EP1797100A1 - Nitrate esters and their use for mitigating cellular damage - Google Patents

Nitrate esters and their use for mitigating cellular damage

Info

Publication number
EP1797100A1
EP1797100A1 EP05787832A EP05787832A EP1797100A1 EP 1797100 A1 EP1797100 A1 EP 1797100A1 EP 05787832 A EP05787832 A EP 05787832A EP 05787832 A EP05787832 A EP 05787832A EP 1797100 A1 EP1797100 A1 EP 1797100A1
Authority
EP
European Patent Office
Prior art keywords
unsubstituted
substituted
group
nitrate
nhr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP05787832A
Other languages
German (de)
French (fr)
Other versions
EP1797100A4 (en
Inventor
Gregory R. J. Thatcher
Brian M. Bennett
James N. Reynolds
Roland J. Boegman
Khem Jhamandas
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Queens University at Kingston
Original Assignee
Queens University at Kingston
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/943,264 external-priority patent/US20050137191A1/en
Application filed by Queens University at Kingston filed Critical Queens University at Kingston
Publication of EP1797100A1 publication Critical patent/EP1797100A1/en
Publication of EP1797100A4 publication Critical patent/EP1797100A4/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • C07D217/16Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/79Acids; Esters
    • C07D213/80Acids; Esters in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/44Benzopyrazines with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/79Benzo [b] furans; Hydrogenated benzo [b] furans with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • C07D311/66Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4 with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/14Radicals substituted by singly bound hetero atoms other than halogen
    • C07D333/16Radicals substituted by singly bound hetero atoms other than halogen by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/30Hetero atoms other than halogen
    • C07D333/32Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/54Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • C07D333/56Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D339/00Heterocyclic compounds containing rings having two sulfur atoms as the only ring hetero atoms
    • C07D339/02Five-membered rings
    • C07D339/04Five-membered rings having the hetero atoms in positions 1 and 2, e.g. lipoic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D339/00Heterocyclic compounds containing rings having two sulfur atoms as the only ring hetero atoms
    • C07D339/08Six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/10Spiro-condensed systems

Definitions

  • This invention relates to nitrate esters and use thereof in mitigating cellular damage. Particularly, this invention relates to selected organic nitrates, most particularly those bearing a sulfur or phosphorus atom ⁇ or ⁇ to the nitrate group, having therapeutic utility as agents that protect tissues from oxidative injury.
  • nitrate ester glyceryl trinitrate (GTN) or nitroglycerin, has been used as a vasodilator in the treatment of angina pectoris for over a hundred years, and the dominant contemporary belief is that GTN exerts its therapeutic effect through in vivo release of nitric oxide (NO).
  • NO nitric oxide
  • Other organic nitrates such as isosorbide dinitrate, have also been identified as effective and clinically important vasodilators.
  • NO itself has been identified as Endothelium Derived Relaxing Factor (EDRF) and several classes of compounds, for example nitrosothiols, in addition to organic nitrates, have been proposed as NO donors or NO prodrugs.
  • EDRF Endothelium Derived Relaxing Factor
  • GT ⁇ is firstly a potent vasodilator and secondly possesses potential neuroprotective properties.
  • GT ⁇ has been found to suppress renal oxidant damage caused by potassium bromate (Rahman et al., Redox Rep. 4: 263-9, 1999).
  • nitrate esters having the formula
  • each of m and n is, independently, an integer from O to 10; each of R , R , R , 17 is, independently, hydrogen, a nitrate group, or A;
  • R 1 is hydrogen or A; where A is selected from: a substituted or unsubstituted aliphatic group having from 1 to 24 carbon atoms in the chain, which optionally contains 1 to 4 O, S, NR 6 , and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted cyclic aliphatic moiety having from 3 to 7 carbon atoms in the aliphatic ring, which optionally contains 1 to 2 O, S, NR 6 , and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted aliphatic moiety comprising a linkage from 0 to 5 carbon atoms between R 1 and R 3 and/or between R 17 and R 4 , which optionally contains 1 to 2 O, S, NR 6 , and/or uns
  • M is H, Na + , K + , NH 4 + , or N + H k R 1 ' ⁇ 4-k) where k is 0 to 3;
  • one of m, n, or p must be equal to at least one.
  • the compound of the invention is:
  • G 1 is Me, OMe, Cl, NO 2 , Br, or H;
  • G 2 is CO 2 Et,
  • the compound is:
  • the compound is:
  • n 2 or 3.
  • the compound is:
  • the compound is:
  • the compound is:
  • the invention features a compound having the formula:
  • nitrate groups containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein each of m and n is, independently, an integer from O to 10;
  • R 1 is a hydrogen or A
  • each of R »2 , n R5 , and R , 18 is, independently, hydrogen or A
  • each of R 3 , R 4 , and R 17 is independently, a hydrogen, a nitrate group, or
  • each of R 6 , R 7 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , and R 16 is, independently, A, a hydrogen, a nitrate group, or a C 1 -C 24 alkyl or acyl group, optionally containing 1-4 ONO 2 substituents or a CpC 6 linkage to R 1 , R 2 , R 3 , or R 4 in cyclic derivatives; each of R 7 and R 11 is, independently, a substituted or unsubstituted C 1 - Cg alkyl or acyl group;
  • each of R 18 and R 3 is, independently, a hydrogen, a nitrate group, or a C 1 -C 4 alkyl chain, which may include one O, linking R 18 and R 3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R 17 and R 4 is, independently, a hydrogen, a nitrate group, a C 1 - C 4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (- C(O)R 5 ); each of R 5 , R 6 , R 8 , R 9 ,
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the invention features a compound having the formula:
  • the invention features a composition that includes a pharmaceutically acceptable carrier and a compound having the general formula:
  • each of m and n is, independently, an integer from O to 10;
  • R 1 is a hydrogen or A;
  • 2 , r R> 5 , and R , 18 is, independently, hydrogen or A;
  • each of R » 3 , ⁇ R4 , and R , 17 is independently, a hydrogen, a nitrate group, or
  • each of R 6 , R 7 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , and R 16 is, independently, A, a hydrogen, a nitrate group, or a C 1 -C 24 alkyl or acyl group, optionally containing 1-4 ONO 2 substituents or a C 1 -C 6 linkage to R 1 , R 2 , R 3 , or R 4 in cyclic derivatives; each of R 7 and R 11 is, independently, a substituted or unsubstituted Q- C 8 alkyl or acyl group;
  • each of R 18 and R 3 is, independently, a hydrogen, a nitrate group, or a C 1 -C 4 alkyl chain, which may include one O, linking R 18 and R 3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R 17 and R 4 is, independently, a hydrogen, a nitrate group, a Q- C 4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (- C(O)R 5 ); each of R 5 , R 6 , R 8 , R 9
  • the invention features a composition that includes a pharmaceutically acceptable carrier and one of the following compounds:
  • the invention features a method for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject by administering to the subject an effective amount of a compound containing at least one aliphatic nitrate group and at least one sulfur atom in proximity to the nitrate group.
  • sulfur atom in proximity or "proximal functional group” is meant a sulfur atom or functional group that is connected through bonds in a ⁇ , ⁇ , or ⁇ relationship to a nitrate ester group (i.e., the atom connectivity is 1,2, or 1,3, or 1,4).
  • the functional group may also be referred to as "proximally located” or “situated in proximity.”
  • Proximal functional groups also include those groups that have a through-space intramolecular juxtaposition with a nitrate group that is within 3 A.
  • the tissue and/or cellular damage can be associated with aging, septic shock, ischemia/reperfusion injury, ulcers, gastritis, ulcerative colitis, Crohn's disease, diabetes, rheumatoid arthritis, asthma, cirrhosis of the liver, allograft rejection, encephalomyelitis, meningitis, pancreatitis, peritonitis, vasculitis, lymphocytic choriomeningitis, glomerulonephritis, uveitis, glaucoma, blepharitis, chalazion, allergic eye disease, corneal ulcer, keratitis, cataracts, age-related macular degeneration, optic neuritis, ileitis, hemorrhagic shock, anaphylactic shock, bacterial infection, viral infection, fungal infection, parasitic infection, hemodialysis, chronic fatigue syndrome, stroke, toxic shock syndrome, adult respiratory distress syndrome, cachexia, myocarditis, ecze
  • the tissue and/or cellular damage can associated with neurological diseases such as, for example, Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amyotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia due to myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; or environmental excitotoxins of plant, animal, or marine origin.
  • neurological diseases such as, for example, Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amyotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia due to myocardial infarction; cerebral vascular occlusion; cerebral vascular hemor
  • tissue and/or cellular damage can be associated with cytokine therapy, wherein a nitrate ester of the invention is administered to the subject before, during, and/or after the administration of the therapeutic cytokine.
  • the compound has the formula:
  • n is an integer from 0 to 10; each of R 3>4>17 is, independently, hydrogen, a nitrate group, or A;
  • R 1 is hydrogen or A
  • A is selected from: a substituted or unsubstituted C 1 -C 24 alkyl group, optionally containing 1 to 4 O, S, NR 6 , and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted cyclic moiety having from 3 to 7 carbon atoms in the ring, which optionally containing 1 to 2 O, S, NR 6 , and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted moiety constituting a linkage from 0 to 5 carbons, to or between any of R 1 , R 3 , R 17 and R 4 ,
  • R 2 , R 5 , R 18 are optionally hydrogen, A, or X-Y;
  • each of R 6 , R 7 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , and R 16 is, independently, a C 1 -C 24 alkyl group, optionally containing 1-4 ONO 2 substituents, a C 1 -C 24 acyl group, optionally containing 1-4 ONO 2 substituents, a CpC 6 ring-forming connection to any of R ! -R 4 , a hydrogen, a nitrate group, or A; and
  • M is H, Na + , K + , NH 4 + , N + H k R 1 ⁇ 4 . k) where k is 0-3;
  • the compound contains at least 2 nitrate groups.
  • the nitrate is beta or gamma to said sulfur atom.
  • the compound in another embodiment, can be any of the compounds that were cited individually as an embodiment of either the first or second aspect of the invention.
  • the compound is:
  • the compound is:
  • alkyl and the prefix “alk-” are inclusive of both straight chain and branched chain saturated or unsaturated groups, and of cyclic groups, i.e., cycloalkyl and cycloalkenyl groups.
  • acyclic alkyl groups are from 1 to 6 carbons and contain at least one C-H bond.
  • the number of carbons in an alkyl group refers to the total number of carbons contained in the group.
  • Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 8 ring carbon atoms.
  • Exemplary cyclic groups include cyclopropyl, cyclopentyl, cyclohexyl, and adamantyl groups.
  • alkyl groups may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halogen, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • aryl is meant a carbocyclic aromatic ring or ring system. Unless otherwise specified, aryl groups are from 6 to 18 carbons. . Examples of aryl groups include phenyl, naphthyl, biphenyl, fluorenyl, and indenyl groups.
  • heterocycle is meant an aromatic or non-aromatic ring system that contains at least one ring heteroatom (e.g., O, S, N).
  • heteroaryl refers to an aromatic heterocyclic ring or ring system that contains at least one ring heteroatom (e.g., O, S, N). Unless otherwise specified, heteroaryl rings contain from 1 to 9 carbons.
  • heteroaryl groups include furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, oxatriazolyl, pyridyl, pyridazyl, pyrimidyl, pyrazyl, triazyl, benzofuranyl, isobenzofuranyl, benzothienyl, indole, indazolyl, indolizinyl, benzisoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, naphtyridinyl, phthalazinyl, phenanthrolinyl, purinyl, and carbazolyl rings or ring systems.
  • non-aromatic heterocyclic groups are from 2 to 9 carbons and can include, for example, dihydropyrrolyl, tetrahydropyrrolyl, piperazinyl, pyranyl, dihydropyranyl, tetrahydropyranyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothiophene, tetrahydrothiophene, and morpholinyl groups.
  • heterocycle specifically excludes ⁇ -lactam rings.
  • aryl, heteroaryl, or heterocyclic groups may be unsubstituted or substituted by one or more substituents selected from the group consisting Of Ci -6 alkyl, hydroxy, halo, nitro, C 1-6 alkoxy, Ci -6 alkylthio, trifluoromethyl, C 1-6 acyl, arylcarbonyl, heteroarylcarbonyl, nitrile, C 1-6 alkoxycarbonyl, arylalkyl (wherein the alkyl group has from 1 to 6 carbon atoms) and heteroarylalkyl (wherein the alkyl group has from 1 to 6 carbon atoms).
  • halide or “halogen” or “halo” is meant bromine, chlorine, iodine, or fluorine.
  • alkyl and the prefix “alk-” are inclusive of both straight chain and branched chain saturated or unsaturated groups, and of cyclic groups, i.e., cycloalkyl and cycloalkenyl groups. Unless otherwise specified, acyclic alkyl groups are from 1 to 6 carbons. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 8 ring carbon atoms. Exemplary cyclic groups include cyclopropyl, cyclopentyl, cyclohexyl, and adamantyl groups.
  • Alkyl groups may be substituted or unsubstituted.
  • substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halogen, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
  • alkoxy is meant a chemical substituent of the formula -OR, where R is an alkyl group.
  • aryloxy is meant a chemical substituent of the formula -OR', where R' is an aryl group.
  • alkaryl is meant a chemical substituent of the formula -RR', where R is an alkyl group and R' is an aryl group,
  • alkheteraryl is meant a chemical substituent of the formula RR", where R is an alkyl group and R" is a heteroaryl group.
  • aliphatic nitrate is a nitrate group that is connected to a non-aromatic carbon.
  • antioxidant is a compound that attenuates or prevents oxidation of a target biomolecule through the inhibition of the initiation or propagation steps that constitute oxidative and peroxidative processes.
  • “Mitigating neurodegeneration” as used herein involves affecting neuroprotection, inhibiting or preventing neurodegeneration, and/or ameliorating the manifestations or impact of neurodegeneration. Such amelioration includes effecting cognition enhancement, as is quantified by tests known in the art (e.g., Venault et al., 1992, incorporated herein by reference).
  • “Modulating” a biological process as used herein encompasses both increasing (positively modulating) and decreasing (negatively modulating) such activity, and thus inhibition, potentiation, agonism, and antagonism of the biological process.
  • Compounds may be constructed according to formulas of the invention in which an S atom is appropriately proximally placed with respect to a nitrate functional group, but in which a carbonyl group intervenes to form a thioester linkage. Such compounds do not form part of the invention if cleavage of this thioester bond produces two entirely separate molecules, one containing the nitrate funtionality, and another the S-functionality.
  • An example of such a compound is one containing the motif, O 2 NOCH 2 C(CH 3 ) 2 C(O)S-(organic radical), which would liberate on thioester cleavage an aliphatic nitrate that does not contain a S-functionality.
  • Figure 1 is a synthetic scheme showing the use of Bunte salt IVd in the preparation of compounds Va, IVr, Vbb, and Vbc.
  • Figure 6 is a graph showing a comparison of 50 ⁇ M GTN (stippled bars) and 50 ⁇ M Va (hatched bars) on coronary perfusion pressure in isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min.
  • Hearts were perfused with drugs for ten min prior to and throughout the period of LCAO.
  • Figure 7 is an immunocytochemical analysis of tyrosine hydroxylase immunoreactivity in the substantia nigra of rats.
  • Figure 7 A is an analysis of a brain section in which the animal was administered the neurotoxic compound 6- hydroxydopamine (6-OHDA).
  • Figure 7B is an analysis of a brain section in which the animal was administered only vehicle control (dimethyl sulfoxide).
  • Figure 7C is an analysis of a brain section in which the animal was treated with compound Va before and after administration of 6-OHDA.
  • Figure 7D is an analysis of a brain section in which the animal was administered only vehicle control.
  • Va varied
  • ImM cysteine
  • Va + PhSH Va + PhSH (1 mM)
  • This invention pertains to methods and compositions useful for treating neurodegeneration or preventing or mitigating tissue and/or cellular damage by administering to a subject a therapeutic nitrate ester.
  • Neuroprotection and/or cognition enhancement can be affected, for example, by modulating an interaction with guanylyl cyclase (GCase, the enzyme responsible for cGMP production in various areas of the brain), modulating a glutamate or non- glutamate neuroreceptor or attenuating free radical damage.
  • GCase guanylyl cyclase
  • the attenuation of free radical concentration by a nitrate ester of the invention can also be useful for preventing or mitigating tissue and/or cellular damage.
  • neurodegeneration is mitigated by stimulating cerebral GCase.
  • One of the major targets for the novel organic nitrates of the invention is GCase activation, resulting in the production of cGMP.
  • Experimental evidence obtained in a number of in vitro model systems supports the notion that elevated levels of cGMP help to prevent apoptotic (programmed) cell death.
  • a cGMP-dependent mechanism significantly increases the survival of trophic factor-deprived PC 12 cells and rat sympathetic neurons (Farinelli et al., 1996), and of primary cultures of rat embryonic motor neurons (Estevez et al., 1998).
  • the mechanism of action for selected organic nitrates in preventing apoptotic cell death may be inhibition of caspase-3 activation indirectly through elevations in cGMP levels or directly via protein S-nitrosylation of the enzyme by an NO-intermediate (Kim et al., 1997).
  • Caspase-3 is a member of the cysteine protease family of enzymes that are essential for the execution step in apoptosis (Cohen, 1997; Nicholson and Thornberry, 1997).
  • caspase-3 Activation of caspase-3 is required for apoptotic cell death in trophic factor-deprived PC 12 cells (Haviv et al., 1997) and in glutamate- mediated apoptotic cell death of cultured cerebellar granule neurons (Du et al., 1997).
  • caspase-3 activity is induced and may be responsible for the apoptotic component of delayed neuronal cell death (Chen et al., 1998; Namura et al., 1998; Ni et al., 1998).
  • Inhibitors of caspase-3 significantly decrease the apoptotic component of delayed neuronal cell death in response to ischemic injury both in vitro (Gottron et al., 1997) and in vivo (Endres et al., 1998).
  • a secreted region of the Alzheimer's disease ⁇ -amyloid precursor protein lowers intracellular calcium levels and provides neuroprotective effects on target cells through increases in cGMP levels and activation of protein kinase G (Barger et al., 1995; Furukawa et al., 1996).
  • nitrated molecules that have the capacity to activate GCase directly or via release of an NO-containing intermediate are used to modulate GCase activity.
  • cognition enhancement is achieved by stimulating cerebral GCase.
  • GCase and cGMP are involved in the formation and retention of new information.
  • cGMP has been directly implicated in both long-term potentiation (LTP) and long-term depression (LTD), which are proposed cellular models for learning and memory (Arancio et al., 1995; Wu et al., 1998).
  • LTP long-term potentiation
  • LTD long-term depression
  • elevation of hippocampal cGMP levels leading to increased protein kinase G activity has been shown to be important for retention and consolidation of new learning (Bernabeu et al., 1996, 1997).
  • stimulation of cerebral GCase activity is expected to improve learning and memory performance in individuals in whom cognitive abilities are impaired by injury, disease, or aging.
  • Cerebral ischemia results in marked increases in the release of the excitatory amino acid glutamate in the affected brain region (Bullock et al., 1998; Huang et al., 1998; Yang et al., 1998).
  • the amount of glutamate released during ischemia is positively correlated with the extent of brain injury.
  • these selected organic nitrates can be used for treatment of conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntingdon's disease; multiple sclerosis; amyotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant, animal, or marine origin; and the like.
  • selected organic nitrates developed to act as modulators of GABA A receptor function, will be to improve memory performance and cognition in patient populations. It will be appreciated, therefore, that these selected organic nitrates can be used for treatment of conditions including but not limited to: stroke; dementias of all type; trauma; drug-induced brain damage; and aging.
  • neurodegeneration is mitigated by inhibition of free radical damage.
  • Reoxygenation and reperfusion after a period of ischemia contributes significantly to the development of brain injury.
  • Oxygen radicals, especially superoxide and peroxynitrite, formed in the period after an ischemic event may initiate processes such as breakdown of membrane lipids (lipid peroxidation), leading to loss of cell membrane integrity and inhibition of mitochondrial function (Macdonald and Stoodley, 1998; Gaetani et al, 1998).
  • Oxidative stress is also believed to be one factor involved in initiation of apoptotic neuronal cell death (Tagami et al., 1998).
  • 6-OHDA is a neurotoxin selectively taken up into dopaminergic neurons, resulting in a selective killing of these neurons, via a mechanism involving oxidative stress that is evident by 4 days after injection of the toxin.
  • MAO-B monoamine oxidase type B
  • deprenyl can prevent 6-OHDA-induced killing of dopaminergic neurons.
  • deprenyl as a positive control in this study.
  • Loss of dopaminergic innervation to the striatum results in an upregulation of postsynaptic dopamine receptors, and the development of dopamine receptor supersensitivity 2-3 weeks after 6-OHDA lesioning of the substantia nigra.
  • the invention features nitrated molecules which have the capacity to inhibit production of free radicals and/or which act as free radical scavengers.
  • Free radical overproduction is associated with a wide range of disease states and/or indications, such as, for example, aging, septic shock, ischemia, overexpression of cytokines, ulcers, inflammatory bowel disease (e.g., gastritis, ulcerative colitis or Crohn's disease), diabetes, arthritis (e.g., rheumatoid arthritis), asthma, cirrhosis, allograft rejection (e.g., transplant rejection), encephalomyelitis, meningitis, pancreatitis, peritonitis, vasculitis, lymphocytic choriomeningitis, glomerulonephritis, ophthalmologic diseases (e.g., uveitis, glaucoma, blepharitis, chalazion, allergic eye disease, corneal ulcer, keratitis, cataract, retinal disorders, age-related macular degeneration, optic neuritis, and the like), ileitis, inflammation induced by overproduction
  • coli infection e.g., HIV
  • fungal e.g., Candidiosis and histoplasmosis
  • parasitic e.g., Leishmaniasis and Schistosomiasis
  • hemodialysis chronic fatigue syndrome, stroke, cancers, including metastatic cancers (e.g., breast cancer, bladder cancer, lung cancer, colon cancer, or cancer of the other organs, or skin or other noncutaneous portions of the body), cardiovascular diseases associated with overproduction of inflammatory cytokines (e.g., heart disease, cardiopulmonary bypass, ischemic/reperfusion injury, and the like), ischemic/reperfusion associated with overproduction of inflammatory cytokines, toxic shock syndrome, adult respiratory distress syndrome, cachexia, myocarditis, autoimmune disorders, eczema, psoriasis, heart failure, dermatitis, urticaria, cerebral ischemia, systemic lupus erythematosis, AIDS, neurodegenerative disorders (e.g., chronic cyto
  • the compounds or methods of the present invention may find use in cytokine therapy (with consequent induction of free radical overproduction) which, for example, is commonly used in the treatment of cancers, including metastatic cancers (e.g., breast cancer, bladder cancer, lung cancer, colon cancer, or cancer of the other organs, or skin or other noncutaneous portions of the body), autoimmune disease, and in AIDS patients.
  • cytokine therapy with consequent induction of free radical overproduction
  • cancers including metastatic cancers (e.g., breast cancer, bladder cancer, lung cancer, colon cancer, or cancer of the other organs, or skin or other noncutaneous portions of the body), autoimmune disease, and in AIDS patients.
  • Systemic hypotension due to the induction of free radical overproduction is a dose-limiting side effect of cytokine therapy.
  • a large patient population exists which will benefit from the invention methods.
  • any organic nitrate in which vasodilatory potency is reduced and neuroprotective potency increased represents a new and useful therapeutic agent for use in neuroprotection, particularly in treatment of conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amylotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever- induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant, animal, or marine origin.
  • GTN itself, proposed as a neuroprotective agent, has no clinical utility as a neuroprotective agent in therapy owing to its extraordinarily high vasodilatory potency.
  • GTN 1,2,3-trinitratopropane
  • cognition enhancement represents a new and useful treatment for cognition enhancement, particularly in treatment of conditions including but not limited to: stroke; dementias of all type, trauma, drug-induced brain damage, and aging.
  • the therapeutic compounds of the invention comprise at least one nitrate group.
  • the nitrate groups(s) can optionally be covalently bound to a carrier moiety or molecule (e.g., an aromatic group, an aliphatic group, peptide, steroid, nucleoside, peptidomimetic, steroidomimetic, or nucleoside analogue, or the like).
  • a carrier moiety or molecule e.g., an aromatic group, an aliphatic group, peptide, steroid, nucleoside, peptidomimetic, steroidomimetic, or nucleoside analogue, or the like.
  • the carrier moiety or molecule can enable the compound to traverse biological membranes and to be biodistributed preferentially, without excessive or premature metabolism.
  • the carrier moiety or molecule can enable the compound to exert amplified neuroprotective effects and/or cognition enhancement through synergism with the nitrate functionality.
  • the invention provides a method of treating a neurological condition and/or preventing an undesirable mental condition (e.g., memory loss) including the step of administering to a subject an effective amount of a therapeutic compound capable of mitigating neurodegeneration which has at least one nitrate group.
  • a therapeutic compound capable of mitigating neurodegeneration which has at least one nitrate group.
  • the therapeutic compound is capable of effecting neuroprotection.
  • the therapeutic compound is capable of effecting cognition enhancement.
  • the therapeutic compound has the formula (Formula I):
  • E, F, G are organic radicals which may contain inorganic counterions; so that a neurological condition is treated.
  • the invention provides a pharmaceutical composition including a physiologically acceptable carrier and a compound having the formula (Formula I):
  • E, F, G are organic radicals which may contain inorganic counterions.
  • the composition is employed for mitigating neurodegeneration, effecting neuroprotection and /or effecting cognition enhancement.
  • the composition may also be employed for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject.
  • therapeutic compounds of the invention that effect neuroprotection and/or effect cognition enhancement in a subject to which the therapeutic compound is administered have the formula (Formula II):
  • compositions comprising a compound of Formula II in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
  • the invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula II to a subject such that said mitigation and /or said neuroprotection an/or cognition enhancement occurs.
  • preferred therapeutic compounds for effecting neuroprotection and/or cognition enhancement in a subject to which the compound is administered include compounds in which R 19 is X-Y.
  • R 19 is X-Y and R 5 , R 6 , R 8 , R 9 , R 12 , R 13 , R 14 , R 15 , R 16 are the same or different alkyl groups containing 1-24 carbon atoms which may contain 1-4 ONO 2 substituents, or C 1 or C 2 connections to R 1 - R 3 in cyclic derivatives;
  • R 1 and R 3 are the same or different and selected from H, C r C 4) alkyl chains, which may inlude one O, linking R 1 and R 3 to form pentosyl, hexosyl, cyclopentyl, or cycohexyl rings, which rings optionally bear hydroxyl substituents;
  • R 2 and R 4 are the same or different and selected from H, a nitrate group, C
  • R 19 is X-Y
  • R 19 is X-Y
  • R 19 is X-Y
  • X and/or Y contains a sulfur-containing functional group.
  • the compound of the invention comprises a heterocyclic functionality, more preferably, a nucleoside or nucleobase.
  • the compound of the invention comprises a carbocyclic functionality, more preferably, a steroidal or carbohydrate moiety.
  • a therapeutic compound of the invention is represented by the formula (Formula III):
  • R 6 - R 9 and R 1 ' - R 16 are the same or different alkyl or acyl groups containing 1-24 carbon atoms which may contain 1-4 ONO 2 substituents, or C 1 - C6 connections to R 1 - R 4 in cyclic derivatives.
  • a therapeutic compound of the invention is represented by the formula III in which the compound contains from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, where each of m and n is, independently, an integer from 0 to 10; R 1 is a hydrogen or A; each of R 2 , R 5 , and R 18 is, independently, hydrogen or A; each of R 3 , R 4 , and R 17 , is independently, a hydrogen, a nitrate group, or A; each of R 6 , R 7 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , and R 16 is, independently, A, a hydrogen, a nitrate group, or a C 1 -C 24 alkyl or acyl group, optionally containing 1-4 ONO 2 substituents or a C 1 -C 6 linkage to R 1 , R 2 ,
  • compositions comprising a compound of Formula III in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
  • the invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula III to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs.
  • a therapeutic compound of the invention can be represented by the formula (Formula IV):
  • R 2 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , Rn, R 12 , Ri 3 , Rj 4 , Ri 5 , and Ri 6 are as defined above.
  • R 2 and R 4 are optionally H, a nitrate group or a connection to R 5 -R 9 and Ri i-Ri 6 in cyclic derivatives.
  • compositions comprising a compound of Formula IV in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
  • the invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula IV to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs.
  • a compound of the invention can be represented by the formula (Formula V):
  • R 2 is optionally H or a connection to R 5 in cyclic derivatives
  • R 4 is H or a nitrate group
  • R 5 is as described above.
  • compositions comprising a compound of Formula V in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
  • the invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula V to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs.
  • Examples and preferred embodiments of compounds of the invention according to formula V are as follows:
  • the invention features one of the following nitrate esters:
  • the invention features a method for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject.
  • the method includes administering to the subject an effective amount of a compound containing at least one aliphatic nitrate group and at least one sulfur atom in proximity to said nitrate, such as, for example, a compound of formula I.
  • the nitrate of this method contains at least 2 nitrate groups.
  • the nitrate is beta or gamma to a sulfur atom.
  • Yet another embodiment features compounds of formula III for preventing or mitigating tissue and/or cellular damage
  • m is an integer from O to 10;
  • n is an integer from O to 10;
  • each of R 3>4>17 is, independently, hydrogen, a nitrate group, or A;
  • R 1 is hydrogen or A, with A is selected from: a substituted or unsubstituted C 1 -C 24 alkyl group, optionally containing 1 to 4 O, S, NR 6 , and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted cyclic moiety having from 3 to 7 carbon atoms in the ring, which optionally contains O, S, NR 6 , and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, Cl
  • R 2 , R 5 , R 18 are optionally hydrogen, A, or X-Y;
  • each of R 6 , R 7 , R 8 , R 9 , R 10 , R 1 ', R 12 , R 13 , R 14 , R 15 , and R 16 is, independently, a C 1 -C 24 alkyl group, optionally containing 1-4 ONO 2 substituents, a CpC 24 acyl group, optionally containing 1-4 ONO 2 substituents, a CpC 6 ring- forming connection to any of R'-R 4 , a hydrogen, a nitrate group, or A; and
  • Preferred compounds include any of the compounds of formulas II, III, IV, and V that have been individually described herein. Other preferred compounds include:
  • Particularly preferred compounds for preventing or mitigating tissue and/or cellular damage are:
  • a therapeutic compound of the invention comprises a cation (i.e., in certain embodiments, one of X or Y includes a cation, e.g., in the compound of formula IVd).
  • M can be a pharmaceutically acceptable alkali metal (e.g. Li, Na, K), ammonium, alkaline earth metal (e.g. Ca, Ba, Mg), higher valency cation, or polycationic counter ion (e.g., polyammonium cation) (see e.g., Berge et al., 1977).
  • alkali metal e.g. Li, Na, K
  • ammonium e.g. Ca, Ba, Mg
  • alkaline earth metal e.g. Ca, Ba, Mg
  • polycationic counter ion e.g., polyammonium cation
  • an anionic portion of the compound to a salt- forming cation will vary depending on the charge of the anionic portion of the compound and the charge of the counterion.
  • Preferred pharmaceutically acceptable salts include a sodium, potassium, or calcium salt, but other salts are also contemplated within their pharmaceutically acceptable range.
  • a therapeutic compound of the invention can be administered in a pharmaceutically acceptable vehicle.
  • pharmaceutically acceptable vehicle includes any and all solvents, excipients, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like which are compatible with the activity of the compound and are physiologically acceptable to the subject.
  • An example of a pharmaceutically acceptable vehicle is buffered normal saline (0.15 M NaCl).
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compound, use thereof in the compositions suitable for pharmaceutical administration is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • Carrier or substituent moieties useful in the present invention may also include moieties which allow the therapeutic compound to be selectively delivered to a target organ.
  • delivery of the therapeutic compound to the brain may be enhanced by a carrier moiety using either active or passive transport (a "targeting moiety").
  • the carrier molecule may be a redox moiety, as described in, for example, U.S. Patents 4,540,654 and 5,389,623, both to Bodor. These patents disclose drugs linked to dihydropyridine moieties which can enter the brain, where they are oxidized to a charged pyridinium species which is trapped in the brain. Thus drugs accumulate in the brain.
  • carrier moieties include compounds, such as amino acids or thyroxine, which can be passively or actively transported in vivo. Such a carrier moiety can be metabolically removed in vivo, or can remain intact as part of an active compound. Structural mimics of amino acids (and other actively transported moieties) including peptidomimetics, are also useful in the invention.
  • peptidomimetic is intended to include peptide analogues which serve as appropriate substitutes for peptides in interactions with, for example, receptors and enzymes. The peptodomimetic must possess not only affinity, but also efficacy and substrate function.
  • a peptidomimetic exhibits functions of a peptide, without restriction of structure to amino acid constituents.
  • Peptidomimetics and methods for their preparation and use are described in Morgan et al. (1989), the contents of which are incorporated herein by reference.
  • Many targeting moieties are known, and include, for example, asialoglycoproteins (see e.g., Wu, U.S. Patent 5,166,320) and other ligands which are transported into cells via receptor-mediated endocytosis (see below for further examples of targeting moieties which may be covalently or non-covalently bound to a target molecule).
  • neurodegeneration in a subject is mitigated, and/or neuroprotection and/or cognition enhancement is effected, by administering a therapeutic compound of the invention to the subject.
  • the invention also features methods for preventing or mitigating tissue and/or cellular damage in a subject by administering a therapeutic compound of the invention to the subject, thereby modulating intercellular and/or intracellular free radical concentration.
  • subject is intended to include living organisms in which the particular neurological condition to be treated can occur. Examples of subjects include humans, apes, monkeys, cows, sheep, goats, dogs, cats, mice, rats, and transgenic species thereof.
  • biochemical cascade initiated by cerebral ischemia is generally accepted to be identical in mammalian species (Mattson and Scheff, 1994; Higashi et al., 1995).
  • pharmacological agents that are neuroprotective in animal models such as those described herein are believed to be predictive of clinical efficacy in humans, after appropriate adjustment of dosage.
  • memory-deficit patterns between brain-damaged rats and humans, which indicates that the rat can serve as an excellent animal model to evaluate the efficacy of pharmacological treatments or brain damage upon memory (Kesner, 1990).
  • tissue plasminogen activator which is administered at a dose of 0.9 mg/kg by intravenous injection (Wittkowsky, 1998).
  • This drug is also effective in protecting the rat brain subjected to cerebral ischemia by occlusion of the middle cerebral artery, when administered at a dose of 10 mg/kg intravenously (Jiang et al., 1998).
  • the rat model of focal cerebral ischemia used in the development of the novel organic nitrate esters described herein has been shown to be shown to be predictive of clinical efficacy with at least one other class of pharmacological agents.
  • the invention further encompasses methods of the invention employed ex vivo or in vitro.
  • the Examples describe studies utilizing tissue homogenates according to the invention.
  • diagnostic tests or studies of efficacy of selected compounds may conveniently be performed ex vivo or in vitro, including in animal models. Such tests, studies and assays are within the scope of the invention.
  • compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A.R. Gennaro, Lippincott Williams & Wilkins, 2000, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • Administration of the compositions of the present invention to a subject to be treated can be carried out using known procedures, at dosages and for periods of time effective to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in the subject.
  • an effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the amount of neurodegeneration that has already occurred at the clinical site in the subject, the age, sex, and weight of the subject, and the ability of the therapeutic compound to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in the subject.
  • Dosage regimens can be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • a non-limiting example of an effective dose range for a therapeutic compound of the invention (e.g., Va) is between 0.5 and 500 mg/kg of body weight per day.
  • preferred concentrations for the active compound are between 5 and 500 mM, more preferably between 10 and 100 mM, and still more preferably between 20 and 50 mM.
  • the therapeutic compounds of the invention can be effective when administered orally. Accordingly, a preferred route of administration is oral administration. Alternatively, the active compound may be administered by other suitable routes such as transdermal, subcutaneous, intraocular, intravenous, intramuscular or intraperitoneal administration, and the like (e.g., by injection). Depending on the route of administration, the active compound may be coated in a material to protect the compound from the action of acids, enzymes and other natural conditions which may inactivate the compound.
  • the compounds of the invention can be formulated to ensure proper distribution in vivo.
  • the blood-brain barrier excludes many highly hydrophilic compounds.
  • the therapeutic compounds of the invention cross the BBB, they can be formulated, for example, in liposomes.
  • liposomes For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811 ; 5,374,548; and 5,399,331.
  • the liposomes may comprise one or more moieties which are selectively transported into specific cells or organs ("targeting moieties"), thus providing targeted drug delivery (see, e.g., Ranade et al., 1989).
  • Exemplary targeting moieties include folate and biotin (see, e.g., U.S. Patent 5,416,016 to Low et al.); mannosides (Umezawa et al., 1988); antibodies (Bloeman et al., 1995; Owais et al., 1995); and surfactant protein A receptor (Briscoe et al., 1995).
  • the therapeutic compounds of the invention are formulated in liposomes; in a more preferred embodiment, the liposomes include a targeting moiety.
  • anionic groups such as phosphonate or carboxylate can be esterified to provide compounds with desirable pharmocokinetic, pharmacodynamic, biodistributive, or other properties.
  • anionic groups such as phosphonate or carboxylate can be esterified to provide compounds with desirable pharmocokinetic, pharmacodynamic, biodistributive, or other properties.
  • Exemplary compounds include IVl and pharmaceutically acceptable salts or esters thereof.
  • the therapeutic compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent.
  • suitable diluents include saline and aqueous buffer solutions.
  • Liposomes include water- in-oil-in- water CGF emulsions as well as conventional liposomes (Strejan et al., 1984).
  • the therapeutic compound may also be administered parenterally (e.g., intramuscularly, intravenously, intraperitoneally, intraspinally, or intracerebrally).
  • Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists.
  • the vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • Sterile injectable solutions can be prepared by incorporating the therapeutic compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the therapeutic compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yield a powder of the active ingredient (i.e., the therapeutic compound) optionally plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the therapeutic compound can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the therapeutic compound and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • the percentage of the therapeutic compound in the compositions and preparations may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of neurological conditions in subjects.
  • Therapeutic compositions can be administered in time-release or depot form, to obtain sustained release of the therapeutic compounds over time.
  • the therapeutic compounds of the invention can also be administered transdermally (e.g., by providing the therapeutic compound, with a suitable carrier, in patch form).
  • Active compounds are administered at a therapeutically effective dosage sufficient to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in a subject.
  • a "therapeutically effective dosage” preferably mitigates neurodegeneration by about 20%, more preferably by about 40%, even more preferably by about 60%, and still more preferably by about 80% relative to untreated subjects.
  • the ability of a compound to mitigate neurodegeneration can be evaluated in model systems that may be predictive of efficacy in mitigating neurodegeneration in human diseases, such as animal model systems known in the art (including, e.g., the method of transient middle cerebral artery occlusion in the rat) or by in vitro methods, (including, e.g., the assays described herein).
  • animal model systems known in the art (including, e.g., the method of transient middle cerebral artery occlusion in the rat) or by in vitro methods, (including, e.g., the assays described herein).
  • the ability of a compound of the invention to mitigate neurodegeneration will, in certain embodiments, be evaluated by observation of one or more symptoms or signs associated with neurodegeneration in vivo.
  • the ability of a compound to mitigate neurodegeneration may be associated with an observable improvement in a clinical manifestation of the underlying neurodegeneration-related disease state or condition, or a slowing or delay in progression of symptoms of the condition.
  • monitoring of clinical manifestations of disease can be useful in evaluating the neurodegeneration-mitigating efficacy of a compound of the invention.
  • neurodegeneration can be associated with conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amylotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant; animal, or marine origin; dementias of all type; trauma; drug-induced brain damage; and aging; or result from surgical procedures such as cardiac bypass.
  • Novel compounds according to the invention can be synthesized by methods set forth herein or in U.S. Patent Nos. 5,807,847; 5,883,122; 6,310,052; and 6,365,579, for example.
  • Various compounds for use in the methods of the invention are commercially available and/or can be synthesized by standard techniques.
  • nitrate esters can be prepared from the corresponding alcohol, oxirane or alkene by standard methods, that include: nitration of alcohols and oxiranes, mixed aqueous/organic solvents using mixtures of nitric and sulfuric acid and/or their salts, with temperature control (see Yang et al., 1996); nitration of alcohols and oxiranes in acetic anhydride using nitric acid or its salts with or without added acid catalyst, with temperature control (see, e.g., Louw et al., 1976); nitration of an alcohol with a nitronium salt, e.g. a tetrafluoroborate; nitration of an alkene with thallium nitrate in an appropriate solvent (see Ouellette et al., 1976).
  • compound IVr proceeded from the Bunte salt, 2,3-dinitrooxypropane-l-thiosulfonate (compound IVd), which was prepared from l,2-dinitrooxy-3-bromopropane as follows: 3- bromopropane-l,2-diol was added dropwise into a cold mixture Of HNO 3 (68- 70%, 4.0eq) and H 2 SO 4 (95%, 4.0 eq) in CH 2 Cl 2 (50 mL) at room temperature over 30 min.
  • HNO 3 68- 70%, 4.0eq
  • H 2 SO 4 95%, 4.0 eq
  • the organic layer was separated, washed, dried and concentrated to yield a yellow oil which was purified by flash chromatography on SiO 2 to give 3-bromopropane-l,2-diol dinitrate in 45% yield (29).
  • the Bunte salt was prepared by reacting 3-bromopropane-l,2-diol dinitrate with an equimolar portion Of Na 2 S 2 O 3 in 3: 1 MeOH/H 2 O at 50°C for 10 hours and subsequently purifying by flash chromatography on SiO 2 (29).
  • the Bunte salt was oxidized with a small molar excess Of H 2 O 2 (30%) in EtOH:H 2 O mixture (1 :1) with a catalytic amount OfH 2 SO 4 for 2 days.
  • Alkyl bromides or alkyl mercaptans were obtained commercially or by adaptation of literature procedures.
  • Bunte salts were obtained from the appropriate alkyl bromide by reaction with sodium thiosulfate, as described above for compound IVd.
  • Bunte salts (9.67 mmoles) were dissolved in distilled water (1OmL).
  • a solution of mercaptan (6.46 mmoles) in IM NaOH (7mL) was added dropwise.
  • the resulting emulsion was stirred for 1 to 15 minutes and then extracted with dichlorometane or ethylacetate.
  • the combined organic extracts were washed with H 2 O, dried over MgSO4 and concentrated under vacuum.
  • Tables 1 and 2 list characterization data determined for other compounds of the invention.
  • Rat hearts were excised and mounted for retrograde aortic perfusion at a constant flow rate of 6-8 mL/min/g heart weight.
  • the coronary perfusion pressure was monitored by a pressure transducer connected to the perfusion line.
  • the left coronary artery was occluded for 45 minutes, after which the occlusion was released and the heart reperfused for 90 minutes.
  • the LCA was re-occluded and 0.5 mL of 1% Evan's Blue dye was slowly infused into the heart, via the aotic cannula, to stain the area of myocardium perfused by the patent right coronary artery.
  • AAR area-at-risk for infarction was determined by negative staining.
  • Acute ischemic damage was assessed by measuring the release of the cytosolic enzyme, LDH, into the perfusate, and by quantitation of infarct size by staining for viable tissue using 2,3,5-triphenyltetrazolium chloride (TTC) followed by computerized planimmetry.
  • Infarct size was expressed as infarct area (negative staining after TTC staining) as a percent of the area-at-risk (negative staining after Evan's Blue dye).
  • Synaptosome Assay The methodology for measurement of thiobarbituric acid reactive substances (TBARS) from synaptosomes was adapted from that of Keller et al. Neuroscience 80: 685-696, 1997.
  • Adult Sprague-Dawley rats 250-30Og were anesthetized with halothane for 20 sec. and decapitated.
  • the brain was removed and the cerebral cortex separated from white matter.
  • the cerebral cortex was homogenized in a solution containing sucrose 0.32 M, EDTA 2mM and TRIS-HCl 10 mM, pH 7.2, using a Teflon pestle.
  • the tissue was 5% w/v in the homogenizing buffer.
  • the homogenate was centrifuged for 10 minutes at 310 g at 4°C. The supernatant was then centrifuged for 10 minutes at 20,000 g at 4°C. The pellet was collected, resuspended in Locke's buffer (154 mM NaCl, 5.6 mM KCl, 2.3 mM CaCl 2 -2H 2 O, 1.0 mM MgCl 2 -6H 2 O, 3.6 mM NaHCO 3 , 5 mM glucose, 5 mM HEPES, pH 7.2) and centrifuged for 10 minutes at 20,000 g at 4°C. The procedure of washing the pellet was repeated 2-3 times in order to reduce transition metal ion concentrations.
  • Locke's buffer 154 mM NaCl, 5.6 mM KCl, 2.3 mM CaCl 2 -2H 2 O, 1.0 mM MgCl 2 -6H 2 O, 3.6 mM NaHCO 3 , 5 mM glucose,
  • Lipid peroxidation synaptosome experimental results. Preliminary lipid peroxidation experiments explored the time course of synaptosome lipid peroxidation, incubating homogenate with FeSO 4 (lO ⁇ M - 150 ⁇ M) in Locke's buffer, for time intervals from 15 minutes to 180 minutes (data not shown). Under these experimental conditions, the level of peroxidation, as measured by TBARS, was seen to be below saturation at 30 minutes using 50 ⁇ M FeSO 4 . The ability of this system to provide concentration dependent lipid peroxidation data was demonstrated using the antioxidant ⁇ -tocopherol ( ⁇ TH), and ascorbic acid (AH), which is known to act as a prooxidant in Fe(II)-induced lipid peroxidation systems (see Figure 8). These conditions thus were chosen for all further synaptosome experiments.
  • ⁇ TH antioxidant ⁇ -tocopherol
  • AH ascorbic acid
  • the Fe/synaptosome/TBARS system was designed to provide concentration-response curves for inhibition of lipid peroxidation, which might be quantified by EC 50 values.
  • Absolute EC 50 values measured in such systems are highly dependent on experimental conditions, and therefore must be benchmarked against well-studied antioxidants, such as Trolox, a water soluble chroman carboxylate derivative of ⁇ TH (see Figure 9).
  • Thiols can display mixed pro- and antioxidant activity towards lipid peroxidation.
  • transition metals either added to lipid preparations, or adventitious metal ions present in tissue homogenates
  • thiols may act as prooxidants.
  • the vic-dithiol, dihydrolipoic acid (LAH 2 , ⁇ , solid line in Figure 10) yielded a concentration dependent prooxidant effect, akin to ascorbic acid, whereas the oxidized disulfide lipoic acid (LA, A , dashed line in Figure 10) showed very modest inhibition of lipid peroxidation at the highest concentration applied.
  • the antioxidant trolox yielded a potent concentration dependent reduction in TBARS products with EC 50 - 6.8 x 10 "5 M (Figure 9), which can be contrasted to the effect of lipoic acid (LA) or dihydrolipoic acid (LAH 2 , see Figure 10).
  • LA lipoic acid
  • LAH 2 dihydrolipoic acid
  • Figure 11 the concentration of GTN have any significant effect in the presence of added LAH 2 (ImM)
  • Figure 11 did not significant effect on lipid peroxidation with any thiol used (e.g. cysteine, PhSH), over the effect of the thiol itself (see GTN with LAH 2 , ⁇ , solid line in Figure 11).
  • compound Va did inhibit lipid peroxidation at higher concentrations in the presence Of LAH 2 .
  • compound Va inhibited TBARS formation with the water soluble thiol, cysteine, and the more lipophilic thiophenol (PhSH) (see Va + cysteine, ⁇ , solid line or Va + PhSH, A, solid line in Figure 12).
  • PhSH water soluble thiol, cysteine, and the more lipophilic thiophenol
  • compound Va showed some prooxidant activity in the presence of cysteine.
  • Concentration response curves were derived from TBARS data for the NO donor NONOate, Sper/NO (spermine NONOate, dashed line), and for DE A/NO (diethylamine NONOate, solid line) (see Figure 14).
  • Gaetani P., A. Pasqualin, R. Rodriguez y Baena, E. Borasio, F. Marzatico, "Oxidative stress in the human brain after subarachnoid hemorrhage", J. Neurosurg. 89 (1998) 748-754.
  • Tagami M, K. Yamagata, K. Ikeda, Y. Nara, H. Fujino, A. Kubota, F. Numano, Y. Yamori, "Vitamin E prevents apoptosis in cortical neurons during hypoxia and oxygen reperfusion", Lab. Invest. 78 (1998) 1415-1429.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Nitrate esters and methods for mitigating neurodegeneration, affecting neuroprotection, affecting cognition enhancement, and/or preventing or mitigating tissue and/or cellular damage in a subject are described. Neurological or cognitive conditions, or damage mediated by free radicals are treated by administering to a subject an effective amount of a therapeutic compound comprising a nitrate ester, or a pharmaceutically acceptable salt thereof.

Description

NITRATE ESTERS AND THEIR USE FOR MITIGATING CELLULAR DAMAGE
BACKGROUND OF THE INVENTION
This invention relates to nitrate esters and use thereof in mitigating cellular damage. Particularly, this invention relates to selected organic nitrates, most particularly those bearing a sulfur or phosphorus atom β or γ to the nitrate group, having therapeutic utility as agents that protect tissues from oxidative injury.
The nitrate ester glyceryl trinitrate (GTN) or nitroglycerin, has been used as a vasodilator in the treatment of angina pectoris for over a hundred years, and the dominant contemporary belief is that GTN exerts its therapeutic effect through in vivo release of nitric oxide (NO). Other organic nitrates, such as isosorbide dinitrate, have also been identified as effective and clinically important vasodilators. NO itself has been identified as Endothelium Derived Relaxing Factor (EDRF) and several classes of compounds, for example nitrosothiols, in addition to organic nitrates, have been proposed as NO donors or NO prodrugs. Endogenous stimulation or exogenous administration of NO have been shown to inhibit production of reactive oxygen species (ROS) and expression of oxidant-mediated molecular or tissue injury. Well-known examples of these classes of compounds and one nitrate, GTN itself, have been suggested to demonstrate neurotoxic or neuroprotective effects by dint of interactions with the redox modulatory site of the N-methyl-D-aspartate (ΝMDA) excitatory amino acid receptor. Thus GTΝ is firstly a potent vasodilator and secondly possesses potential neuroprotective properties. In addition, GTΝ has been found to suppress renal oxidant damage caused by potassium bromate (Rahman et al., Redox Rep. 4: 263-9, 1999). Several attempts have been made to increase the efficacy or potency of alternative organic nitrates as vasodilators relative to GTΝ, for example, by incorporation of propanolamine or cysteine functionalities. However, no attempt has been made to separately regulate the vasodilatory and cytoprotective effects of GTΝ. Indeed, postural hypotension and weakness are signs of cerebral ischemia, and are adverse effects associated with the vasodilatory effects of GTΝ. Observed in treatment, these effects are highly contraindicative of GTΝ itself, and by extrapolation GTΝ derivatives (1,2,3-trinitratopropane derivatives), as clinically useful protective therapeutic agents.
SUMMARY OF THE INVENTION
In as much as the potent vasodilatory effects of organic nitrates may prove (a) deleterious to, or alternatively (b) synergistic with the protective effects of GTN, it is postulated herein that regulation of these two effects is required for development of new and useful protective therapeutic agents. Further, it is postulated that such regulation may be achieved through use of an appropriate organic nitrate, such as, for example, nitrate esters incorporating sulfur-containing or phosphorus-containing functionalities into the structure of the nitrate esters. Accordingly, a first aspect the invention features nitrate esters having the
formula: , or a pharmaceutically acceptable salt thereof, wherein: each of m and n is, independently, an integer from O to 10; each of R , R , R , 17 is, independently, hydrogen, a nitrate group, or A;
R1 is hydrogen or A; where A is selected from: a substituted or unsubstituted aliphatic group having from 1 to 24 carbon atoms in the chain, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted cyclic aliphatic moiety having from 3 to 7 carbon atoms in the aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted aliphatic moiety comprising a linkage from 0 to 5 carbon atoms between R1 and R3 and/or between R17 and R4, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; a substituted or unsubstituted aliphatic group having from 1 to 24 carbon atoms in the chain, containing linkages selected from C=O, C=S, and C=NOH, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; a substituted or unsubstituted aryl group; a heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group; each of R2, R5, R18 is, independently, hydrogen, A, or X-Y; where X is F, Br, Cl, NO2 CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7, or SSR5;
Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16)(OM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7, or SSR5, or does not exist; each of R6, R7, R8, R9, R11, R12, R13, R14, R15, R16 is, independently, an alkyl or acyl group containing 1-24 carbon atoms, which may contain 1-4 ONO2 substituents; a C1-C6 connection to R'-R4 in a cyclic derivative; a hydrogen, a nitrate group, or A; and
M is H, Na+, K+, NH4 +, or N+H14R1 \4.k) where k is O to 3, or other pharmaceutically acceptable counterion; with the proviso that: when m = O; n = 1 ; each of R18 and R3 is, independently, H, a nitrate group, or a C1-C4 alkyl chain, which may include one O linking R18 and R3 to form a pentosyl, hexosyl, cyclopentyl, or cyclohexyl ring, which ring optionally bears a hydroxyl substituent; each of R17 and R4 is, independently H, a nitrate group, a Ci -C4 alkyl, optionally bearing 1 to 3 nitrate groups, or an acyl group (-C(O)R5); each of R5, R6, R8, R9, R12, R13, R14, R15, R16 is, independently, an alkyl group containing 1 to 12 carbon atoms, which may contain 1 to 4 ONO2 substituents; or a C1 or C2 connection to R18, R17, or R3 in a cyclic derivative; each of R7 and R11 is, independently, a Ci to C8 alkyl or acyl group;
M is H, Na+, K+, NH4 +, or N+HkR1 '{4-k) where k is 0 to 3; and
X is CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), or SSR4; then Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15)(OR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
In accordance with the present invention, one skilled in the recognizes that one of m, n, or p must be equal to at least one.
In one embodiment, the compound of the invention is:
wherein G1 is Me, OMe, Cl, NO2, Br, or H; G2 is CO2Et,
CO2H, CO2Me, CONH2, or CO(CH2)2NEt2; and G3 is Cl, OMe, or CONH2
In another embodiment, the compound is:
In another embodiment, the compound is:
wherein n is 2 or 3.
In another embodiment, the compound is:
In another embodiment, the compound is:
;or In yet another embodiment, the compound is:
;
ONO2 [iiiam]; .
In a second aspect, the invention features a compound having the formula:
, or a pharmaceutically acceptable salt thereof, containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein each of m and n is, independently, an integer from O to 10;
R1 is a hydrogen or A; each of R »2 , n R5 , and R , 18 is, independently, hydrogen or A; each of R3, R4, and R17, is independently, a hydrogen, a nitrate group, or
A; each of R6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, A, a hydrogen, a nitrate group, or a C1-C24 alkyl or acyl group, optionally containing 1-4 ONO2 substituents or a CpC6 linkage to R1, R2, R3, or R4 in cyclic derivatives; each of R7 and R11 is, independently, a substituted or unsubstituted C1- Cg alkyl or acyl group;
A is selected from: a CpC24 alkyl group, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups, or 1-2 nitrate groups; a C3-C24 alkyl group, containing 1-5 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the carbon chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups; a C3-C7 linkage to any of R1, R2, R3, R4, or R17, forming an aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 6 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CrC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a C0-C5 linkage to or between any of R1, R3, R4, or R17, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, bearing two or more substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted Cr C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; an unsubstituted Co-C5 linkage to or between any of R1, R3, and R4, which optionally contains 1 to 2 non-adjacent O, S, NR6, and/or unsaturations in the linkage; a C1-C5 linkage to or between any of R1, R3, R4, and R17 containing 1 to 2 C-O, C=S, or C=NOR7 linkages, which optionally contains O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted Q -C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heteroaryl group; a substituted or unsubstituted heterocyclic group; an amino, cyclic amino, diamino, triamino, alkylamino, dialkylamino, arylamino, diarylamino, or alkylarylamino group; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7, or does not exist;
Y is F, Br, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, NHOH, N2H3, N2H2R13, N2HR13R14, N3, SC(=NH)N(RI5)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR7, SO2M, S(O)R8, S(O)R5, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)(OR13), C(O)(SR13), C(S)R5, C(S)R12, C(S)OR12, or does not exist; and
M is H, Na+, K+, NH4 +, N+HkR1V-10 where k is 0"3> or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l ; each of R18 and R3 is, independently, a hydrogen, a nitrate group, or a C1-C4 alkyl chain, which may include one O, linking R18 and R3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R17 and R4 is, independently, a hydrogen, a nitrate group, a C1- C4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (- C(O)R5); each of R5, R6, R8, R9, R12, R13, R14, R15, and R16 is, independently, a C1- C12 alkyl group, optionally bearing from 1-4 ONO2 substituents, or a C1-C2 alkyl linkage to R18, R17, or R3;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(0)(0R16)(0M), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
In one embodiment of the second aspect, the compound is:
In another embodiment, the compound is:
In another embodiment, the compound is:
In another embodiment the compound is:
In another embodiment, the compound is:
or In another embodiment, the compound is:
In yet another embodiment of the second aspect, the compound is:
In a third aspect, the invention features a compound having the formula:
In a fourth aspect, the invention features a composition that includes a pharmaceutically acceptable carrier and a compound having the general formula:
or a pharmaceutically acceptable salt thereof, containing 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein each of m and n is, independently, an integer from O to 10; R1 is a hydrogen or A; each of R | 2 , r R> 5 , and R , 18 is, independently, hydrogen or A; each of R » 3 , π R4 , and R , 17 , is independently, a hydrogen, a nitrate group, or
A; each of R6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, A, a hydrogen, a nitrate group, or a C1-C24 alkyl or acyl group, optionally containing 1-4 ONO2 substituents or a C1-C6 linkage to R1, R2, R3, or R4 in cyclic derivatives; each of R7 and R11 is, independently, a substituted or unsubstituted Q- C8 alkyl or acyl group;
A is selected from: a C1-C24 alkyl group, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups, or 1-2 nitrate groups; a C3-C24 alkyl group, containing 1-5 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the carbon chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups; a C3-C7 linkage to any of R1, R2, R3, R4, or R17, forming an aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 6 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CpC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a C0-C5 linkage to or between any of R1, R3, R4, or R17, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, bearing two or more substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1- C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; an unsubstituted C0-C5 linkage to or between any of R1, R3, and R4, which optionally contains 1 to 2 non-adjacent O, S, NR6, and/or unsaturations in the linkage; a CpC5 linkage to or between any of R1, R3, R4, and R17 containing 1 to 2 C=O, C=S, or C=NOR7 linkages, which optionally contains O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted Ci -C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heteroaryl group; a substituted or unsubstituted heterocyclic group; an amino, cyclic amino, diamino, triamino, alkylamino, dialkylamino, arylamino, diarylamino, or alkylarylamino group; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCC=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7, or does not exist;
Y is F, Br, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, NHOH, N2H3, N2H2R13, N2HR13R14, N3, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR7, SO2M, S(O)R8, S(O)R5, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)(OR13), C(O)(SR13), C(S)R5, C(S)R12, C(S)OR12, or does not exist; and
M is H, Na+, K+, NH4 +, N+HkR1 \4.k) where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l ; each of R18 and R3 is, independently, a hydrogen, a nitrate group, or a C1-C4 alkyl chain, which may include one O, linking R18 and R3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R17 and R4 is, independently, a hydrogen, a nitrate group, a Q- C4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (- C(O)R5); each of R5, R6, R8 , R9, R12, R13, R14, R15, and R16 is, independently, a C1- C12 alkyl group, optionally bearing from 1-4 ONO2 substituents, or a Q-C2 alkyl linkage to R18, R17, or R3;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN,SC(=NH)N(R15)2, SQ=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
In a fifth aspect, the invention features a composition that includes a pharmaceutically acceptable carrier and one of the following compounds:
In a sixth aspect, the invention features a method for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject by administering to the subject an effective amount of a compound containing at least one aliphatic nitrate group and at least one sulfur atom in proximity to the nitrate group. By "sulfur atom in proximity" or "proximal functional group" is meant a sulfur atom or functional group that is connected through bonds in a β, γ, or δ relationship to a nitrate ester group (i.e., the atom connectivity is 1,2, or 1,3, or 1,4). The functional group may also be referred to as "proximally located" or "situated in proximity." Proximal functional groups also include those groups that have a through-space intramolecular juxtaposition with a nitrate group that is within 3 A.
In one embodiment, the tissue and/or cellular damage can be associated with aging, septic shock, ischemia/reperfusion injury, ulcers, gastritis, ulcerative colitis, Crohn's disease, diabetes, rheumatoid arthritis, asthma, cirrhosis of the liver, allograft rejection, encephalomyelitis, meningitis, pancreatitis, peritonitis, vasculitis, lymphocytic choriomeningitis, glomerulonephritis, uveitis, glaucoma, blepharitis, chalazion, allergic eye disease, corneal ulcer, keratitis, cataracts, age-related macular degeneration, optic neuritis, ileitis, hemorrhagic shock, anaphylactic shock, bacterial infection, viral infection, fungal infection, parasitic infection, hemodialysis, chronic fatigue syndrome, stroke, toxic shock syndrome, adult respiratory distress syndrome, cachexia, myocarditis, eczema, psoriasis, dermatitis, urticaria, cerebral ischemia, systemic lupus erythematosis, chronic neurodegenerative disease, priapism, cystic fibrosis, schizophrenia, depression, premenstrual syndrome, anxiety, addiction, migraine, gastrointestinal motility disorders, obesity, hyperphagia, hematologic cancers, myelofibrosis, graft- versus-host disease, CNS trauma, hepatitis, renal failure, chronic hepatitis C, drug-induced lung injury (e.g., paraquat), bacterial translocation, circulatory shock, traumatic shock, vascular aneurysm, metastatic cancer, or myocardial infarction.
In another embodiment, the tissue and/or cellular damage can associated with neurological diseases such as, for example, Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amyotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia due to myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; or environmental excitotoxins of plant, animal, or marine origin.
In yet another embodiment, the tissue and/or cellular damage can be associated with cytokine therapy, wherein a nitrate ester of the invention is administered to the subject before, during, and/or after the administration of the therapeutic cytokine.
In another embodiment of the sixth aspect, the compound has the formula:
, containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein m is an integer from O to 10; n is an integer from 0 to 10; each of R3>4>17 is, independently, hydrogen, a nitrate group, or A;
R1 is hydrogen or A;
A is selected from: a substituted or unsubstituted C1-C24 alkyl group, optionally containing 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted cyclic moiety having from 3 to 7 carbon atoms in the ring, which optionally containing 1 to 2 O, S, NR6, and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted moiety constituting a linkage from 0 to 5 carbons, to or between any of R1, R3, R17 and R4, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the linkage, and optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted C1-C24 alkyl group, containing 1-4 linkages selected from C=O, C=S, and C=NOH, which optionally contains O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
R2, R5, R18, are optionally hydrogen, A, or X-Y;
X is F, Br, Cl, NO2, CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SCC=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)R5, S(O)2R5, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7 or SSR5, SS, or does not exist;
Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)R5, S(O)2R5, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R5,
C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7 or SSR5, C(S)R5, C(S)R12, C(S)OR12, or does not exist; each of R6, R7, R8, R9, R11, R12, R13, R14, R15, and R16 is, independently, a C1-C24 alkyl group, optionally containing 1-4 ONO2 substituents, a C1-C24 acyl group, optionally containing 1-4 ONO2 substituents, a CpC6 ring-forming connection to any of R!-R4, a hydrogen, a nitrate group, or A; and
M is H, Na+, K+, NH4 +, N+HkR1 \4.k) where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when nr=0; n=l; each of R18 and R3 is, individually, H, a nitrate group,or a C1 - C4 alkyl group, which may include one O, linking R18 and R3 to form pentosyl, hexosyl, cyclopentyl, or cyclohexyl rings, which optionally bears hydroxyl substituents; each of R17 and R4 is, individually, H, a nitrate group, a C1-C4 alkyl group, optionally bearing from 1-3 nitrate groups, or -C(O)R5; each of R5, R6, R8 , R9, R12, R13, R14, R15, R16 is, individually, a C1-C12 alkyl group, optionally containing 1-4 ONO2 substituents or a C1-C2 ring- forming connections to R18, R17, or R3; each of R7 and R11 is, independently, a C)-C8 alkyl group or a C1-C8 acyl group;
M is H, Na+, K+, NH4 +, N+HkR1 \4.k) where k is 0-3;
X is CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, or SSR4; and Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SCC=NH)N(R15)2, SCt=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H,
CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
In another embodiment, the compound contains at least 2 nitrate groups. In another embodiment, the nitrate is beta or gamma to said sulfur atom.
In another embodiment of the sixth aspect, the compound can be any of the compounds that were cited individually as an embodiment of either the first or second aspect of the invention.
In another embodiment, the compound is:
In yet another embodiment of the sixth aspect, the compound is:
As used herein, the terms "alkyl" and the prefix "alk-" are inclusive of both straight chain and branched chain saturated or unsaturated groups, and of cyclic groups, i.e., cycloalkyl and cycloalkenyl groups. Unless otherwise specified, acyclic alkyl groups are from 1 to 6 carbons and contain at least one C-H bond. The number of carbons in an alkyl group refers to the total number of carbons contained in the group. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 8 ring carbon atoms. Exemplary cyclic groups include cyclopropyl, cyclopentyl, cyclohexyl, and adamantyl groups. Unless otherwise indicated, alkyl groups may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halogen, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
By "aryl" is meant a carbocyclic aromatic ring or ring system. Unless otherwise specified, aryl groups are from 6 to 18 carbons. . Examples of aryl groups include phenyl, naphthyl, biphenyl, fluorenyl, and indenyl groups.
By "heterocycle" is meant an aromatic or non-aromatic ring system that contains at least one ring heteroatom (e.g., O, S, N). The term "heteroaryl" refers to an aromatic heterocyclic ring or ring system that contains at least one ring heteroatom (e.g., O, S, N). Unless otherwise specified, heteroaryl rings contain from 1 to 9 carbons. Exemplary heteroaryl groups include furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, oxatriazolyl, pyridyl, pyridazyl, pyrimidyl, pyrazyl, triazyl, benzofuranyl, isobenzofuranyl, benzothienyl, indole, indazolyl, indolizinyl, benzisoxazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, naphtyridinyl, phthalazinyl, phenanthrolinyl, purinyl, and carbazolyl rings or ring systems. Unless otherwise specified, non-aromatic heterocyclic groups are from 2 to 9 carbons and can include, for example, dihydropyrrolyl, tetrahydropyrrolyl, piperazinyl, pyranyl, dihydropyranyl, tetrahydropyranyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothiophene, tetrahydrothiophene, and morpholinyl groups. As defined in the present application, the definition of heterocycle specifically excludes β-lactam rings.
Unless otherwise specified, aryl, heteroaryl, or heterocyclic groups may be unsubstituted or substituted by one or more substituents selected from the group consisting Of Ci-6 alkyl, hydroxy, halo, nitro, C1-6 alkoxy, Ci-6 alkylthio, trifluoromethyl, C1-6 acyl, arylcarbonyl, heteroarylcarbonyl, nitrile, C1-6 alkoxycarbonyl, arylalkyl (wherein the alkyl group has from 1 to 6 carbon atoms) and heteroarylalkyl (wherein the alkyl group has from 1 to 6 carbon atoms).
By "halide" or "halogen" or "halo" is meant bromine, chlorine, iodine, or fluorine. As used herein, the terms "alkyl" and the prefix "alk-" are inclusive of both straight chain and branched chain saturated or unsaturated groups, and of cyclic groups, i.e., cycloalkyl and cycloalkenyl groups. Unless otherwise specified, acyclic alkyl groups are from 1 to 6 carbons. Cyclic groups can be monocyclic or polycyclic and preferably have from 3 to 8 ring carbon atoms. Exemplary cyclic groups include cyclopropyl, cyclopentyl, cyclohexyl, and adamantyl groups. Alkyl groups may be substituted or unsubstituted. Exemplary substituents include alkoxy, aryloxy, sulfhydryl, alkylthio, arylthio, halogen, hydroxyl, fluoroalkyl, perfluoralkyl, amino, aminoalkyl, disubstituted amino, quaternary amino, hydroxyalkyl, carboxyalkyl, and carboxyl groups.
By "alkoxy" is meant a chemical substituent of the formula -OR, where R is an alkyl group. By "aryloxy" is meant a chemical substituent of the formula -OR', where R' is an aryl group. By "alkaryl" is meant a chemical substituent of the formula -RR', where R is an alkyl group and R' is an aryl group, By "alkheteraryl" is meant a chemical substituent of the formula RR", where R is an alkyl group and R" is a heteroaryl group.
What is meant by "aliphatic nitrate" is a nitrate group that is connected to a non-aromatic carbon.
What is meant by "antioxidant" is a compound that attenuates or prevents oxidation of a target biomolecule through the inhibition of the initiation or propagation steps that constitute oxidative and peroxidative processes.
"Mitigating neurodegeneration" as used herein involves affecting neuroprotection, inhibiting or preventing neurodegeneration, and/or ameliorating the manifestations or impact of neurodegeneration. Such amelioration includes effecting cognition enhancement, as is quantified by tests known in the art (e.g., Venault et al., 1992, incorporated herein by reference). "Modulating" a biological process as used herein (for example, modulating activity of the non-glutamate neuroreceptors), encompasses both increasing (positively modulating) and decreasing (negatively modulating) such activity, and thus inhibition, potentiation, agonism, and antagonism of the biological process.
It is an object of the present invention to provide provide novel organic nitrates, including aliphatic nitrate esters bearing a sulfur or phosphorus moiety β or γ to a nitrate group, or congeners thereof. Another object of the present invention is to provide methods for making such novel organic nitrates. Another object of the invention is to provide methods for affecting neuroprotection, mitigating neurodegeneration, affecting cognition enhancement, and/or protecting tissues from oxidative injury employing selected organic nitrates. Another object of the present invention is to provide novel drugs as neuroprotective agents. Another another object of the present invention is to provide novel drugs for use in cognition enhancement. Another object of the invention is to provide novel drugs for use in protecting tissues from oxidative injury.
It will be understood by those skilled in the art, that when organic radicals, such as Rn, X, or Y, are represented as "does not exist," that the valency of the carbon bound to the radical is adjusted accordingly (i.e., by a point of unsaturation).
Compounds may be constructed according to formulas of the invention in which an S atom is appropriately proximally placed with respect to a nitrate functional group, but in which a carbonyl group intervenes to form a thioester linkage. Such compounds do not form part of the invention if cleavage of this thioester bond produces two entirely separate molecules, one containing the nitrate funtionality, and another the S-functionality. An example of such a compound is one containing the motif, O2NOCH2C(CH3)2C(O)S-(organic radical), which would liberate on thioester cleavage an aliphatic nitrate that does not contain a S-functionality. It is understood by one skilled in the art that facile cleavage of a thioester in an aqueous biological milieu will render an aliphatic nitrate that does not contain a proximal S, and therefore, such a thioester does not comprise part of the current invention.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a synthetic scheme showing the use of Bunte salt IVd in the preparation of compounds Va, IVr, Vbb, and Vbc.
Figure 2 is a graph showing a comparison of 50 μM GTN (stippled bars), 10 μM Va (cross-hatched bars), 30 μM Va (upward hatched bars) and 50 μM Va (downward hatched bars) on lactate dehydrogenase (LDH) release from isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min. Hearts were perfused with drugs for ten min prior to and throughout the period of LCAO. Data are the mean ± standard errors (n= 3-6).
Figure 3 is a graph showing a comparison of 50 μM GTN (stippled bars), 50 μM Va (hatched bars) and 50 μM compound IIIam (cross-hatched bars) on lactate dehydrogenase (LDH) release from isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min. Hearts were perfused with drugs for ten min prior to and throughout the period of reperfusion. Data are the mean ± standard errors (n= 4-6).
Figure 4 is a graph showing a comparison of 50 μM GTN (open bar) and 50 μM Va (hatched bar) on infarct size in isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min. Hearts were perfused with drugs for ten min prior to and throughout the period of LCAO. Data are the mean ± standard errors (n= 5-6).
Figure 5 is a graph showing a comparison of 50 μM GTN (open bar), 50 μM Va (hatched bar) and 50 μM compound IIIam (cross-hatched bar) on infarct size in isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min. Hearts were perfused with drugs for ten min prior to and throughout the period of reperfusion. Data are the mean ± standard errors (n = 3-6).
Figure 6 is a graph showing a comparison of 50 μM GTN (stippled bars) and 50 μM Va (hatched bars) on coronary perfusion pressure in isolated perfused rat heart after left coronary artery occlusion (LCAO) for 45 min followed by reperfusion for 90 min. Hearts were perfused with drugs for ten min prior to and throughout the period of LCAO. Coronary perfusion pressure was recorded just prior to LCAO (baseline), at the beginning of reperfusion (0 min reperfusion) and at the end of the 90 min reperfusion period (90 min reperfusion). Data are the mean ± standard errors (n = 6).
Figure 7 is an immunocytochemical analysis of tyrosine hydroxylase immunoreactivity in the substantia nigra of rats. Figure 7 A is an analysis of a brain section in which the animal was administered the neurotoxic compound 6- hydroxydopamine (6-OHDA). Figure 7B is an analysis of a brain section in which the animal was administered only vehicle control (dimethyl sulfoxide). Figure 7C is an analysis of a brain section in which the animal was treated with compound Va before and after administration of 6-OHDA. Figure 7D is an analysis of a brain section in which the animal was administered only vehicle control.
Figure 8 is a graph showing the inhibition and potentiation of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS (thiobarbituric acid reactive substances) determination, on incubation of rat brain synaptosomes with ascorbic acid or α-tocopherol (n=3). Percentage TBARS detected is given relative to TBARS in presence of FeSO4 (100%). "Control" experiment is in the presence of vehicle and absence Of FeSO4. Error bars show S.E.M.
Figure 9 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with Trolox (n=3). Percentage inhibition is given relative to: control TBARS in presence of vehicle and absence Of FeSO4 (100%); TBARS in presence of vehicle and FeSO4 (0%). Error bars show S.E.M; data is fitted to a sigmoidal curve (EC50 = 6.8 x 10"5 M). Figure 10 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with lipoic acid (LA, dashed line) or dihydrolipoic acid (LAH2, solid line) (n=3). Percentage inhibition relative to: control TBARS in presence of vehicle and absence of FeSO4 (100%); TBARS in presence of vehicle and FeSO4 (0%). Error bars show S.E.M.
Figure 11 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with: GTN (varied) + LAH2 (ImM) (■, solid line); Va (T, dashed line); or Va (varied) + LAH2 (ImM) (A, dashed line), (n=3). Percentage inhibition relative to: control TBARS in presence of vehicle and absence Of FeSO4 (100%); TBARS in presence of vehicle, adjuvants and FeSO4 (0%). In experiments with adjuvant LAH2, 0% inhibition corresponds to TBARS in presence of vehicle containing LAH2. Error bars show S.E.M.
Figure 12 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with: Va (varied) + cysteine (ImM) (■, solid line); or Va + PhSH (1 mM) (A, solid), (n=3). For comparison, inhibition curve with NONOate, DET A/NO (in the absence of adjuvants; •, dashed line) is shown. Percentage inhibition relative to controls :TBARS in presence of vehicle and absence of FeSO4 (100%); TBARS in presence of vehicle, adjuvant and FeSO4 (0%). In both experiments with adjuvant thiols, 0% inhibition corresponds to TBARS in presence of vehicle containing thiol (ImM). Error bars show S.E.M.
Figure 13 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with: IVr (varied) (■); or Va + PhSH (1 mM) (A), (n=3). Percentage inhibition relative to: control TBARS in presence of vehicle and absence Of FeSO4 (100%); TBARS in presence of vehicle, adjuvant, and FeSO4 (0%). In experiments with thiol, 0% inhibition corresponds to TBARS in presence of vehicle containing adjuvant PhSH. Error bars show S.E.M.
Figure 14 is a graph showing the effects of NO-donors. Inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with: (a) DEA/NO (diethylamine NONOate, ■, solid line); and Sper/NO (spermine NONOate, D, dashed lines), (n=3). Percentage inhibition relative to: control TBARS in presence of vehicle, adjuvant, and absence of FeSO4 (100%); TBARS in presence of vehicle and FeSO4 (0%). Error bars show S.E.M. Data is fitted to sigmoidal curves (EC50: DEA/NO = 7 x 10"5 M; Sper/NO = 2 x 10"4 M).
Figure 15 is a graph showing the inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with z'-amyl nitrite (IAN, ■ solid line) (n-3). Percentage inhibition relative to: control TBARS in presence of vehicle and absence of FeSO4 (100%); TBARS in presence Of FeSO4 (0%). Data is fitted to a sigmoidal curve: EC50 = 1.6 x 10"4 M.
Figure 16 is a graph showing the comparison of inhibition by nitrate IVs in: (1) FeSO4 (50μM) induced lipid peroxidation in rat brain synaptosomes assessed by TBARS determination (O dashed line); and, (2) ABAP induced lipid peroxidation in liposomes assessed by BODIPY peroxidation (• solid line) (n=3). Percentage inhibition relative to: control response in presence of vehicle and absence of initiator (100%); response in presence of vehicle and initiator (0%). Data is fitted to a sigmoidal curve: EC50 (synaptosomes) = 1.0 x 10'3 M, (liposomes) = 1.3 x 10"4 M.
Figure 17 is a graph showing the comparison of inhibition of lipid peroxidation induced by FeSO4 (50μM) as assessed by TBARS determination, on incubation of rat brain synaptosomes with nitrate Vbb (T solid line) and nitrate Vbc (■ solid line), compared to Trolox (□ dashed line) (n=3). Percentage inhibition relative to: control TBARS in presence of vehicle and absence Of FeSO4 (100%); TBARS in presence Of FeSO4 (0%). Data is fitted to a sigmoidal curve: EC50 Vbb = 2.0 x 10"5 M; EC50 Vbc = 7 x 10"7 M and 7 x 10" 5 M).
DETAILED DESCRIPTION OF THE INVENTION This invention pertains to methods and compositions useful for treating neurodegeneration or preventing or mitigating tissue and/or cellular damage by administering to a subject a therapeutic nitrate ester. Neuroprotection and/or cognition enhancement can be affected, for example, by modulating an interaction with guanylyl cyclase (GCase, the enzyme responsible for cGMP production in various areas of the brain), modulating a glutamate or non- glutamate neuroreceptor or attenuating free radical damage. The attenuation of free radical concentration by a nitrate ester of the invention can also be useful for preventing or mitigating tissue and/or cellular damage.
According to certain aspects of the invention, neurodegeneration is mitigated by stimulating cerebral GCase. One of the major targets for the novel organic nitrates of the invention is GCase activation, resulting in the production of cGMP. Experimental evidence obtained in a number of in vitro model systems supports the notion that elevated levels of cGMP help to prevent apoptotic (programmed) cell death. Thus, a cGMP-dependent mechanism significantly increases the survival of trophic factor-deprived PC 12 cells and rat sympathetic neurons (Farinelli et al., 1996), and of primary cultures of rat embryonic motor neurons (Estevez et al., 1998). The mechanism of action for selected organic nitrates in preventing apoptotic cell death may be inhibition of caspase-3 activation indirectly through elevations in cGMP levels or directly via protein S-nitrosylation of the enzyme by an NO-intermediate (Kim et al., 1997). Caspase-3 is a member of the cysteine protease family of enzymes that are essential for the execution step in apoptosis (Cohen, 1997; Nicholson and Thornberry, 1997). Activation of caspase-3 is required for apoptotic cell death in trophic factor-deprived PC 12 cells (Haviv et al., 1997) and in glutamate- mediated apoptotic cell death of cultured cerebellar granule neurons (Du et al., 1997). In animal models of cerebral ischemia, caspase-3 activity is induced and may be responsible for the apoptotic component of delayed neuronal cell death (Chen et al., 1998; Namura et al., 1998; Ni et al., 1998). Inhibitors of caspase-3 significantly decrease the apoptotic component of delayed neuronal cell death in response to ischemic injury both in vitro (Gottron et al., 1997) and in vivo (Endres et al., 1998). A secreted region of the Alzheimer's disease β-amyloid precursor protein lowers intracellular calcium levels and provides neuroprotective effects on target cells through increases in cGMP levels and activation of protein kinase G (Barger et al., 1995; Furukawa et al., 1996). In preferred embodiments of the methods of the invention, nitrated molecules that have the capacity to activate GCase directly or via release of an NO-containing intermediate are used to modulate GCase activity.
According to certain other aspects of the invention, cognition enhancement (e.g., improved memory performance) is achieved by stimulating cerebral GCase. Several lines of experimental evidence support the notion that GCase and cGMP are involved in the formation and retention of new information. cGMP has been directly implicated in both long-term potentiation (LTP) and long-term depression (LTD), which are proposed cellular models for learning and memory (Arancio et al., 1995; Wu et al., 1998). In animal models, elevation of hippocampal cGMP levels leading to increased protein kinase G activity has been shown to be important for retention and consolidation of new learning (Bernabeu et al., 1996, 1997). Thus, stimulation of cerebral GCase activity is expected to improve learning and memory performance in individuals in whom cognitive abilities are impaired by injury, disease, or aging.
We have shown that novel organic nitrate esters have differential effects to activate soluble GCase and to cause cGMP accumulation in vascular and brain tissue. There is a clear dissociation between the vascular relaxation effects of organic nitrate esters and ability to affect neuroprotection. Activation of GCase and accumulation of cGMP have been shown to be important in the neuroprotection of hippocampal brain slices subjected to a period of in vitro ischemia.
Cerebral ischemia results in marked increases in the release of the excitatory amino acid glutamate in the affected brain region (Bullock et al., 1998; Huang et al., 1998; Yang et al., 1998). In both humans (Bullock et al., 1998) and experimental animals (Huang et al., 1998; Goda et al., 1998; Yang et al., 1998), the amount of glutamate released during ischemia is positively correlated with the extent of brain injury. In experimental animal models of cerebral ischemia, decreased release of glutamate during ischemia (Goda et al., 1998) or blockade of glutamate receptors with antagonists (Ibarrola et al., 1998; O'Neill et al., 1998; Umemura et al., 1997) significantly reduces the extent of brain injury. However, these interventions are only effective when given prior to or during the ischemic insult. To be broadly useful, a therapeutic intervention is preferably effective when administered after the period of ischemia. We have designed a class of novel organic nitrate esters having high efficacy in effecting neuroprotection in vivo in models of transient global and focal cerebral ischemia when given after the ischemic insult. It will be appreciated, therefore, that these selected organic nitrates can be used for treatment of conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntingdon's disease; multiple sclerosis; amyotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant, animal, or marine origin; and the like.
The direct effects of selected organic nitrates on amino acid neurotransmitter receptors has been tested using the Xenopus oocyte expression system and two-electrode voltage-clamp recording methods. Selected organic nitrates were found to have direct, modulatory effects on GABAA receptor function (see Working Examples below). These allosteric modulatory effects of selected organic nitrates were not shared by direct NO-generating compounds, indicating a novel mechanism of action for selected organic nitrates to interact with GABAA receptors. In behavioural models of learning and memory, drugs which decrease GABAA receptor function improve performance on learning and memory tasks (Venaμlt et al., 1992). Thus, the behavioural effect of selected organic nitrates, developed to act as modulators of GABAA receptor function, will be to improve memory performance and cognition in patient populations. It will be appreciated, therefore, that these selected organic nitrates can be used for treatment of conditions including but not limited to: stroke; dementias of all type; trauma; drug-induced brain damage; and aging.
According to certain aspects of the invention, neurodegeneration is mitigated by inhibition of free radical damage. Reoxygenation and reperfusion after a period of ischemia contributes significantly to the development of brain injury. Oxygen radicals, especially superoxide and peroxynitrite, formed in the period after an ischemic event may initiate processes such as breakdown of membrane lipids (lipid peroxidation), leading to loss of cell membrane integrity and inhibition of mitochondrial function (Macdonald and Stoodley, 1998; Gaetani et al, 1998). Oxidative stress is also believed to be one factor involved in initiation of apoptotic neuronal cell death (Tagami et al., 1998). In experimental animal models of ischemic brain injury, free radical scavengers and enhanced activity of superoxide dismutase have been found to reduce the extent of neuronal injury and cell death (Chan et al., 1998; Mizuno et al., 1998; Tagami et al., 1998).
It has been shown that 2,3-dinitrooxy-(2,3-bis-nitrooxy-propyldisulfanyl)- propane, compound Va, improves task acquisition in cognitively impaired animals (see Smith et al., NeuroReport 11 : 3883, 2000), suggesting to us that novel nitrates administered to an intact animal can have direct effects on the brain. These observations also lead us to postulate that the nitrates of the present invention could protect tissues against oxidative injury. Accordingly, we tested the effects of compound Va in the 6-hydroxydopamine (6-OHDA) model of Parkinson's disease. 6-OHDA is a neurotoxin selectively taken up into dopaminergic neurons, resulting in a selective killing of these neurons, via a mechanism involving oxidative stress that is evident by 4 days after injection of the toxin. Previous studies have demonstrated that the monoamine oxidase type B (MAO-B) inhibitor, deprenyl, can prevent 6-OHDA-induced killing of dopaminergic neurons. We therefore employed deprenyl as a positive control in this study. Loss of dopaminergic innervation to the striatum results in an upregulation of postsynaptic dopamine receptors, and the development of dopamine receptor supersensitivity 2-3 weeks after 6-OHDA lesioning of the substantia nigra. This supersensitivity to dopamine can be unmasked by challenging the animal with a dopamine receptor agonist, such as apomorphine. Since the substantia nigra pars compacta is a bilateral structure, unilateral destruction of the substantia nigra will induce a dopamine receptor imbalance, which can manifest behaviourally as apomorphine-induced turning in the direction contralateral to the lesion. This behavioural manifestation of 6- OHDA toxicity provides a convenient, quantifiable index of neuronal injury and neuroprotection. Immunocytochemical analysis confirmed that compound Va preserved TH-positive neurons in the substantia nigra of 6-OHD A- injected rats (see Example 3 and Figure 7). These data demonstrate that compound Va is a very effective neuroprotective agent against 6-OHD A-induced killing of dopaminergic neurons in the rat substantia nigra pars compacta.
Accordingly, in certain aspects and embodiments of the invention, the invention features nitrated molecules which have the capacity to inhibit production of free radicals and/or which act as free radical scavengers.
Free radical overproduction is associated with a wide range of disease states and/or indications, such as, for example, aging, septic shock, ischemia, overexpression of cytokines, ulcers, inflammatory bowel disease (e.g., gastritis, ulcerative colitis or Crohn's disease), diabetes, arthritis (e.g., rheumatoid arthritis), asthma, cirrhosis, allograft rejection (e.g., transplant rejection), encephalomyelitis, meningitis, pancreatitis, peritonitis, vasculitis, lymphocytic choriomeningitis, glomerulonephritis, ophthalmologic diseases (e.g., uveitis, glaucoma, blepharitis, chalazion, allergic eye disease, corneal ulcer, keratitis, cataract, retinal disorders, age-related macular degeneration, optic neuritis, and the like), ileitis, inflammation induced by overproduction of inflammatory cytokines (e.g., liver inflammation, renal inflammation, airway inflammation, and the like), hemorrhagic shock, anaphylactic shock, burn, infection leading to the overproduction of inflammatory cytokines (including bacterial (e.g., E. coli infection), viral (e.g., HIV), fungal (e.g., Candidiosis and histoplasmosis) and parasitic (e.g., Leishmaniasis and Schistosomiasis) infections), hemodialysis, chronic fatigue syndrome, stroke, cancers, including metastatic cancers (e.g., breast cancer, bladder cancer, lung cancer, colon cancer, or cancer of the other organs, or skin or other noncutaneous portions of the body), cardiovascular diseases associated with overproduction of inflammatory cytokines (e.g., heart disease, cardiopulmonary bypass, ischemic/reperfusion injury, and the like), ischemic/reperfusion associated with overproduction of inflammatory cytokines, toxic shock syndrome, adult respiratory distress syndrome, cachexia, myocarditis, autoimmune disorders, eczema, psoriasis, heart failure, dermatitis, urticaria, cerebral ischemia, systemic lupus erythematosis, AIDS, neurodegenerative disorders (e.g., chronic neurodegenerative disease), chronic pain, priapism, cystic fibrosis, schizophrenia, depression, premenstrual syndrome, anxiety, addiction, migraine, gastrointestinal motility disorders, obesity, hyperphagia, solid tumors (e.g., neuroblastoma), malaria, hematologic cancers, myelofibrosis, lung injury, graft-versus-host disease, head injury, CNS trauma, hepatitis, renal failure, liver disease (e.g., chronic hepatitis C), drug- induced lung injury (e.g., paraquat), transplant rejection and preservation, fertility enhancement, bacterial translocation, circulatory shock, traumatic shock, and vascular aneurysm (e.g., aortic aneurysm), ileus, or myocardial infarction.
In addition, the compounds or methods of the present invention may find use in cytokine therapy (with consequent induction of free radical overproduction) which, for example, is commonly used in the treatment of cancers, including metastatic cancers (e.g., breast cancer, bladder cancer, lung cancer, colon cancer, or cancer of the other organs, or skin or other noncutaneous portions of the body), autoimmune disease, and in AIDS patients. Systemic hypotension due to the induction of free radical overproduction is a dose-limiting side effect of cytokine therapy. Thus, a large patient population exists which will benefit from the invention methods.
It will also be appreciated by those skilled in the art, that any organic nitrate in which vasodilatory potency is reduced and neuroprotective potency increased, represents a new and useful therapeutic agent for use in neuroprotection, particularly in treatment of conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amylotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever- induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant, animal, or marine origin. GTN itself, proposed as a neuroprotective agent, has no clinical utility as a neuroprotective agent in therapy owing to its extraordinarily high vasodilatory potency. Similarly, by extrapolation, 1,2,3-trinitratopropane (GTN) derivatives are not expected to have clinical utility as neuroprotective agents in therapy owing to their especially high vasodilatory potency.
It will additionally be appreciated by those skilled in the art, that the use in therapy of any organic nitrate in cognition enhancment, represents a new and useful treatment for cognition enhancement, particularly in treatment of conditions including but not limited to: stroke; dementias of all type, trauma, drug-induced brain damage, and aging.
In particular, the therapeutic compounds of the invention comprise at least one nitrate group. The nitrate groups(s) can optionally be covalently bound to a carrier moiety or molecule (e.g., an aromatic group, an aliphatic group, peptide, steroid, nucleoside, peptidomimetic, steroidomimetic, or nucleoside analogue, or the like). In addition to functioning as a carrier for the nitrate functionality, the carrier moiety or molecule can enable the compound to traverse biological membranes and to be biodistributed preferentially, without excessive or premature metabolism. Further, in addition to functioning as a carrier for the nitrate functionality, the carrier moiety or molecule can enable the compound to exert amplified neuroprotective effects and/or cognition enhancement through synergism with the nitrate functionality.
In one aspect, the invention provides a method of treating a neurological condition and/or preventing an undesirable mental condition (e.g., memory loss) including the step of administering to a subject an effective amount of a therapeutic compound capable of mitigating neurodegeneration which has at least one nitrate group. In one embodiment, the therapeutic compound is capable of effecting neuroprotection. In another embodiment of the invention, the therapeutic compound is capable of effecting cognition enhancement. The therapeutic compound has the formula (Formula I):
wherein E, F, G are organic radicals which may contain inorganic counterions; so that a neurological condition is treated.
In another aspect, the invention provides a pharmaceutical composition including a physiologically acceptable carrier and a compound having the formula (Formula I):
wherein: E, F, G are organic radicals which may contain inorganic counterions. The composition is employed for mitigating neurodegeneration, effecting neuroprotection and /or effecting cognition enhancement. The composition may also be employed for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject.
In another aspect, therapeutic compounds of the invention that effect neuroprotection and/or effect cognition enhancement in a subject to which the therapeutic compound is administered have the formula (Formula II):
in which: m, n, p are integers from O to 10; R3'17 are each, independently, hydrogen, a nitrate group, or A; R1' 4 are each, independently, hydrogen or A, where A is selected from: a substituted or unsubstituted aliphatic group (preferably a branched, or straight-chain aliphatic moiety having from 1 to 24 carbon atoms in the chain, which optionally contains 1 to 4 O, S, NR6 ; and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino or aryl, or heterocyclic groups; an unsubstituted or substituted cyclic aliphatic moiety having from 3 to 7 carbon atoms in the aliphatic ring, which optionally contains 1 to 2 O, S, NR6 , and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, amino or aryl, or heterocyclic groups; an unsubstituted or substituted aliphatic moiety constituting a linkage from 0 to 5 carbons, between R1 and R3 and/or between R17 and R4, which optionally contains 1 to 2 O, S, NR6 > and/or unsaturations in the linkage, and optionally bearing from 1 to 4 hydroxy, nitrate, amino or aryl, or heterocyclic groups); a substituted or unsubstituted aliphatic group (preferably a branched, cyclic or straight-chain aliphatic moiety having from 1 to 24 carbon atoms in the chain), containing linkages selected from C=O, C=S, and C=NOH, which optionally contains 1 to 4 O, S, NR6 ; and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino or aryl, or heterocyclic groups; a substituted or unsubstituted aryl group; a heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group; R2, R5, R18, R19 are optionally hydrogen, A, or X-Y; where X is F, Br, Cl, NO2, CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SCC=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R", C(O), C(O)R12, C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7 or SSR5; Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7 or SSR5, or does not exist; R6, R7, R8 , R9, R11, R12, R13, R14, R15, R16 are the same or different alkyl or acyl groups containing 1-24 carbon atoms which may contain 1-4 ONO2 substituents; or C1 - C6 connections to R1 - R4 in cyclic derivatives; or are each independently hydrogen, a nitrate group, or A; M is H, Na+, K+, NH4 +, N+HkR11^-1C) where k is 0-3, or other pharmaceutically acceptable counterion.
Pharmaceutical compostions comprising a compound of Formula II in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention. The invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula II to a subject such that said mitigation and /or said neuroprotection an/or cognition enhancement occurs.
According to this aspect of the invention, preferred therapeutic compounds for effecting neuroprotection and/or cognition enhancement in a subject to which the compound is administered include compounds in which R19 is X-Y. In some preferred embodiments: R19 is X-Y and R5, R6, R8 , R9, R12, R13, R14, R15, R16 are the same or different alkyl groups containing 1-24 carbon atoms which may contain 1-4 ONO2 substituents, or C1 or C2 connections to R1 - R3 in cyclic derivatives; R1 and R3 are the same or different and selected from H, CrC4) alkyl chains, which may inlude one O, linking R1 and R3 to form pentosyl, hexosyl, cyclopentyl, or cycohexyl rings, which rings optionally bear hydroxyl substituents; R2 and R4, are the same or different and selected from H, a nitrate group, C1-C4 alkyl optionally bearing 1-3 nitrate group, and acyl groups (-C(O)R5); and R7, R1 ' are the same or different C1 - C8, alkyl or acyl.
In certain embodiments in which R19 is X-Y, m, p = 1, and n = O. In other embodiments in which R19 is X-Y, X is selected from: CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(^NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R11,
C(O), C(O)R12, C(O)(OR13), PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SSR4. In certain other embodiments in which R19 is X-Y, Y is selected from CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SR4, SO2M, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R11, C(O)R12, C(O)(SR13), SR5, SSR5, or does not exist. In some embodiments, X and/or Y contains a sulfur-containing functional group. In certain embodiments, the compound of the invention comprises a heterocyclic functionality, more preferably, a nucleoside or nucleobase. In other embodiments, the compound of the invention comprises a carbocyclic functionality, more preferably, a steroidal or carbohydrate moiety. In another aspect of the invention, a therapeutic compound of the invention is represented by the formula (Formula III):
in which: m, n are 1-10; R , 1- "9 , R 11-18 , X, and Y have the meaning as defined above. In certain preferred embodiments, R6 - R9 and R1 ' - R16 are the same or different alkyl or acyl groups containing 1-24 carbon atoms which may contain 1-4 ONO2 substituents, or C1 - C6 connections to R1 - R4 in cyclic derivatives. In certain preferred embodiments, R18 is A and m = n = 1.
Examples and preferred embodiments of this aspect include:
O9
(HIb); (me);
O2
[HIr];
[πis]. [πiu]. [Illag];
[Illai]; and [Illam]. In another aspect of the invention, a therapeutic compound of the invention is represented by the formula III in which the compound contains from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, where each of m and n is, independently, an integer from 0 to 10; R1 is a hydrogen or A; each of R2, R5, and R18 is, independently, hydrogen or A; each of R3, R4, and R17, is independently, a hydrogen, a nitrate group, or A; each of R6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, A, a hydrogen, a nitrate group, or a C1-C24 alkyl or acyl group, optionally containing 1-4 ONO2 substituents or a C1-C6 linkage to R1, R2, R3, or R4 in cyclic derivatives; each of R7 and R11 is, independently, a substituted or unsubstituted C1-Cg alkyl or acyl group;
A is a CpC24 alkyl group, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups, or 1-2 nitrate groups; a C3- C24 alkyl group, containing 1-5 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the carbon chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups; a C3-C7 linkage to any of R1, R2, R3, R4, or R17, forming an aliphatic ring, which optionally contains O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 6 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted CpC4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted CpC4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a C0-C5 linkage to or between any of R1, R3, R4, or R17, which optionally contains O, S, NR6, and/or unsaturations in the linkage, bearing two or more substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CpC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted CpC4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted CpC4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; an unsubstituted Co-C5 linkage to or between any of R1, R3, and R4, which optionally contains 1 to 2 non-adjacent O, S, NR6, and/or unsaturations in the linkage; a CpC5 linkage to or between any of R1, R3, R4, and R17 containing 1 to 2 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 substiruents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heteroaryl group; a substituted or unsubstituted heterocyclic group; an amino, cyclic amino, diamino, triamino, alkylamino, dialkylamino, arylamino, diarylamino, or alkylarylamino group; a hydroxy group; an alkoxy group; or a substituted or unsubstituted aryloxy group;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7, or does not exist;
Y is F, Br, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, NHOH, N2H3, N2H2R13, N2HR13R14, N3, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR7, SO2M, S(O)R8, S(O)R5, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)(OR13), C(O)(SR13), C(S)R5, C(S)R12, C(S)OR12, or does not exist; and M is H, Na+, K+, NH4 +, N+H1Jt1 V10 where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l, each of R18 and R3 is, independently, a hydrogen, a nitrate group, or a Cj-C4 alkyl chain, which may include one O, linking R18 and R3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R17 and R4 is, independently, a hydrogen, a nitrate group, a C1-C4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (-C(O)R5); each of R5, R6, R8 , R9, R10, R12, R13, R14, R15, and R16 is, independently, a C1-C12 alkyl group, optionally bearing from 1-4 ONO2 substituents, or a C1-C2 alkyl linkage to R18, R17, or R3; X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCC=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7; and Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
Examples and preferred embodiments of this aspect include:
.
3
IVt]; [Vz];
[IVr];
[Vam];
Pharmaceutical compostions comprising a compound of Formula III in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention. The invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula III to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs.
In another aspect of the invention, a therapeutic compound of the invention can be represented by the formula (Formula IV):
in which n = 0, X is CH2 or does not exist, and Y is selected from F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(^=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16)(OM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O)R12, C(O)(OR13), C(O)(SR13), SR5,
SSR7 or SSR5. R2, R4, R5, R6, R7, R8, R9, Rn, R12, Ri3, Rj4, Ri5, and Ri6 are as defined above. In certain preferred embodiments, R2 and R4 are optionally H, a nitrate group or a connection to R5-R9 and Ri i-Ri6in cyclic derivatives.
Pharmaceutical compositions comprising a compound of Formula IV in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
The invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula IV to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs.
Examples and preferred embodiments of compounds of the invention according to Formula IV are as follows:
a
(IVd);
(IVh); .
In yet another aspect of the invention, a compound of the invention can be represented by the formula (Formula V):
in which R2 is optionally H or a connection to R5 in cyclic derivatives, R4 is H or a nitrate group, and R5 is as described above.
Pharmaceutical compositions comprising a compound of Formula V in admixture with a pharmaceutically acceptable carrier therefor are provided by the invention.
The invention further provides methods of mitigating neurodegeneration, effecting neuroprotection and/or effecting cognition enhancement in a subject comprising the step of administering a compound of Formula V to a subject such that said mitigation and /or said neuroprotection and/or cognition enhancement occurs. Examples and preferred embodiments of compounds of the invention according to formula V (Formulae Va-c) are as follows:
(Va); O2N NO2 (Vb); or O2N H
(Vc).
In another aspect, the invention features one of the following nitrate esters:
[Iliad];
[Illah];
In another aspect, the invention features a method for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in the subject. The method includes administering to the subject an effective amount of a compound containing at least one aliphatic nitrate group and at least one sulfur atom in proximity to said nitrate, such as, for example, a compound of formula I. In one embodiment, the nitrate of this method contains at least 2 nitrate groups. In another embodiment, the nitrate is beta or gamma to a sulfur atom.
Yet another embodiment features compounds of formula III for preventing or mitigating tissue and/or cellular damage where m is an integer from O to 10; n is an integer from O to 10; each of R3>4>17 is, independently, hydrogen, a nitrate group, or A; R1 is hydrogen or A, with A is selected from: a substituted or unsubstituted C1-C24 alkyl group, optionally containing 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted cyclic moiety having from 3 to 7 carbon atoms in the ring, which optionally contains O, S, NR6, and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted moiety constituting a linkage from 0 to 5 carbons, to or between any of R1, R3, R17 and R4, which optionally contains O, S, NR6, and/or unsaturations in the linkage, and optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted C1-C24 alkyl group, containing 1-4 linkages selected from C=O, C=S, and C=NOH, which optionally contains O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino;a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
R2, R5, R18, are optionally hydrogen, A, or X-Y;
X is F, Br, Cl, NO2, CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SCC=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)R5, S(O)2R5, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7 or SSR5, SS, or does not exist;
Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)R5, S(O)2R5, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5,
C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7 or SSR5, C(S)R5, C(S)R12, C(S)OR12, or does not exist; each of R6, R7, R8, R9, R10, R1 ', R12, R13, R14, R15, and R16 is, independently, a C1-C24 alkyl group, optionally containing 1-4 ONO2 substituents, a CpC24 acyl group, optionally containing 1-4 ONO2 substituents, a CpC6 ring- forming connection to any of R'-R4, a hydrogen, a nitrate group, or A; and
M is H, Na+, K+, NH4 +, N+H1^R11^ where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l, each of R18 and R3 is, individually, H, a nitrate group,or a C1 - C4 alkyl group, which may include one O, linking R18 and R3 to form pentosyl, hexosyl, cyclopentyl, or cyclohexyl rings, which optionally bears hydroxyl substituents; each of R17 and R4 is, individually, H, a nitrate group, a C1-C4 alkyl group, optionally bearing from 1- 3 nitrate groups, or -C(O)R5; each of R5, R6, R8 , R9, R12, R13, R14, R15, R16 is, individually, a C1-Cj2 alkyl group, optionally containing 1-4 ONO2 substituents or a C1-C2 ring-forming connections to R18,R17, or R3; each of R7 and R1 ' is, independently, a C1-C8 alkyl group or a C1-C8 acyl group; M is H, Na+, K+, NH4 +, N+HkR1 ' (4-k) where k is 0-3 ;
X is CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15j2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M,
CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, or SSR4; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist. Preferred compounds include any of the compounds of formulas II, III, IV, and V that have been individually described herein. Other preferred compounds include:
-
Particularly preferred compounds for preventing or mitigating tissue and/or cellular damage are:
It will be noted that the structure of some of the compounds of this invention includes asymmetric carbon atoms. It is to be understood accordingly that the isomers (e.g., enantiomers, diastereomers) arising from such asymmetry are included within the scope of this invention. Such isomers can be obtained in substantially pure form by classical separation techniques and by asymmetric synthesis. For the purposes of this application, unless expressly noted to the contrary, a compound shall be construed to include both the R and S stereoisomers at each stereogenic center. In certain embodiments, a therapeutic compound of the invention comprises a cation (i.e., in certain embodiments, one of X or Y includes a cation, e.g., in the compound of formula IVd). If the cationic group is a proton, then the compound is considered an acid. If the proton is replaced by a metal ion or its equivalent, the compound is a salt. Pharmaceutically acceptable salts of the therapeutic compound are within the scope of the invention. For example, M can be a pharmaceutically acceptable alkali metal (e.g. Li, Na, K), ammonium, alkaline earth metal (e.g. Ca, Ba, Mg), higher valency cation, or polycationic counter ion (e.g., polyammonium cation) (see e.g., Berge et al., 1977). It will be appreciated that the stoichiometry of an anionic portion of the compound to a salt- forming cation will vary depending on the charge of the anionic portion of the compound and the charge of the counterion. Preferred pharmaceutically acceptable salts include a sodium, potassium, or calcium salt, but other salts are also contemplated within their pharmaceutically acceptable range.
A therapeutic compound of the invention.can be administered in a pharmaceutically acceptable vehicle. As used herein "pharmaceutically acceptable vehicle" includes any and all solvents, excipients, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like which are compatible with the activity of the compound and are physiologically acceptable to the subject. An example of a pharmaceutically acceptable vehicle is buffered normal saline (0.15 M NaCl). The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compound, use thereof in the compositions suitable for pharmaceutical administration is contemplated. Supplementary active compounds can also be incorporated into the compositions. Carrier or substituent moieties useful in the present invention may also include moieties which allow the therapeutic compound to be selectively delivered to a target organ. For example, delivery of the therapeutic compound to the brain may be enhanced by a carrier moiety using either active or passive transport (a "targeting moiety"). Illustratively, the carrier molecule may be a redox moiety, as described in, for example, U.S. Patents 4,540,654 and 5,389,623, both to Bodor. These patents disclose drugs linked to dihydropyridine moieties which can enter the brain, where they are oxidized to a charged pyridinium species which is trapped in the brain. Thus drugs accumulate in the brain. Other carrier moieties include compounds, such as amino acids or thyroxine, which can be passively or actively transported in vivo. Such a carrier moiety can be metabolically removed in vivo, or can remain intact as part of an active compound. Structural mimics of amino acids (and other actively transported moieties) including peptidomimetics, are also useful in the invention. As used herein, the term "peptidomimetic" is intended to include peptide analogues which serve as appropriate substitutes for peptides in interactions with, for example, receptors and enzymes. The peptodomimetic must possess not only affinity, but also efficacy and substrate function. That is, a peptidomimetic exhibits functions of a peptide, without restriction of structure to amino acid constituents. Peptidomimetics and methods for their preparation and use are described in Morgan et al. (1989), the contents of which are incorporated herein by reference. Many targeting moieties are known, and include, for example, asialoglycoproteins (see e.g., Wu, U.S. Patent 5,166,320) and other ligands which are transported into cells via receptor-mediated endocytosis (see below for further examples of targeting moieties which may be covalently or non-covalently bound to a target molecule).
In the methods of the invention, neurodegeneration in a subject is mitigated, and/or neuroprotection and/or cognition enhancement is effected, by administering a therapeutic compound of the invention to the subject. The invention also features methods for preventing or mitigating tissue and/or cellular damage in a subject by administering a therapeutic compound of the invention to the subject, thereby modulating intercellular and/or intracellular free radical concentration. The term "subject" is intended to include living organisms in which the particular neurological condition to be treated can occur. Examples of subjects include humans, apes, monkeys, cows, sheep, goats, dogs, cats, mice, rats, and transgenic species thereof. As would be apparent to a person of skill in the art, the animal subjects employed in the working examples set forth below are reasonable models for human subjects with respect to the tissues and biochemical pathways in question, and consequently the methods, therapeutic compounds and pharmaceutical compositions directed to same. As evidenced by Mordenti (1986) and similar articles, dosage forms for animals such as, for example, rats can be and are widely used directly to establish dosage levels in therapeutic applications in higher mammals, including humans.
In particular, the biochemical cascade initiated by cerebral ischemia is generally accepted to be identical in mammalian species (Mattson and Scheff, 1994; Higashi et al., 1995). In light of this, pharmacological agents that are neuroprotective in animal models such as those described herein are believed to be predictive of clinical efficacy in humans, after appropriate adjustment of dosage. Specifically, there are comparable memory-deficit patterns between brain-damaged rats and humans, which indicates that the rat can serve as an excellent animal model to evaluate the efficacy of pharmacological treatments or brain damage upon memory (Kesner, 1990). The only approved drug for the clinical treatment of occlusive stroke in humans is tissue plasminogen activator, which is administered at a dose of 0.9 mg/kg by intravenous injection (Wittkowsky, 1998). This drug is also effective in protecting the rat brain subjected to cerebral ischemia by occlusion of the middle cerebral artery, when administered at a dose of 10 mg/kg intravenously (Jiang et al., 1998). Thus, the rat model of focal cerebral ischemia used in the development of the novel organic nitrate esters described herein has been shown to be shown to be predictive of clinical efficacy with at least one other class of pharmacological agents.
As would also be apparent to a person skilled in the art, the invention further encompasses methods of the invention employed ex vivo or in vitro. For example, the Examples describe studies utilizing tissue homogenates according to the invention. Furthermore, diagnostic tests or studies of efficacy of selected compounds may conveniently be performed ex vivo or in vitro, including in animal models. Such tests, studies and assays are within the scope of the invention.
The pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A.R. Gennaro, Lippincott Williams & Wilkins, 2000, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York). Administration of the compositions of the present invention to a subject to be treated can be carried out using known procedures, at dosages and for periods of time effective to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in the subject. An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the amount of neurodegeneration that has already occurred at the clinical site in the subject, the age, sex, and weight of the subject, and the ability of the therapeutic compound to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in the subject. Dosage regimens can be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. A non-limiting example of an effective dose range for a therapeutic compound of the invention (e.g., Va) is between 0.5 and 500 mg/kg of body weight per day. In an aqueous composition, preferred concentrations for the active compound (i.e., the therapeutic compound that can mitigate neurodegeneration, effect neuroprotection, effect cognition enhancement, and/or prevent or mitigate tissue and/or cellular damage) are between 5 and 500 mM, more preferably between 10 and 100 mM, and still more preferably between 20 and 50 mM.
The therapeutic compounds of the invention can be effective when administered orally. Accordingly, a preferred route of administration is oral administration. Alternatively, the active compound may be administered by other suitable routes such as transdermal, subcutaneous, intraocular, intravenous, intramuscular or intraperitoneal administration, and the like (e.g., by injection). Depending on the route of administration, the active compound may be coated in a material to protect the compound from the action of acids, enzymes and other natural conditions which may inactivate the compound.
The compounds of the invention can be formulated to ensure proper distribution in vivo. For example, the blood-brain barrier (BBB) excludes many highly hydrophilic compounds. To ensure that the therapeutic compounds of the invention cross the BBB, they can be formulated, for example, in liposomes. For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811 ; 5,374,548; and 5,399,331. The liposomes may comprise one or more moieties which are selectively transported into specific cells or organs ("targeting moieties"), thus providing targeted drug delivery (see, e.g., Ranade et al., 1989). Exemplary targeting moieties include folate and biotin (see, e.g., U.S. Patent 5,416,016 to Low et al.); mannosides (Umezawa et al., 1988); antibodies (Bloeman et al., 1995; Owais et al., 1995); and surfactant protein A receptor (Briscoe et al., 1995). In a preferred embodiment, the therapeutic compounds of the invention are formulated in liposomes; in a more preferred embodiment, the liposomes include a targeting moiety.
Delivery and in vivo distribution can also be affected by alteration of an anionic group of compounds of the invention. For example, anionic groups such as phosphonate or carboxylate can be esterified to provide compounds with desirable pharmocokinetic, pharmacodynamic, biodistributive, or other properties. Exemplary compounds include IVl and pharmaceutically acceptable salts or esters thereof.
To administer the therapeutic compound by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the therapeutic compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water- in-oil-in- water CGF emulsions as well as conventional liposomes (Strejan et al., 1984).
The therapeutic compound may also be administered parenterally (e.g., intramuscularly, intravenously, intraperitoneally, intraspinally, or intracerebrally). Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In some cases, it will be preferable to include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
Sterile injectable solutions can be prepared by incorporating the therapeutic compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the therapeutic compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yield a powder of the active ingredient (i.e., the therapeutic compound) optionally plus any additional desired ingredient from a previously sterile- filtered solution thereof. The therapeutic compound can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The therapeutic compound and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The percentage of the therapeutic compound in the compositions and preparations may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of neurological conditions in subjects.
Therapeutic compositions can be administered in time-release or depot form, to obtain sustained release of the therapeutic compounds over time. The therapeutic compounds of the invention can also be administered transdermally (e.g., by providing the therapeutic compound, with a suitable carrier, in patch form). Active compounds are administered at a therapeutically effective dosage sufficient to mitigate neurodegeneration, to effect neuroprotection, to effect cognition enhancement, and/or to prevent or mitigate tissue and/or cellular damage in a subject. A "therapeutically effective dosage" preferably mitigates neurodegeneration by about 20%, more preferably by about 40%, even more preferably by about 60%, and still more preferably by about 80% relative to untreated subjects. The ability of a compound to mitigate neurodegeneration can be evaluated in model systems that may be predictive of efficacy in mitigating neurodegeneration in human diseases, such as animal model systems known in the art (including, e.g., the method of transient middle cerebral artery occlusion in the rat) or by in vitro methods, (including, e.g., the assays described herein).
It will be appreciated that the ability of a compound of the invention to mitigate neurodegeneration will, in certain embodiments, be evaluated by observation of one or more symptoms or signs associated with neurodegeneration in vivo. Thus, for example, the ability of a compound to mitigate neurodegeneration may be associated with an observable improvement in a clinical manifestation of the underlying neurodegeneration-related disease state or condition, or a slowing or delay in progression of symptoms of the condition. Thus, monitoring of clinical manifestations of disease can be useful in evaluating the neurodegeneration-mitigating efficacy of a compound of the invention.
The method of the invention is useful for treating neurodegeneration associated with any disease in which neurodegeneration occurs. Clinically, neurodegeneration can be associated with conditions including but not limited to: stroke; Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amylotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia; myocardial infarction; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; environmental excitotoxins of plant; animal, or marine origin; dementias of all type; trauma; drug-induced brain damage; and aging; or result from surgical procedures such as cardiac bypass.
Novel compounds according to the invention can be synthesized by methods set forth herein or in U.S. Patent Nos. 5,807,847; 5,883,122; 6,310,052; and 6,365,579, for example. Various compounds for use in the methods of the invention are commercially available and/or can be synthesized by standard techniques. In general, nitrate esters can be prepared from the corresponding alcohol, oxirane or alkene by standard methods, that include: nitration of alcohols and oxiranes, mixed aqueous/organic solvents using mixtures of nitric and sulfuric acid and/or their salts, with temperature control (see Yang et al., 1996); nitration of alcohols and oxiranes in acetic anhydride using nitric acid or its salts with or without added acid catalyst, with temperature control (see, e.g., Louw et al., 1976); nitration of an alcohol with a nitronium salt, e.g. a tetrafluoroborate; nitration of an alkene with thallium nitrate in an appropriate solvent (see Ouellette et al., 1976).
The following Examples further illustrate the present invention and are not intended to be limiting in any respect. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims. Example 1. Preparation of Nitrate Esters
Synthesis of Compound IVr
As shown in Figure 1 , the synthesis of compound IVr proceeded from the Bunte salt, 2,3-dinitrooxypropane-l-thiosulfonate (compound IVd), which was prepared from l,2-dinitrooxy-3-bromopropane as follows: 3- bromopropane-l,2-diol was added dropwise into a cold mixture Of HNO3 (68- 70%, 4.0eq) and H2SO4 (95%, 4.0 eq) in CH2Cl2 (50 mL) at room temperature over 30 min. The organic layer was separated, washed, dried and concentrated to yield a yellow oil which was purified by flash chromatography on SiO2 to give 3-bromopropane-l,2-diol dinitrate in 45% yield (29). The Bunte salt was prepared by reacting 3-bromopropane-l,2-diol dinitrate with an equimolar portion Of Na2S2O3 in 3: 1 MeOH/H2O at 50°C for 10 hours and subsequently purifying by flash chromatography on SiO2 (29). The Bunte salt was oxidized with a small molar excess Of H2O2 (30%) in EtOH:H2O mixture (1 :1) with a catalytic amount OfH2SO4 for 2 days. Extraction with CH2Cl2, concentration, and purification by flash chromatography on SiO2 yielded compound IVr as a yellow oil (Rf = 0.65; CH2Cl2:hexane = 65:35; 5%). 1H-NMR (CDCl3, 400 MHz): 5.55-5.65 (m, IH), 4.87-4.94 (dd, IH, J 12.94, 2.94), 4.62-4.70 (m, IH, J 12.88), 3.13-3.30 (m, 2H). 13C-NMR: (CDCl3, 100 MHz): 76.74, 69.46/69.42, 36.65/36.63. Mass spec, (m/z, EI+): 380.1 (M-NO2)+ 290%; 426.1 (M)+ 100%; 427.1 (M+l)+ 10%; 428.1 (M+2)+ 17%; 429.2 (M+3)+ 1.5%; 430.3 (M+4)+ 1.3%; calculated for C6H10N4O12S3 426.0. Elemental analysis: calculated for C6H10N4O14S2: C, 16.90; H, 2.36; S, 15.04; calculated for C6H10N4O12S3 : C, 16.90; H, 2.36; S, 22.56; found: C, 17.27; H, 2.38; S, 21.68.
Synthesis of Compound Va, Vbb, and Vbc
As shown in Figure 1, the syntheses of compounds Va, Vbb, and Vbc proceeded from the Bunte salt, compound IVd. A round-bottomed flask equipped with a dropping funnel, a thermometer, and a mechanical stirrer was charged with a solution of compound IVd and cooled to 0° with the aid of an ice-salt bath. A cold solution of the thiol precursor of compounds Va, Vbb, or Vbc was added rapidly, with vigorous stirring for 3 minutes, followed by the addition of aqueous saturated NaCl. The mixture was warmed to 50C and stirring stopped after 10 minutes. The crude disulfides Va, Vbb, or Vbc, were extracted 3x with diethyl ether. The extracts were combined, dried over calcium sulfate, and filtered through a glass-wool plug. Removal of the solvent leaves disulfide product, which can be further purified by silica gel chromatography.
Synthesis of Compounds Ve, Vf Vg, Vh, Vi, Vj, and Vu
Alkyl bromides or alkyl mercaptans were obtained commercially or by adaptation of literature procedures. Bunte salts were obtained from the appropriate alkyl bromide by reaction with sodium thiosulfate, as described above for compound IVd. Bunte salts (9.67 mmoles) were dissolved in distilled water (1OmL). To this solution, a solution of mercaptan (6.46 mmoles) in IM NaOH (7mL) was added dropwise. The resulting emulsion was stirred for 1 to 15 minutes and then extracted with dichlorometane or ethylacetate. The combined organic extracts were washed with H2O, dried over MgSO4 and concentrated under vacuum. The residue was purified by flash column chromatography on silica gel to give the following: Compound Vj (53% yield), 13C NMR (75Mhz, CDCl3) 36.9, 69.8, 77.6, 128.6, 129.5, 129.8, 136.0; Compound Vi (43% yield), 13C NMR (75Mhz, CDCl3) 36.9, 69.8, 77.4, 122.7, 130.9, 132.9, 135.1 ; Compound Vh (9% yield), 13C NMR (75Mhz, CDCl3) 36.9, 69.6, 77.6, 124.5, 126.9, 144.7, 147.0; Compound Vg (81% yield), 13C NMR (75Mhz, CDCl3) 36.8, 69.8, 77.5, 129.9, 130.8, 134.5, 134.8; Compound Vf (56% yield), 13C NMR (75Mhz, CDCl3) 36.5, 55.8, 69.7, 77.8, 115.4, 126.7, 133.7, 160.9; Compound Ve (52% yield), 13C NMR (75Mhz, CDCl3) 21.5, 36.8, 69.8, 77.7, 130.5, 130.6, 132.5, 139.2; and Compound Vuj (61% yield), 13C NMR (75Mhz, CDCl3) 36.7, 69.9, 77.6, 116.8-117.1(d), 131.3- 131.3(d), 132.5-132.6(d), 161.6-164.9(d).
Synthesis of Compound Vb l,4-dibromo-2,3-dinitrobutanediol (8.88 mmol) and Na2S2O3.5H2O (2.81 g ; 18 mmol) were dissolved in a mixture of 100 mL of MeOH and 45 mL of H2O. The resulting solution was heated during 4 days at 40-45°. After this time the reaction mixture was partially evaporated to reduce the volume of solvents. The resulting mixture was extracted with 4x50 mL of Et2O. The extracts were combined, washed (H2O), dried (MgSO4) and concentrated. The residue was purified by flash column chromatography on silica gel (hexane:EtOAc 85:15), yielding the title compound (10%) 13C (CDCl3): 83.9, 31.7.
Tables 1 and 2 list characterization data determined for other compounds of the invention.
Example 2
Characterization of Cardioprotection in Isolated, Perfused Heart
In order to test for potential cardioprotective properties, the effects of Va and compound IIIam were tested in an in vitro model of cardiac ischemia, in which isolated, perfused rat hearts were subjected to transient left coronary artery occlusion (LCAO) followed by reperfusion. Drug treatments [DMSO (drug vehicle), GTN, compound Va or compound IIIam were initiated at two distinct time points: (i) prior to and throughout the 45 minute period of LCAO (protection) or (ii) prior to and throughout the 90 minute reperfusion period (salvage). Drug-induced reduction of lactate dehydrogenase (LDH) release and reduction of infarct size were assessed as measures of cardioprotection. Rat hearts were excised and mounted for retrograde aortic perfusion at a constant flow rate of 6-8 mL/min/g heart weight. The coronary perfusion pressure was monitored by a pressure transducer connected to the perfusion line. To induce regional ischemia, the left coronary artery was occluded for 45 minutes, after which the occlusion was released and the heart reperfused for 90 minutes. At the end of the reperfusion period, the LCA was re-occluded and 0.5 mL of 1% Evan's Blue dye was slowly infused into the heart, via the aotic cannula, to stain the area of myocardium perfused by the patent right coronary artery. Thus the area-at-risk (AAR) for infarction was determined by negative staining. Acute ischemic damage was assessed by measuring the release of the cytosolic enzyme, LDH, into the perfusate, and by quantitation of infarct size by staining for viable tissue using 2,3,5-triphenyltetrazolium chloride (TTC) followed by computerized planimmetry. Infarct size was expressed as infarct area (negative staining after TTC staining) as a percent of the area-at-risk (negative staining after Evan's Blue dye). Left coronary artery occlusion was associated with a 5- 10 fold increase in LDH release (Figures 2 and 4, solid bars), whereas LDH release was not increased in non-occluded hearts (Figures 2 and 3, open bars). When administered prior to and during the period of LCA occlusion (protection protocol), compound Va, in a concentration-dependent manner, significantly reduced LDH release during the subsequent period of reperfusion (Figure 2, * P < 0.01 vs. LCAO + DMSO, one-way ANOVA, Tukey-Kramer post-hoc test), whereas GTN had no effect. Using the same drug infusion protocol, compound Va significantly reduced the size of the myocardial infarct after LCAO and reperfusion (Figure 4, * P < 0.01 vs DMSO-treated hearts, one-way ANOVA, Tukey-Kramer post-hoc test) whereas GTN had no effect. As a measure of the functional recovery of the heart after ischemia/reperfusion injury, the increase in perfusion pressure during the reperfusion period was monitored (Figure 6). In hearts subjected to LCAO, perfusion pressure was increased by about 2-fold by the end of the reperfusion period. This increase was markedly reduced in hearts treated with compound Va, whereas GTN had no effect. When GTN, compound Va and compound IIIam were infused prior to and throughout the reperfusion period (salvage protocol), all three drugs decreased LDH release during the reperfusion period (Figure 3, * P < 0.01 vs. LCAO + DMSO, one- way ANOVA, Tukey-Kramer post-hoc test). However, only compound Va and compound IIIam reduced infarct size when administered prior to and throughout the reperfusion period (Figure 5). These results indicate that compound Va reduces the severity of ischemia-reperfusion injury when administered prior to and throughout an acute ischemic insult, and that compounds Va and compound IIIam reduce the severity of ischemia- reperfusion injury when administered just prior to and throughout reperfusion, after a prolonged ischemic insult. However, the prototypical nitrate, GTN, is ineffective at protecting the heart from acute ischemic insult.
Example 3
Neuroprotection against 6-hydroxydopamine-induced killing of dopaminergic neurons in the rat substantia nigra pars compacta
Male Long-Evans rats were anesthetized with sodium pentobarbital, and received stereotaxic, unilateral injections of 6-OHDA (6 μg in 2 μl) into the right substantia nigra pars compacta. Vehicle (dimethylsulfoxide, DMSO) or compound Va were given by subcutaneous injection every hour for 6 hours, beginning 30 minutes before 6-OHDA. Each dose of compound Va was 200 μmol/kg. Two weeks after the administration of 6-OHDA or vehicle, the rats received a single injection of apomorphine (1 mg/kg, s.c), and contralateral rotations were counted at 15 minute intervals for 60 minutes. In some animals, the brains were fixed, and frozen sections cut for immunocytochemical analysis of tyrosine hydroxylase (TH).
In vehicle-treated animals, apomorphine induced rotations contralateral to the lesion that persisted for the entire 60 minute observation period. In contrast, animals that received compound Va exhibited essentially no rotations over 60 minutes in response to apomorphine injection, suggesting virtually complete protection against the neurotoxic effects of 6-OHDA (Table 3). Immunocytochemical analysis confirmed that compound Va preserved TH- positive neurons in the substantia nigra of 6-OHDA-injected rats (Figure 7). These data demonstrate that compound Va is a very effective neuroprotective agent against 6-OHDA-induced killing of dopaminergic neurons in the rat substantia nigra pars compacta.
*, p < 0.001 compared to vehicle control, One-way analysis of variance with Bonferroni's Multiple Comparison's test.
Synaptosome Assay The methodology for measurement of thiobarbituric acid reactive substances (TBARS) from synaptosomes was adapted from that of Keller et al. Neuroscience 80: 685-696, 1997. Adult Sprague-Dawley rats (250-30Og) were anesthetized with halothane for 20 sec. and decapitated. The brain was removed and the cerebral cortex separated from white matter. The cerebral cortex was homogenized in a solution containing sucrose 0.32 M, EDTA 2mM and TRIS-HCl 10 mM, pH 7.2, using a Teflon pestle. The tissue was 5% w/v in the homogenizing buffer. The homogenate was centrifuged for 10 minutes at 310 g at 4°C. The supernatant was then centrifuged for 10 minutes at 20,000 g at 4°C. The pellet was collected, resuspended in Locke's buffer (154 mM NaCl, 5.6 mM KCl, 2.3 mM CaCl2-2H2O, 1.0 mM MgCl2-6H2O, 3.6 mM NaHCO3, 5 mM glucose, 5 mM HEPES, pH 7.2) and centrifuged for 10 minutes at 20,000 g at 4°C. The procedure of washing the pellet was repeated 2-3 times in order to reduce transition metal ion concentrations. Finally, the pellet was resuspended in Locke's buffer for use in the lipid peroxidation assay. All assays were performed in triplicate and on three separate synaptosome preparations from different animals. Potential antioxidants and prooxidants were freshly prepared: in Locke's buffer [FeSO4; ascorbic acid (AA); Trolox; cysteine]; in 10 mM NaOH [NONOates]; or in organic solvent, such as methanol or DMSO, [α-tocopherol (αTH); nitrates; nitrites; PhSH; lipoic acid (LA); dihydrolipoic acid (LAH2)]; final dilution <2.5% (v/v) organic component). Pro/antioxidants (or solvent vehicle in control experiments) were added to the synaptosome preparation, followed immediately by freshly prepared aqueous FeSO4 (or buffer in control experiments) and incubated for 30 minutes at 37°C (air equilibrated). After incubation, TBA reagent (TBA 0.375% w/v, Cl3CCO2H 15% w/v, HCl IM 25% v/v) was added to the homogenate (homogenate: TBA reagent, 1 :2 v/v), and the sealed samples were boiled for 15 minutes at approx. 96-1000C. The cooled samples were then centrifuged for 10 minutes at 9,000 g at room temperature. The pink supernatant was transferred into microplates and the absorbance was measured at 530 nm on a Dynex MRX microplate reader. TBA reagent solutions were freshly made and calibrated using solutions of authentic malondialdehyde.
Lipid peroxidation: synaptosome experimental results. Preliminary lipid peroxidation experiments explored the time course of synaptosome lipid peroxidation, incubating homogenate with FeSO4 (lOμM - 150μM) in Locke's buffer, for time intervals from 15 minutes to 180 minutes (data not shown). Under these experimental conditions, the level of peroxidation, as measured by TBARS, was seen to be below saturation at 30 minutes using 50μM FeSO4. The ability of this system to provide concentration dependent lipid peroxidation data was demonstrated using the antioxidant α-tocopherol (αTH), and ascorbic acid (AH), which is known to act as a prooxidant in Fe(II)-induced lipid peroxidation systems (see Figure 8). These conditions thus were chosen for all further synaptosome experiments.
The Fe/synaptosome/TBARS system was designed to provide concentration-response curves for inhibition of lipid peroxidation, which might be quantified by EC50 values. Absolute EC50 values measured in such systems are highly dependent on experimental conditions, and therefore must be benchmarked against well-studied antioxidants, such as Trolox, a water soluble chroman carboxylate derivative of αTH (see Figure 9).
Thiols can display mixed pro- and antioxidant activity towards lipid peroxidation. In particular, in the presence of transition metals, either added to lipid preparations, or adventitious metal ions present in tissue homogenates, thiols may act as prooxidants. The vic-dithiol, dihydrolipoic acid (LAH2, ■, solid line in Figure 10) yielded a concentration dependent prooxidant effect, akin to ascorbic acid, whereas the oxidized disulfide lipoic acid (LA, A , dashed line in Figure 10) showed very modest inhibition of lipid peroxidation at the highest concentration applied. Cysteine (1 mM) was a prooxidant in the presence Of FeSO4 giving 117% of the lipid peroxidation seen in the presence Of FeSO4 alone, whereas PhSH (1 mM) in the presence Of FeSO4, gave 110% of the lipid peroxidation seen in the presence Of FeSO4 alone (data not shown). Of further consideration in analysis of lipid peroxidation data is the requirement for added thiols in experiments with nitrates. Clearly, any antioxidant effect of nitrates may be masked by the prooxidant effect of the adjuvant thiol. Thus, it was chosen to present the data as "percentage inhibition of lipid peroxidation", by normalizing TBARS measurements to: (i) 100% inhibition of lipid peroxidation (corresponding to TBARS in the presence of vehicle and absence of FeSO4); and, (ii) 0% inhibition of lipid peroxidation (corresponding to TBARS in the presence of vehicle, FeSO4, and any added thiol). This methodology and protocol was applied uniformly to all experiments graphed in Figures 9-17.
The antioxidant trolox yielded a potent concentration dependent reduction in TBARS products with EC50 - 6.8 x 10"5 M (Figure 9), which can be contrasted to the effect of lipoic acid (LA) or dihydrolipoic acid (LAH2, see Figure 10). GTN alone had no effect on Fe-induced lipid peroxidation (data not shown), nor did varied concentrations of GTN have any significant effect in the presence of added LAH2 (ImM) (Figure 11). Indeed, GTN produced no significant effect on lipid peroxidation with any thiol used (e.g. cysteine, PhSH), over the effect of the thiol itself (see GTN with LAH2, ■, solid line in Figure 11). However, compound Va (A, dashed line, Figure 11) did inhibit lipid peroxidation at higher concentrations in the presence Of LAH2. Further, in contrast to GTN, compound Va inhibited TBARS formation with the water soluble thiol, cysteine, and the more lipophilic thiophenol (PhSH) (see Va + cysteine, ■, solid line or Va + PhSH, A, solid line in Figure 12). Addition of PhSH (ImM) yielded a concentration dependent inhibition curve for lipid peroxidation: EC5O = 1.4 x 10~5 M. At high millimolar concentrations, compound Va showed some prooxidant activity in the presence of cysteine.
Data for inhibition of iron-induced lipid peroxidation by compound IVr can be fit to a curve leading to 100% efficacy with an EC50 of 1.2 x 10"4 M (or fitted without constraint on efficacy to EC50 = 2.7 x 10"4 M; efficacy = 78%). Moreover, compound Va, in the absence of thiol, showed a modest inhibition of lipid peroxidation, more pronounced at lower concentrations (see Figure 13). This antioxidant effect is clearly not an ubiquitous property of disulfides, since lipoic acid (LA) does not show such properties in the identical assay (Figure 10).
Concentration response curves were derived from TBARS data for the NO donor NONOate, Sper/NO (spermine NONOate, dashed line), and for DE A/NO (diethylamine NONOate, solid line) (see Figure 14). The potency and efficacy of inhibition of lipid peroxidation by Sper/NO and by IAN (see Figure 15) were both observed to be approximately identical (EC50 IAN = 1.6 x 10"4 M, Sper/NO = 2 x 10'4 M). TBARS measured for the compound IVs (dashed lines in Figure 16), in the absence of any adjuvants, revealed similar efficacy for this compound compared to Sper/NO and IAN, but a potency lower by an order of magnitud (EC50 (synaptosomes) = 1.0 x 10"3 M, (liposomes) = 1.3 x 10'4 M). The observed behavior of compounds IVs and IVr was similar, but measurements on compound IVr could not be extended to higher concentrations because of solubility. The potency and efficacy of inhibition of lipid peroxidation by compounds Vbb (T, solid line in Figure 17) and Vbc (■, solid line in Figure 17) were also measured and found to be 2.0 x 10~5 M for Vbb and and 7 x lO'7 M and 7 x 10"5 M for Vbc.
REFERENCES
Abe, K., C. Takeyama, K. Yoshimura, "Effects of S-8510, a novel benzodiazepine receptor partial inverse agonist, on basal forebrain lesioning- induced dysfunction in rats", Eur. J. Pharmacol. 347 (1998) 145-152.
Arancio, O., E.R. Kandel, R.D. Hawkins, "Activity-dependent long-term enhancement of transmitter release by presynaptic 3 ',5 '-cyclic GMP in cultured hippocampal neurons", Nature 376 (1995) 74-80.
Barger, S.W., R.R. Fiscus, P. Ruth, F. Hofmann, M.P. Mattson, "Role of cyclic GMP in the regulation of neuronal calcium and survival by secreted forms of β- amyloid precursor protein", J. Neurochem. 64 (1995) 2087-2096.
Berge et al., "Pharmaceutical Salts", J. Pharm. Sci. 66 (1977) 1-19.
Bernabeu, R., N. Schroder, J. Quevedo, M. Cammarota, I. Izquierdo, J.H. Medina, "Further evidence for the involvement of a hippocampal cGMP/cGMP-dependent protein kinase cascade in memory consolidation", NeuroReport 8 (1997) 2221-2224.
Bernabeu, R., P. Schmitz, M..P Faillace, I. Izquierdo, J.H. Medina, "Hippocampal cGMP and cAMP are differentially invloved in memory processing of inhibitory avoidance learning", NeuroReport 7 (1996) 585-588.
Bloeman, P.G. et al, FEBS Lett, 357 (1995) 140. Briscoe et al., Am. J Physiol. 1233 (1995) 134.
Bullock, R., A Zauner, JJ. Woodward, J. Myseros, S.C. Choi, J.D. Ward, A. Marmarou, H.F. Young, "Factors affecting excitatory amino acid release following severe human head injury", J. Neurosurg. 89 (1998) 507-518.
Chan, P.H., M. Kawase, K. Murakami, S.F. Chen, Y. Li, B. Calagui, L. Reola, E. Carlson, CJ. Epstein, "Overexpression of SODl in transgenic rats protects vulnerable neurons against ischemic damage after global cerebral ischemia and reperfusion", J. Neurosci. 18 (1998) 8292-8299.
Chen, J., T. Nagayama, K. Jin, R.A. Stetler, R.L. Zhu, S.H. Graham, R.P. Simon, "Induction of caspase-3-like protease may mediate delayed neuronal death in the hippocampus after transient cerebral ischemia", J. Neurosci. 18 (1998) 4914-4928.
Cohen, G.M., "Caspases: the executioners of aopotosis", Biochem. J. 326 (1997) 1-16.
Du, Y., K.R. Bales, R.C. Dodel, E. Hamilton-Byrd, J.W. Horn, D.L. Czilli, L.K. Simmons, B. Ni, S.M. Paul, "Activation of a caspase-3 -related cysteine protease is required for glutamate-mediated apoptosis of cultured cerebellar granule neurons", Proc. Natl. Acad. Sci. USA 94 (1997) 11657-11662.
Endres, M., S. Namura, M. Shimizu-Sasamata, C. Waeber, L. Zhang, T. Gomez-Isla, B.T. Hyman, M.A. Moskowitz, "Attenuation of delayed neuronal death after mild focal ischemia in mice by inhibition of the caspase family", J. Cereb. Blood Flow Metab. 18 (1998) 238-247.
Estevez, A.G., N. Spear, J.A. Thompson, T.L. Cornwell, R. Radi, L. Barbeito, J. S. Beckman, "Nitric oxide-dependent production of cGMP supports the survival of rat embryonic motor neurons cultured with brain-derived neurotrophic factor", J. Neurosci. 18 (1998) 3708-3714. Farinelli, S.E., D. S. Park, L.A. Greene, "Nitric oxide delays the death of trophic factor-deprived PC 12 cells and sympathetic neurons by a cGMP-mediated mechanism", J. Neurosci. 16 (1996) 23-25-2334.
Furukawa, K., S.W. Barger, E.M. Blalock, M.P. Mattson, "Activation of K+ channels and suppression of neuronal activity by secreted β-amyloid precursor protein", Nature 379 (1996) 74-78.
Gaetani, P., A. Pasqualin, R. Rodriguez y Baena, E. Borasio, F. Marzatico, "Oxidative stress in the human brain after subarachnoid hemorrhage", J. Neurosurg. 89 (1998) 748-754.
Goda, H., H. Ooboshi, H. Nakane, S. Ibayashi, S. Sadoshima, M. Fujishima, "Modulation of ischemia-evoked release of excitatory and inhibitory amino acids by adenosine Al receptor agonist", Eur. J. Pharmacol. 357 (1998) 149- 155.
Gottron, F.J., H.S. Ying, D.W. Choi, "Caspase inhibition selectively reduces the apoptotic component of oxygen-glucose deprivation-induced cortical neuronal cell death", MoI. Cell. Neurosci. 9 (1997) 159-169.
Haviv, R., L. Lindenboim, H. Li, J. Yuan, R. Stein, "Need for caspases in apoptosis of trophic factor-deprived PC 12 cells", J. Neurosci. Res. 50 (1997) 69-80.
Higashi et al., Neuropathol. Appl. Neurobiol. 21 (1995) 480-483.
Huang, F.P., L.F. Zhou, G. Y. Yang, "Effects of mild hypothermia on the release of regional glutamate and glycine during extended transient focal cerebral ischemia in rats", Neurochem. Res. 23 (1998) 991-996.
Ibarrola, D., H. Seegers, A. Jaillard, M. Hommel, M. Decorps, R. Massarelli, "The effect of eliprodil on the evolution of a focal cerebral ischaemia in v/vo", Eur. J. Pharmacol. 352 (1998) 29-35. Jiang et al., J. Cereb. Blood Flow Metab. 18 (1998) 758-767.
Kesner, NIDA Res. Monographs 97 (1990) 22-36.
Kim, Y.M., R.V. Talanian, T.R. Billiar, "Nitric oxide inhibits apoptosis by preventing increases in caspase-3-like activity via two distinct mechanisms", J. Biol. Chem. 272 (1997) 31138-31148.
Louw, R., H.P.W. Vermeeren, JJ.A. Van Asten, WJ. Ultee, J.Chem.Soc, Chem.Comm. (1976) 496-497
Macdonald, R.L., M. Stoodley, "Pathophysiology of cerebral ischemia", Neurol. Med. Chir. (Tokyo) 38 (1998) 1-11.
Mattson and Scheff, J. Neurotrauma 11(1994) 3-33.
Mizuno, A., K. Umemura, M. Nakashima, "Inhibitory effect of MCI- 186, a free radical scavenger, on cerebral ischemia following rat middle cerebral artery occlusion", Gen. Pharmacol. 30 (1998) 575-578.
Mordenti, "Man versus beast: Pharmacokinetic scaling in mammals", J. Pharm. Sci. 75 (1986) 1028-1040.
Morgan et al., "Approaches to the discovery of non-peptide ligands for peptide receptors and peptidases", In Ann. Rep. Med. Chem. (Virick FJ. , et al.) (1989) pp. 243-253, Academic Press, San Diego, CA.
Namura, S., J. Zhu, K. Fink, M. Endres, A Srinivasan, KJ. Tomaselli, J. Yuan, M. A. Moskowitz, "Activation and cleavage of caspase-3 in apoptosis induced by experimental cerebral ischemia", J. Neurosci. 18 (1998) 3659-3668.
Ni, B., X .Wu, Y. Su, D. Stephenson, E.B. Smalstig, J. Clemens, S.M. Paul, "Transient global forebrain ischemia induces a prolonged expression of the caspase-3 mRNA in rat hippocampal CAl pyramidal neurons", J. Cereb. Blood Flow Metab. 18 (1998) 248-256. Nicholson, D.W., N.A. Thornberry, "Caspases: killer proteases", Trends Biochem. Sci. 22 (1997) 299-306.
O'Neill, MJ, A. Bond, P.L. Ornstein, M.A. Ward, CA. Hicks, K. Hoo, D. Bleakman, D. Lodge, "Decahydroisoquinolines: novel competitive AMPA/kainate antagonists with neuroprotective effects in global cerebral ischaemia", Neuropharmacol. 37 (1998) 1211-1222.
Ouellette, RJ, RJ. Bertsch, J. Org. Chem. 41 (1976) 2782-2783.
Owais, M. et al, Antimicrob. Agents Chemother. 39 (1995) 180.
Pallares, M, M. Darnaudery, J. Day, M. Le Moal, W. Mayo, "The neurosteroid pregnenolone sulfate infused into the nucleus basalis increases both acetylcholine release in the frontal cortex or amygdala and spatial memory", Neurosci. 87 (1998) 551-558.
Ranade, V.V, J. Clin. Pharmacol. 29 (1989) 685.
Strejan et al, J. Neuroimmunol. 7 (1984) 27.
Tagami, M, K. Yamagata, K. Ikeda, Y. Nara, H. Fujino, A. Kubota, F. Numano, Y. Yamori, "Vitamin E prevents apoptosis in cortical neurons during hypoxia and oxygen reperfusion", Lab. Invest. 78 (1998) 1415-1429.
Umemura, K, A. Shimakura, M. Nakashima, "Neuroprotective effect of a novel AMPA receptor antagonist, YM90K, in a rat focal cerebral ischaemia", Brain Res. 773 (1997) 61-65.
Umezawa et al, Biochem. Biophys. Res. Commun. 153 (1988) 1038.
Venault, P.G, Chapouthier, L, Prado de Carvalho and Rossier, J, Encephale, 18 (1992) 655.
Wittkowsky, Pharmacotherapy 18 (1998) 945-1005. Wu, J., Y. Wang, M.J. Rowan, R. Anwyl, "Evidence for involvement of the cGMP -protein kinase G signaling system in the induction of long-term depression, but not long-term potentiation, in the dentate gyrus in vitro", J. Neurosci. 18 (1998) 3589-3596.
Yang, K., J.D. Ariz, J. Lock, C. Sanchez, B.M. Bennett, A.B. Fraser, G.R. Thatcher, J.Chem.Soc, Perkin Trans.1 (1996) 1073-1075.
Yang, Y.L., W.H. Pan, T.H. Chiu, M.T. Lin, "Striatal glutamate release is important for development of ischemic damage to strital neurons during rat heatstroke", Brain Res. 795 (1998) 121-127.
All publications and patents cited in this specification are hereby incorporated by reference herein as if each individual publication or patent were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

CLAIMS 1. A nitrate ester having the general formula:
or a pharmaceutically acceptable salt thereof, wherein: each of m and n is, independently, an integer from 0 to 10; each of R3, R4, R17 is, independently, hydrogen, a nitrate group, or A;
R1 is hydrogen or A; where A is selected from: a substituted or unsubstituted aliphatic group having from 1 to 24 carbon atoms in the chain, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted cyclic aliphatic moiety having from 3 to 7 carbon atoms in the aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; an unsubstituted or substituted aliphatic moiety comprising a linkage from 0 to 5 carbon atoms between R1 and R3 and/or between R17 and R4, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; a substituted or unsubstituted aliphatic group having from 1 to 24 carbon atoms in the chain, containing linkages selected from C=O, C=S, and C=NOH, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, amino, aryl, or heterocyclic groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group; each of R2, R5, R18 is, independently, hydrogen, A, or X-Y; where X is F, Br, Cl, NO2 CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SCC=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15XOR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7, or SSR5;
Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R1 1, C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7, or SSR5, or does not exist; each of R6, R7, R8, R9, R11, R12, R13, R14, R15, R16 is, independently, an alkyl or acyl group containing 1-24 carbon atoms, which may contain 1-4 ONO2 substituents; a CpC6 connection to R'-R4 in a cyclic derivative; a hydrogen, a nitrate group, or A; and
M is H, Na+, K+, NH4 +, or N+H14R1 '(4-k) where k is O to 3, or other pharmaceutically acceptable counterion; with the proviso that: when m = 0; n = 1; each of R18 and R3 is, independently, H, a nitrate group, or a C1-C4 alkyl chain, which may include one O linking R18 and R3 to form a pentosyl, hexosyl, cyclopentyl, or cyclohexyl ring, which ring optionally bears a hydroxyl substituent; each of R17 and R4 is, independently H, a nitrate group, a Ci -C4 alkyl, optionally bearing 1 to 3 nitrate groups, or an acyl group (-C(O)R5); each of R5, R6, R8, R9, R12, R13, R14, R15, R16 is, independently, an alkyl group containing 1 to 12 carbon atoms, which may contain 1 to 4 ONO2 substituents; or a C1 or C2 connection to R18, R17, or R3 in a cyclic derivative; each of R7 and R1 ' is, independently, a C1 to C8 alkyl or acyl group;
M is H, Na+, K+, NH4 +, or N+HkR1 '(4-k) where k is 0 to 3; and
X is CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), or SSR4; then Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2, SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(0)(0R16)(0M), P(O)(OM)R15, CO2M, CO2H, CO2R", C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
2. The compound of claim 1 selected from the following:
and G1 is Me, OMe, Cl, NO2, Br, or H; G2 is CO2Et, CO2H, CO2Me, CONH2, or CO(CH2)2NEt2, and G3 is Cl, OMe, or CONH2.
3. The compound of claim 1 selected from the following:
4. The compound of claim 1 selected from the following:
, wherein n is 2 or 3.
The compound of claim 1 selected from the following:
6. The compound of claim 1 selected from the following:
and
7. The compound of claim 1 selected from the following:
O2NO. % \ S i . O2N0 QNO2 ;
H
8. A nitrate ester having the general formula:
or a pharmaceutically acceptable salt thereof, containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein each of m and n is, independently, an integer from 0 to 10;
R1 is a hydrogen or A; each of R2, R5, and R18 is, independently, hydrogen or A; each of R3, R4, and R17, is independently, a hydrogen, a nitrate group, or A; each of R6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, A, a hydrogen, a nitrate group, or a C1-C24 alkyl or acyl group, optionally containing 1-4 ONO2 substituents or a C1-C6 linkage to R1, R2, R3, or R4 in cyclic derivatives; each of R7 and R11 is, independently, a substituted or unsubstituted C1- C8 alkyl or acyl group;
A is selected from: a CpC24 alkyl group, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups, or 1-2 nitrate groups; a C3-C24 alkyl group, containing 1-5 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the carbon chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups; a C3-C7 linkage to any of R1, R2, R3, R4, or R17, forming an aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 6 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CpC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a C0-C5 linkage to or between any of R1, R3, R4, or R17, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, bearing two or more substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted Cj-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; an unsubstituted Co-C5 linkage to or between any of R1, R3, and R4, which optionally contains 1 to 2 non-adjacent O, S, NR6, and/or unsaturations in the linkage; a C1-C5 linkage to or between any of R1, R3, R4, and R17 containing 1 to 2 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CpC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heteroaryl group; a substituted or unsubstituted heterocyclic group; an amino, cyclic amino, diamino, triamino, alkylamino, dialkylamino, arylamino, diarylamino, or alkylarylamino group; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group; X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7, or does not exist;
Y is F, Br, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, NHOH, N2H3, N2H2R13, N2HR13R14, N3, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR7, SO2M, S(O)R8, S(O)R5, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)(OR13), C(O)(SR13), C(S)R5, C(S)R12, C(S)OR12, or does not exist; and
M is H, Na+, K+, NH4 +, N+HkR1 Vk) where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l ; each of R18 and R3 is, independently, a hydrogen, a nitrate group, or a C1-C4 alkyl chain, which may include one O, linking R18 and R3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substiruents; each of R17 and R4 is, independently, a hydrogen, a nitrate group, a Cr C4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (-C(O)R5); each of R5, R6, R8 , R9, R12, R13, R14, R15, and R16 is, independently, a C1- C]2 alkyl group, optionally bearing from 1-4 ONO2 substiruents, or a Ci-C2 alkyl linkage to R18, R17, or R3;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15)(OR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
9. The compound of claim 8 selected from the following:
10. The compound of claim 8 selected from the following:
11. The compound of claim 8 selected from the following:
12. The compound of claim 8 selected from the following:
13. The compound of claim 8 selected from the following:
Ill
14. The compound of claim 8 selected from the following:
15. The compound of claim 8 selected from the following:
16. A nitrate ester selected from the following:
17. A composition comprising a pharmaceutically acceptable carrier and a compound having the general formula:
Y
or a pharmaceutically acceptable salt thereof, containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein each of m and n is, independently, an integer from O to 10;
R1 is a hydrogen or A; each of R , R , and R is, independently, hydrogen or A; each of R3, R4, and R17, is independently, a hydrogen, a nitrate group, or A; each of R6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, A, a hydrogen, a nitrate group, or a CpC24 alkyl or acyl group, optionally containing 1-4 ONO2 substituents or a CpC6 linkage to R1, R2, R3, or R4 in cyclic derivatives; each of R7 and R11 is, independently, a substituted or unsubstituted C1- Cg alkyl or acyl group;
A is selected from: a C1-C24 alkyl group, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups, or 1-2 nitrate groups; a C3-C24 alkyl group, containing 1-5 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 4 O, S, NR6, and/or unsaturations in the carbon chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino, unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or unsubstituted or substituted heterocyclic groups; a C3-C7 linkage to any of R1, R2, R3, R4, or R17, forming an aliphatic ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 6 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted C1-C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted CpC4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a C0-C5 linkage to or between any of R1, R3, R4, or R17, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, bearing two or more substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted Ci -C4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted C1-C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted Ci-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; an unsubstituted C0-C5 linkage to or between any of R1, R3, and R4, which optionally contains 1 to 2 non-adjacent O, S, NR6, and/or unsaturations in the linkage; a CpC5 linkage to or between any of R1, R3, R4, and R17 containing 1 to 2 C=O, C=S, or C=NOR7 linkages, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, optionally bearing from 1 to 4 substituents, independently selected from unsubstituted or substituted alkyl, unsubstituted or substituted aryl, unsubstituted or substituted CpC4 alkaryl, unsubstituted or substituted heteroaryl, unsubstituted or substituted Ci -C4 alkheteroaryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C1-C4 alkheterocyclic, hydroxy, nitrate, Cl, F, and amino groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heteroaryl group; a substituted or unsubstituted heterocyclic group; an amino, cyclic amino, diamino, triamino, alkylamino, dialkylamino, arylamino, diarylamino, or alkylarylamino group; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16)(OM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7, or does not exist; Y is F, Br, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, NHOH, N2H3, N2H2R13, N2HR13R14, N3, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR7, SO2M, S(O)R8, S(O)R5, PO2HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)(OR13), C(O)(SR13), C(S)R5, C(S)R12, C(S)OR12, or does not exist; and
M is H, Na+, K+, NH4 +, N+HkR1 \4.k) where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0 and n=l ; each of R18 and R3 is, independently, a hydrogen, a nitrate group, or a Ci-C4 alkyl chain, which may include one O, linking R18 and R3 together to form a pentosyl, a hexosyl, a cyclopentyl, or a cyclohexyl ring, said ring optionally bearing from 1-4 hydroxyl substituents; each of R17 and R4 is, independently, a hydrogen, a nitrate group, a Cp C4 alkyl group, optionally bearing from 1-3 nitrate groups, or an acyl group (-C(O)R5); each of R5, R6, R8 , R9, R12, R13, R14, R15, and R16 is, independently, a C1- C12 alkyl group, optionally bearing from 1-4 ONO2 substituents, or a Ci -C2 alkyl linkage to R18, R17, or R3;
X is F, Br, NO2, CH2 CF2, O, NH, NMe, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SH, SR5, SR7, S(O)R8, S(O)R5, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)(OR13), PO2H, PO2M, P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SR7; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR1J), SR4, SR5, or SSR5, or Y does not exist.
18. A composition comprising a pharmaceutically acceptable carrier and a compound selected from:
19. A method for preventing or mitigating tissue and/or cellular damage in a subject by modulating intercellular and/or intracellular free radical concentration in said subject, said method comprising administering to said subject an effective amount of a compound containing at least one aliphatic nitrate group and at least one sulfur atom in proximity to said nitrate.
20. The method of claim 19, wherein said compound has the formula:
containing from 1 to 3 nitrate groups and an S atom in proximity to a nitrate group, wherein m is an integer from O to 10; n is an integer from 0 to 10; each of R3'4'17 is, independently, hydrogen, a nitrate group, or A;
R1 is hydrogen or A;
A is selected from: a substituted or unsubstituted C1-C24 alkyl group, optionally containing 1 to 4 O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted cyclic moiety having from 3 to 7 carbon atoms in the ring, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the ring, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; an unsubstituted or substituted moiety constituting a linkage from 0 to 5 carbons, to or between any of R1, R3, R17 and R4, which optionally contains 1 to 2 O, S, NR6, and/or unsaturations in the linkage, and optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted Ci-C24 alkyl group, containing 1-4 linkages selected from C=O, C=S, and C=NOH, which optionally contains O, S, NR6, and/or unsaturations in the chain, optionally bearing from 1 to 4 hydroxy, nitrate, Cl, F, amino or unsubstituted or substituted aryl, or unsubstituted or substituted heterocyclic groups; a substituted or unsubstituted aryl group; a substituted or unsubstituted heterocyclic group; an amino group selected from alkylamino, dialkylamino, cyclic amino, cyclic diamino, cyclic triamino, arylamino, diarylamino, and alkyarylamino; a hydroxy group; an alkoxy group; and a substituted or unsubstituted aryloxy group;
R2, R5, R18, are optionally hydrogen, A, or X-Y;
X is F, Br, Cl, NO2, CH2, CF2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)R5, S(O)2R5, S(O)OR8, S(O)2OR9, PO2HM, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2H, PO2M,
P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, SSR7 or SSR5, SS, or does not exist;
Y is F, Br, Cl, CH3, CF2H, CF3, OH, NH2, NHR6, NR6R7, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)R5, S(O)2R5, S(O)2OR9, PO2HM, PO3M2, P(O)(OR15)(OR16), P(O)(OR16XOM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(OR13), C(O)(SR13), SR5, SSR7 or
SSR5, C(S)R5, C(S)R12, C(S)OR12, or does not exist; each ofR6, R7, R8 , R9, R11, R12, R13, R14, R15, and R16 is, independently, a CrC24 alkyl group, optionally containing 1-4 ONO2 substituents, a C] -C24 acyl group, optionally containing 1-4 ONO2 substituents, a C1-C6 ring- forming connection to any of R'-R4, a hydrogen, a nitrate group, or A; and
M is H, Na+, K+, NH4 +, N+HkR1V10 where k is 0-3, or other pharmaceutically acceptable counterion; and with the proviso that, when m=0; n=l; each of R18 and R3 is, individually, H, a nitrate group,or a Ci - C4 alkyl group, which may include one O, linking R and R to form pentosyl, hexosyl, cyclopentyl, or cyclohexyl rings, which optionally bears hydroxyl substituents; each of R17 and R4 is, individually, H, a nitrate group, a C1-C4 alkyl group, optionally bearing from 1-3 nitrate groups, or -C(O)R5; each of R5, R6, R8 , R9, R12, R13, R14, R15, R16 is, individually, a C1-C12 alkyl group, optionally containing 1-4 ONO2 substituents or a CpC2 ring- forming connections to R18,R17, or R3; each of R7 and R11 is, independently, a C1-C8 alkyl group or a Ci-C8 acyl group;
M is H, Na+, K+, NH4 +, N+HkR1V10 where k is °"3;
X is CH2, O, NH, NMe, CN, NHOH, N2H3, N2H2R13, N2HR13R14, N3, S, SCN, SC(=NH)N(R15)2, SC(=NH)NHR15, SC(O)N(R15)2 , SC(O)NHR15, SO3M, SH, SR7, SO2M, S(O)R8, S(O)2R9, S(O)OR8, S(O)2OR9, PO3HM, PO3M2, P(O)(OR15XOR16), P(O)(OR16)(OM), P(O)(R15)(OR8), P(O)(OM)R15, CO2M, CO2H, CO2R11, C(O), C(O)R12, C(O)(OR13), PO2M,
P(O)(OR14), P(O)(R13), SO, SO2, C(O)(SR13), SR5, or SSR4; and
Y is not CN, N2H2R13, N2HR13R14, N3, SCN, SC(^NH)N(R15)2, SC(=NH)NHR15, SC(O)NHR15, SO3M, SH, SO2M, PO3M2, PO3HM, P(O)(OR15XOR16), P(O)(OR16XOM), P(O)(OM)R15, CO2M, CO2H, CO2R5, C(O)R12, C(O)(SR13), SR4, SR5, or SSR5, or Y does not exist.
21. The method of claim 19, wherein said nitrate contains at least 2 nitrate groups.
22. The method of claim 19, wherein said nitrate is beta or gamma to said sulfur atom.
23. The method of claim 19, wherein said compound is selected from:
wherein G1 is Me, OMe, Cl, NO2, Br, or H; G2 is CO2Et, CO2H, CO2Me, CONH2, or CO(CH2)2NEt2, and G3 is Cl, OMe, or CONH2;
24. The method of claim 19, wherein said compound is selected from:
5. The method of claim 19, wherein said compound is selected from:
26. The method of claim 19, wherein said compound is selected from:
r
27. The method of claim 19, wherein said compound is:
28. The method of claim 19, wherein said tissue and/or cellular damage is associated with aging, septic shock, ischemia/reperfusion injury, ulcers, gastritis, ulcerative colitis, Crohn's disease, diabetes, rheumatoid arthritis, asthma, cirrhosis of the liver, allograft rejection, encephalomyelitis, meningitis, pancreatitis, peritonitis, vasculitis, lymphocytic choriomeningitis, glomerulonephritis, uveitis, glaucoma, blepharitis, chalazion, allergic eye disease, corneal ulcer, keratitis, cataracts, age-related macular degeneration, optic neuritis, ileitis, hemorrhagic shock, anaphylactic shock, bacterial infection, viral infection, fungal infection, parasitic infection, hemodialysis, chronic fatigue syndrome, stroke, toxic shock syndrome, adult respiratory distress syndrome, cachexia, myocarditis, eczema, psoriasis, dermatitis, urticaria, cerebral ischemia, systemic lupus erythematosis, chronic neurodegenerative disease, priapism, cystic fibrosis, schizophrenia, depression, premenstrual syndrome, anxiety, addiction, migraine, gastrointestinal motility disorders, obesity, hyperphagia, hematologic cancers, myelofibrosis, graft-versus-host disease, CNS trauma, hepatitis, renal failure, chronic hepatitis C, drug-induced lung injury, fertility enhancement, bacterial translocation, circulatory shock, traumatic shock, myocardial infarction, or vascular aneurysm.
29. The method of claim 19, wherein said tissue and/or cellular damage is associated with Parkinson's disease; Alzheimer's disease; Huntington's disease; multiple sclerosis; amylotrophic lateral sclerosis; AIDS-induced dementia; epilepsy; alcoholism; alcohol withdrawal; drug-induced seizures; viral/bacterial/fever-induced seizures; trauma to the head; hypoglycemia; hypoxia due to myocardial infarct; cerebral vascular occlusion; cerebral vascular hemorrhage; hemorrhage; or environmental excitotoxins of plant, animal, or marine origin
30. The method of claim 19, wherein said tissue and/or cellular damage is associated with cytokine therapy, wherein said compound is administered to said subject before, during, and/or after the administration of said cytokine to said subject.
EP05787832A 2004-09-17 2005-09-16 Nitrate esters and their use for mitigating cellular damage Pending EP1797100A4 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/943,264 US20050137191A1 (en) 1996-06-04 2004-09-17 Nitrate esters and their use for mitigating cellular damage
PCT/CA2005/001417 WO2006029532A1 (en) 1996-06-04 2005-09-16 Nitrate esters and their use for mitigating cellular damage

Publications (2)

Publication Number Publication Date
EP1797100A1 true EP1797100A1 (en) 2007-06-20
EP1797100A4 EP1797100A4 (en) 2009-05-13

Family

ID=38024540

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05787832A Pending EP1797100A4 (en) 2004-09-17 2005-09-16 Nitrate esters and their use for mitigating cellular damage

Country Status (3)

Country Link
EP (1) EP1797100A4 (en)
AU (1) AU2005284573A1 (en)
CA (1) CA2580627A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000054756A2 (en) * 1999-03-15 2000-09-21 Queen's University At Kingston Nitrate esters and their use for introducing neuroprotection and cognition enhancement
WO2001049275A2 (en) * 1999-12-29 2001-07-12 Queen's University At Kingston Methods and compositions for mitigating pain using nitrate esters

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000054756A2 (en) * 1999-03-15 2000-09-21 Queen's University At Kingston Nitrate esters and their use for introducing neuroprotection and cognition enhancement
WO2001049275A2 (en) * 1999-12-29 2001-07-12 Queen's University At Kingston Methods and compositions for mitigating pain using nitrate esters

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO2006029532A1 *

Also Published As

Publication number Publication date
CA2580627A1 (en) 2006-03-23
EP1797100A4 (en) 2009-05-13
AU2005284573A1 (en) 2006-03-23

Similar Documents

Publication Publication Date Title
WO2006029532A1 (en) Nitrate esters and their use for mitigating cellular damage
US6677374B2 (en) Nitrate esters and methods of making same
CA2791738C (en) Aryl imidazolyl compounds for the treatment of cancer
US7572781B2 (en) Prodrugs for use as ophthalmic agents
EP1246625B1 (en) Uses and compositions of nitrate esters for providing sedation
DK2943467T3 (en) Benzylidenguanidinderivativer and therapeutic use for the treatment of diseases associated with improper protein folding
JP2008523145A (en) Spiro derivatives as lipoxygenase inhibitors
JP2008531558A (en) Novel lipooxygenase inhibitors
CA2807968C (en) 2-(4-methylthiazol-5-yl)ethyl nitrate maleate salt compound and uses thereof
CA2798272C (en) Compounds and methods of treating brain disorders
EP1797100A1 (en) Nitrate esters and their use for mitigating cellular damage
NZ577311A (en) The oxalate salt of 3-benzyl-2-methyl-2,3,3a,4,5,6,7,7a-octahydrobenzo[d]isoxazol-4-one
EP1569648A1 (en) Prodrugs for use a ophthalmic agents
KR20100097940A (en) Novel ck2 inhibitor and method of preparing the same
WO2014163643A1 (en) Gga and gga derivatives compositions thereof and methods for treating neurodegenerative diseases including paralysis including them

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070413

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1103740

Country of ref document: HK

A4 Supplementary search report drawn up and despatched

Effective date: 20090409

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 209/44 20060101ALI20090404BHEP

Ipc: C07D 241/44 20060101ALI20090404BHEP

Ipc: A61K 31/21 20060101ALI20090404BHEP

Ipc: C07D 333/16 20060101ALI20090404BHEP

Ipc: A61K 31/38 20060101ALI20090404BHEP

Ipc: A61K 31/395 20060101ALI20090404BHEP

Ipc: C07D 217/16 20060101ALI20090404BHEP

Ipc: C07D 339/04 20060101ALI20090404BHEP

Ipc: C07D 307/79 20060101ALI20090404BHEP

Ipc: C07D 495/10 20060101AFI20060405BHEP

Ipc: C07D 311/66 20060101ALI20090404BHEP

Ipc: C07D 311/16 20060101ALI20090404BHEP

Ipc: C07D 333/56 20060101ALI20090404BHEP

Ipc: C07D 213/80 20060101ALI20090404BHEP

Ipc: C07D 409/12 20060101ALI20090404BHEP

Ipc: A61K 31/095 20060101ALI20090404BHEP

Ipc: C07D 333/32 20060101ALI20090404BHEP

Ipc: C07D 491/052 20060101ALI20090404BHEP

Ipc: C07D 417/06 20060101ALI20090404BHEP

Ipc: C07D 487/04 20060101ALI20090404BHEP

Ipc: C07D 207/16 20060101ALI20090404BHEP

Ipc: C07D 339/08 20060101ALI20090404BHEP

Ipc: C07C 321/14 20060101ALI20090404BHEP

D18D Application deemed to be withdrawn (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090401

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1103740

Country of ref document: HK