WO2010094090A2 - Synucleinopathies - Google Patents

Synucleinopathies Download PDF

Info

Publication number
WO2010094090A2
WO2010094090A2 PCT/BE2010/000013 BE2010000013W WO2010094090A2 WO 2010094090 A2 WO2010094090 A2 WO 2010094090A2 BE 2010000013 W BE2010000013 W BE 2010000013W WO 2010094090 A2 WO2010094090 A2 WO 2010094090A2
Authority
WO
WIPO (PCT)
Prior art keywords
guanidine
furan
ylcarbonyl
carbonyl
pyrazole
Prior art date
Application number
PCT/BE2010/000013
Other languages
French (fr)
Other versions
WO2010094090A3 (en
Inventor
Veerle Baekelandt
Sabrina Buettner
Melanie Gerard
Frank Madeo
Joris Winderucjx
Original Assignee
Katholleke Universiteit Leuven
University Of Graz
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 GB0902662A external-priority patent/GB0902662D0/en
Priority claimed from GB0902781A external-priority patent/GB0902781D0/en
Application filed by Katholleke Universiteit Leuven, University Of Graz filed Critical Katholleke Universiteit Leuven
Publication of WO2010094090A2 publication Critical patent/WO2010094090A2/en
Publication of WO2010094090A3 publication Critical patent/WO2010094090A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three 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
    • C07D307/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention concerns compounds for inhibiting ⁇ -synuclein toxicity.
  • Such compounds can be used in the treatment or prevention of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).
  • PD Parkinson's disease
  • DLB dementia with Lewy bodies
  • PAF pure autonomic failure
  • MSA multiple system atrophy
  • the present invention relates to sodium-hydrogen exchanger type 1 (Nhe- 1) inhibitors and most particular furancarbonylguanidine derivative compounds with Nhe-1 activity.
  • Nhe- 1 sodium-hydrogen exchanger type 1
  • Such compounds can be used in the treatment or prevention of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).
  • synucleinopathies such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).
  • the invention further relates to pharmaceutical compositions, to pharmaceutical compositions using a combination, or a pharmaceutical composition of such combination, of a sodium/hydrogen exchange type-1 (Nhe-1) inhibitor and a composition for the treatment of of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).
  • PD Parkinson's disease
  • DLB dementia with Lewy bodies
  • PAF pure autonomic failure
  • MSA multiple system atrophy
  • the invention further provides kits directed to such combinations.
  • Neurodegenerative diseases are symptoms of systematic degeneration and drop out of neurons and a lot of severe diseases are known, e.g. Alzheimer's disease, Parkinson's disease, Parkinson syndrome, dementia, amyotrophic lateral sclerosis, progressive supranuclear palsy, Huntington's disease, spinocerebellar ataxia, etc. A lot of molecules are involved with the mechanism of neurodegenerative death and it is assumed that they are overexpressed or functional disorder occurs in them. However, almost nothing is known on the molecular pathology and an effective method for the treatment is not established yet.
  • Neurodegenerative diseases symptoms include tremor, rigidity, akinesia, bradykinesia, slow movement, postural reflex disorder, pulsion, gait disorder, depression, dysmnesia, amyotrophy, muscle weakness, dysfunctions of upper extremities, dysarthria, dysphagia, respiratory obstruction, palsy, paralysis, etc. It is well known in the art that numerous therapeutic regimens have been developed, for example, the use of compounds that inhibit the sodium/hydrogen exchange type-1 (NHE- 1) transport system.
  • NHE-1 sodium/hydrogen exchange type-1
  • NHE-1 inhibitors elicit protective effects against ischemia, particularly that affecting the myocardium, consists of a reduction in the increased sodium ion influx which is caused in reperfused/hypoperfused tissues due to intracellular acidification and subsequent activation of the sodium/hydrogen exchange transport system. This results in a delay of sodium overload of the tissue. Since sodium and calcium ion transport are coupled in cardiac tissue, this also prevents the life threatening calcium overload of myocardial cells.
  • the present inventions relates to the surprising finding that the inhibition if Nhe-1 affords a mechanism for treatment and prevention of synucleinopathies.
  • the present invention relates to a furancarbonylguanidine derivative represented by the Formula (I) and/or pharmaceutically acceptable salts, solvates and isomers thereof, for use in a treatment to cure or to prevent synucleinopathy, and wherein
  • R 1 and R 2 are each independently selected from H; F; Cl; Br; I; CF 3 ; SO 2 CH 3 ; NO 2 ; NH 2 ; C 1 -C 5 straight or branched alkyl; or OR 4 ;
  • R 3 is selected from H, NH 2 , C 1 -C 5 straight or branched alkyl, NH(COCH 3 ), N(CH 3 )COCH 3 , methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
  • R 4 is selected from H, CF 3 , C 1 -C 5 straight or branched alkyl, or phenyl.
  • the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
  • present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative represented by Formula (I) and/or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein
  • R 2 are each independently selected from H; F; Cl; Br; I; CF 3 ; SO 2 CH 3 ; NO 2 ; NH 2 ; C 1 -C 5 straight or branched alkyl; or OR 4 ;
  • R 3 is selected from H, NH 2 , C 1 -C 5 straight or branched alkyl, NH(COCH 3 ), N(CH 3 )COCH 3 , methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
  • R 4 is selected from H, CF 3 , C 1 -C 5 straight or branched alkyl, or phenyl.
  • the pharmaceutical composition according to present invention comprises a furancarbonylguanidine derivative and/or pharmaceutically acceptable salts, solvates and isomers thereof selected from the group consisting of:
  • present invention further relates to the use of a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
  • a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
  • the use of the compounds of present invention includes the use of particular furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
  • the present invention relates to a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy and the Nhe-1 inhibitor is selected from the group consisting of: a) a NHE-1 inhibiting carbonylguanidine compound of Formula (I)
  • - Z is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination with up to three substituents independently selected from R 1 , R 2 and R 3 ; or
  • - Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ; or
  • - Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ; or - Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R 1 , R 2 and R 3 ; or
  • - Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxy(C r C 4 )alkyl, (C 1 - C 4 )alkyl, (C r C 4 )alkylthio, (C 3 -C 4 )cycloalkyl, (C 3 -C 7 )cycloalkyl(C r C 4 )alkyl, (C r C 4 )alkoxy, (Ci-C 4 )alkoxy(Ci-C 4 )alkyl, mono-N- or di-N,N-(CrC 4 )alkylcarbamoyl, M or M(C r C 4 )alkyl, any of said previous (C r C 4 )alkyl moieties optionally having from one to nine fluorines; said (C r C 4 )alkyl or (C 3 -C 4 )cycloalkyl optionally mono-or di-substituted independently with
  • present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy.
  • the pharmaceutical composition comprises an effective amount of a carbonylguanidine Nhe-1 inhibitor selected from the group consisting of: a) a compound of Formula (I)
  • - Z is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination with up to three substituents independently selected from R 1 , R 2 and R 3 ; or
  • - Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ; or
  • - Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ; or - Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R 1 , R 2 and R 3 ; or
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxy(CrC 4 )alkyl, (C 1 - C 4 )alkyl, (C 1 -C 4 )alkylthio, (C 3 -C 4 )cycloalkyl, (C 3 -C 7 )cycloalkyl(C 1 -C 4 )alkyl, (C r C 4 )alkoxy, (CrOOalkoxyCCrOOalkyl, mono-N- or di-N.N-CCrC ⁇ alkylcarbamoyl, M or M ⁇ -C ⁇ alky!, any of said previous (d-C 4 )alkyl moieties optionally having from one to nine fluor
  • M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R 6 , R 7 and R 8 , wherein one of R 6 , R 7 and R 8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (C 1 -C 4 )alkyl and additionally R 6 , R 7 and R 8 are optionally hydroxy, nitro, halo, (C 1
  • the pharmaceutical composition further comprises a composition for the treatment of Parkinson's disease.
  • the pharmaceutical composition further comprises a composition for the treatment of Parkinson's disease comprises a compound selected from the group consisting of:
  • a dopaminergic effective amount of a compound and further comprising a pharmaceutically acceptable carrier.
  • present invention also relates to a kit comprising an amount of a
  • Nhe-1 inhibitor of present invention and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of
  • Parkinson's disease and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • the kit of present invention comprises an amount of a Nhe-1 inhibitor of present invention, a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • Embodiment 1 concerns a furancarbonylguanidine derivative represented by
  • Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy and wherein:
  • R 1 and R 2 are each independently selected from H; F; Cl; Br; I; CF 3 ; SO 2 CH 3 ; NO 2 ; NH 2 ; C 1 -Cs straight or branched alkyl; or OR 4 ; 0 - R 3 is selected from H, NH 2 , C 1 -C 5 straight or branched alkyl, NH(COCH 3 ), N(CH 3 )COCH 3 , methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
  • R 4 is selected from H, CF 3 , C 1 -C 5 straight or branched alkyl, or phenyl.
  • furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof according to embodiment 1 , wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: 0 - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
  • a pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
  • R 1 and R 2 are each independently selected from H; F; Cl; Br; I; CF 3 ; SO 2 CH 3 ; NO 2 ; NH 2 ; 15 Ci-C 5 straight or branched alkyl; or OR 4 ;
  • R 3 is selected from H, NH 2 , C 1 -C 5 straight or branched alkyl, NH(COCH 3 ), N(CH 3 )COCH 3 , methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
  • R 4 is selected from H, CF 3 , C 1 -C 5 straight or branched alkyl, or phenyl.
  • composition according to embodiment 4 wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
  • composition according to any of embodiments 4 to 5, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
  • furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
  • Parkinson's disease selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
  • Group 1 concerns a Nhe-1 inhibitor, for instance a NHE-1 inhibiting carbonylguanidine, for use in a treatment to cure or to prevent synucleinopathy.
  • Nhe-1 inhibitor for instance a NHE-1 inhibiting carbonylguanidine, for use in a treatment to cure or to prevent synucleinopathy.
  • the Nhe-1 inhibitor of group 1 wherein said Nhe-1 inhibitor is selected from the group consisting of:
  • Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5-
  • Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and
  • Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R 1 , R 2 and R 3 ; or Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxy(CrC 4 )alkyl, (C 1 - C 4 )alkyl, (d-C ⁇ alkylthio, (C 3 -C 4 )cycloalkyl, (C 3 -C 7 )cycloalkyl(C 1 -C 4 )alkyl, (C r C 4 )alkoxy, (C ⁇ C 4 )alkoxy(C ⁇ C 4 )alkyl, mono-N- or di-N.N-CCrC ⁇ alkylcarbamoyl, M or M(C 1 -C 4 )alkyl, any of said previous (Ci-C 4 )alkyl moieties optionally having from one to nine fluorines; said (CrC 4 )aIkyl or (C 3 -C 4 )cycloalkyl optionally mono-or di-substituted independently with hydroxy, (C r C
  • cariporide N-(4-lsopropyl-3-methanesulfonyl-benzoyl)-guanidine, or a pharmaceutically acceptable salt, solvate or isomer thereof;
  • SM-15681 (5-(N-ethyl-N-isopropyl)amiloride,5-(ethylisopropyl)amiloride), or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • Nhe-1 inhibitor of group 2 wherein said Nhe-1 inhibitor is selected from the group consisting of:
  • Nhe-1 inhibitor of any of groups 1 to 3 wherein said synucleinopathy is selected from the group consisting of Parkinson's disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy.
  • a pharmaceutical composition comprising an effective amount of a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy. 6.
  • the pharmaceutical composition of group 5 wherein said Nhe-1 inhibitor is selected from the group consisting of:
  • Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5
  • Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group 30 of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5
  • - Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three
  • - Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxy(C r C 4 )alkyl, (C 1 - 0 C 4 )alkyl, (C r C 4 )alkylthio, (C 3 -C 4 )cycloalkyl, (C 3 -C 7 )cycloalkyl(C 1 -C 4 )alkyl, (C r C 4 )alkoxy, (CrC 4 )alkoxy(Ci-C 4 )alkyl, mono-N- or di-N.N- ⁇ -OOalkylcarbamoyl, M or M(C 1 -C 4 )alkyl, any of said previous (C- ⁇ -C 4 )alkyl moieties optionally having from one to nine fluorines; said (C 1 -C 4 )alkyl or (C 3 -C 4 )cycloalkyl optionally mono-or di-substituted independently with
  • composition according to any of groups 5 to 6, wherein said pharmaceutical composition is selected from the group consisting of:
  • synucleinopathy is selected from the group consisting of Parkinson's disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy
  • composition according to any of groups 5 to 8, further comprising a composition for the treatment of Parkinson's disease.
  • compositions according to group 9 further comprising a composition for the treatment of Parkinson's disease, wherein said composition for the treatment of Parkinson's disease comprises a compound selected from the group consisting of: - L-DOPA;
  • a kit comprising an amount of a Nhe-1 inhibitor according to group 1, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of Parkinson's disease, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • kits according to group 11 wherein said composition for the treatment of Parkinson's disease is selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • R 3 is selected from H, NH 2 , C 1 -C 5 straight or branched alkyl, NH(COCH 3 ), N(CH 3 )COCH 3 , methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
  • R 4 is selected from H, CF 3 , C 1 -C 5 straight or branched alkyl, or phenyl.
  • (ll) is a 3 to 8 membered ring.
  • - is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination;
  • - is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan; or
  • - is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole; or
  • - is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens;
  • - is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring;
  • a pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative of any of the previous claims 13 to 15 or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy. 5. Any of the previous claims 13 to 16, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
  • furancarbonylguanidine derivative is of the group consisting of cariporide (N-(4-lsopropyl-3-methanesulfonyl- benzoyl)-guanidine), BIIB-513 (benzamide ⁇ N-(aminominomethyl)-4[4-(2- furanylcarbonyl)- 1-piperazinyl]-3-(methylsulfonyl), methanesulfonate ⁇ ), TY-12533 ( ⁇ .y. ⁇ j ⁇ -tetrahydro ⁇ -methyl- ⁇ H-cycloheptal ⁇ pyridine-S-carbonylguanidine maleate) and SM-15681 (5-(N-ethyl-N-isopropyl)amiloride,5-(ethylisopropyl)amiloride) or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • cariporide N-(4-lsopropyl-3-methanesulfonyl
  • Figure 1 shows the growth of a wild type yeast strain or a nud deletion mutant without or with expression of human EndoG as expressed by the Optical Density (OD600nm relative units). Samples were taken at 3 days after inuculation of the cultures. The values shown represent mean + SEM.
  • Figure 2 shows the growth of wild type yeast cells expressing human ⁇ -synuclein and human EndoG on selective medium without or supplemented with of [5-(2-Methyl-5- Fluorophenyl)furan-2-ylcarbonyl]guanidine (Sigma-Aldrich K4389) at the concentrations indicated as expressed by the Optical Density (OD ⁇ OOnm relative units). The values shown represent mean + SEM.
  • Figure 3 concerns the effect of compound K4389 on a-synuclein aggregation and late apoptosis. It shows: (A) Effect of different concentrations of [5-(2-Methyl-5- fluorophenyl)furan-2-ylcarbonyl]guanidine (Sigma-Aldrich K4389) on ⁇ -Synuclein aggregation in SHSY5Y cells induced by oxidative stress. Stock concentrations in DMSO are diluted 100x until final concentration shown on graph. D : p ⁇ 0.01 (Kruskal-Wallis test followed by Dunnet's post hoc test). (B) Effect of different concentrations of K4389 on nucleus condensation, a marker for late apoptosis. All percentages are relative to negative control condition: DMSO, 100%.
  • Figure 4 concerns the inhibition of the yeast homolog of NHE-1 decreases alpha- synuclein toxicity. It displays (A) Quantification of ROS-accumulation using the conversion of non-fluorescent dihydroethidium (DHE) to fluorescent ethidium (Eth) in wild type yeast cells expressing human alpha-synuclein ( ⁇ Syn) for 48 h or harbouring the empty vector control. Cells were treated with indicated concentrations of the NHE-1 inhibitor K4389 (solved in DMSO) or equivalent concentrations of DMSO alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control.
  • DHE non-fluorescent dihydroethidium
  • ⁇ Syn human alpha-synuclein
  • Aliphatic refers to a group selected from the set of "Alkyl”, “Alkenyl” and “Alkynyl”.
  • saturated aliphatic refers to "Alkyl”
  • Unsaturated aliphatic refers to a group selected from the set of "Alkenyl” and “Alkynyl”.
  • Alkyl refers to a straight or branched chain hydrocarbon containing from 1 or 2 to 10 or 20 or more carbon atoms (e.g., C2, C3, C4, C5, C6, C7, C8, C9, CIO, Cl 1, C12, C13, C14, C15, etc.).
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n- heptyl, n-octyl, n- nonyl, n-decyl, and the like.
  • alkyl groups as described herein are optionally substituted (e.g., from 1to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
  • substituted indicates that the specified group is either unsubstituted, or substituted by one or more suitable substituents.
  • a "substituent” is an atom or atoms substituted in place of a hydrogen atom on the parent chain or cycle of an organic molecule, for example, H, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
  • Alkenyl refers to a straight or branched chain hydrocarbon containing from 1 or 2 to 10 or 20 or more carbons, and containing at least one carbon-carbon double bond, formed structurally, for example, by the replacement of two hydrogens.
  • alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl-l-heptenyl, 3- decenyl and the like.
  • alkenyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
  • Alkynyl refers to a straight or branched chain hydrocarbon group containing from 1 or 2 to 10 or 20 or more carbon atoms, and containing at least one carbon- carbon triple bond.
  • Representative examples of alkynyl include, but are not limited, to acetylenyl, 1-propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, 1-butynyl and the like.
  • alkynyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
  • cycloaliphatic refers to a saturated or unsaturated cyclic hydrocarbon group containing from 3 to 8 carbons or more, which is not aromatic.
  • Representative examples of cycloalkyl include, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • cycloalkyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
  • Heterocyclic refers to a monocyclic or a bicyclic ring system.
  • Monocyclic heterocyclic ring systems are exemplified by any 5 or 6 member ring containing 1 , 2, 3, or 4 heteroatoms independently selected from the group consisting of: O, N, and S.
  • the 5 member ring has from 0 to 2 double bonds, and the 6 member ring has from 0-3 double bonds.
  • monocyclic ring systems include, but are not limited to, azetidine, azepine, aziridine, diazepine, 1 ,3-dioxolane, dioxane, dithiane, furan, imidazole, imidazoline, imidazolidine, isothiazole, isothiazoline, isothiazolidine, isoxazole, isoxazoline, isoxazolidine, morpholine, oxadiazole, oxadiazoline, oxadiazolidine, oxazole, oxazoline, oxazolidine, piperazine, piperidine, pyran, pyrazine, pyrazole, pyrazoline, pyrazolidine, pyridine, pyrimidine, pyridazine, pyrrole, pyrroline, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, tetraz
  • Bicyclic ring systems are exemplified by any of the above monocyclic ring systems fused to an aryl group as defined herein, a cycloalkyl group as defined herein, or another monocyclic ring system as defined herein.
  • Representative examples of bicyclic ring systems include but are not limited to, for example, benzimidazole, benzothiazole, benzothiadiazole, benzothiophene, benzoxadiazole, benzoxazole, benzofuran, benzopyran, benzothiopyran, benzodioxine, 1 ,3- benzodioxole, cinnoline, indazole, indole, indoline, indolizine, naphthyridine, isobenzofuran, isobenzothiophene, isoindole, isoindoline, isoquinoline, phthalazine, pyranopyridine, quinoline, quinolizine
  • Aromatic refers to a fused ring system having one or more aromatic rings.
  • Representative examples of aryl include, azulenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like.
  • aromatic groups of this invention can be substituted with 1 , 2, 3, 4, or 5 substituents independently selected from alkenyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylsulfinyl, alkylsulfonyl, alkylthio, alkynyl, aryl, aryloxy, azido, arylalkoxy, arylalkyl, aryloxy, carboxy, cyano, formyl, halogen, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, mercapto, nitro, sulfamyl, sulfo, sulfonate, -NR1R" (wherein, R1 and R" are independently selected from hydrogen, alkyl, alkylcarbonyl, aryl, arylalkyl and formyl), and -C
  • Heteroaromatic means a cyclic, aromatic hydrocarbon in which one or more carbon atoms have been replaced with heteroatoms. If the heteroaromatic group contains more than one heteroatom, the heteroatoms may be the same or different.
  • heteroaryl groups include pyridyl, pyrimidinyl, imidazolyl, thienyl, furyl, pyrazinyl, pyrrolyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, indolyl, isoindolyl, indolizinyl, triazolyl, pyridazinyl, indazolyl, purinyl, quinolizinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxalinyl, isothiazolyl, and benzo[b]thienyl.
  • Preferred heteroaryl groups are five and six membered rings and contain from one to three heteroatoms independently selected from the group consisting of: O, N, and S.
  • the heteroaryl group, including each heteroatom can be unsubstituted or substituted with from 1 to 4 suitable substituents, as chemically feasible.
  • ⁇ -synuclein refers to one in a family of structurally related proteins that are prominently expressed in the central nervous system. Aggregated ⁇ -synuclein proteins form brain lesions that are hallmarks of some neurodegenerative diseases (synucleinopathies).
  • the gene for ⁇ -synuclein which is called SNCA, is on chromosome 4q21.
  • Alpha-synuclein is a member of the synuclein family, which also includes beta- and gamma-synuclein. Synucleins are abundantly expressed in the brain and alpha- and beta- synuclein inhibit phospholipase D2 selectively. Ueda et al.
  • Aza is in the meaning of containing nitrogen in place of carbon.
  • synucleinopathies is used to name a group of neurodegenerative disorders characterized by fibrillary aggregates of alpha-synuclein protein in the cytoplasm of selective populations of neurons and glia. These disorders include Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), multiple system atrophy (MSA), neurodegeneration with brain iron accumulation, type 1 (e.g. Hallervorden- Spatz syndrome, neuroaxonal dystrophy), and other diseases that may have synuclein- immunoreactive lesions (e.g. Traumatic brain injury, Pick disease, Amyotrophic lateral sclerosis).
  • Parkinson's disease PD
  • DLB dementia with Lewy bodies
  • PAF pure autonomic failure
  • MSA multiple system atrophy
  • neurodegeneration with brain iron accumulation e.g. Hallervorden- Spatz syndrome, neuroaxonal dystrophy
  • type 1 e.g. Hallervorden- Spatz syndrome, neuroaxonal dys
  • ⁇ -synuclein toxicity associated disease refers to "synucleinopathies” and is used as a synonym.
  • MSA Multiple system atrophy
  • OPCA olivopontocerebellar atrophy
  • SND striatonigral degeneration
  • SDS Shy-Drager syndrome
  • the formation of alpha-synuclein aggregates is a critical event in the pathogenesis of multiple system atrophy (MSA).
  • the histopathological hallmark is the formation of ⁇ -synuclein-positive glial cytoplasmic inclusions (GCIs) in oligodendroglia.
  • GCIs ⁇ -synuclein-positive glial cytoplasmic inclusions
  • Parkinson's disease is the second most common age-associated neurodegenerative disease. Parkinson's disease can e.g. be sporadic, familial with ⁇ -synuclein mutations, or familial with mutations other than ⁇ -synuclein.
  • missense mutations in the ⁇ -synuclein gene are linked to early-onset dominant familial PD. More recently, overexpression of wild-type ⁇ -synuclein due to gene duplication or triplication was found to be sufficient to cause a familial form of PD.
  • Dementia with Lewy bodies is the second most frequent cause of hospitalization for dementia, after Alzheimer's disease. Current estimates are that about 60-to-75% of diagnosed dementias are of the Alzheimer's and mixed (Alzheimer's and vascular dementia) type, 10-to-15% are Lewy Bodies type, with the remaining types being of an entire spectrum of dementias including frontotemporal lobar degeneration, alcoholic dementia, pure vascular dementia.
  • Dementia with Lewy bodies includes e.g. "pure" Lewy body dementia, Lewy body variant of Alzheimer disease, familial Alzheimer disease with APP mutations, familial Alzheimer disease with PS-1 mutations, familial Alzheimer disease with PS-2 mutations, and Down syndrome.
  • Pure autonomic failure also known as Bradbury-Eggleston syndrome or idiopathic orthostatic hypotension
  • the symptoms concern a degenerative disease of the peripheral nervous system, symptoms include dizziness and fainting (caused by orthostatic hypotension), visual disturbances and neck pain. Chest pain, fatigue and sexual dysfunction are less common symptoms that may also occur. Symptoms are worse when standing; sometimes one may relieve symptoms by laying down. Accumulation of alpha-synuclein in autonomic nerves causes pure autonomic failure (Horacio Kaufmann et al. Neurology 2001 ;56:980-981).
  • the present invention addresses the following unmet medical needs, inter alia; 1) providing compounds and pharmaceutical compositions capable of effectively controlling, in particular preventing or treating synucleinopathy such as Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy. 2) providing compounds and pharmaceutical compositions capable of effectively controlling, in particular preventing or treating synucleinopathy such as but not limited to Parkinson's disease.
  • Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R 1 , R 2 and R 3 ; or Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ; wherein R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxyCCrC ⁇ alkyl, (C 1 - C 4 )alkyl, (d-C ⁇ alkylthio, (C 3 -C 4 )cycloalkyl, (C3-C 7 )cycloalkyl(d
  • cariporide or a pharmaceutically acceptable salt, solvate or isomer thereof
  • BIIB-513 or a pharmaceutically acceptable salt, solvate or isomer thereof
  • TY-12533 or a pharmaceutically acceptable salt, solvate or isomer thereof
  • SM-15681 or a pharmaceutically acceptable salt, solvate or isomer thereof, the prodrugs thereof, and the pharmaceutically acceptable salts, solvates or isomers of the compounds and prodrugs, may be prepared as disclosed in the aforementioned, commonly-assigned PCT International Application Publication No. WO 99/43663, the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor cariporide i.e. N-(aminoiminomethyl)-4-(1-methylethyl)-3- (methylsulfonyl)-benzamide
  • the preferred NHE-1 inhibitor BIIB-513 i.e. N-(aminoiminomethyl)4-(4-(2-furanylcarbonyl)-1-piperazinyl)-3- (methylsulfonyl)-benzamide, may be prepared as disclosed in U.S. Pat. No. 6,114,335, the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor TY-12533 i.e. ⁇ .T. ⁇ . ⁇ -tetrahydro ⁇ -methyl- ⁇ H-cycloheptaEbJpyridine-S-carbonylguanidine, may be prepared as disclosed in PCT International Application Publication No. WO 98/39300, the disclosure of which is incorporated herein by reference.
  • the preferred NHE- 1 inhibitor SM-15681 i.e. N-(aminoiminomethyl)-1-methyl-1H-indole-2-carboxamide, may be prepared as disclosed in EPO 0 708 091 , the disclosure of which is incorporated herein by reference.
  • the ability of a compound to function as an NHE-1 inhibitor may be determined according to the protocols described in detail herein below.
  • Example 1 A yeast strain for identifying compounds that reduce ⁇ -synuclein toxicity
  • a yeast strain suitable for studying synucleinopathy was created.
  • a wild type BY4741 yeast strain (MATa his3Ai leu2 ⁇ 0 met15AO ura3 ⁇ 0) obtained from Euroscarf was transformed with a plasmid allowing for constitutive expression of human ⁇ -synuclein, as well as with human endonuclease G which is an effector of the ⁇ -synuclein-induced toxic effect.
  • fibroblasts stably expressing human Nhe-1 are plated onto collagen coated 96 well plates (50,000/well) and grown to confluence in growth media (DMEM high glucose, 10% fetal bovine serum, 50 u/ml penicillin and streptomycin). Confluent plates are incubated for 30 min at 37° C. with the pH sensitive fluorescent probe BCECF (5 ⁇ M; Molecular Probes, Eugene, Oreg.). BCECF loaded cells are incubated for 30 min at 37° C.
  • Intracellular acidification is initiated via rapid replacement of acid loading media with recovery media (120 mM NaCl, 5 mM KCI, 1 mM MgCI 2, 1.8 mM CaCI 2, 5 mM glucose, 10 mM HEPES, pH 7.5) ⁇ test compound, and Nhe-mediated recovery of intracellular pH is monitored as the subsequent time-dependent increase BCECF fluorescence.
  • the potency of human Nhe-1 inhibitors is calculated as the concentration that reduces recovery of intracellular pH by 50% (IC 50 ).
  • Example 4 - Assessment of a yeast-based model in high-throughput screens selection of Nhe-1 inhibitors that suppress ⁇ -svnuclein toxicity in yeast.
  • yeast model expressing human ⁇ -synuclein and human endonuclease G is examined in high-throughput screens. To this end, 500 compounds are tested individually to address their capacity to modulate ⁇ -synuclein toxicity in yeast cells.
  • Yeast cells are grown in 96-well microtiter plates and growth is monitored by measuring OD at 600 nm. The compounds are dissolved in DMSO and added to the growth medium at a final concentration of 10 ⁇ g/ml. Every microtiter plate contains positive and negative controls for calculating the Z'-factor in order to assess the quality of the assay of each microtiter plate.
  • Positive and negative growth controls are wild-type yeast cells transformed with ⁇ -synuclein and human endonuclease G, respectively. 500 compounds are screened in duplicate (screen 1 and 2). The OD ⁇ oo values are normalised to the negative control and thus represent the growth relative to yeast cells that produce ⁇ - synuclein. The assay is done in duplicate and the effect of each compound on growth is determined by measuring optical density at 600 nm (OD 6 O 0 ). The OD 600 measurements are normalised and depicted as a scatter graph. The Z'-factor (Zhang et al. (1999) J. Biomol. Screen. 4, 67-73) is calculated for each microtiter plate in order to obtain a quantitative score of the assay quality.
  • the average Z'-factor of each plate is found at least 0.70 or higher indicating that the assay is robust and reproducible and therefore very suitable for high-throughput screening purposes. Testing 500 compounds consistently results in highly reproducible OD 600 values for each compound. Importantly, this assay allows selection of compounds that significantly and reproducibly improve growth of the yeast strain. Hence the ⁇ -synuclein toxicity is overcome.
  • the compound (5-(2-methyl-5- fluorophenyl)furan-2-ylcarbonyl)guanidine (abbreviated as K4389) is dissolved in DMSO, therefore negative control condition is with DMSO only.
  • aggregates are visualized with thioflavin S and the percentage of aggregate positive cells is determined (Figure 2).
  • the percentage of cells in late apoptosis is also determined based on nucleus condensation, visualized with a nuclear DAPI staining.
  • the compound K4389 clearly has a concentration-dependent effect on ⁇ -Synuclein aggregation with a statistically significant effect from 1 nM (non parametric Kruskal-Wallis test followed by Dunn's post hoc test) (Figure 2A).
  • the same trend is also noticed in the effect on ⁇ -Synuclein-induced toxicity (effect on late apoptosis) although differences are not statistically significant (Figure 2B).
  • Example 6 The 3-Substituted-(5-arylfuran-2-ylcarbonvQguanidines NHE-1 inhibitors reduces ⁇ -synuclein toxicity in yeast
  • Example 7 Inhibition of the yeast homolog of NHE-1 decreases alpha-svnuclein toxicity.
  • Figure 4 displays (A) Quantification of ROS-accumulation using the conversion of non- fluorescent dihydroethidium (DHE) to fluorescent ethidium (Eth) in wild type yeast cells expressing human alpha-synuclein ( ⁇ Syn) for 48 h or harbouring the empty vector control. Cells were treated with indicated concentrations of the NHE-1 inhibitor K4389 (solved in DMSO) or equivalent concentrations of DMSO alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control.
  • DHE non- fluorescent dihydroethidium
  • ⁇ Syn human alpha-synuclein
  • the present invention also relates to pharmaceutical compositions or formulations which comprise one or more of the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention in admixture with one or more pharmaceutically acceptable excipients, diluents or carriers, the latter being for instance as described hereinafter.
  • excipient and “carrier” are used interchangeably throughout the description of the present invention and said terms are defined herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition.”
  • excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means for achieving effective absorption by the recipient of the active ingredient.
  • An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach.
  • the formulator can also take advantage of the fact the compounds of the present invention have improved cellular potency, pharmacokinetic properties, as well as improved oral bioavailability.
  • furancarbonylguanidine derivative Nhe-1 inhibitors of this invention and/or pharmaceutically acceptable salts, solvates and isomers thereof, when intended to be included in a pharmaceutical composition may be formulated with conventional carriers and excipients, which can be selected in accordance with ordinary pharmaceutical practice.
  • tablets may contain excipients, glidants, fillers, binders and the like.
  • Aqueous formulations are preferably prepared in sterile form and, when intended for delivery by other than oral administration, are usually isotonic.
  • compositions optionally contain excipients such as those set forth in the " Handbook of Pharmaceutical Excipients " (1986) and may include ascorbic acid and/or other antioxidants, chelating agents, carbohydrates such as dextrin-containing compounds (e.g. maltodextrins and/or cyclodextrins), hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like.
  • excipients such as those set forth in the " Handbook of Pharmaceutical Excipients " (1986) and may include ascorbic acid and/or other antioxidants, chelating agents, carbohydrates such as dextrin-containing compounds (e.g. maltodextrins and/or cyclodextrins), hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like.
  • pharmaceutically acceptable carrier means any material or substance with which the active ingredient is formulated in order to facilitate its application or dissemination to the locus to be treated, for instance by dissolving, dispersing or diffusing the said composition, and/or to facilitate its storage, transport or handling without impairing its effectiveness.
  • the pharmaceutically acceptable carrier may be a solid or a liquid or a gas which has been compressed to form a liquid, i.e. the pharmaceutical compositions of this invention can suitably be in the form of concentrates, emulsions, solutions, granulates, dusts, sprays, aerosols, suspensions, ointments, creams, tablets, pellets or powders.
  • Suitable pharmaceutical carriers for use in the pharmaceutical compositions and formulations of the invention are well known to those skilled in the art. They also include additives such as, but not limited to, wetting agents, dispersing agents, stickers, adhesives, emulsifying agents, solvents, coatings, antibacterial and antifungal agents (for example phenol, sorbic acid, chlorobutanol), isotonic agents (such as sugars or sodium chloride) and the like, provided the same are consistent with pharmaceutical practice, i.e. carriers and additives which do not create permanent damage to mammals.
  • additives such as, but not limited to, wetting agents, dispersing agents, stickers, adhesives, emulsifying agents, solvents, coatings, antibacterial and antifungal agents (for example phenol, sorbic acid, chlorobutanol), isotonic agents (such as sugars or sodium chloride) and the like, provided the same are consistent with pharmaceutical practice, i.e. carriers and additives which do not create permanent damage to mammals.
  • compositions of the present invention may be prepared in any known manner, for instance by homogeneously mixing, coating and/or grinding the active ingredient, in a one-step or multi-steps procedure, with the selected one or more carrier materials and, where appropriate, the other additives such as surface-active agents. They may also be prepared by micronisation, for instance in view to obtain them in the form of microspheres usually having a diameter of about 1 to 10 ⁇ m, namely for the manufacture of microcapsules for controlled or sustained release of the active ingredient.
  • Suitable surface-active agents also known as emulsifiers, that can be used in the pharmaceutical compositions of the present invention include non-ionic, cationic and/or anionic materials having good emulsifying, dispersing and/or wetting properties.
  • Suitable anionic surfactants include both water-soluble soaps and water-soluble synthetic surface- active agents.
  • Suitable soaps are alkaline or alkaline-earth metal salts, solvates and isomers, unsubstituted or substituted ammonium salts, solvates and isomers of higher (CiO-C 22 ) fatty acids, e.g.
  • Synthetic surfactants include sodium or calcium salts, solvates and isomers of polyacrylic acids; fatty sulphonates and sulphates; sulphonated benzimi-dazole derivatives preferably having 8 to 22 carbon atoms; and alkylarylsulphonates.
  • Fatty sulphonates or sulphates are usually in the form of alkaline or alkaline-earth metal salts, solvates and isomers, unsubstituted ammonium salts, solvates and isomers or ammonium salts, solvates and isomers substituted with an alkyl or acyl radical having from 8 to 22 carbon atoms, e.g.
  • alkylarylsulphonates are the sodium, calcium or alcanolamine salts, solvates and isomers of dodecylbenzene sulphonic acid or dibutyl-naphtalenesulphonic acid or a naphtalenesulphonic acid/formaldehyde condensation product.
  • phosphates e.g. salts, solvates and isomers of phosphoric acid ester and an adduct of p-nonylphenol with ethylene and/or propylene oxide, or phospholipids.
  • Suitable phospholipids for this purpose include natural (originating from animal or plant cells) or synthetic phospholipids of the cephalin or lecithin type such as, but not limited to, phosphatidylethanoiamine, phosphatidylserine, phosphatidylglycerine, lyso-lecithin, cardiolipin, dioctanylphosphatidylcholine, dipalmitoylphoshatidyl-choline and mixtures thereof in various proportions.
  • Suitable non-ionic surfactants include polyethoxylated and polypropoxy-lated derivatives of alkylphenols, fatty alcohols, fatty acids, aliphatic amines or amides containing at least
  • alkylarene-sulphonates and dialkylsulphosuccinates such as, but not limited to, polyglycol ether derivatives of aliphatic and cycloaliphatic alcohols, saturated and unsaturated fatty acids and alkylphenols, said derivatives preferably containing from 3 to 10 glycol ether groups and from 8 to 20 carbon atoms in the (aliphatic) hydrocarbon moiety and/or from 6 to 18 carbon atoms in the alkyl moiety of the alkylphenol.
  • non-ionic surfactants are water-soluble adducts of polyethylene oxide with poylypropylene glycol, ethylenediaminopolypropylene glycol containing from 1 to 10 carbon atoms in the alkyl chain, which adducts contain from about
  • non-ionic surfactants are nonylphenol- polyethoxyethanol, castor oil polyglycolic ethers, polypropy-lene/polyethylene oxide adducts, tributylphenoxypolyethoxyethanol, polyethyleneglycol and octylphenoxypolyethoxyethanol.
  • Fatty acid esters of polyethylene sorbitan such as polyoxyethylene sorbitan trioleate
  • glycerol sorbitan
  • sucrose and pentaerythritol are also suitable non-ionic surfactants.
  • Suitable cationic surfactants include quaternary ammonium salts, solvates and isomers, particularly halides, having 4 hydrocarbon radicals optionally substituted with halo, phenyl, substituted phenyl or hydroxy; for instance quaternary ammonium salts, solvates and isomers containing as N-substituent at least one C 8 -C 22 alky] radical (e.g. cetyl, lauryl, palmityl, myristyl, oleyl and the like) and, as further substituents, unsubstituted or halogenated C 1 -C 4 alkyl, benzyl and/or hydroxy C 1 -C 4 alkyl radicals.
  • C 8 -C 22 alky e.g. cetyl, lauryl, palmityl, myristyl, oleyl and the like
  • the compound of this invention may be administered by any route appropriate for the bone disorder to be treated. Suitable routes include, but are not limited to, oral, rectal, nasal, topical (including transdermal ⁇ , ocular, buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intra-arterially, intradermal, intrathecal and epidural) routes.
  • routes include, but are not limited to, oral, rectal, nasal, topical (including transdermal ⁇ , ocular, buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intra-arterially, intradermal, intrathecal and epidural) routes.
  • the preferred route of administration may vary in accordance with certain clinical parameters, for example with the condition of the patient to be treated.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • a suitable tablet may be made by compression or molding, optionally with one or more pharmaceutically acceptable inactive ingredients such as described hereinabove.
  • Compressed tablets may be prepared by compressing, in a suitable compressing machine, the active ingredient in a free-flowing form such as a powder, granules, beads or pellets, optionally admixed with one or more pharmaceutically acceptable excipients such as binders, lubricants, inert diluents, preservatives, and surface-active or dispersing agents.
  • Molded tablets may be made by molding, in a suitable molding machine, a mixture of the powdered active compound moistened with suitable amounts of one or more inert liquid diluents.
  • the tablets may optionally be further coated, and may be formulated so as to provide slow or controlled release of the active ingredient therein.
  • the pharmaceutical formulations of the invention are optionally in the form of a topical ointment or cream.
  • the active ingredient may be admixed with a paraffinic or water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water cream base.
  • the aqueous phase of the cream base may include, for example, at least 30 % by weight of a polyhydric alcohol, i.e.
  • Topical formulations may desirably include at least a compound which enhances absorption or penetration of the active ingredient through the skin or other areas of application.
  • dermal penetration enhancers include, but are not limited to, dimethylsulfoxide and related analogues. The solubility of the active compound of this invention in most pharmaceutically acceptable oils is quite high.
  • a cream formulation may suitably include one or more straight or branched chain, mono-or dibasic alkyl esters such as, but not limited to, di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP, the three latter being preferred esters.
  • high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns, which may be administered by rapid inhalation through the nasal passage from a container.
  • Suitable formulations wherein the carrier is a liquid, for administration as for example a nasal spray or as nasal drops include oily solutions of the active ingredient.
  • the active compound of the invention may be delivered into the systemic circulation through the vaginal mucosa from a vaginal device incorporated with a transmucosal vaginal composition.
  • Said composition may be formulated and incorporated into the device as a suppository, cream, spray, gel, film, powder, foam, ointment, microcapsules, nanocapsules or a capsule containing microparticles or nanoparticles; and said vaginal device may be a vaginal tampon, vaginal ring, vaginal strip, vaginal capsule, vaginal tablet, vaginal pessary, vaginal cup or vaginal sponge; and said composition is delivered to the vaginal mucosa by inserting said device into the vagina.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the patient; and aqueous and non-aqueous sterile suspensions which may include one or more suspending agents and/or thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or a weekly dose, as described herein, or an appropriate fraction thereof, of the active ingredient of this invention.
  • the present invention also provides controlled release pharmaceutical formulations, containing as an active ingredient the novel compound of the invention, in which the release of the active ingredient is controlled and regulated to allow less frequency dosing or to improve the pharmacokinetic or toxicity profile of said compound.
  • Controlled release formulations adapted for oral administration in which discrete units comprising the compound of the invention can be prepared according to conventional methods in the art. Additional ingredients may be included in order to control the duration of action of the active ingredient in the composition.
  • Control release compositions may thus be achieved by selecting appropriate polymer carriers such as for example polyesters, polyamino- acids, polyvinyl pyrrolidone, ethylene-vinyl acetate copolymers, methylcellulose, carboxymethylcellulose, protamine sulfate and the like.
  • the rate of drug release and duration of action may also be controlled by incorporating the active ingredient into particles, e.g. microcapsules, of a polymeric substance such as hydrogels, polylactic acid, hydroxymethylcellulose, polymethylmethacrylate and other similar polymers.
  • a polymeric substance such as hydrogels, polylactic acid, hydroxymethylcellulose, polymethylmethacrylate and other similar polymers.
  • Such compositions include colloid drug delivery systems like liposomes, microspheres, microemulsions, nanoparticles, nanocapsules and the like.
  • the pharmaceutical composition may require protective coatings.
  • the effective dosage of the compounds of present invention can be determined according to age, weight, gender, administration method, health condition and severity of a disease.
  • the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 0.1 mg/day to about 1000 mg/day. More preferably, the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 1 mg/day to about 500 mg/day. Alternatively, the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 5 mg/day to about 300 mg/day.
  • the administration times are determined by a doctor or a pharmacist to be once a day or a few times a day.
  • compositions according to the present invention include: a) from about 0.1 mg/day to about 1000 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients.
  • Another embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 1000 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients.
  • a further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 500 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients.
  • a further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 250 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients.
  • compositions according to the present invention further include: a) from about 0.1 mg/day to about 1000 mg/day of the carbonylguanidine NH, in particular the furancarbonylguanidine compound, for instance [5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients.
  • compositions a) from about 1 mg/day to about 1000 mg/day of the carbonylguanidine, in particular the furancarbonylguanidine compound, for instance [5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients.
  • compositions a) from about 1 mg/day to about 500 mg/day of the furancarbonylguanidine compound for instance [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients.
  • a further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 250 mg/day of the furancarbonylguanidine compound for instance 5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients.
  • compositions of the present invention can be administered as frequently as necessary to achieve a suitable therapeutic response in the patient being treated.
  • a first aspect of "therapeutic amount” relates to compositions which deliver a compound according to the present invention wherein the plasma level of said the furancarbonylguanidine compound for instance is from about 0.1 pg/mL to about 100 mg/mL in humans or higher mammals.
  • Another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said furancarbonylguanidine compound is from about 0.1 pg/mL to about 1 mg/mL in humans or higher mammals.
  • Another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 pg/mL to about 1 mg/mL in humans or higher mammals.
  • compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 pg/mL to about 10 ⁇ g mg/mL in humans or higher mammals.
  • compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 ng/mL to about 25 mg/mL in humans or higher mammals.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) one or more pharmaceutically acceptable excipients.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine effective for preventing synucleinopathy; b) one or more pharmaceutically acceptable excipients.
  • excipient and “carrier” are used interchangeably throughout the description of the present invention and said terms are defined herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition.”
  • excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means for achieving effective absorption by the recipient of the active ingredient.
  • An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach.
  • the formulator can also take advantage of the fact the compounds of the present invention have improved cellular potency, pharmaco-kinetic properties, as well as improved oral bioavailability.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a composition for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a composition for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of L-DOPA for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a neuroprotective amount of a compound for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
  • compositions which provide protection against synucleinopathy comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a dopaminergic effective amount of a compound for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
  • the present invention also relates to the use of a Nhe-1 inhibitor and/or pharmaceutically acceptable salts, solvates and isomers thereof of the present invention in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
  • the present invention also relates to the use of a carbonylguanidine, more particularly a furancarbonylguanidine derivative and/or pharmaceutically acceptable salts, solvates and isomers thereof of the present invention in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
  • the compounds of the present invention can be used in the manufacture of one or more medicaments, non-limiting examples of which are: i) a compound for use in the manufacture of a medicament for the treatment of Parkinson's disease; ii) a compound for use in the manufacture of a medicament for the treatment of dementia with Lewy bodies; iii) a compound for use in the manufacture of a medicament for the treatment of pure autonomic failure; iv) a compound for use in the manufacture of a medicament for the treatment of osteoporosis without the side effect of inducing multiple system atrophy.
  • KIT Present invention relates to a kit comprising an amount of a Nhe-1 inhibitor of present invention, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of Parkinson's disease, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • the kit of present invention comprises an amount of a Nhe-1 inhibitor of present invention, a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • the kit will also include directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage levels, or when titration of the individual components of the combination is desired by the prescribing physician.
  • Blister packs are well known in the packaging industry and are used widely for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally comprise a sheet of relatively rigid material covered with a foil of a preferably transparent plastic material. During the packaging process, recesses (e.g., blisters) are formed in the plastic foil. The recesses generally conform to the size and shape of the tablets or capsules to be contained therein. Next, the tablets or capsules are placed in the recesses and the sheet of relatively rigid material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses are formed.
  • recesses e.g., blisters
  • the tablets or capsules are captively retained and sealed inside the recesses between the plastic foil and the sheet.
  • the strength of the sheet is such that the tablets or capsules may be removed from the blister pack by the application of manual pressure on the outer surface of the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed through the formed opening.
  • a memory aid on the pack for example, in the form of numbers or similar indicia proximate to the tablets or capsules whereby the indicia correspond to the days of the regimen which the dosage form so specified is to be ingested.
  • a “daily dose” can be a single tablet or capsule, or multiple tablets or capsules, or tablets or capsules to be ingested on a given day.
  • a daily dose of the sodium-hydrogen exchanger type 1 (NHE-1) inhibitor can consist of a single tablet or capsule, while a daily dose of the second compound, selected from the group consisting of (a) a compliment modulator, (b) a metabolic modulator, (c) an anti- apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na +/Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, can consist of multiple tablets or capsules, and vice-versa.
  • the memory aid should reflect this.
  • a pack designed to dispense the daily doses one at a time in the order of their intended use is provided.
  • the pack is equipped with a memory aid, so as to further facilitate compliance with the dosage regimen.
  • a memory aid comprises a mechanical counter that indicates the number of daily doses to be dispensed
  • Another example of such a memory aid comprises a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date and time that the last daily dose has been taken and/or reminds the patient when the next dose is to be taken.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Present inventions demonstrates that NHE-1 inhibiting carbonylguanidine derivatives inhibit α-synuclein toxicity. Such compounds can be used in the treatment or prevention of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).

Description

SYNUCLEINOPATHIES
FIELD OF THE INVENTION
The present invention concerns compounds for inhibiting α-synuclein toxicity. Such compounds can be used in the treatment or prevention of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA).
More particular the present invention relates to sodium-hydrogen exchanger type 1 (Nhe- 1) inhibitors and most particular furancarbonylguanidine derivative compounds with Nhe-1 activity. Such compounds can be used in the treatment or prevention of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA). The invention further relates to pharmaceutical compositions, to pharmaceutical compositions using a combination, or a pharmaceutical composition of such combination, of a sodium/hydrogen exchange type-1 (Nhe-1) inhibitor and a composition for the treatment of of synucleinopathies, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), and multiple system atrophy (MSA). The invention further provides kits directed to such combinations.
BACKGROUND OF THE INVENTION
Neurodegenerative diseases are symptoms of systematic degeneration and drop out of neurons and a lot of severe diseases are known, e.g. Alzheimer's disease, Parkinson's disease, Parkinson syndrome, dementia, amyotrophic lateral sclerosis, progressive supranuclear palsy, Huntington's disease, spinocerebellar ataxia, etc. A lot of molecules are involved with the mechanism of neurodegenerative death and it is assumed that they are overexpressed or functional disorder occurs in them. However, almost nothing is known on the molecular pathology and an effective method for the treatment is not established yet.
Neurodegenerative diseases symptoms include tremor, rigidity, akinesia, bradykinesia, slow movement, postural reflex disorder, pulsion, gait disorder, depression, dysmnesia, amyotrophy, muscle weakness, dysfunctions of upper extremities, dysarthria, dysphagia, respiratory obstruction, palsy, paralysis, etc. It is well known in the art that numerous therapeutic regimens have been developed, for example, the use of compounds that inhibit the sodium/hydrogen exchange type-1 (NHE- 1) transport system. The mechanism by which NHE-1 inhibitors elicit protective effects against ischemia, particularly that affecting the myocardium, consists of a reduction in the increased sodium ion influx which is caused in reperfused/hypoperfused tissues due to intracellular acidification and subsequent activation of the sodium/hydrogen exchange transport system. This results in a delay of sodium overload of the tissue. Since sodium and calcium ion transport are coupled in cardiac tissue, this also prevents the life threatening calcium overload of myocardial cells.
The present inventions relates to the surprising finding that the inhibition if Nhe-1 affords a mechanism for treatment and prevention of synucleinopathies.
There is a long felt need in the art for compounds which have the ability to slow down the progression of synucleinopathies, and there is an urgent need in the art to develop a safe and effective therapy against synucleinopathies.
SUMMARY OF THE INVENTION
In one embodiment, the present invention relates to a furancarbonylguanidine derivative represented by the Formula (I) and/or pharmaceutically acceptable salts, solvates and isomers thereof, for use in a treatment to cure or to prevent synucleinopathy, and wherein
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
(I)
In a particular embodiment of present invention, the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-phenylfuran-2-ylcarbonyl]guanidine, - [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methoxyphenyl)furan-2-ylcarbonyI]guanidine,
- [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-aminophenyl)furan-2-yIcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyI] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyl)furan-2-ylcarbonylJguanidine,
- [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyI]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and - [5-(2-isopropoxy-5-chlorophenyl)furan-2-yIcarbonyl]guanidine.
In another embodiment, present invention also relates to a pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative represented by Formula (I) and/or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein
- Ri and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
In a particular embodiment of present invention, the pharmaceutical composition according to present invention comprises a furancarbonylguanidine derivative and/or pharmaceutically acceptable salts, solvates and isomers thereof selected from the group consisting of:
- [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-πitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furaπ-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine,
- [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-difluorophenyl)furan-2-yIcarbonyl]guanidine, - [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichIorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethyIphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and - [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine.
In another embodiment, present invention further relates to the use of a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
In a particular embodiment of present invention, the use of the compounds of present invention includes the use of particular furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluσrophenyl)furan-2-ylcarbonyl]guanidine, - [5-phenylfuran-2-ylcarbonyl]guanidine, - [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [δ-CS-chlorophenyOfuran^-ylcarbonyljguanidine,
- [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[4-(trifluoromethyl)phenyl]furan~2-ylcarbonyl]guanidine,
- [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine, - [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furaπ-2-ylcarbonyl]guanidine,
- [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and - [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine. In particular embodiment, the present invention relates to a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy and the Nhe-1 inhibitor is selected from the group consisting of: a) a NHE-1 inhibiting carbonylguanidine compound of Formula (I)
Figure imgf000009_0001
(I) a prodrug thereof, or a pharmaceutically acceptable salt, solvate or isomer of the compound or the prodrug thereof; wherein:
- Z is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination with up to three substituents independently selected from R1, R2 and R3; or
- Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5; or
- Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5; or - Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R1, R2 and R3; or
- Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R4 and R5;
Wherein R1, R2, R3, R4 and R5 are each independently hydrogen, hydroxy(CrC4)alkyl, (C1- C4)alkyl, (CrC4)alkylthio, (C3-C4)cycloalkyl, (C3-C7)cycloalkyl(CrC4)alkyl, (CrC4)alkoxy, (Ci-C4)alkoxy(Ci-C4)alkyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl, M or M(CrC4)alkyl, any of said previous (CrC4)alkyl moieties optionally having from one to nine fluorines; said (CrC4)alkyl or (C3-C4)cycloalkyl optionally mono-or di-substituted independently with hydroxy, (CrC4)alkoxy,
Figure imgf000009_0002
(Ci-C4)alkylsutfonyl, (C1- C4)alkyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl or mono-N- or CH-N1N-(C1- C4)alkylaminosulfonyl; and said (C3-C4)cycloalkyl optionally having from one to seven fluorines; wherein M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R6, R7 and R8, wherein one of R6, R7 and R8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (CrC4)alkyl and additionally R6, R7 and R8 are optionally hydroxy, nitro, halo, (Ci-C4)a!koxy, (C^C^alkoxycarbonyl, (d-C^alkyl, formyl, (Ci-C4)alkanoyl, (C1-C4)alkanoyloxy, (C-i-C4)alkanoylamino, (Ci-C4)alkoxycarbonylamino, sulfonamido, (CrC4)alkylsulfonamido, amino, mono-N- or di-N.N-tCrCOalkylamino, carbamoyl, mono-N- or di-N.N-td-GOalkylcarbamoyl, cyano, thiol,
Figure imgf000010_0001
(C1- C4)alkylsulfinyl, (CrC4)alkylsulfonyl, mono-N- or di-N.N^CrC^alkylaminosulfonyl, (C2- C4)alkenyl, (C2-C4)alkynyl or (C5-C7)cycloalkenyl, wherein said (CrC4)alkoxy, (CrC4)alkyl, (Ci-C7)alkanoyl, (CrC4)alkylthio, mono-N- or di- N,N-(CrC4)alkylamino or (C3-C7)cycloalkyl R6, R7 and R8 substituents are optionally mono- substituted independently with hydroxy, (CrC4)alkoxycarbonyl, (C3-C7)cycloalkyl, (Ci-C4)alkanoyl, (CrC^alkanoylamino, (CrC4)alkanoyloxy, (CrC^alkoxycarbonylamino, sulfonamido, (CrC^alkylsulfonamido, amino, mono-N- or di-N.N-^rC^alkylamino, carbamoyl, mono-N- or di-N.N-^-C^alkylcarbamoyl, cyano, thiol, nitro, (CrC4)alkylthio, (CrC^alkylsulfinyl, (CrC4)alkylsulfonyl or mono-N- or di-N,N-(Ci-C4)alkylaminosulfonyl or optionally substituted with one to nine fluorines; b) cariporide, or a pharmaceutically acceptable salt, solvate or isomer thereof; c) BIIB-513, or a pharmaceutically acceptable salt, solvate or isomer thereof; d) TY-12533, or a pharmaceutically acceptable salt, solvate or isomer thereof; and e) SM-15681, or a pharmaceutically acceptable salt, solvate or isomer thereof. In a preferred embodiment of present invention, the Nhe-1 inhibitor is selected from the group consisting of:
- [1 -(2-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl]guanidine; - [5-methyl-1 -(2-trif luoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-phenyl-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyl-i -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-i-phenyl-i H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-1-(2,6-dichlorophenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(quinolin-6-yl)-1 H-pyrazole-4-carbonyl]guanidine; - [5-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyl-1-(quinolin-5-yl)-1H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-i -(quinolin-8-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -phenyl-1 H-pyrazoIe-4-carbonyl]guanidine;
- [3-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine; - [3-methyl-1-(isoquinolin-5-yl)-1 H-pyrazole-4-carbonyI]guanidine;
- [2-methyl-5-phenyl-2H-pyrazole-3-carbonyl]guanidine; - [2-methyl-5-(naphthalen-1-yl)-2H-pyrazole-3-carbonyl]guanidine;
- [5-methyl-2-phenyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [5-methyl-2-(3-methoxyphenyl)-2H-1,2,3-triazole-4-carbonyl]guanidine; - [2-(3-bromophenyl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [2-(naphthalen-1 -yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [2-(isoquinolin-5-yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [5-methyl-2-(quinolin-5-yl)-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [1 -(naphthalen-1 -yO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine; - [1 -(2-chloro-4-methylsulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(2-trifluoromethyl-4-fluorophenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-bromophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-f luorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(2-chloro-5-methoxyphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-4-methylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4- carbonyl]guanidine; - [1-(2,5-dichlorophenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1-(2,3-dichlorophenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine; - [1 -(2-chloro-5-aminocarbonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(2-chloro-5-aminosulfonylphenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-f luoro-6-trif luoromethylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 ^-chloro-δ-methylsulfonylphenyO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine; - [1-(2-chloro-5-dimethylaminosulfonylphenyl)-5-cyclopropyl-1H-pyrazole-4- carbonyl]guanidine;
- [1 -(2-trifluoromethyl-4-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(8-bromoquinolin-5-yl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(β-chloroquinolin-δ-yO-δ-cyclopropyM H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-7-yl)-δ-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzimidazol-δ-yO-δ-cyclopropyM H-pyrazole-4-carbonyl]guanidine; - [1 -(1 -isoquinolyl)~δ-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [δ-cyclopropyl-1 -(4-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(indazol-6-yl)-δ-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(indazol-δ-yl)-δ-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzimidazol-δ-yΙ)-δ-ethyM H-pyrazole-4-carbonyl]guanidine; - [1-(1-methylbenzimidazol-6-yl)-δ-ethyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(δ-quinolinyl)-δ-n-propyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(δ-quinolinyl)-δ-isopropyM H-pyrazole-4-carbonyl]guanidine;
- [δ-ethyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-methylbenzimidazol-δ-yl)-δ-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(1 ,4-benzodioxan-6-yl)-δ-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzotriazol-δ-yl)-δ-ethyl-i H-pyrazoIe-4-carbonyl]guanidine;
- [1 -(3-chloroindazol-δ-yl)-δ-ethyl-1 H-pyrazole-4-carbonyI]guanidine;
- [1-(δ-quinolinyl)-δ-butyl-1 H-pyrazole-4-carbonyl]guanidine;
- [δ-propyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine; - [δ-isopropyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(indazol-7-yl)-3-methyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2,1 ,3-benzothiadiazol-4-yl)-3-methyl-1H-pyrazole-4-carbonyl]guanidine; and
- [3-methyl-1 -(quinolin-δ-yl)-i H-pyrazole-4-carbonyl]guanidine; the prodrugs thereof, and the pharmaceutically acceptable salt, solvate or isomer of said compounds and prodrugs thereof.
In another embodiment, present invention also relates to a pharmaceutical composition comprising an effective amount of a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy.
In a particular embodiment of present invention, the pharmaceutical composition comprises an effective amount of a carbonylguanidine Nhe-1 inhibitor selected from the group consisting of: a) a compound of Formula (I)
Figure imgf000012_0001
(I) a prodrug thereof, or a pharmaceutically acceptable salt, solvate or isomer of the compound or the prodrug thereof; wherein:
- Z is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination with up to three substituents independently selected from R1, R2 and R3; or
- Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5; or
- Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5; or - Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R1, R2 and R3; or
- Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R4 and R5; wherein R1, R2, R3, R4 and R5 are each independently hydrogen, hydroxy(CrC4)alkyl, (C1- C4)alkyl, (C1-C4)alkylthio, (C3-C4)cycloalkyl, (C3-C7)cycloalkyl(C1-C4)alkyl, (CrC4)alkoxy, (CrOOalkoxyCCrOOalkyl, mono-N- or di-N.N-CCrC^alkylcarbamoyl, M or M^-C^alky!, any of said previous (d-C4)alkyl moieties optionally having from one to nine fluorines; said (CrC4)alkyl or (C3-C4)cycloalkyl optionally mono-or di-substituted independently with hydroxy, (C.,-C4)alkoxy, (CrC4)alklthio, (CrC4)alkylsulfinyl, (d-C^alkylsulfonyl, (C1- C4)alkyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl or mono-N- or di-N, N-(C1- C4)alkylaminosulfonyl; and said (C3-C4)cycloalkyl optionally having from one to seven fluorines;
wherein M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R6, R7 and R8, wherein one of R6, R7 and R8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (C1-C4)alkyl and additionally R6, R7 and R8 are optionally hydroxy, nitro, halo, (C1-C4)BIkOXy, (C^C^alkoxycarbonyl, (Ci-C4)alkyl, formyl, (CrC4)alkanoyl, (Ci-C4)alkanoyloxy, (Cϊ-C^alkanoylamino, (Ci-C4)alkoxycarbonylamino, sulfonamido, (Ci-C4)alkylsulfonamido, amino, mono-N- or di-N^-^-C^alkylamino, carbamoyl, mono-N- or di-N.NHCrC^alkylcarbamoyl, cyano, thiol, (C1-C4)alkylthio, (C1- C4)alkylsulfinyl, (CrC4)alkylsulfonyl, mono-N- or di-N.N-td-C^alkylaminosulfonyl, (C2- C4)alkenyl, (C2-C4)alkynyl or (C5-C7)cycloalkenyl, wherein said (CrC4)alkoxy, (Ci-C4)alkyl, (CrC7)alkanoyl, (Ci-C4)alkylthio, mono-N- or di- N,N-(CrC4)alkylamino or (C3-C7)cycloalkyl R6, R7 and R8 substituents are optionally mono- substituted independently with hydroxy, (CrC^alkoxycarbonyl, (C3-C7)cycloalkyl, (CrC-Oalkanoyl, (C1-C4)alkanoylamino, (CrC^alkanoyloxy, (CrC^alkoxycarbonylamino, sulfonamido,
Figure imgf000014_0001
carbamoyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl, cyano, thiol, nitro, (Ci-C4)alkyfthio, (CrC^alkylsulfinyl, (C1-C4)alkylsulfonyl or mono-N- or di-N,N-(C1-C4)alkylaminosulfonyl or optionally substituted with one to nine fluorines; b) cariporide, or a pharmaceutically acceptable salt, solvate or isomer thereof; c) BIIB-513, or a pharmaceutically acceptable salt, solvate or isomer thereof; d) TY-12533, or a pharmaceutically acceptable salt, solvate or isomer thereof; and e) SM-15681, or a pharmaceutically acceptable salt, solvate or isomer thereof In a preferred embodiment of present invention, the pharmaceutical composition comprises an effective amount of a Nhe-1 inhibitor selected from the group consisting of:
- [1 -(2-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-phenyl-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyl-i -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-i-phenyM H-pyrazole-4-carbonyl]guanidine;
- [δ-cyclopropyl-1 -(2,6-dichlorophenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(quinolin-6-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyM -(quinolin-5-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-1-(quinolin-8-yl)-1H-pyrazole-4-carbonyl]guanidine; - [3-methyl-1 -phenyl-1 H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine; - [3-methyl-1-(isoquinolin-5-yl)-1H-pyrazole-4-carbonyl]guanidine; - [2-methyl-5-phenyl-2H-pyrazole-3-carbonyl]guanidine; - [2-methyl-5-(naphthalen-1 -yl)-2H-pyrazole-3-carbonyl]guanidine;
- [5-methyl-2-phenyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [5-methyl-2-(3-methoxyphenyl)-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [2-(3-bromophenyl)-5-methyl-2H-1,2,3-triazole-4-carbonyl]guanidine; - [2-(naphthalen-1-yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [2~(isoquinolin-5-yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [5-methyl-2-(quinolin-5-yl)-2H-1 ,2,3-triazole-4-carbonyI]guanidine;
- [1 -(naphthalen-1 -yO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-4-methylsulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2-trifluoromethyl-4-f luorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-bromophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-fluorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-methoxyphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 ^-chloro^-methylaminosulfonylphenyO-δ-cyclopropyl-i H-pyrazole-4- carbonyljguanidine;
- [1 -(2,5-dichlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(2,3-dichlorophenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-aminocarbonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-aminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2-fluoro-6-trifluoromethylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-methylsulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-dimethylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4- carbonyl]guanidine;
- [1 -(2-trifluoromethyl-4-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(8-bromoquinolin-5-yl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(6-chloroquinolin-5-yl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-7-yl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzimidazol-δ-yO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 -isoquinolyO-δ-cycloprOpyM H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyl-i ~(4-quinolinyI)-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(indazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(indazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzimidazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(1-methylbeπzimidazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-n-propyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(5-quinolinyI)-5-isopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-methylbenzimidazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 ,4-benzodioxan-6-yI)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(benzotriazol-5-yl)-5-ethyl-1H-pyrazo[e-4-carbonyl]guanidine; - [1 -(3-chloroindazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(5-quinolinyl)-5-butyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-propyl-1 -(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidiπe;
- [5-isopropyl-1 -(6-quinolinyI)-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-7-yl)-3-methyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2,1 ,3-benzothiadiazol-4-yl)-3-methyl-1H-pyrazole-4-carbonyI]guanidine; and
- [3-methyl-1 -(quinolin-5-yl)-1 H-pyrazole-4-carbonyl]guanidine; the prodrugs thereof, and the pharmaceutically acceptable salt, solvate or isomer of said compounds and prodrugs.
In a particular embodiment of present invention, the pharmaceutical composition further comprises a composition for the treatment of Parkinson's disease.
In a preferred embodiment of present invention, the pharmaceutical composition further comprises a composition for the treatment of Parkinson's disease comprises a compound selected from the group consisting of:
- L-DOPA; - a neuroprotective amount of a compound; and
- a dopaminergic effective amount of a compound; and further comprising a pharmaceutically acceptable carrier.
In another embodiment, present invention also relates to a kit comprising an amount of a
Nhe-1 inhibitor of present invention, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of
Parkinson's disease, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
In a particular embodiment of present invention, the kit of present invention comprises an amount of a Nhe-1 inhibitor of present invention, a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
Some embodiments of the invention are set forth in claim format directly below:
51. Embodiment 1 concerns a furancarbonylguanidine derivative represented by
Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-Cs straight or branched alkyl; or OR4; 0 - R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000017_0001
5 (I)
2. The furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof according to embodiment 1 , wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: 0 - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-phenylfuran-2~ylcarbonyl]guanidine, - [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-aminophenyI)furan-2-ylcarbonyl]guanidine,
- [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyI)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methyl-5-fluorophenyl)furan-2-yIcarbonyI]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-5-trifluoromethylphenyl)furan-2-yIcarbonyl]guanidine,
- [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and
5 - [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine.
3. The furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof according to any of embodiments 1 to 2, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
104. A pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; 15 Ci-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000020_0001
(I)
5. The pharmaceutical composition according to embodiment 4, wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [δ-β-chlorophenyOfuran^-ylcarbonyllguanidine,
- [δ-^-chlorophenyOfuran^-ylcarbonylføuanidine,
- [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine,
- [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyI)furan-2-ylcarbonyl]guanidine,
- [5-(2-phenoxyphenyl)furaπ-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-chlorophenyl)furan-2-yIcarbonyl]guanidine, - [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guaniciine,
- [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and
- [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine.
6. The pharmaceutical composition according to any of embodiments 4 to 5, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
7. Use of a furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
8. The use according to embodiment 7, wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-chIorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methylphenyI)furan-2-ylcarbonyl]guanidine, - [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[3-(trifluoromethyl)phenyI]furan-2-ylcarbonyl]guanidine,
- [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyI]guanidine,
- [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyI)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine,
- [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-difluorophenyl)furan-2-ylcarbony|]guanidine, - [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluoropheπyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-6-fluorophenyl)furan-2-yIcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyI)furan-2-ylcarbonyl]guanidine, - [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-yIcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyI]guanidine, - [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- f5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyI)furan-2-yIcarbonyl]guanidine, and
- [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine.
9. The use according to any of embodiments 7 to 8, wherein said synucleinopathy is
5 selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
Some embodiment groups of the invention are set forth in claim format directly below:
1. Group 1 concerns a Nhe-1 inhibitor, for instance a NHE-1 inhibiting carbonylguanidine, for use in a treatment to cure or to prevent synucleinopathy. 102. The Nhe-1 inhibitor of group 1 wherein said Nhe-1 inhibitor is selected from the group consisting of:
(a) a compound of Formula (I)
Figure imgf000026_0001
15 a prodrug thereof, or a pharmaceutically acceptable salt, solvate or isomer of the compound or the prodrug thereof; wherein:
- - Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5-
20 Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5
- - Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and
25 R5
Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R1, R2 and R3; or Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R4 and R5;
wherein R1, R2, R3, R4 and R5 are each independently hydrogen, hydroxy(CrC4)alkyl, (C1- C4)alkyl, (d-C^alkylthio, (C3-C4)cycloalkyl, (C3-C7)cycloalkyl(C1-C4)alkyl, (CrC4)alkoxy, (CτC4)alkoxy(CτC4)alkyl, mono-N- or di-N.N-CCrC^alkylcarbamoyl, M or M(C1-C4)alkyl, any of said previous (Ci-C4)alkyl moieties optionally having from one to nine fluorines; said (CrC4)aIkyl or (C3-C4)cycloalkyl optionally mono-or di-substituted independently with hydroxy, (CrC4)alkoxy, (d-C4)alklthio, (d-C4)alkylsulfinyl, (d-C4)alkylsulfonyl, (C1- C4)alkyl, mono-N- or di-N, N-(C1 -C4)alkylcarbamoyl or mono-N- or di-N, N-(C1- C4)alkylaminosulfonyl; and said (C3-C4)cycloalkyl optionally having from one to seven fluorines; wherein M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R6, R7 and R8, wherein one of R6, R7 and R8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (C-|-C4)alkyl and additionally R6, R7 and R8 are optionally hydroxy, nitro, halo, (C1-C4JaIkOXy, (d-C4)alkoxycarbonyl, (Ci-C4)alkyl, formyl, (Ci-C4)alkanoyl, (CrC4)alkanoyloxy, (CrC^alkanoylamino, (d-C^alkoxycarbonylamino, sulfonamido, (C1-C4)alkylsulfonamido, amino, mono-N- or di-N.N-td-C^alkylamino, carbamoyl, mono-N- or di-N.N^d-C^alkylcarbamoyl, cyano, thiol, (d-C4)alkylthio, (C1- C4)alkylsulfinyl, (d-C4)alkylsulfonyl, mono-N- or di-N,N-(C-i-C4)alkylaminosulfonyl, (C2- C4)alkenyl, (C2-C4)alkynyl or (C5-C7)cycloalkenyl, wherein said (C1-C4JaIkOXy, (Ci-C4)a!kyl, (CrC7)alkanoyl, (d-C4)alkylthio, mono-N- or di- N.N-tCrGOalkylamino or (C3-C7)cycloalkyl R6, R7 and R8 substituents are optionally mono- substituted independently with hydroxy, (C,-C4)alkoxycarbonyl, (C3-C7)cycloalkyl, (Ci-C4)alkanoyl, (CrC4)alkanoylamino, (d-C4)alkanoyloxy, (CrC4)alkoxycarbonylamino, sulfonamido, (CrC4)alkylsulfonamido, amino, mono-N- or di-N,N-(CrC4)alkylamino, carbamoyl, mono-N- or di-N,N-(C-ι-C4)alkylcarbamoyl, cyano, thiol, nitro, (CrC4)alkylthio, (Ci-C4)alkylsulfinyl, (CrC^alkylsulfonyl or mono-N- or di-N,N-(CrC4)alkylaminosulfonyl or optionally substituted with one to nine fluorines;
(b) cariporide (N-(4-lsopropyl-3-methanesulfonyl-benzoyl)-guanidine), or a pharmaceutically acceptable salt, solvate or isomer thereof;
(c) BIIB-513 (benzamide {N-(aminominomethyl)-4[4-(2-furanylcarbonyl)-
1-piperazinyl]-3-(methylsulfonyl), methanesulfonate}), or a pharmaceutically acceptable salt, solvate or isomer thereof;
(d) TY-12533 (β.Z.S.Θ-tetrahydro^-methyl-δH-cycloheptatbjpyridine-S-carbonylguanidine maleate) , or a pharmaceutically acceptable salt, solvate or isomer thereof; and
(e) SM-15681 (5-(N-ethyl-N-isopropyl)amiloride,5-(ethylisopropyl)amiloride), or a pharmaceutically acceptable salt, solvate or isomer thereof.
3. The Nhe-1 inhibitor of group 2 wherein said Nhe-1 inhibitor is selected from the group consisting of:
- [1 -(2-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-phenyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-i -(2-trif luoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [δ-cyclopropyM-phenyl-i H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyM -(2,6-dichlorophenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [δ-methyl-1 -(quinolin-6-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyl-1-(quinolin-5-yl)-1H-pyrazole-4-carbonyl]guanidine;
- [δ-cyclopropyl-1 -(quinolin-8-yl)-1 H-pyrazole-4-carbonyl]guanidine; - [3-methyl-1 -phenyl- 1H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -(isoquinolin-5-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [2-methyl-5-phenyl-2H-pyrazole-3-carbonyl]guanidine; - ^-methyl-δ-Cnaphthalen-i-yO^H-pyrazole-S-carbonyllguanidine; - [δ-methyl-2-phenyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [δ-methyl-2-(3-methoxyphenyl)-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [2-(3-bromophenyl)-5-methyl-2H-1 ,2,3-triazoIe-4-carbonyI]guaniciine; - [2-(naphthalen-1-yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [2-(isoquinolin-5-yl)-5-methyl-2H-1,2,3-triazole-4-carbonyl]guanidine; - [δ-methyl^-Cquinolin-S-yO^H-I^.S-triazole^-carbonylJguanicline; - [1 -(naphthalen-1 -yO-δ-cyclopropyM H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-4-methylsulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-trifluoromethyl-4-fluorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-bromophenyI)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(2-fluorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-methoxyphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-4-methylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4- carbonyl]guanidine;
- [1 -(2,5-dichlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2,3-dichlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-aminocarbonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(2-chloro-5-aminosulfonylphenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-fluoro-6-trifluoromethylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-methylsulfonyIphenyI)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(2-chloro-5-dimethylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4- carbonyl]guanidine; - [1-(2-trifluoromethyl-4-chlorophenyl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(8-bromoquinolin-5-yl)-5-cyclopiOpyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(δ-chloroquinolin-δ-yO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine; - [1 -(iπdazol-Z-yO-δ-cyclopropyl-i H-pyrazo!e-4-carbonyl]guanidine;
- [1 -(benzimidazol-5-yl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 -isoquinolyO-S-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyl-1-(4-quinolinyl)-1H-pyrazo!e-4-carbonyl]guanidine;
- [1-(indazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(indazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzimidazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 -methylbenzimidazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinoliny!)-5-n-propyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-isopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [5-ethyl-1 -(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-methylbenzimidazol-5-yl)-5-ethyl-i H-pyrazole-4-carbonyl]guanidine; - [1 -(1 ,4-benzodioxan-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(benzotriazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(3-chloroindazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-butyl-1 H-pyrazole-4-carbonyl]guanidine; 5 - [5-propyl-1 -(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-isopropyl-1-(6-quinolinyl)-1H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-7-yl)-3-methyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2,1,3-benzothiadiazol-4-yI)-3-methyl-1H-pyrazole-4-carbonyl]guanidine; and
- [3-methyl-1 -(quinolin-5-yI)-1 H-pyrazole-4-carbonyl]guanidine;
10 the prodrugs thereof, and the pharmaceutically acceptable salt, solvate or isomer of said compounds and prodrugs thereof.
4. The Nhe-1 inhibitor of any of groups 1 to 3 wherein said synucleinopathy is selected from the group consisting of Parkinson's disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy. 155. A pharmaceutical composition comprising an effective amount of a Nhe-1 inhibitor for use in a treatment to cure or to prevent synucleinopathy. 6. The pharmaceutical composition of group 5 wherein said Nhe-1 inhibitor is selected from the group consisting of:
(a) a compound of Formula (I) 20
Figure imgf000030_0001
a prodrug thereof, or a pharmaceutically acceptable salt, solvate or isomer of the compound or the prodrug thereof; wherein:
25 Z is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5
- Z is a five-membered rings with as single heteroatom nitrogen with Z being of the group 30 of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole which Z can be mono- or di-substituted with up to two substituents independently selected from R4 and R5
- Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three
5 substituents independently selected from R1, R2 and R3; or
- Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R4 and R5;
wherein R1, R2, R3, R4 and R5 are each independently hydrogen, hydroxy(CrC4)alkyl, (C1- 0 C4)alkyl, (CrC4)alkylthio, (C3-C4)cycloalkyl, (C3-C7)cycloalkyl(C1-C4)alkyl, (CrC4)alkoxy, (CrC4)alkoxy(Ci-C4)alkyl, mono-N- or di-N.N-^-OOalkylcarbamoyl, M or M(C1-C4)alkyl, any of said previous (C-ι-C4)alkyl moieties optionally having from one to nine fluorines; said (C1-C4)alkyl or (C3-C4)cycloalkyl optionally mono-or di-substituted independently with hydroxy, (C1-OOaIkOXy, (CrC4)alklthio, (CrC4)alky!sulfinyl,
Figure imgf000031_0001
(C1- 5 C4)alkyl, mono-N- or di-N,N-(C1-C4)alkylcarbamoyl or mono-N- or di-N,N-(Cr C4)alkylaminosulfonyl; and said (C3-C4)cycloalkyl optionally having from one to seven fluorines; wherein M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from 0 oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both 5 rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R6, R7 and R8, wherein one of R6, R7 and R8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (C^C^alkyl and additionally R6, R7 and R8 are 0 optionally hydroxy, nitro, halo, (CrC4)alkoxy, (CrC4)alkoxycarbonyl, (C1-C4)alkyl, formyl, (CrC^alkanoyl, (C^C^alkanoyloxy, (CrC^alkanoylamino, (CrC^alkoxycarbonylamino, sulfonamido, (CrC^alkylsulfonamido, amino, mono-N- or di-N.N-^rC^alkylamino, carbamoyl, mono-N- or di-N.N-^rC^alkylcarbamoyl, cyano, thiol, (CrC4)alkylthio, (C1- C4)alkylsulfinyl, (Ci-C4)alkylsulfonyl, mono-N- or di-N,N-(Ci-C4)alkylaminosulfonyl, (C2- 5 C4)alkenyl, (C2-C4)alkynyl or (C5-C7)cycloalkenyl, wherein said (d-C4)alkoxy, (CrC4)alkyl, (CrC7)alkanoyl, (CrC4)alkylthio, mono-N- or di- N,N-(C1-C4)alkylamino or (C3-C7)cycloalkyl R6, R7 and R8 substituents are optionally mono- substituted independently with hydroxy, (C^^Jalkoxycarbonyl, (C3-C7)cycloalkyl, (Ci-C4)alkanoyl, (CrC4)alkanoylamino, (CrC4)alkanoyloxy, (Ci-C4)alkoxycarbonylamino, sulfonamido, (d^Jalkylsulfonamido, amino, mono-N- or di-N,N-(C1-C4)alkylamino, carbamoyl, mono-N- or di-N, N-(C1 -C4)alkylcarbamoyl, cyano, thiol, nitro, (d-C/Oalkylthio, (CrC4)alkylsulfinyl, (C1-C4)alkylsulfonyl or mono-N- or di-N,N-(Ci-C4)alkylaminosulfonyl or optionally substituted with one to nine fluorines;
(b) cariporide, or a pharmaceutically acceptable salt, solvate or isomer thereof;
(c) BIIB-513, or a pharmaceutically acceptable salt, solvate or isomer thereof;
(d) TY-12533, or a pharmaceutically acceptable salt, solvate or isomer thereof; and
(e) SM-15681 , or a pharmaceutically acceptable salt, solvate or isomer thereof.
7. The pharmaceutical composition according to any of groups 5 to 6, wherein said pharmaceutical composition is selected from the group consisting of:
- [1 -(2-chlorophenyl)-5-methyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-phenyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyM -(2-trifluoromethylphenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyM -phenyl-1 H-pyrazole-4-carbonyl]guanidine; - [5-cyclopropyM -(2,6-dichlorophenyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(quinolin-6-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyM -(quinolin-5-yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [5-cyclopropyM -(quinolin-8-yl)-1 H-pyrazole-4-carbonyl]guanidine; - [3-methyl-1 -phenyl-1 H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -(naphthalen-1 -yl)-1 H-pyrazole-4-carbonyl]guanidine;
- [3-methyl-1 -(isoquinolin-5-yl)-1 H-pyrazole-4-carbonyl]guanidine; - [2-methyl-5-phenyl-2H-pyrazole-3-carbonyl]guanidine;
- [2-methyl-5-(naphthalen-1-yl)-2H-pyrazole-3-carbonyl]guanidine; - [5-methyl-2-phenyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine;
- [5-methyl-2-(3-methoxyphenyl)-2H-1,2,3-triazole-4-carbonyl]guanidine; - [2-(3-bromophenyl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [2-(naphthalen-1 ~yl)-5-methyl-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [2-(isoquinolin-5-yl)-5-methyl-2H-1 ,2,3-tria2ole-4-carbonyl]guanidine; - [5-methyl-2-(quinolin-5-yl)-2H-1 ,2,3-triazole-4-carbonyl]guanidine; - [1-(naphthalen-1-yl)-5-cyclopropyl-1H-pyrazole-4-carbonyl]guanidine; - [1 -(2-chloro-4-methylsulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyi]guanidine;
- [1 -(2-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-trifluoromethyl-4-fluorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-bromophenyI)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-fluorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2-chloro-5-methoxyphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-4-methylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazoIe-4- carbonyljguanidine;
- [1 -(2,5-dichlorophenyI)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2,3-dichlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2-chloro-5-aminocarbonylphenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-aminosulfonylphenyl)-5-cyclopropyI-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-f luoro-θ-trifluoromethylphenyO-S-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-methylsulfonylphenyl)-5-cyclopropyI-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(2-chloro-5-dimethylaminosulfonylphenyl)-5-cyclopropyl-1 H-pyrazole-4- carbonyl]guanidine;
- [1 -(2-trifluoromethyl-4-chlorophenyl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(δ-bromoquinolin-δ-yO-S-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(β-chloroquinolin-S-yO-S-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-7-yl)-5-cyclopropyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(benzimidazol-δ-yO-δ-cyclopropyl-i H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 -isoquinolyO-δ-cyclopropyM H-pyrazole-4-carbonyi]guanidine;
- [5-cyclopropyl-i -(4-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(indazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1-(benzimidazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(1 -methylbenzimidazol-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-n-propyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-isopropyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-ethyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine; - [1 -(2-methylbenzimidazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(1 ,4-benzodioxan-6-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine; - [1 ~(benzotriazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(3-chloroindazol-5-yl)-5-ethyl-1 H-pyrazole-4-carbonyl]guanidine;
- [1 -(5-quinolinyl)-5-butyl-1 H-pyrazole-4-carbonyl]guanidine;
- [5-propyl-1-(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine; - [5-isopropyl-1 -(6-quinolinyl)-1 H-pyrazole-4-carbonyl]guanidine;
- [1-(indazol-7-yl)-3-methyl-1 H-pyrazo!e-4-carbonyl]guanidine;
- [1 -(2,1 ,3-benzothiadiazol-4-yl)-3-methyl-1H-pyrazole-4-carbonyl]guanidine; and
- [3-methyl-1 -(quinolin-5-yl)-1 H-pyrazoIe-4-carbonyl]guanidine; the prodrugs thereof, and the pharmaceutically acceptable salt, solvate or isomer of said compounds and prodrugs.
8. The pharmaceutical composition according to any of groups 5 to 7, wherein said synucleinopathy is selected from the group consisting of Parkinson's disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy
9. The pharmaceutical composition according to any of groups 5 to 8, further comprising a composition for the treatment of Parkinson's disease.
10. The pharmaceutical composition according to group 9, further comprising a composition for the treatment of Parkinson's disease, wherein said composition for the treatment of Parkinson's disease comprises a compound selected from the group consisting of: - L-DOPA;
- a neuroprotective amount of a compound; and
- a dopaminergic effective amount of a compound; and a pharmaceutically acceptable carrier. v
11. A kit comprising an amount of a Nhe-1 inhibitor according to group 1, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of Parkinson's disease, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
12. The kit according to group 11 , wherein said composition for the treatment of Parkinson's disease is selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container. These and other objects, features, and advantages will become apparent to those of ordinary skill in the art from a reading of the following detailed description and the appended claims.
Some embodiment groups of the invention are set forth in claim format directly below:
1. A furancarbonylguanidine NHE-1 inhibitor represented by Formula (IV) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathv. and wherein : - R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; CrC5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000035_0001
(IV)
in which the cyclic group (A) presented by Formula (II)
Figure imgf000035_0002
(ll) is a 3 to 8 membered ring.
and wherein the cyclic group (B) represented by the Fomula (V)
Figure imgf000036_0001
(V)
Is:
- is an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination; or
- is a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5- Dihydrofuran) and unsaturated furan; or
- is a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole; or
- is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens; or
- is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring;
2. The furancarbonylguanidine derivative of claim 13, wherein the cyclic group presented by Formule (II) is an aromatic ring for instance an aryl.
3. The furancarbonylguanidine derivative of claim 13, wherein the cyclic group presented by Formule (II) is a carbocycle. 4. A pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative of any of the previous claims 13 to 15 or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy. 5. Any of the previous claims 13 to 16, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
6. Any of the previous claims 13 to 17, wherein said furancarbonylguanidine derivative is of the group consisting of cariporide (N-(4-lsopropyl-3-methanesulfonyl- benzoyl)-guanidine), BIIB-513 (benzamide {N-(aminominomethyl)-4[4-(2- furanylcarbonyl)- 1-piperazinyl]-3-(methylsulfonyl), methanesulfonate}), TY-12533 (β.y.δjθ-tetrahydro^-methyl-δH-cycloheptal^pyridine-S-carbonylguanidine maleate) and SM-15681 (5-(N-ethyl-N-isopropyl)amiloride,5-(ethylisopropyl)amiloride) or a pharmaceutically acceptable salt, solvate or isomer thereof.
Each of the claims set out a particular embodiment of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention will become more fully understood from the detailed description given herein below and the accompanying drawings which are given by way of illustration only, and thus are not limitative of the present invention.
Figure 1 shows the growth of a wild type yeast strain or a nud deletion mutant without or with expression of human EndoG as expressed by the Optical Density (OD600nm relative units). Samples were taken at 3 days after inuculation of the cultures. The values shown represent mean + SEM.
Figure 2 shows the growth of wild type yeast cells expressing human α-synuclein and human EndoG on selective medium without or supplemented with of [5-(2-Methyl-5- Fluorophenyl)furan-2-ylcarbonyl]guanidine (Sigma-Aldrich K4389) at the concentrations indicated as expressed by the Optical Density (ODΘOOnm relative units). The values shown represent mean + SEM.
Figure 3 concerns the effect of compound K4389 on a-synuclein aggregation and late apoptosis. It shows: (A) Effect of different concentrations of [5-(2-Methyl-5- fluorophenyl)furan-2-ylcarbonyl]guanidine (Sigma-Aldrich K4389) on α-Synuclein aggregation in SHSY5Y cells induced by oxidative stress. Stock concentrations in DMSO are diluted 100x until final concentration shown on graph. D : p<0.01 (Kruskal-Wallis test followed by Dunnet's post hoc test). (B) Effect of different concentrations of K4389 on nucleus condensation, a marker for late apoptosis. All percentages are relative to negative control condition: DMSO, 100%.
Figure 4: concerns the inhibition of the yeast homolog of NHE-1 decreases alpha- synuclein toxicity. It displays (A) Quantification of ROS-accumulation using the conversion of non-fluorescent dihydroethidium (DHE) to fluorescent ethidium (Eth) in wild type yeast cells expressing human alpha-synuclein (αSyn) for 48 h or harbouring the empty vector control. Cells were treated with indicated concentrations of the NHE-1 inhibitor K4389 (solved in DMSO) or equivalent concentrations of DMSO alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control. (B) Quantification of ROS-accumulation in wild type yeast cells expressing human alpha-synuclein for 16 h (αSyn) or harbouring the empty vector control upon treatment with 0.25 mM of the NHE-1 inhibitor K4389, Synonym: [5-(2-Methyl-5- fluorophenyl)furan-2-ylcarbonyl]guanidine / CAS Number: 852146-73-7 / Linear Formula: C13H12FN3O2, olved in MeOH) and equivalent concentration of MeOH alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control. (C) Quantification of ROS-accumulation in wild type cells and cells lacking the homolog of human NHE-1 (Δnhxi) expressing human alpha-synuclein (αSyn) for 16 h or harbouring the empty vector control. Positive stained cells were quantified using flow cytometry.
DETAILED DESCRIPTION OF THE INVENTION
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. All patents, applications, published applications and other publications are incorporated by reference in their entirety. In the event that there is a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.
"Aliphatic" as used herein refers to a group selected from the set of "Alkyl", "Alkenyl" and "Alkynyl".
"Saturated aliphatic" as used herein refers to "Alkyl" "Unsaturated aliphatic" as used herein refers to a group selected from the set of "Alkenyl" and "Alkynyl".
"Alkyl", as used herein, refers to a straight or branched chain hydrocarbon containing from 1 or 2 to 10 or 20 or more carbon atoms (e.g., C2, C3, C4, C5, C6, C7, C8, C9, CIO, Cl 1, C12, C13, C14, C15, etc.). Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n- heptyl, n-octyl, n- nonyl, n-decyl, and the like. In some embodiments, alkyl groups as described herein are optionally substituted (e.g., from 1to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
The term "optionally substituted" indicates that the specified group is either unsubstituted, or substituted by one or more suitable substituents. A "substituent" is an atom or atoms substituted in place of a hydrogen atom on the parent chain or cycle of an organic molecule, for example, H, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
"Alkenyl," as used herein, refers to a straight or branched chain hydrocarbon containing from 1 or 2 to 10 or 20 or more carbons, and containing at least one carbon-carbon double bond, formed structurally, for example, by the replacement of two hydrogens. Representative examples of "alkenyl" include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl-l-heptenyl, 3- decenyl and the like. In some embodiments, alkenyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
"Alkynyl," as used herein, refers to a straight or branched chain hydrocarbon group containing from 1 or 2 to 10 or 20 or more carbon atoms, and containing at least one carbon- carbon triple bond. Representative examples of alkynyl include, but are not limited, to acetylenyl, 1-propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, 1-butynyl and the like. In some embodiments, alkynyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloaliphatic, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
The term "cycloaliphatic", as used herein, refers to a saturated or unsaturated cyclic hydrocarbon group containing from 3 to 8 carbons or more, which is not aromatic. Representative examples of cycloalkyl include, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some embodiments, cycloalkyl groups as described herein are optionally substituted (e.g., from 1 to 3 or 4 times) with independently selected H, halo, hydroxy, acyl, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclo, aryl, heteroaryl, alkoxy, amino, amide, thiol, sulfone, sulfoxide, oxo, oxy, nitro, carbonyl, carboxy, amino acid sidechain, amino acid and peptide.
"Heterocyclic", as used herein, refers to a monocyclic or a bicyclic ring system.
Monocyclic heterocyclic ring systems are exemplified by any 5 or 6 member ring containing 1 , 2, 3, or 4 heteroatoms independently selected from the group consisting of: O, N, and S. The 5 member ring has from 0 to 2 double bonds, and the 6 member ring has from 0-3 double bonds. Representative examples of monocyclic ring systems include, but are not limited to, azetidine, azepine, aziridine, diazepine, 1 ,3-dioxolane, dioxane, dithiane, furan, imidazole, imidazoline, imidazolidine, isothiazole, isothiazoline, isothiazolidine, isoxazole, isoxazoline, isoxazolidine, morpholine, oxadiazole, oxadiazoline, oxadiazolidine, oxazole, oxazoline, oxazolidine, piperazine, piperidine, pyran, pyrazine, pyrazole, pyrazoline, pyrazolidine, pyridine, pyrimidine, pyridazine, pyrrole, pyrroline, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, tetrazine, tetrazole, thiadiazole, thiadiazoline, thiadiazolidine, thiazole, thiazoline, thiazolidine, thiophene, thiomorpholine, thiomorpholine sulfone, sulfoxide, thiopyran, triazine, triazole, trithiane, and the like. Bicyclic ring systems are exemplified by any of the above monocyclic ring systems fused to an aryl group as defined herein, a cycloalkyl group as defined herein, or another monocyclic ring system as defined herein. Representative examples of bicyclic ring systems include but are not limited to, for example, benzimidazole, benzothiazole, benzothiadiazole, benzothiophene, benzoxadiazole, benzoxazole, benzofuran, benzopyran, benzothiopyran, benzodioxine, 1 ,3- benzodioxole, cinnoline, indazole, indole, indoline, indolizine, naphthyridine, isobenzofuran, isobenzothiophene, isoindole, isoindoline, isoquinoline, phthalazine, pyranopyridine, quinoline, quinolizine, quinoxaline, quinazoline, tetrahydroisoquinoline, tetrahydroquinoline, thiopyranopyridine, and the like.
"Aromatic" as used herein refers to a fused ring system having one or more aromatic rings. Representative examples of aryl include, azulenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like. The aromatic groups of this invention can be substituted with 1 , 2, 3, 4, or 5 substituents independently selected from alkenyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylsulfinyl, alkylsulfonyl, alkylthio, alkynyl, aryl, aryloxy, azido, arylalkoxy, arylalkyl, aryloxy, carboxy, cyano, formyl, halogen, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, mercapto, nitro, sulfamyl, sulfo, sulfonate, -NR1R" (wherein, R1 and R" are independently selected from hydrogen, alkyl, alkylcarbonyl, aryl, arylalkyl and formyl), and -C(O)NRIR" (wherein R' and R" are independently selected from hydrogen, alkyl, alkylcarbonyl, aryl, arylalkyl, and formyl). "Homoaromatic" means a cyclic, aromatic hydrocarbon in which no carbon atoms have been replaced with heteroatoms. The homoaromatic group can be unsubstituted or substituted with from 1 to 5 suitable substituents.
"Heteroaromatic" means a cyclic, aromatic hydrocarbon in which one or more carbon atoms have been replaced with heteroatoms. If the heteroaromatic group contains more than one heteroatom, the heteroatoms may be the same or different. Examples of heteroaryl groups include pyridyl, pyrimidinyl, imidazolyl, thienyl, furyl, pyrazinyl, pyrrolyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, indolyl, isoindolyl, indolizinyl, triazolyl, pyridazinyl, indazolyl, purinyl, quinolizinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinoxalinyl, isothiazolyl, and benzo[b]thienyl. Preferred heteroaryl groups are five and six membered rings and contain from one to three heteroatoms independently selected from the group consisting of: O, N, and S. The heteroaryl group, including each heteroatom, can be unsubstituted or substituted with from 1 to 4 suitable substituents, as chemically feasible. For example, the heteroatom S may be substituted with one or two oxo groups, which may be shown as =0.
As used herein, α-synuclein refers to one in a family of structurally related proteins that are prominently expressed in the central nervous system. Aggregated α-synuclein proteins form brain lesions that are hallmarks of some neurodegenerative diseases (synucleinopathies). The gene for α-synuclein, which is called SNCA, is on chromosome 4q21. Alpha-synuclein is a member of the synuclein family, which also includes beta- and gamma-synuclein. Synucleins are abundantly expressed in the brain and alpha- and beta- synuclein inhibit phospholipase D2 selectively. Ueda et al. (Ueda, K.; et al. Proc. Nat. Acad. Sci. 90: 11282-11286, 1993) isolated an apparently full-length cDNA encoding a 140-amino acid protein within which 2 previously unreported amyloid sequences were encoded in tandem in the mouse hydrophobic domain. Campion, D.; et al. Genomics 26: 254-257, 1995. cloned 3 alternatively spliced transcripts in lymphocytes derived from a normal subject, while Jakes, R.;et al (FEBS Lett. 345: 27-32, 1994) identified two distinct synucleins from human brain and Beyer, K.; et al. (Neurogenetics 9: 15-23, 2008.) identified and characterized a new alpha-synuclein isoform and its role in Lewy body diseases. These defined structures are hereby incorporated into the definition of α- synuclein.
"Aza" is in the meaning of containing nitrogen in place of carbon.
The term "synucleinopathies" is used to name a group of neurodegenerative disorders characterized by fibrillary aggregates of alpha-synuclein protein in the cytoplasm of selective populations of neurons and glia. These disorders include Parkinson's disease (PD), dementia with Lewy bodies (DLB), pure autonomic failure (PAF), multiple system atrophy (MSA), neurodegeneration with brain iron accumulation, type 1 (e.g. Hallervorden- Spatz syndrome, neuroaxonal dystrophy), and other diseases that may have synuclein- immunoreactive lesions (e.g. Traumatic brain injury, Pick disease, Amyotrophic lateral sclerosis). Clinically, they are characterized by a chronic and progressive decline in motor, cognitive, behavioural, and autonomic functions, depending on the distribution of the lesions. Because of clinical overlap, differential diagnosis is sometimes very difficult. The term "α-synuclein toxicity associated disease" refers to "synucleinopathies" and is used as a synonym.
Multiple system atrophy (MSA) is a sporadic neurodegenerative disorder that encompasses olivopontocerebellar atrophy (OPCA), striatonigral degeneration (SND) and Shy-Drager syndrome (SDS). The formation of alpha-synuclein aggregates is a critical event in the pathogenesis of multiple system atrophy (MSA). The histopathological hallmark is the formation of α-synuclein-positive glial cytoplasmic inclusions (GCIs) in oligodendroglia. α-synuclein aggregation is also found in glial nuclear inclusions, neuronal cytoplasmic inclusions (NCIs), neuronal nuclear inclusions (NNIs) and dystrophic neuritis. Parkinson's disease (PD) is the second most common age-associated neurodegenerative disease. Parkinson's disease can e.g. be sporadic, familial with α-synuclein mutations, or familial with mutations other than α-synuclein. Several observations in the art suggest malfunctioning of the protein α-synuclein to be a toxic trigger of the neurodegenerative process during PD. In addition, three missense mutations (A30P, A53T and E46K) in the α-synuclein gene are linked to early-onset dominant familial PD. More recently, overexpression of wild-type α-synuclein due to gene duplication or triplication was found to be sufficient to cause a familial form of PD.
Dementia with Lewy bodies is the second most frequent cause of hospitalization for dementia, after Alzheimer's disease. Current estimates are that about 60-to-75% of diagnosed dementias are of the Alzheimer's and mixed (Alzheimer's and vascular dementia) type, 10-to-15% are Lewy Bodies type, with the remaining types being of an entire spectrum of dementias including frontotemporal lobar degeneration, alcoholic dementia, pure vascular dementia. Dementia with Lewy bodies includes e.g. "pure" Lewy body dementia, Lewy body variant of Alzheimer disease, familial Alzheimer disease with APP mutations, familial Alzheimer disease with PS-1 mutations, familial Alzheimer disease with PS-2 mutations, and Down syndrome.
Pure autonomic failure, also known as Bradbury-Eggleston syndrome or idiopathic orthostatic hypotension, is a form of dysautonomia that first occurs in middle age or later in life; men are affected more often than women. It is one of three diseases classified as primary autonomic failure. The symptoms concern a degenerative disease of the peripheral nervous system, symptoms include dizziness and fainting (caused by orthostatic hypotension), visual disturbances and neck pain. Chest pain, fatigue and sexual dysfunction are less common symptoms that may also occur. Symptoms are worse when standing; sometimes one may relieve symptoms by laying down. Accumulation of alpha-synuclein in autonomic nerves causes pure autonomic failure (Horacio Kaufmann et al. Neurology 2001 ;56:980-981).
The present invention addresses the following unmet medical needs, inter alia; 1) providing compounds and pharmaceutical compositions capable of effectively controlling, in particular preventing or treating synucleinopathy such as Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy. 2) providing compounds and pharmaceutical compositions capable of effectively controlling, in particular preventing or treating synucleinopathy such as but not limited to Parkinson's disease.
Methods for the preparation of particular furancarbonylguanidine derivative and/or pharmaceutically acceptable salts, solvates and isomers for use of present invention are selected from the group consisting of:
- [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [δ-CS-chlorophenyOfuran^-ylcarbonyriguanidine,
- [5-(4-chlorophenyl)furan-2-ylcarbony!]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine, - [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine,
- [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethylphenyl)furan-2-y!carbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and
- [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, have been disclosed in detail in WO 2005/063727.
The compounds of Formula :
Figure imgf000045_0001
a prodrug thereof, or a pharmaceutically acceptable salt, solvate or isomer of the compound or the prodrug thereof; wherein: Z is carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R1, R2 and R3; or Z is carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R4 and R5; wherein R1, R2, R3, R4 and R5 are each independently hydrogen, hydroxyCCrC^alkyl, (C1- C4)alkyl, (d-C^alkylthio, (C3-C4)cycloalkyl, (C3-C7)cycloalkyl(d-C4)alkyl, (d-C4)alkoxy, (Ci-C4)alkoxy(Ci-C4)alkyl, mono-N- or di-N.N^d-dJalkylcarbamoyl, M or M(C1-C4)alkyl, any of said previous (d-C4)alkyl moieties optionally having from one to nine fluorines; said (d-d)alkyl or (C3-C4)cycloalkyl optionally mono-or di-substituted independently with hydroxy, (CrC4)alkoxy, (C1-C4JaIkItNo, (CrC4)alkylsulfinyl, (CrC4)alkylsulfonyl, (C1- C4)alkyl, mono-N- or di-N,N-(C-ι-C4)alkylcarbamoyl or mono-N- or di-N, N-(C1- C4)alkylaminosu!fonyl; and said (C3-C4)cycloalkyl optionally having from one to seven fluorines; wherein M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R6, R7 and R8, wherein one of R6, Rr and R8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (CrC4)alkyl and additionally R6, R7 and R8 are optionally hydroxy, nitro, halo, (d~C4)alkoxy, (d-C4)alkoxycarbonyl, (C1-C4)alkyl, formyl, (d-C4)alkanoyl, (d-C4)alkanoyloxy, (d-C4)alkanoyIamino, (d-C4)alkoxycarbonylamino, sulfonamido, (d-C4)alkylsulfonamido, amino, mono-N- or di-N,N-(d-C4)alkylamino, carbamoyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl, cyano, thiol, (C1-C4)alkylthio, (C1- C4)alkylsulfinyl, (CrC4)alkylsulfonyl, mono-N- or di-N,N-(d-C4)alkylaminosulfonyl, (C2- C4)alkenyl, (C2-C4)alkynyl or (C5-C7)cycloalkenyl, wherein said (CrC4)alkoxy, (Ci-C4)alkyl, (CrC7)alkanoyl, (d-C4)alkylthio, mono-N- or di- N.N-td-C^alkylamino or (C3-C7)cycloalkyl R6, R7 and R8 substituents are optionally mono- substituted independently with hydroxy, (d-C4)alkoxycarbonyl, (C3-C7)cycloalkyl, (d-C4)alkanoyl, (d-C4)alkanoylamino, (CrC4)alkanoyloxy, (d-C4)alkoxycarbonylamino, sulfonamido, (CrC4)alkylsulfonamido, amino, mono-N- or di-N,N-(d-C4)alkylamino, carbamoyl, mono-N- or di-N,N-(CrC4)alkylcarbamoyl, cyano, thiol, nitro, (CrC4)alkylthio, (Ci-C4)alkylsulfinyl, (C1-C4)alkylsulfonyi or mono-N- or di-N,N-(CrC4)alkylaminosulfonyl or optionally substituted with one to nine fluorines;
(b) cariporide, or a pharmaceutically acceptable salt, solvate or isomer thereof; (c) BIIB-513, or a pharmaceutically acceptable salt, solvate or isomer thereof; (d) TY-12533, or a pharmaceutically acceptable salt, solvate or isomer thereof; and (e) SM-15681 , or a pharmaceutically acceptable salt, solvate or isomer thereof, the prodrugs thereof, and the pharmaceutically acceptable salts, solvates or isomers of the compounds and prodrugs, may be prepared as disclosed in the aforementioned, commonly-assigned PCT International Application Publication No. WO 99/43663, the disclosure of which is incorporated herein by reference.
The preferred NHE-1 inhibitor cariporide, i.e. N-(aminoiminomethyl)-4-(1-methylethyl)-3- (methylsulfonyl)-benzamide, may be prepared as disclosed in U.S. Pat. No. 5,591 ,754, the disclosure of which is incorporated herein by reference. The preferred NHE-1 inhibitor BIIB-513, i.e. N-(aminoiminomethyl)4-(4-(2-furanylcarbonyl)-1-piperazinyl)-3- (methylsulfonyl)-benzamide, may be prepared as disclosed in U.S. Pat. No. 6,114,335, the disclosure of which is incorporated herein by reference. The preferred NHE-1 inhibitor TY-12533, i.e. δ.T.δ.θ-tetrahydro^-methyl-δH-cycloheptaEbJpyridine-S-carbonylguanidine, may be prepared as disclosed in PCT International Application Publication No. WO 98/39300, the disclosure of which is incorporated herein by reference. The preferred NHE- 1 inhibitor SM-15681, i.e. N-(aminoiminomethyl)-1-methyl-1H-indole-2-carboxamide, may be prepared as disclosed in EPO 0 708 091 , the disclosure of which is incorporated herein by reference.
The ability of a compound to function as an NHE-1 inhibitor may be determined according to the protocols described in detail herein below.
Example 1 - A yeast strain for identifying compounds that reduce α-synuclein toxicity A yeast strain suitable for studying synucleinopathy was created. In particular, a wild type BY4741 yeast strain (MATa his3Ai leu2Δ0 met15AO ura3Δ0) obtained from Euroscarf was transformed with a plasmid allowing for constitutive expression of human α-synuclein, as well as with human endonuclease G which is an effector of the α-synuclein-induced toxic effect.
Here we show that when overexpressed the human endonuclease G can complement its yeast orthologue Nuc1 as demonstrated by reduced growth in wild type cells and alleviation of the growth defect of yeast mutant cells lacking Nuc1 (Figure 3). The combined expression of human α-synuclein and human endonuclease G in wild type yeast cells provides a system that allows selection or identification of compounds that potentially reduce toxicity mediated by these proteins.
Example 2 - r5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyllguanidine reduces α- synuclein toxicity in yeast
Experiments are carried out in BY4741 (MATa his3A1 leu2Δ0 met15Δ0 ura3Δ0) expressing human α-synuclein and human endonuclease G. The combined expression of human α-synuclein and human endonuclease G in wild type yeast cells provides a system that allows selection or identification of compounds that potentially reduce toxicity mediated by these proteins. All strains are grown on SC medium containing 0.17% yeast nitrogen base (Difco), 0.5% (NhU)2SO4 and 30 mg/l of all amino acids (except 80 mg/l histidine and 200 mg/l leucine), 30 mg/l adenine, and 320 mg/l uracil with 2% glucose (SCD). The above mentioned SC medium was without or supplemented with [5-(2-methyl- 5-fluorophenyl)furan-2-ylcarbonyl]guanidine at concentrations of 0.02, 0.2, 2.0, 20.0 μM. Our data surprisingly show that already at micromolar concentrations this compound improves the growth of yeast cells expressing both human proteins on minimal selective medium (Figure 1). Example 3 - Measurement of human Nhe-1 inhibitory activity Methodologies for measurement of human Nhe-1 activity and inhibitor potency are predicated on those published by Watson et al., Am. J. Physiol., 24;G222g-G238, 1991), where Nhe-1 mediated recovery of intracellular pH is measured following intracellular acidification. Thus, fibroblasts stably expressing human Nhe-1 (Counillon, L. et al., MoI. Pharmacol., 44:1041-1045 (1993) are plated onto collagen coated 96 well plates (50,000/well) and grown to confluence in growth media (DMEM high glucose, 10% fetal bovine serum, 50 u/ml penicillin and streptomycin). Confluent plates are incubated for 30 min at 37° C. with the pH sensitive fluorescent probe BCECF (5 μM; Molecular Probes, Eugene, Oreg.). BCECF loaded cells are incubated for 30 min at 37° C. in acid loading media (70 mM choline chloride, 50 mM NHCI 4, 5 mM KCI, 1 mM MgCI 2, 1.8 mM CaCI 2, 5 mM glucose, 10 mM HEPES, pH 7.5), and then placed in a Fluorescent Imaging Plate Reader (Molecular Devices, CA). BCECF fluorescence is monitored using excitation and emission wavelengths of 485 nM and 525 nM, respectively. Intracellular acidification is initiated via rapid replacement of acid loading media with recovery media (120 mM NaCl, 5 mM KCI, 1 mM MgCI 2, 1.8 mM CaCI 2, 5 mM glucose, 10 mM HEPES, pH 7.5)±test compound, and Nhe-mediated recovery of intracellular pH is monitored as the subsequent time-dependent increase BCECF fluorescence. The potency of human Nhe-1 inhibitors is calculated as the concentration that reduces recovery of intracellular pH by 50% (IC50). Example 4 - Assessment of a yeast-based model in high-throughput screens: selection of Nhe-1 inhibitors that suppress α-svnuclein toxicity in yeast. The usability of the yeast model expressing human α-synuclein and human endonuclease G is examined in high-throughput screens. To this end, 500 compounds are tested individually to address their capacity to modulate α-synuclein toxicity in yeast cells. Yeast cells are grown in 96-well microtiter plates and growth is monitored by measuring OD at 600 nm. The compounds are dissolved in DMSO and added to the growth medium at a final concentration of 10 μg/ml. Every microtiter plate contains positive and negative controls for calculating the Z'-factor in order to assess the quality of the assay of each microtiter plate. Positive and negative growth controls are wild-type yeast cells transformed with α-synuclein and human endonuclease G, respectively. 500 compounds are screened in duplicate (screen 1 and 2). The ODβoo values are normalised to the negative control and thus represent the growth relative to yeast cells that produce α- synuclein. The assay is done in duplicate and the effect of each compound on growth is determined by measuring optical density at 600 nm (OD6O0). The OD600 measurements are normalised and depicted as a scatter graph. The Z'-factor (Zhang et al. (1999) J. Biomol. Screen. 4, 67-73) is calculated for each microtiter plate in order to obtain a quantitative score of the assay quality. The average Z'-factor of each plate is found at least 0.70 or higher indicating that the assay is robust and reproducible and therefore very suitable for high-throughput screening purposes. Testing 500 compounds consistently results in highly reproducible OD600 values for each compound. Importantly, this assay allows selection of compounds that significantly and reproducibly improve growth of the yeast strain. Hence the α-synuclein toxicity is overcome.
Example 5 - Visualisation of alpha-svnuclein aggregation.
Methods to enhance and monitor α-Synuclein aggregation have been described previously (WO2005109004; Ostrerova -Golts et al. J. Neurosci. 20: 6048-6054, 2000 and Gerard et al. J Neurosci. 2010 in press). For induction of α-Synuclein aggregation to evaluate the effect of Nhe-1 inhibition on α-Synuclein aggregation, 10 mM FeCI2, 100μM H2O2 and different compound concentrations are added to α-Synuclein overexpressing SHSY5Y cells and cells are incubated for three days. The compound (5-(2-methyl-5- fluorophenyl)furan-2-ylcarbonyl)guanidine (abbreviated as K4389) is dissolved in DMSO, therefore negative control condition is with DMSO only. After fixation, aggregates are visualized with thioflavin S and the percentage of aggregate positive cells is determined (Figure 2). The percentage of cells in late apoptosis is also determined based on nucleus condensation, visualized with a nuclear DAPI staining. The compound K4389 clearly has a concentration-dependent effect on α-Synuclein aggregation with a statistically significant effect from 1 nM (non parametric Kruskal-Wallis test followed by Dunn's post hoc test) (Figure 2A). The same trend is also noticed in the effect on α-Synuclein-induced toxicity (effect on late apoptosis) although differences are not statistically significant (Figure 2B).
Example 6: The 3-Substituted-(5-arylfuran-2-ylcarbonvQguanidines NHE-1 inhibitors reduces α-synuclein toxicity in yeast
The combined expression of human α-synuclein and human endonuclease G in wild type yeast cells provides a system that allows selection or identification of compounds that potentially reduce toxicity mediated by these proteins. Strains BY4741 (MATa his3Δ1 leu2Δ0 met15Δ0 ura3Δ0) expressing human α-synuclein and human endonuclease G are grown on SC medium containing 0.17% yeast nitrogen base (Difco), 0.5% (NHU)2SO4 and 30 mg/l of all amino acids (except 80 mg/l histidine and 200 mg/l leucine), 30 mg/l adenine, and 320 mg/l uracil with 2% glucose (SCD). Micromolar concentrations of the compounds N-(aminoiminomethyl)-5-(5-chloro-2-methoxyphenyl)-3-phenyl-, 2-
Furancarboxamide, N-(aminoiminomethyl)-5-(3-chlorophenyl)-3-ethyl-, 2-
Furancarboxamide, N-(aminoiminomethyl)-3-methyl-5-phenyl-, 2-Furancarboxamide, N- (aminoiminomethyl)-5-(2,5-dichlorophenyl)-3-methyl-, 2-Furancarboxamide, N- (aminoiminomethyl)-5-(2,5-difluorophenyl)-3-methyl- and 2-Furancarboxamide, N- (aminoiminomethyl)-5-(5-chloro-2-methoxyphenyl)-3-methyl- improve the growth of yeast cells expressing both human proteins on minimal selective medium.
Example 7. Inhibition of the yeast homolog of NHE-1 decreases alpha-svnuclein toxicity. Figure 4 displays (A) Quantification of ROS-accumulation using the conversion of non- fluorescent dihydroethidium (DHE) to fluorescent ethidium (Eth) in wild type yeast cells expressing human alpha-synuclein (αSyn) for 48 h or harbouring the empty vector control. Cells were treated with indicated concentrations of the NHE-1 inhibitor K4389 (solved in DMSO) or equivalent concentrations of DMSO alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control. (B) Quantification of ROS-accumulation in wild type yeast cells expressing human alpha-synuclein for 16 h (αSyn) or harbouring the empty vector control upon treatment with 0.25 mM of the NHE-1 inhibitor K4389, Synonym: [5-(2-Methyl~5-fluorophenyl)furan- 2-ylcarbonyl]guanidine / CAS Number: 852146-73-7 / Linear Formula: C13H12FN3O2, olved in MeOH) and equivalent concentration of MeOH alone. Fluorescence intensity was quantified using a TECAN plate reader and data represents fold of corresponding vector control. (C) Quantification of ROS-accumulation in wild type cells and cells lacking the homolog of human NHE-1 (Δnhxi) expressing human alpha-synuclein (αSyn) for 16 h or harbouring the empty vector control. Positive stained cells were quantified using flow cytometry.
PHARMACEUTICAL FORMULATIONS
The present invention also relates to pharmaceutical compositions or formulations which comprise one or more of the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention in admixture with one or more pharmaceutically acceptable excipients, diluents or carriers, the latter being for instance as described hereinafter.
For the purposes of the present invention the term "excipient" and "carrier" are used interchangeably throughout the description of the present invention and said terms are defined herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition." The formulator will understand that excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means for achieving effective absorption by the recipient of the active ingredient. An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach. The formulator can also take advantage of the fact the compounds of the present invention have improved cellular potency, pharmacokinetic properties, as well as improved oral bioavailability.
The furancarbonylguanidine derivative Nhe-1 inhibitors of this invention and/or pharmaceutically acceptable salts, solvates and isomers thereof, when intended to be included in a pharmaceutical composition, may be formulated with conventional carriers and excipients, which can be selected in accordance with ordinary pharmaceutical practice. For instance, tablets may contain excipients, glidants, fillers, binders and the like. Aqueous formulations are preferably prepared in sterile form and, when intended for delivery by other than oral administration, are usually isotonic. Pharmaceutical formulations optionally contain excipients such as those set forth in the " Handbook of Pharmaceutical Excipients " (1986) and may include ascorbic acid and/or other antioxidants, chelating agents, carbohydrates such as dextrin-containing compounds (e.g. maltodextrins and/or cyclodextrins), hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like.
The term "pharmaceutically acceptable carrier" as used herein means any material or substance with which the active ingredient is formulated in order to facilitate its application or dissemination to the locus to be treated, for instance by dissolving, dispersing or diffusing the said composition, and/or to facilitate its storage, transport or handling without impairing its effectiveness.
The pharmaceutically acceptable carrier may be a solid or a liquid or a gas which has been compressed to form a liquid, i.e. the pharmaceutical compositions of this invention can suitably be in the form of concentrates, emulsions, solutions, granulates, dusts, sprays, aerosols, suspensions, ointments, creams, tablets, pellets or powders.
Suitable pharmaceutical carriers for use in the pharmaceutical compositions and formulations of the invention are well known to those skilled in the art. They also include additives such as, but not limited to, wetting agents, dispersing agents, stickers, adhesives, emulsifying agents, solvents, coatings, antibacterial and antifungal agents (for example phenol, sorbic acid, chlorobutanol), isotonic agents (such as sugars or sodium chloride) and the like, provided the same are consistent with pharmaceutical practice, i.e. carriers and additives which do not create permanent damage to mammals.
The pharmaceutical compositions of the present invention may be prepared in any known manner, for instance by homogeneously mixing, coating and/or grinding the active ingredient, in a one-step or multi-steps procedure, with the selected one or more carrier materials and, where appropriate, the other additives such as surface-active agents. They may also be prepared by micronisation, for instance in view to obtain them in the form of microspheres usually having a diameter of about 1 to 10 μm, namely for the manufacture of microcapsules for controlled or sustained release of the active ingredient.
Suitable surface-active agents, also known as emulsifiers, that can be used in the pharmaceutical compositions of the present invention include non-ionic, cationic and/or anionic materials having good emulsifying, dispersing and/or wetting properties. Suitable anionic surfactants include both water-soluble soaps and water-soluble synthetic surface- active agents. Suitable soaps are alkaline or alkaline-earth metal salts, solvates and isomers, unsubstituted or substituted ammonium salts, solvates and isomers of higher (CiO-C22) fatty acids, e.g. the sodium or potassium salts, solvates and isomers of oleic acid, stearic acid or natural fatty acid mixtures obtainable from coconut oil or tallow oil. Synthetic surfactants include sodium or calcium salts, solvates and isomers of polyacrylic acids; fatty sulphonates and sulphates; sulphonated benzimi-dazole derivatives preferably having 8 to 22 carbon atoms; and alkylarylsulphonates. Fatty sulphonates or sulphates are usually in the form of alkaline or alkaline-earth metal salts, solvates and isomers, unsubstituted ammonium salts, solvates and isomers or ammonium salts, solvates and isomers substituted with an alkyl or acyl radical having from 8 to 22 carbon atoms, e.g. the sodium or calcium salt of lignosulphonic acid or dodecylsulphonic acid or a mixture of fatty alcohol sulphates obtained from natural fatty acids, alkaline or alkaline-earth metal salts, solvates and isomers of sulphuric or sulphonic acid esters (such as sodium lauryl sulphate) and sulphonic acids of fatty alcohol/ethylene oxide abducts. Examples of alkylarylsulphonates are the sodium, calcium or alcanolamine salts, solvates and isomers of dodecylbenzene sulphonic acid or dibutyl-naphtalenesulphonic acid or a naphtalenesulphonic acid/formaldehyde condensation product. Also suitable are the corresponding phosphates, e.g. salts, solvates and isomers of phosphoric acid ester and an adduct of p-nonylphenol with ethylene and/or propylene oxide, or phospholipids. Suitable phospholipids for this purpose include natural (originating from animal or plant cells) or synthetic phospholipids of the cephalin or lecithin type such as, but not limited to, phosphatidylethanoiamine, phosphatidylserine, phosphatidylglycerine, lyso-lecithin, cardiolipin, dioctanylphosphatidylcholine, dipalmitoylphoshatidyl-choline and mixtures thereof in various proportions.
Suitable non-ionic surfactants include polyethoxylated and polypropoxy-lated derivatives of alkylphenols, fatty alcohols, fatty acids, aliphatic amines or amides containing at least
12 carbon atoms in the molecule, alkylarene-sulphonates and dialkylsulphosuccinates such as, but not limited to, polyglycol ether derivatives of aliphatic and cycloaliphatic alcohols, saturated and unsaturated fatty acids and alkylphenols, said derivatives preferably containing from 3 to 10 glycol ether groups and from 8 to 20 carbon atoms in the (aliphatic) hydrocarbon moiety and/or from 6 to 18 carbon atoms in the alkyl moiety of the alkylphenol. Further suitable non-ionic surfactants are water-soluble adducts of polyethylene oxide with poylypropylene glycol, ethylenediaminopolypropylene glycol containing from 1 to 10 carbon atoms in the alkyl chain, which adducts contain from about
20 to 250 ethyleneglycol ether groups and/or from 10 to 100 propyleneglycol ether groups. Such compounds usually contain from 1 to 5 ethyleneglycol units per propyleneglycol unit. Representative examples of non-ionic surfactants are nonylphenol- polyethoxyethanol, castor oil polyglycolic ethers, polypropy-lene/polyethylene oxide adducts, tributylphenoxypolyethoxyethanol, polyethyleneglycol and octylphenoxypolyethoxyethanol. Fatty acid esters of polyethylene sorbitan (such as polyoxyethylene sorbitan trioleate), glycerol, sorbitan, sucrose and pentaerythritol are also suitable non-ionic surfactants.
Suitable cationic surfactants include quaternary ammonium salts, solvates and isomers, particularly halides, having 4 hydrocarbon radicals optionally substituted with halo, phenyl, substituted phenyl or hydroxy; for instance quaternary ammonium salts, solvates and isomers containing as N-substituent at least one C8-C22 alky] radical (e.g. cetyl, lauryl, palmityl, myristyl, oleyl and the like) and, as further substituents, unsubstituted or halogenated C1-C4 alkyl, benzyl and/or hydroxy C1-C4 alkyl radicals.
A more detailed description of surface-active agents suitable for this purpose may be found for instance in "McCutcheon's Detergents and Emulsifiers Annual" (MC Publishing Group, Ridgewood, New Jersey, 1981), "Tensid-Taschenbuch", 2nd ed. (Hanser Verlag, Vienna, 1981) and "Encyclopaedia of Surfactants" (Chemical Publishing Co., New York, 1981).
The compound of this invention may be administered by any route appropriate for the bone disorder to be treated. Suitable routes include, but are not limited to, oral, rectal, nasal, topical (including transdermal^, ocular, buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intra-arterially, intradermal, intrathecal and epidural) routes. The preferred route of administration may vary in accordance with certain clinical parameters, for example with the condition of the patient to be treated.
Pharmaceutical formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. A suitable tablet may be made by compression or molding, optionally with one or more pharmaceutically acceptable inactive ingredients such as described hereinabove. Compressed tablets may be prepared by compressing, in a suitable compressing machine, the active ingredient in a free-flowing form such as a powder, granules, beads or pellets, optionally admixed with one or more pharmaceutically acceptable excipients such as binders, lubricants, inert diluents, preservatives, and surface-active or dispersing agents. Molded tablets may be made by molding, in a suitable molding machine, a mixture of the powdered active compound moistened with suitable amounts of one or more inert liquid diluents. The tablets may optionally be further coated, and may be formulated so as to provide slow or controlled release of the active ingredient therein.
The pharmaceutical formulations of the invention are optionally in the form of a topical ointment or cream. When formulated in an ointment, the active ingredient may be admixed with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base may include, for example, at least 30 % by weight of a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups either in a monomeric form such as, but not limited to, propylene glycol, butane-1 ,3-diol, mannitol, sorbitol and glycerol, or in apolymeric form such as polyethylene glycol (with various numbers of repeating units, including PEG 400) and mixtures thereof. Topical formulations may desirably include at least a compound which enhances absorption or penetration of the active ingredient through the skin or other areas of application. Examples of such dermal penetration enhancers include, but are not limited to, dimethylsulfoxide and related analogues. The solubility of the active compound of this invention in most pharmaceutically acceptable oils is quite high. Thus a cream formulation may suitably include one or more straight or branched chain, mono-or dibasic alkyl esters such as, but not limited to, di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP, the three latter being preferred esters. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate. Formulations suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size for example in the range 20 to 500 microns, which may be administered by rapid inhalation through the nasal passage from a container. Suitable formulations wherein the carrier is a liquid, for administration as for example a nasal spray or as nasal drops, include oily solutions of the active ingredient. When the pharmaceutical formulation is intended for a female patient, the active compound of the invention may be delivered into the systemic circulation through the vaginal mucosa from a vaginal device incorporated with a transmucosal vaginal composition. Said composition may be formulated and incorporated into the device as a suppository, cream, spray, gel, film, powder, foam, ointment, microcapsules, nanocapsules or a capsule containing microparticles or nanoparticles; and said vaginal device may be a vaginal tampon, vaginal ring, vaginal strip, vaginal capsule, vaginal tablet, vaginal pessary, vaginal cup or vaginal sponge; and said composition is delivered to the vaginal mucosa by inserting said device into the vagina.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the patient; and aqueous and non-aqueous sterile suspensions which may include one or more suspending agents and/or thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or a weekly dose, as described herein, or an appropriate fraction thereof, of the active ingredient of this invention.
The present invention also provides controlled release pharmaceutical formulations, containing as an active ingredient the novel compound of the invention, in which the release of the active ingredient is controlled and regulated to allow less frequency dosing or to improve the pharmacokinetic or toxicity profile of said compound. Controlled release formulations adapted for oral administration in which discrete units comprising the compound of the invention can be prepared according to conventional methods in the art. Additional ingredients may be included in order to control the duration of action of the active ingredient in the composition. Control release compositions may thus be achieved by selecting appropriate polymer carriers such as for example polyesters, polyamino- acids, polyvinyl pyrrolidone, ethylene-vinyl acetate copolymers, methylcellulose, carboxymethylcellulose, protamine sulfate and the like. The rate of drug release and duration of action may also be controlled by incorporating the active ingredient into particles, e.g. microcapsules, of a polymeric substance such as hydrogels, polylactic acid, hydroxymethylcellulose, polymethylmethacrylate and other similar polymers. Such compositions include colloid drug delivery systems like liposomes, microspheres, microemulsions, nanoparticles, nanocapsules and the like. Depending upon the selected route of administration, the pharmaceutical composition may require protective coatings.
The effective dosage of the compounds of present invention can be determined according to age, weight, gender, administration method, health condition and severity of a disease. For example, the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 0.1 mg/day to about 1000 mg/day. More preferably, the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 1 mg/day to about 500 mg/day. Alternatively, the effective dose of the compound for an adult patient having the weight of 70 kg might be from about 5 mg/day to about 300 mg/day. The administration times are determined by a doctor or a pharmacist to be once a day or a few times a day.
Non-limiting examples of compositions according to the present invention include: a) from about 0.1 mg/day to about 1000 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients. Another embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 1000 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients. A further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 500 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients. A further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 250 mg/day of the compounds of present invention; and b) one or more pharmaceutical excipients.
Non-limiting examples of compositions according to the present invention further include: a) from about 0.1 mg/day to about 1000 mg/day of the carbonylguanidine NH, in particular the furancarbonylguanidine compound, for instance [5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients. Another embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 1000 mg/day of the carbonylguanidine, in particular the furancarbonylguanidine compound, for instance [5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients. A further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 500 mg/day of the furancarbonylguanidine compound for instance [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients. A further embodiment according to the present invention relates to the following compositions: a) from about 1 mg/day to about 250 mg/day of the furancarbonylguanidine compound for instance 5-(2-methyl-5-fluorophenyl)furan-2- ylcarbonyl]guanidine; and b) one or more pharmaceutical excipients.
The terms "effective amount" and "therapeutic amount" as used herein may vary according to a range of factors known in the art, such as the disease state, age, sex, and weight of the human or animal being treated.
Although particular dosage regimes are described herein, the person skilled in the art appreciates that these dosage regimes may be altered, after due consideration of said factors for each class of patients or each individual patient, in order to provide optimum therapeutic response. For example, several divided doses of the furancarbonylguanidine compound for instance may be administered daily or the dose may be proportionally reduced as indicated by the requirements of the therapeutic situation. In addition, the compositions of the present invention can be administered as frequently as necessary to achieve a suitable therapeutic response in the patient being treated. For the purposes of the present invention a first aspect of "therapeutic amount" relates to compositions which deliver a compound according to the present invention wherein the plasma level of said the furancarbonylguanidine compound for instance is from about 0.1 pg/mL to about 100 mg/mL in humans or higher mammals. Another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said furancarbonylguanidine compound is from about 0.1 pg/mL to about 1 mg/mL in humans or higher mammals. Another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 pg/mL to about 1 mg/mL in humans or higher mammals. Yet another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 pg/mL to about 10 μg mg/mL in humans or higher mammals. Another aspect relates to compositions which deliver a compound according to the present invention wherein said plasma level of said compound is from about 1 ng/mL to about 25 mg/mL in humans or higher mammals.
Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the furancarbonylguanidine derivatives and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) one or more pharmaceutically acceptable excipients.
Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine effective for preventing synucleinopathy; b) one or more pharmaceutically acceptable excipients.
For the purposes of the present invention the term "excipient" and "carrier" are used interchangeably throughout the description of the present invention and said terms are defined herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition." The formulator will understand that excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means for achieving effective absorption by the recipient of the active ingredient. An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach. The formulator can also take advantage of the fact the compounds of the present invention have improved cellular potency, pharmaco-kinetic properties, as well as improved oral bioavailability.
The term "dopaminergic effective amount", as used herein, refers to an amount of a compound sufficient to inhibit the binding of dopamine to a dopamine receptor with the effect of altering (i. e., increasing or decreasing) dopamine mediated neurotransmission. Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a composition for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients. Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a composition for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients. Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of L-DOPA for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients. Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a neuroprotective amount of a compound for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients. Another aspect of the present invention relates to compositions which provide protection against synucleinopathy, said compositions comprising: a) a therapeutically effective amount of the Nhe-1 inhibitors and/or pharmaceutically acceptable salts, solvates and isomers thereof according to the present invention effective for preventing synucleinopathy; b) a therapeutically effective amount of a dopaminergic effective amount of a compound for the treatment of Parkinson's disease; c) one or more pharmaceutically acceptable excipients.
USE OF COMPOUNDS
The present invention also relates to the use of a Nhe-1 inhibitor and/or pharmaceutically acceptable salts, solvates and isomers thereof of the present invention in the manufacture of a medicament for the treatment or prevention of synucleinopathy. The present invention also relates to the use of a carbonylguanidine, more particularly a furancarbonylguanidine derivative and/or pharmaceutically acceptable salts, solvates and isomers thereof of the present invention in the manufacture of a medicament for the treatment or prevention of synucleinopathy.
The compounds of the present invention can be used in the manufacture of one or more medicaments, non-limiting examples of which are: i) a compound for use in the manufacture of a medicament for the treatment of Parkinson's disease; ii) a compound for use in the manufacture of a medicament for the treatment of dementia with Lewy bodies; iii) a compound for use in the manufacture of a medicament for the treatment of pure autonomic failure; iv) a compound for use in the manufacture of a medicament for the treatment of osteoporosis without the side effect of inducing multiple system atrophy.
KIT Present invention relates to a kit comprising an amount of a Nhe-1 inhibitor of present invention, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a composition for the treatment of Parkinson's disease, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container. In a particular embodiment of present invention, the kit of present invention comprises an amount of a Nhe-1 inhibitor of present invention, a composition for the treatment of Parkinson's disease selected from the group consisting of (a) L-DOPA, (b) a neuroprotective amount of a compound, and (c) a dopaminergic effective amount of a compound, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
Normally, the kit will also include directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage levels, or when titration of the individual components of the combination is desired by the prescribing physician.
One example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are used widely for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally comprise a sheet of relatively rigid material covered with a foil of a preferably transparent plastic material. During the packaging process, recesses (e.g., blisters) are formed in the plastic foil. The recesses generally conform to the size and shape of the tablets or capsules to be contained therein. Next, the tablets or capsules are placed in the recesses and the sheet of relatively rigid material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses are formed. As a result, the tablets or capsules are captively retained and sealed inside the recesses between the plastic foil and the sheet. Preferably, the strength of the sheet is such that the tablets or capsules may be removed from the blister pack by the application of manual pressure on the outer surface of the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed through the formed opening. It is further desirable to provide a memory aid on the pack, for example, in the form of numbers or similar indicia proximate to the tablets or capsules whereby the indicia correspond to the days of the regimen which the dosage form so specified is to be ingested. An additional example of such a memory aid is a calendar printed on the pack, for example, as follows: "First Week, Monday, Tuesday, ... etc. Second Week, Monday, Tuesday, ..." etc. In light of the instant disclosure, other variations will be readily apparent to one of ordinary skill in the art. A "daily dose" can be a single tablet or capsule, or multiple tablets or capsules, or tablets or capsules to be ingested on a given day. Also, a daily dose of the sodium-hydrogen exchanger type 1 (NHE-1) inhibitor can consist of a single tablet or capsule, while a daily dose of the second compound, selected from the group consisting of (a) a compliment modulator, (b) a metabolic modulator, (c) an anti- apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na +/Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, can consist of multiple tablets or capsules, and vice-versa. The memory aid should reflect this.
In another specific embodiment of the invention, a pack designed to dispense the daily doses one at a time in the order of their intended use is provided. Preferably, the pack is equipped with a memory aid, so as to further facilitate compliance with the dosage regimen. An example of such a memory aid comprises a mechanical counter that indicates the number of daily doses to be dispensed, Another example of such a memory aid comprises a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date and time that the last daily dose has been taken and/or reminds the patient when the next dose is to be taken.
While particular embodiments of the present invention have been illustrated and described, it is obvious to those skilled in the art that various other changes and modifications can be made without departing from the spirit and scope of the invention. It is therefore intended to cover in the appended claims all such changes and modifications that are within the scope of this invention.
Each of the claims set out a particular embodiment of the invention.

Claims

SYNUCLEINOPATHIESCLAIMS
1. A furancarbonylguanidine derivative represented by Formula (I) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, wherein :
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000064_0001
in which the cyclic group (A) presented by Formula (II)
Figure imgf000064_0002
(II) is a 3 to 8 membered ring.
2. The furancarbonylguanidine derivative of claim 1 , wherein the cyclic group presented by Formule (II) is an aromatic ring, for instance an aryl.
3. The furancarbonylguanidine derivative of claim 1 , wherein the cyclic group presented by Formule (II) is a carbocycle.
4. The furancarbonylguanidine derivative of claim 1, represented by Formula (III) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000065_0001
(Ml)
The furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof according to any one of the claim 1 , 2, 3 or 4, wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of:
- [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine, ~ [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-phenylfuran-2-ylcarbonyl]guanidine, - [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine, ~ [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, ~ [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methylphenyl)furan-2~ylcarbonyl]guanidine, - [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonylJguanidine, - [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine, - [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyI]guanidine,
- [5-(2-aminophenyl)furaπ-2-ylcarbonyl]guanidine, - [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-yIcarbonyi]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarboπyl] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5~(2-isopropoxyphenyl)furan-2-yIcarbonyl]guanidine,
- [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methyi-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyI]guanidine,
- [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,
5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and - [δ-^-isopropoxy-δ-chlorophenyOfuran^-ylcarbonylføuanidine.
6. The furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof according any one of the claim 1, 2, 3 or 4,, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
7. A pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative represented by Formula (III) or by Formula I or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alky!; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000067_0001
(I)
8. The pharmaceutical composition according to claim 7, wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of: - [5-(2-fluorophenyl)furan-2-yicarbonyl]guanidine, - [5-(3-fluorophenyl)furan-2-yIcarbonyl]guanidine,
- [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5~phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methylphenyl)furan-2-ylcarbonyl]guaπidine, - [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine,
- [5~(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyi]guanidine,
- [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [S-CS.δ-dichlorophenyOfuran^-ylcarbonyllguanidine, - [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,5-dimethylphenyl)furan-2-yIcarbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,6-dimethoxyphenyI)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidiπe, - [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-hydroxy-5-chIorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and
- [5-(2-isopropoxy-5-chloropheny!)furan-2-ylcarbonyl]guanidine.
9. The pharmaceutical composition according to any of the claims 7 to 8, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
10. Use of a furancarbonylguanidine derivative represented by Formula (III) or by Formula
(I) or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof in the manufacture of a medicament for the treatment or prevention of synucleinopathy and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4;
- R3 is selected from H, NH2, C1-C5 straight or branched alkyl, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, CrC5 straight or branched alkyl, or phenyl.
Figure imgf000069_0001
(111)
Figure imgf000070_0001
(D
11. The use according to claim 10, wherein said furancarbonylguanidine derivative or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof are selected from the group consisting of:
- [5-(2-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-fluorophenyl)furan-2-ylcarbonyl]guanidine,
~ [5-(4-fluorophenyl)furan-2-ylcarbonyl]guanidine, - [5-phenylfuran-2-ylcarbonyl]guanidine,
- [5-(2-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [δ-β-chlorophenyOfuran^-ylcarbonyllguanidine,
- [5-(4-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(3-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(4-methylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-[2-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[3-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-[4-(trifluoromethyl)phenyl]furan-2-ylcarbonyl]guanidine, - [5-(2-methoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-methoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-nitrophenyl)furan-2-ylcarbonyl]guanine,
- [5-(3-nitrophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-nitrophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3-aminophenyl)furan-2-ylcarbonyl]guanidine, - [5-(4-aminophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethylphenyl)furan-2-ylcarbonyl] guanidine, - [5-(2-ethoxyphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-isopropoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-phenoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-difluorophenyl)furan-2-ylcarbonyI]guanidine, - [5-(2,4-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-difluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-6-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-fluoro-5-methylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methyl-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-fluorophenyl)furan-2-ylcarbonyl]guanidine,
- [δ-β.δ-dichlorophenyOfuran^-ylcarbonyltøuanidine,
- [5-(2,3-dichlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dichlorophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-methoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-chloro-5-trifluoromethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(3,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethylphenyl)furan-2-ylcarbonyl]guanidine, - [5-(2,3-dimethylphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,6-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,3-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2,5-dimethoxyphenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-methoxy-5-bromophenyl)furan-2-ylcarbonyl]guanidine, - [5-(2-hydroxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine,
- [5-(2-ethoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine, and
- [5-(2-isopropoxy-5-chlorophenyl)furan-2-ylcarbonyl]guanidine.
12. The use according to any of claims 10 to 11, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
13. A furancarbonylguanidine NHE-1 inhibitor represented by Formula (IV) or pharmaceutically acceptable salts, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy, and wherein:
- R1 and R2 are each independently selected from H; F; Cl; Br; I; CF3; SO2CH3; NO2; NH2; C1-C5 straight or branched alkyl; or OR4; - R3 is selected from H, NH2, C1-C5 straight or branched alky!, NH(COCH3), N(CH3)COCH3, methylamino, acetylamino, acetylmethylamino, dimethylamino, phenyl, pyrrol, piperidinyl, or pyrrolidinyl,
- R4 is selected from H, CF3, C1-C5 straight or branched alkyl, or phenyl.
Figure imgf000072_0001
in which the cyclic group (A) presented by Formula (II)
Figure imgf000072_0002
is a 3 to 8 membered ring. and wherein the cyclic group (B) represented by the Fomula (V)
Figure imgf000072_0003
(V) is:
- an heterocyclic five-membered rings containing two or three heteroatoms whereby the heteroatoms are nitrogen, oxygen or a combination; or
- a five-membered rings with as single heteroatom oxygen with Z being of the group of saturated tetrahydrofuran, unsaturated dihydrofuran (2,3-Dihydrofuran or 2,5-
Dihydrofuran) and unsaturated furan; or
- a five-membered rings with as single heteroatom nitrogen with Z being of the group of saturated pyrrolidine, unsaturated Pyrroline and unsaturated Pyrrole; or
- carbon connected and is a five-membered, diaza, di-unsaturated ring having two contiguous nitrogens; or - carbon connected and is a five-membered, triaza, di-unsaturated ring, said ring;
14. The furancarbonylguanidine derivative of claim 13, wherein the cyclic group presented by Formule (II) is an aromatic ring for instance an aryl.
15. The furancarbonylguanidine derivative of claim 13, wherein the cyclic group presented by Formule (II) is a carbocycle.
16. A pharmaceutical composition comprising an effective amount of a furancarbonylguanidine derivative of any of the previous claims 13 to 15 or pharmaceutically acceptable salts, solvates and isomers, solvates and isomers thereof for use in a treatment to cure or to prevent synucleinopathy.
17. Any of the previous claims 13 to 16, wherein said synucleinopathy is selected from Parkinson's disease, dementia with Lewy bodies, pure autonomic failure or multiple system atrophy.
18. Any of the previous claims 13 to 17, wherein said furancarbonylguanidine derivative is of the group consisting of cariporide (N-(4-lsopropyl-3-methanesulfonyl- benzoyl)-guanidine), BIIB-513 (benzamide {N-(aminominomethyl)-4[4-(2- furanylcarbonyl)- 1-piperazinyl]-3-(methylsulfonyl), methanesulfonate}), TY-12533 (β.y.δ.θ-tetrahydro^-methyl-δH-cycloheptafbJpyridine-S-carbonylguanidine maleate) and SM-15681 (5-(N-ethyl-N-isopropyl)amiloride,5-(ethylisopropyl)amiloride) or a pharmaceutically acceptable salt, solvate or isomer thereof.
PCT/BE2010/000013 2009-02-18 2010-02-18 Synucleinopathies WO2010094090A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0902662A GB0902662D0 (en) 2009-02-18 2009-02-18 Reduction of alpha synuclein toxicity
GB0902662.6 2009-02-18
GB0902781.4 2009-02-19
GB0902781A GB0902781D0 (en) 2009-02-19 2009-02-19 Nhe-1 innhibitors

Publications (2)

Publication Number Publication Date
WO2010094090A2 true WO2010094090A2 (en) 2010-08-26
WO2010094090A3 WO2010094090A3 (en) 2010-10-14

Family

ID=42341548

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/BE2010/000013 WO2010094090A2 (en) 2009-02-18 2010-02-18 Synucleinopathies

Country Status (1)

Country Link
WO (1) WO2010094090A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10300155B2 (en) 2015-12-31 2019-05-28 Washington University Alpha-synuclein ligands

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA003603B1 (en) * 1998-02-27 2003-06-26 Пфайзер Продактс Инк. N-[(substituted five-membered di- or triaza diunsaturated ring)carbonyl] guanidine derivatives for the treatment of ischemia
KR100518184B1 (en) * 2003-12-27 2005-10-04 한국화학연구원 Furancarbonylguanidine derivatives, their preparation and pharmaceutical compositions containing them
CN101460161A (en) * 2006-03-29 2009-06-17 弗尔德里克斯制药股份有限公司 Inhibition of alpha-synuclein toxicity

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BEYER, K. ET AL., NEUROGENETICS, vol. 9, 2008, pages 15 - 23
CAMPION, D. ET AL., GENOMICS, vol. 26, 1995, pages 254 - 257
COUNILLON, L. ET AL., MOL. PHARMACOL., vol. 44, 1993, pages 1041 - 1045
HORACIO KAUFMANN ET AL., NEUROLOGY, vol. 56, 2001, pages 980 - 981
JAKES, R., FEBS LETT., vol. 345, 1994, pages 27 - 32
OSTREROVA -GOLTS ET AL., J. NEUROSCI., vol. 20, 2000, pages 6048 - 6054
UEDA, K. ET AL., PROC. NAT. ACAD. SCI., vol. 90, 1993, pages 11282 - 11286
WATSON ET AL., AM. J. PHYSIOL., vol. 24, 1991, pages G222G - G238
ZHANG ET AL., J. BIOMOL. SCREEN., vol. 4, 1999, pages 67 - 73

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10300155B2 (en) 2015-12-31 2019-05-28 Washington University Alpha-synuclein ligands

Also Published As

Publication number Publication date
WO2010094090A3 (en) 2010-10-14

Similar Documents

Publication Publication Date Title
US11130779B2 (en) PPAR agonists and methods of use thereof
US10550071B2 (en) PPAR agonists
ES2206351T3 (en) DERIVATIVES OF BENZAZOL AND ITS USE AS MODULATORS OF JNK.
AU2007245625B2 (en) N- ( 2-thiazolyl) -amide derivatives as GSK-3 inhibitors
KR101471999B1 (en) -3 gsk-3 inhibitors
ES2606043T3 (en) Biaryl derivatives as nAChR modulators
CN104718205B (en) Sodium channel inhibitor, its preparation method and its purposes
US20170114060A1 (en) Novel effective antiviral compounds and methods using same
US20210261554A1 (en) Ripk2 inhibitors and method of treating cancer with same
US20210395238A1 (en) Nadph oxidase 4 inhibitors
MXPA03005777A (en) Substituted triazole diamine derivatives as kinase inhibitors.
AU2018202898A1 (en) Therapeutics targeting truncated Adenomatous Polyposis Coli (APC) proteins
DE60215139T2 (en) MALONYL COA-DECARBOXYLASE INHIBITORS AS METABOLIC MODULATORS
KR20060099513A (en) Phenyl-piperazine derivatives as modulators of muscarinic receptors
US20240009169A9 (en) PPARG Modulators for the Treatment of Osteoporosis
KR20110115571A (en) Toluidine sulfonamides and their use
KR20060123452A (en) Naphthyridine derivatives and their use as modulators of muscarinic receptors
EP2991963B1 (en) Inhibitors of nhr2 and/or runx1/eto-tetramerization
ES2812800T3 (en) New therapeutic uses of benzylidenguanidine derivatives for the treatment of proteopathies
WO2010094090A2 (en) Synucleinopathies
US20170029388A1 (en) Treatment of Neurodegenerative Diseases with Asparagine Endopeptidase (AEP) Inhibitors and Compositions Related Thereto
WO2011146101A1 (en) Chemical agents for the prevention or inhibition of tumor metastasis
DE60209886T2 (en) TIE2 RECEPTOR KINASE INHIBITORS FOR THE TREATMENT OF ANGIOGENIC DISEASES
US9944597B2 (en) Polysubstituted pyrroles having microtubule-disrupting, cytotoxic and antitumor activities and methods of use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10721280

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10721280

Country of ref document: EP

Kind code of ref document: A2