ R20 \ R21 x
R18—
\=N\ \ \
S N/st
+ N_N\ N\/
__ J—
R197 LN R22 N 0r
7 7 7
JVVV"
R24\N/ s
\:+/ .
R5, R6, R7, R8, R8’, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22, R23, and R24
are each independently hydrogen, n, —CN, —CF3, —OH, —NH2, —COOH, —COOR25,
—CH2COOR25, —CH2COOH, an unsubstituted or tuted alkyl, unsubstituted or substituted
heteroalkyl, unsubstituted or substituted cycloalkyl, unsubstituted or substituted
heterocycloalkyl, unsubstituted or substituted aryl, or an unsubstituted or substituted heteroaryl;
R25 is an unsubstituted alkyl, and M is a metal; b) a compound having the structure of one of
ae (X)-(XV),
WO 30150
wherein R13, R23, R33, and R4a are independently -(CH2)mCHZOX1 0r -(CH2CHZO)nX1; m is 1-6; n
is 3—50; X1 is substituted or unsubstituted €1-12 alkyl; M is a metal; and each A is,
independently, hydrogen or an electron withdrawing group; and c) a compound having the
structure of one of Formulae (XVI)—(XVII),
wherein at least one of R”, or R16, R2], or R26, R3b or R36, and R4], or R4c is, independently,
—(CH2)pCHZOX2 or -(CH2CHZO)qX2; the other one of R”, or R16, R2], or ch, R3b or R3c, and R4],
or R40 is, independently, a C142 alkyl (straight chain or branched); p is 1-6; q is 3-50; X2 is
substituted or unsubstituted C142 alkyl; M is a metal; and each A is, independently, hydrogen or
an electron withdrawing group; n said al threat agent is an anti-cholinesterase
agent, a GABA-agent or a metabolic poison.
[0211] Embodiment 2. The method according to embodiment 1, n said compound
has the structure of Formula (I) or Formula (II).
Embodiment 3. The method according to ment 2, wherein said compound
has the structure of Formula (II).
Embodiment 4. The method according to embodiment 3, wherein said metal is
manganese, iron, cobalt, copper, nickel, or zinc.
Embodiment 5. The method according to embodiment 4, wherein said metal is
manganese.
Embodiment 6. The according to ment 2, wherein R1, R2, R3, and R4 are
R5—N /+ N—R6
each \=/
; and R5 and R6 are ndently unsubstituted alkyl.
Embodiment 7. The method according to embodiment 6, wherein said compound
has the structure of Formula (VII)
\_N/T/\NJ
1 r
E: \J
2 k
N4 N /—U—\ (V11).
Embodiment 8. The method according to embodiment 1, wherein said compound
has the structure of one of Formulae (X)—(XV).
Embodiment 9. The method according to embodiment 8, wherein said metal is
manganese, iron, cobalt, copper, nickel, or zinc.
Embodiment 10. The method according to embodiment 9, wherein said metal is
manganese.
[0220] Embodiment 11. The method according to embodiment 1, wherein said nd
has the structure of one of Formulae (XVI)—(XVII).
Embodiment 12. The method according to embodiment 11, n said metal is
manganese, iron, cobalt, copper, nickel, or zinc.
ment 13. The method according to embodiment 12, wherein said metal is
manganese.
Embodiment 14. The method according to any one of ments 1 to 13, n
said chemical threat agent causes seizures and neuropathology.
Embodiment 15. The method according to ment 14, wherein said chemical
threat agent is a nerve agent.
[0225] ment 16. The method according to embodiment 15, wherein said nerve agent
disrupts nerve signal by inhibiting acetylcholinesterase.
Embodiment 17. The method according to embodiment 14, n said chemical
threat agent is sarin, parathion, rb or tetramine (TETS).
Embodiment 18. The method according to any one of embodiments 1 to 13, wherein
said chemical threat agent targets the blood.
[0228] Embodiment 19. The method according to embodiment 18, wherein said chemical
threat agent is cyanide, sodium acetate, arsenic trioxide or strychnine.
Embodiment 20. The method according to embodiment 1, wherein the effect of said
treating a subject suffering from exposure to a chemical threat agent lasts for at least 90 minutes
ing said administration.
[0230] Embodiment 21. A method for reducing brain injury in a subject in need thereof,
comprising stering to said subject an effective amount of a compound selected from: a) a
compound having the structure of Formula (I) or Formula (11),
R4 R2
R4 R2
Rs R3
(1) (11),
wherein R1, R2, R3, and R4 are each independently —CF3, -C02R8, —CORg’,
JVVV‘ mm
JWV‘
_ R12\N/ N/R13 / S
/N+ N+ R17—N N
R11 R15 R14 R16 \—J—
7 7 7 ,
mm W
JVVV‘ mm
7 7 7 701-
R24\N/ S
R5, R6, R7, R8, R8’, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22, R23, and R24
are each independently hydrogen, halogen, —CN, —CF3, —OH, —NH2, —COOH, —COOR25,
—CH2COOR25, —CH2COOH, an unsubstituted or substituted alkyl, unsubstituted or tuted
heteroalkyl, unsubstituted or substituted cycloalkyl, unsubstituted or tuted
heterocycloalkyl, unsubstituted or substituted aryl, or an unsubstituted or substituted heteroaryl;
R25 is an unsubstituted alkyl; and M is a metal; b) a compound having the structure of one of
Formulae (X)—(XV),
wherein R13, R23, R33, and R4a are independently -(CH2)mCHZOX1 or -(CH2CHZO)nX1; m is 1-6; n
is 3—50; X1 is substituted or unsubstituted €1-12 alkyl; M is a metal; and each A is,
independently, hydrogen or an electron awing group; and c) a compound haVing the
structure of one of Formulae (XVI)—(XVII),
wherein at least one of R”, or R16, R2], or R26, R3b or R36, and R4b or R4c is, independently,
—(CH2)pCHZOX2 or -(CH2CHZO)qX2; the other one of R”, or R16, R2], or ch, R3b or R3c, and R4b
or R40 is, independently, a C142 alkyl (straight chain or branched); p is 1-6; q is 3-50; X2 is
substituted or unsubstituted C142 alkyl; M is a metal; and each A is, independently, hydrogen or
an electron awing group.
Embodiment 22. The method according to embodiment 21, wherein said brain
injury results from seizure.
Embodiment 23. The method according to embodiment 22, wherein said brain
injury is cognitive dysfunction.
ment 24. The method according to embodiment 22, wherein said seizure
results from exposure to a chemical threat agent.
Embodiment 25. The method according to any one of embodiments l or 24, wherein
said chemical threat agent is an anti-cholinesterase agent.
[0235] Embodiment 26. The method according to any one of embodiments l or 24, r
comprising administering to said t an anticholinergic agent.
Embodiment 27. The method according any one of embodiments l or 24, r
sing administering to said subject an anti-seizure agent.
Embodiment 28. The method according to embodiment 27, wherein said anti-
seizure agent is a benzodiazepine.
Embodiment 29. The method according any one of embodiments 1 or 24, further
comprising administering to said t an anticholinergic agent and an anti-seizure agent.
Embodiment 30. The method according any one of ments 1 or 24, further
comprising administering to said subject an acetylcholinesterase reactivating agent.
Embodiment 31. The method ing to embodiment 30, wherein said
acetylcholinesterase reactivating agent is pralidoxime.
Embodiment 32. The method according any one of embodiments 1 or 24, n
said administering occurs prior to said exposure to said chemical threat agent.
Embodiment 33. The method according to embodiment 32, wherein said
administering occurs at least 30 minutes prior to said exposure to said chemical threat agent.
Embodiment 34. The method according to embodiment 21, wherein the effect of
said reducing brain injury lasts for at least 90 minutes following said administration.
Embodiment 35. The method ing to embodiment 21, wherein said compound
has the structure of Formula (I) or Formula (II).
[0245] Embodiment 36. The method ing to embodiment 35, wherein said compound
has the structure of Formula (II).
Embodiment 37. The method ing to embodiment 36, wherein said metal is
ese, iron, cobalt, copper, nickel, or zinc.
Embodiment 38. The method according to embodiment 37, wherein said metal is
manganese.
Embodiment 39. The according to embodiment 35, wherein R1, R2, R3, and R4 are
R5—N /+ N—R6
each \:/
; and R5 and R6 are independently unsubstituted alkyl.
Embodiment 40. The method according to embodiment 39, wherein said nd
\_N/_\JJr/N
N + N—\
has the structure of Formula (VII) / \—J (VII).
Claims (4)
- WHAT IS CLAIMED IS:1 1. Use of a compound in the manufacture of a medicament for treating a2 subject suffering from re to a chemical threat agent, said compound selected from:3 a) a compound having the structure of Formula (I) or Formula (II),N NNH NR4 M+ R2R4 R2N NN HN4 R3 R3(I) (II),5 wherein6 R1, R2, R3, and R4 are each independently -CF3, -CO2R8, -COR8’,R5 N + N R6 O R77 , OH , ,R12 R13N NN S N8 , R11 , R15 R14 ,S R18 NN+ R17 N N N S9 R16 , , R19 , NR20 R21R23 R24N N N N N S10 , R22 , N , or11 ;12 R5, R6, R7, R8, R8’, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22,13 R23, and R24 are each independently en,14 halogen, -CN, -CF3, -OH, -NH2, -COOH, -COOR25, -CH2COOR25, -CH2C15 OOH, an unsubstituted or substituted alkyl, unsubstituted or substituted16 heteroalkyl, unsubstituted or substituted cycloalkyl, unsubstituted or17 substituted heterocycloalkyl, unsubstituted or substituted aryl, or an18 unsubstituted or substituted heteroaryl;19 R25 is an unsubstituted alkyl; and20 M is a metal;21 b) a compound having the ure of one of Formulae (X)-(XV),R1a R1aN+ N+A A A AA A A AN N NH NR4a N+ M+ N+ R2a R4a N+ N+ R2aN N N HNA A A AA A A AX XIN+ N+22 R3a R3aR1a R1aN+ N+A A A AA A R2a A A R2aN N N+ NH N N+N+ N N N+ N HNR4a A A R4a A AA A AXII AXIIIN+ N+23 R3a R3aN+ N+R1a R1aA A A AA AR2a A R2aN N N+ NH N N+N+ N N N+ N HNR4a R4a AA A AA A A AR3a R3aN+ XVXIV N+24 ,25 wherein26 R1a, R2a, R3a, and R4a are independently -(CH2)mCH2OX1 or -(CH2CH2O)nX1;27 m is 1-6;28 n is 3-50;29 X1 is substituted or unsubstituted C1-12 alkyl;30 M is a metal; and31 each A is, independently, hydrogen or an on withdrawing group; and32 c) a nd having the structure of one of Formulae (XVII),N+ NR1c R1b N+ NR1c R1bA A A AA A A AR4b R2c R4b R2cN NN N+ NH NN N+N+ N N N N+ N HN NR4c R2b R4c R2bA A A AA A A AR3b R3c XVI R3c XVIIN N+ R3b N N+33 ,34 wherein35 at least one of R1b or R1c, R2b or R2c, R3b or R3c, and R4b or R4c is,36 independently, -(CH2)pCH2OX2 or -(CH2CH2O)qX2;37 the other one of R1b or R1c, R2b or R2c, R3b or R3c, and R4b or R4c is, independently,38 a C1-12 alkyl (straight chain or ed);39 p is 1-6;40 q is 3-50;41 X2 is substituted or unsubstituted C1-12 alkyl;42 M is a metal; and43 each A is, independently, hydrogen or an electron withdrawing group;44 wherein said chemical threat agent is an anti-cholinesterase agent, a gent,45 cyanide, sodium fluoroacetate, arsenic trioxide or strychnine; and wherein said46 chemical threat agents contemplated herein do not include sulfur mustard,47 chlorine gas, phosgene, or 2-chloroethyl ethyl e (CEES).1 2. The use according to claim 1, wherein said compound has the structure of
- 2 Formula (I) or Formula (II).1 3. The use according to claim 2, wherein said compound has the structure of2 Formula (II).1 4. The use according to claim 3, wherein said metal is manganese, iron,2 cobalt, copper, nickel, or zinc.1 5. The use according to claim 4, wherein said metal is manganese.1 6. The use ing to claim 2,2 whereinR5 N + N R6
- 3 R1, R2, R3, and R4 are each ; and
- 4 R5 and R6 are independently unsubstituted alkyl.1 7. The use according to claim 6, wherein said compound has the structure of2 Formula (VII)N NN NN NMn+ +N NN NN + N3 (VII).1 8. The use according to claim 1, wherein said compound has the structure of2 one of Formulae (X)-(XV).1 9. The use according to claim 8, wherein said metal is ese, iron,2 cobalt, copper, nickel, or zinc.1 10. The use according to claim 9, wherein said metal is ese.1 11. The use according to claim 1, wherein said compound has the structure of2 one of Formulae (XVI)-(XVII).1 12. The use according to claim 11, wherein said metal is manganese, iron,2 cobalt, copper, nickel, or zinc.1 13. The use according to claim 12, wherein said metal is manganese.1 14. The use according to any one of claims 1 to 13, wherein said chemical2 threat agent causes seizures and neuropathology.1 15. The use according to claim 14, wherein said chemical threat agent is a2 nerve agent.1 16. The use according to claim 15, wherein said nerve agent disrupts nerve2 signal by inhibiting acetylcholinesterase.1 17. The use ing to claim 14, wherein said chemical threat agent is sarin,2 parathion, aldicarb or ine (TETS).1 18. The use according to any one of claims 1 to 13, wherein said chemical2 threat agent targets the blood.1 19. The use according to claim 18, wherein said al threat agent is2 cyanide, sodium fluoroacetate, arsenic trioxide or strychnine.1 20. The use ing to claim 1, wherein the effect of said medicament in a2 subject ing from exposure to a chemical threat agent lasts for at least 90 minutes following3 administration.1 21. Use of a compound in the manufacture of a medicament for reducing brain2 injury in a subject in need thereof, said compound selected from:3 a) a compound having the structure of Formula (I) or Formula (II),N NNH NR4 M+ R2R4 R2N NN HN4 R3 (I) (II),5 wherein6 R1, R2, R3, and R4 are each independently -CF3, -CO2R8, -COR8’,R5 N + N R6 O R77 , OH , ,R12 R13N NN S N8 , R11 , R15 R14 ,S R18 NN+ R17 N N N S9 R16 , , R19 , NR20 R21R23 R24N N N N N S10 , R22 , N , or11 ;12 R5, R6, R7, R8, R8’, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22,13 R23, and R24 are each independently hydrogen,14 halogen, -CN, -CF3, -OH, -NH2, -COOH, -COOR25, -CH2COOR25, -CH2C15 OOH, an unsubstituted or substituted alkyl, tituted or substituted16 heteroalkyl, unsubstituted or substituted cycloalkyl, unsubstituted or17 substituted heterocycloalkyl, unsubstituted or substituted aryl, or an18 unsubstituted or tuted heteroaryl;19 R25 is an unsubstituted alkyl; and20 M is a metal;21 b) a nd having the structure of one of Formulae (X)-(XV),R1a R1aN+ N+A A A AA A A AN N NH NR4a N+ M+ N+ R2a R4a N+ N+ R2aN N N HNA A A AA A A AX XIN+ N+22 R3a R3aR1a R1aN+ N+A A A AA A R2a A A R2aN N N+ NH N N+N+ N N N+ N HNR4a A A R4a A AA A A AXII XIIIN+ N+23 R3a R3aN+ N+R1a R1aA A AA AR2a A R2aN N N+ NH N N+N+ N N N+ N HNR4a R4a AA A AA A A AR3a R3aN+ XVXIV N+24 ,25 wherein26 R1a, R2a, R3a, and R4a are independently -(CH2)mCH2OX1 or -(CH2CH2O)nX1;27 m is 1-6;28 n is 3-50;29 X1 is substituted or unsubstituted C1-12 alkyl;30 M is a metal; and31 each A is, independently, hydrogen or an electron withdrawing group; and32 c) a compound having the structure of one of Formulae (XVI)-(XVII),N+ NR1c R1b N+ NR1c R1bA A A AA A A AR4b R2c R4b R2cN NN N+ NH NN N+N+ N N N N+ N HN NR4c R2b R4c R2bA A A AA A A AR3b R3c XVI R3c XVIIN N+ R3b N N+33 ,34 wherein35 at least one of R1b or R1c, R2b or R2c, R3b or R3c, and R4b or R4c is,36 independently, -(CH2)pCH2OX2 or -(CH2CH2O)qX2;37 the other one of R1b or R1c, R2b or R2c, R3b or R3c, and R4b or R4c is, independently,38 a C1-12 alkyl (straight chain or branched);39 p is 1-6;40 q is 3-50;41 X2 is substituted or unsubstituted C1-12 alkyl;42 M is a metal; and43 each A is, independently, hydrogen or an electron withdrawing group;44 n said brain injury results from exposure to a chemical threat agent,45 wherein said chemical threat agent is an anti-cholinesterase agent, a46 GABA-agent, e, sodium fluoroacetate, c trioxide, or47 strychnine, and n said chemical threat agents contemplated herein48 do not include sulfur mustard, chlorine gas, phosgene, or 2-chloroethyl49 ethyl sulfide (CEES).1 22. The use according to claim 21, wherein said brain injury s from2 seizure.1 23. The use according to claim 22, wherein said brain injury is cognitive2 dysfunction.1 24. The use according to claim 22, wherein said seizure results from exposure2 to a chemical threat agent.1 25. The use according to any one of claims 1 or 24, wherein said chemical2 threat agent is an anti-cholinesterase agent.1 26. The use according to any one of claims 1 or 24, wherein said treating2 further ses administration of an olinergic agent.1 27. The use according any one of claims 1 or 24, wherein said treating further2 comprises administration of an anti-seizure agent.1 28. The use according to claim 27, wherein said anti-seizure agent is a2 benzodiazepine.1 29. The use according any one of claims 1 or 24, wherein said treating r2 ses administration of an anticholinergic agent and an anti-seizure agent.1 30. The use according any one of claims 1 or 24, n said treating further2 comprises administration of an acetylcholinesterase reactivating agent.1 31. The use according to claim 30, wherein said acetylcholinesterase2 reactivating agent is pralidoxime.1 32. The use according any one of claims 1 or 24, wherein said treating occurs2 prior to said exposure to said chemical threat agent.1 33. The use according to claim 32, wherein said ng occurs at least 302 minutes prior to said exposure to said chemical threat agent.1 34. The use according to claim 21, wherein the effect of said medicament in2 treating brain injury lasts for at least 90 minutes following administration.1 35. The use according to claim 21, wherein said compound has the structure2 of Formula (I) or Formula (II).1 36. The use according to claim 35, wherein said nd has the structure2 of Formula (II).1 37. The use according to claim 36, wherein said metal is manganese, iron,2 cobalt, copper, nickel, or zinc.1 38. The use according to claim 37, wherein said metal is manganese.1 39. The use according to claim 35,2 whereinR5 N + N R63 R1, R2, R3, and R4 are each ; and4 R5 and R6 are ndently unsubstituted alkyl.1 40. The use according to claim 39, n said compound has the structure2 of Formula (VII)N NN NN NMn+ +N NN NN + N3 (VII).4 41. The use according to claim 1 or claim 21 substantially as herein described.
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US201161566530P | 2011-12-02 | 2011-12-02 | |
| US61/566,530 | 2011-12-02 | ||
| PCT/US2012/067633 WO2013130150A2 (en) | 2011-12-02 | 2012-12-03 | Metalloporphyrin neurological treatments |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| NZ625066A NZ625066A (en) | 2016-05-27 |
| NZ625066B2 true NZ625066B2 (en) | 2016-08-30 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Aquilonius et al. | Clinical pharmacokinetics of cholinesterase inhibitors | |
| ES2215305T3 (en) | ANALGESIC METHODS USING THE SYNTHETIC CATALYSTS FOR THE DISMUTATION OF SUPEROXIDE RADICALS. | |
| EP3452040B1 (en) | Formulations and treatments employing hydroxypyridonate actinide/lanthanide decorporation agents | |
| ES2374399T3 (en) | PIRIDOXAMINE FOR USE IN THE TREATMENT OF DIABETIC NEPHROPATHY IN TYPE II DIABETES. | |
| US9295674B2 (en) | Metalloporphyrin neurological treatments | |
| Kim et al. | Effects of combinational prophylactics composed of physostigmine and procyclidine on soman-induced lethality, seizures and brain injuries | |
| US20070093518A1 (en) | Pharmaceutical compositions for the treatment of organophosphate poisoning | |
| CA2076816A1 (en) | Nerve gas antidote | |
| Bajgar et al. | Some possibilities to study new prophylactics against nerve agents | |
| Wu et al. | Regulation of kynurenic acid synthesis studied by microdialysis in the dorsal hippocampus of unanesthetized rats | |
| Taysse et al. | Comparative efficacy of diazepam and avizafone against sarin-induced neuropathology and respiratory failure in guinea pigs: influence of atropine dose | |
| NZ625066B2 (en) | Metalloporphyrin neurological treatments | |
| US10314857B2 (en) | Compositions and methods for treating carbon monoxide and/or cyanide poisoning | |
| Harris et al. | Reactivation of VX-inhibited cholinesterase by 2-PAM and HS-6 in rats | |
| Myhrer et al. | Supralethal poisoning by any of the classical nerve agents is effectively counteracted by procyclidine regimens in rats | |
| US6660762B2 (en) | Methods and compositions to treat lung diseases | |
| Taysse et al. | Protection against soman-induced neuropathology and respiratory failure: a comparison of the efficacy of diazepam and avizafone in guinea pig | |
| Krejcova et al. | Cyclosarin-an organophosphate nerve agent | |
| Barker et al. | The Kv7 modulator, retigabine, is an efficacious antiseizure drug for delayed treatment of organophosphate-induced status epilepticus | |
| Karalliedde et al. | Management of organophosphorus compound poisoning | |
| EP2568807B1 (en) | Phenoxyalkyl pyridinium oxime therapeutics for treatment of organophosphate poisoning | |
| Mak et al. | Experimental chemotherapy of lymphatic filariasis. A review | |
| Wolthuis et al. | Search for a therapy against soman-intoxication | |
| EP1087776A1 (en) | A process for the treatment of organophosphate poisoning | |
| Kassa et al. | The influence of oxime selection on the efficacy of antidotal treatment of soman-poisoned rats |