Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ENANTIOMERS OF 8-HYDROXY QUINOLINE DERIVATIVES AND THE SYNTHESIS THEREOF
Document Type and Number:
WIPO Patent Application WO/2016/162706
Kind Code:
A1
Abstract:
Our invention relates to novel enantiomer derivatives of 8-hydroxyquinoline derivatives with general formula (I) and (II) and the synthesis thereof and pharmaceutically acceptable salts and metal complexes and thereof, and the medicinal and/or pharmaceutical compositions comprising these compounds. The essence of the subject matter of the invention relates to the fact that prior art discloses the biological effect and characteristics only of the racemic products, the novel enantiomer derivatives according to the invention appear in our application for the first. The subject matter of the invention furthermore relates to a novel, stereo selective synthesis for the preparation of the novel enantiomer derivatives according to the invention. The novel medicinal and/or pharmaceutical compositions comprising these enantiomers are suitable for the treatment of the named diseases, and the enenatiomers are used for manufacture of these compositions. These applications for manufacture of the compositions are also the subject matters of the invention. The compounds according to the invention can be used preferably as cytoprotective, neuroprotective, cardioprotective, anxiolytic and antidepressant agent for treatment of neuropsychiatric and neurologic diseases and diseases in connections with transplantations and with ischemia and reperfusion injuries thereof, and inhibition of organ, advantageously skin graft rejection. According to our studies the R-enantiomer has either sole or high biological effect in some cases.

Inventors:
PUSKÁS LÁSZLÓ (HU)
KANIZSAI IVÁN (HU)
PILLOT THIERRY (FR)
GYURIS MÁRIÓ (HU)
SZABÓ ANDRÁS (HU)
TAKÁCS FERENC (HU)
HACKLER LÁSZLÓ (HU)
Application Number:
PCT/HU2016/000012
Publication Date:
October 13, 2016
Filing Date:
March 07, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AVIDIN CO LTD (HU)
SONEAS RES CO LTD (HU)
SYNAGING SAS (HU)
International Classes:
C07D401/12; A61K31/4709; A61K31/506; A61P25/00; A61P37/00
Domestic Patent References:
WO2008116092A12008-09-25
WO2011148208A12011-12-01
WO2016020892A12016-02-11
WO2011148208A12011-12-01
WO2004007461A12004-01-22
Foreign References:
US20030180321A12003-09-25
CN104744439A2015-07-01
US20080293699A12008-11-27
Other References:
CIMARELLI C ET AL: "SOLVENT-FREE ASYMMETRIC AMINOALKYLATION OF ELECTRON-RICH AROMATIC COMPOUNDS: STEREOSELECTIVE SYNTHESIS OF AMINOALKYLNAPHTHOLS BY CRYSTALLIZATION-INDUCED ASYMMETRIC TRANSFORMATION", THE JOURNAL OF ORGANIC CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 66, no. 14, 1 January 2001 (2001-01-01), pages 4759 - 4765, XP008038874, ISSN: 0022-3263, DOI: 10.1021/JO0101205
WANG YU ET AL: "Predicting dual-targeting anti-influenza agents using multi-models", MOLECULAR DIVERSITY, ESCOM SCIENCE PUBLISHERS, LEIDEN, NL, vol. 19, no. 1, 2 October 2014 (2014-10-02), pages 123 - 134, XP035433703, ISSN: 1381-1991, [retrieved on 20141002], DOI: 10.1007/S11030-014-9552-4
JOHN H. CARDELLINA ET AL: "Separation of Betti Reaction Product Enantiomers: Absolute Configuration and Inhibition of Botulinum Neurotoxin A", ACS MEDICINAL CHEMISTRY LETTERS, vol. 2, no. 5, 12 May 2011 (2011-05-12), United States, pages 396 - 401, XP055290924, ISSN: 1948-5875, DOI: 10.1021/ml200028z
RINO RAGNO ET AL: "Identification of Small-Molecule Inhibitors of the XendoU Endoribonucleases Family", CHEMMEDCHEM, vol. 6, no. 10, 29 July 2011 (2011-07-29), DE, pages 1797 - 1805, XP055290997, ISSN: 1860-7179, DOI: 10.1002/cmdc.201100281
YIPIN LU ET AL: "Discovery of a nanomolar inhibitor of the human murine double minute 2 (MDM2)-p53 interaction through an integrated, virtual database screening strategy", JOURNAL OF MEDICINAL CHEMISTRY, vol. 49, no. 13, 26 May 2006 (2006-05-26), US, pages 3759 - 3762, XP055291420, ISSN: 0022-2623, DOI: 10.1021/jm060023+
CHI LIU ET AL: "Novel Hybrid Virtual Screening Protocol Based on Molecular Docking and Structure-Based Pharmacophore for Discovery of Methionyl-tRNA Synthetase Inhibitors as Antibacterial Agents", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES 2012 MDPI AG CHE, vol. 14, no. 7, 9 July 2013 (2013-07-09), pages 14225 - 14239, XP055290991, ISSN: 1661-6596, DOI: 10.3390/ijms140714225
XIAOLUN WANG ET AL: "Conformation-Specific Effects of Raf Kinase Inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, vol. 55, no. 17, 13 September 2012 (2012-09-13), US, pages 7332 - 7341, XP055290992, ISSN: 0022-2623, DOI: 10.1021/jm300613w
TOMLINSON M L ET AL: "A Chemical Genomic Approach Identifies Matrix Metalloproteinases as Playing an Essential and Specific Role in Xenopus Melanophore Migration", CHEMISTRY AND BIOLOGY, CURRENT BIOLOGY, LONDON, GB, vol. 16, no. 1, 30 January 2009 (2009-01-30), pages 93 - 104, XP025897064, ISSN: 1074-5521, [retrieved on 20090130], DOI: 10.1016/J.CHEMBIOL.2008.12.005
LEE M; PARK J: "Regulation of NFAT activation: a potential therapeutic target for immunosuppression", MOL CELLS, vol. 22, no. 1, 31 August 2006 (2006-08-31), pages 1 - 7
MITTL PRE; MARCO SD; KREBS JF; KARANEWSKY DS; PRIESTLE JP; TOMASELLI KJ; GRUT TER MG: "Structure of Recombinant Human CPP32 in Complex with the Tetrapeptide Acetyl-Asp-Val-Ala-Asp Fluoromethyl Ketone", J BIOL CHEM., vol. 272, 1997, pages 6539 - 6547
EMMEL EA; VERWEIJ CL; DURAND DB; HIGGINS KM; LACY E; CRABTREE GR: "Cyclosporin A specifically inhibits function of the nuclear proteins involved in T cell activation", SCIENCE, vol. 246, 1989, pages 1617 1620
KARTTUNEN J; SHASTRI N: "Measurement of ligand-induced activation in single viable T cells using the lacZ reporter gene", PROC NATL ACAD SCI USA., vol. 88, 1991, pages 3972
"Protocols for Neural Cell Culture", 2001, article "Ruth Cole, 'Jean de Vellis. Preparation of Astrocyte, Oligodendrocyte, and Microglia Cultures from Primary Rat Cerebral Cultures", pages: 117 - 127
GREGORY J.- GAGE; DARYL R. KIPKE; WILLIAM SHAM, J. VIS. EXP., 2012, pages E3564
ABDUL HM; SAMA MA; FURMAN JL; MATHIS DM; BECKETT TL; WEIDNER AM; PATEL ES; BAIG I; MURPHY MP; LEVINE H 3RD: "Cognitive decline in Alzheimer's disease is associated with selective changes in calcineurin/NFAT signaling", J NEUROSCI., vol. 29, 2009, pages 12957 - 12969
ARABPOOR Z; HAMIDI G; RASHIDI B; SHABRANG M; ALAEI H; SHARIFI MR; SALAMI M; DOLATABADI HR; REISI P.: "Erythropoietin improves neuronal proliferation in dentate gyrus of hippocampal'formation in an animal model of Alzheimer's disease", ADV BIOMED RES., vol. 1, 2012, pages 50
ASAI A; QIU JH; NARITA Y; CHI S; SAITO N; SHINOURA N; HAMADA H; KUCHINO Y; KIRINO T: "High level calcineunn activity predisposes neuronal cells to apoptosis", J BIOL CHEM., vol. 274, 1999, pages 34450 - 34458
BARTUS, R.T: "On neurodegenerative-diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis", EXP. NEUROL., vol. 163, 2000, pages 495E529
BACH FH: "Heme Oxygenase-1 and Transplantation Tolerance Human", IMMUNOLOGY, vol. 67, 2006, pages 430 - 432
BETTI, M., GAZZ. CHIM. ITAL., vol. 30 II, 1900, pages 301
BETTI, M, GAZZ. CHIM ITAL., vol. 33 II, 1903, pages 2
BRESGEN N; KARLHUB2R G; KRIZBAI I; BAUER H ET AL.: "Oxidative stress in cultured cerebral endothelial cells induces chromosomal abe rrations, micronuclei, and apoptosis", J. NEUROSCI. RES., vol. 72, 2003, pages 327 - 333
BRITTON RS; LEICESTER KL; BACON BR: "Iron toxicity and chelation therapy", INT J HEMATOL., vol. 76, 2002, pages 219 - 228
CALABRESE V; BUTTERFIELD DA; STELLA AM: "Nutritional antioxidants and the heme oxygenase pathway of stress tolerance: novel targets for neuroprotection in Alzheimer's disease", ITAL. J. BIOCHEM., vol. 52, 2003, pages 177 - 181
JAZWA A; CUADRADO A: "Targeting heme oxygenase-1 for neuroprotection and neuromflammation in neurodegenerative diseases", CURR DRUG'TARGETS., vol. 11, 2010, pages 1517 - 1531
CHEN K; GUNTER K; MAINES MD: "Neurons overexpressing heme oxygenase-1 resist oxidative stress-mediated cell death", J. NEUROCHEM, vol. 75, 2000, pages 304 - 313
CHONG ZZ; SHANG YC; MU Y; CUI S; YAO Q; MA ESE K: "Targeting erythropoietin for chronic neurodegenerative diseases", EXPERT OPIN THER TARGETS., vol. 17, 2013, pages 707 - 720
CHOWDHURY R; HARDY A; SCHOFIELD CJ: "The human oxygen sensing machinery and its manipulation", CHEM SOC REV., vol. 37, 2008, pages 1308 - 1319
CHU J; LI JG; CEBALLOS-DIAZ ,C; GOLDE T; PRATICÒ D: "The influence of 5-lipoxygenase- on Alzheimer's disease-related tau pathology: in vivo and'in vitro evidence", BIOL PSYCHIATRY, vol. 74, 2013, pages 321 - 328
RUTH COLE; JEAN DE VELLIS: "Preparation of Astrocyte, Oligodendrocyte, and Microglia Cultures from Primary Rat Cerebral Cultures", PROTOCOLS FOR NEURAL CELL CULTURE KÖNYV, vol. 9, 2001, pages 117 - 127
DE CALIGNON A; FOX LM; PITSTICK R; CARLSON GA; BA'CSKA BJ; SPIRES-JONES TL; HYMAN BT: "Caspase activation precedes and leads to tangles", NATURE, vol. 464, 2010, pages 1201 - 1204
DEGTEREV A; YUAN J: "Expansion and evolution of cell death programmes", NAT REV MOL CELL BIOL., vol. 9, 2008, pages 378 - 390
ECKERT A; MARQUES CA; KEIL U; SCHUSSEL K; MULLER WE: "Increased apoptotic cell death in sporadic and genetic Alzheimer's disease", ANN N Y ACAD SCI., vol. 1010, 2003, pages 604 - 609
EMMEL EA; VERWEIJ CL; DURAND DB; HIGGINS KM; LACY E; CRABTREE GR: "Cyclosporin A specifically inhibits function of the nuclear proteins involved in T cell activation", SCIENCE, vol. 246, 1989, pages 1617 - 1620
FIRUZI 0; ZHUO J; CHINNICI CM; WISNIEWSKI T: "Pratico D.5-Lipoxygenase gene disruption reduces amyloid-beta pathology in a mouse model of Alzheimer's disease", FASEB J, vol. 22, 2008, pages 1169 - 1178
FREDERICKSON CJ; KOH JY; BUSH AI: "The neurobiology of zinc in health and disease", NAT REV NEUROSCI., vol. 6, 2005, pages 449 - 462
GAGE GJ, DR; SHAIN W: "Whole Animal Perfusion Fixation 'for Rodents", J. VIS. EXP., vol. 65, 2012, pages E3564
FRISCH MJ; TRUCKS GW; SCHLEGEL HB; SCUSERIA GE ET AL.: "Gaussian 09 rev. B01", 2010, GAUSSIAN, INC.
HAGEMEYER N; BORETIUS S; OTT C; VON. STREITBERG A; WELPMGHUS H; SPERLING S; FRAHM J; SIMONS M; GHEZZI P; EHRENREICH H: "Erythropoietin attenuates neurological and histological consequences of toxic demyelination in mice", MOL MED., vol. 18, 2012, pages 628 - 635
HENGARTNER MO: "The biochemistry of apoptosis.", NATURE, vol. 407, 2000, pages 770 - 776
HUDRY E; WU HY; ARBEL-ORNATH M; HASHIMOTO T; MATSOUAKA R; FAN Z; SPIRES-JONES TL; BETENSKY RA; BACSKAI BJ; HYMAN BT: "Inhibition of the NFAT pathway alleviates amyloid ? neurotoxicity in a mouse model of Alzheimer's disease", J NEUROSCI., vol. 32, 2012, pages 3176 - 3192
IDRISS NK; BLANN AD; LIP GY: "Hemoxygenase-1 in cardiovascular disease", J AM COLL CARDIOL., vol. 52, 2008, pages 971 - 978
JANCIAUSKIENE S; WRIGHT HT; LINDGREN S: "Fibrillar Alzheimer's amyloid peptide Abeta(1-42) stimulates low density lipoprotein binding and cell association, free radical production and cell cytotoxicity in PC12 cells", NEUROPEPTIDES, vol. 33, 1999, pages 510 - 516
JAZWA A; CUADRADO A: "Targeting heme oxygenase-1 for neuroprotection and neuroinflammation in neurodegenerative diseases", CURR DRUG TARGETS., vol. 11, 2010, pages 1517 - 1531
KARTTUNEN J; SHASTRI N.: "Measurement of ligand-induced activation in single viable T cells using ''the lacZ reporter", GENE. PROC NATL ACAD SCI USA., vol. 88, 1991, pages 3972
KOH JY; SUH SW; GWAG BJ; HE YY; HSU CY; CHOI DW: "The role of zinc in selective neuronal death after transient global cerebral ischemia", SCIENCE, vol. 272, 1996, pages 1013 - 1016
KORKMAZ S; BARNUCZ E; LOGANATHAN S; LI S; RADOVITS T; HEGEDUSP; ZUBAREVICH A; HIRSCHBERG K; WEYMANN A; PUSKÁS LG: "Q50, an iron-chelating. and zinc-complexing-agent, improves cardiac function in rat, models of ischemia/reperfusion-induced myocardial injury", CIRC J., vol. 77, 2013, pages 1817 - 1826
LEWÉN. A; MATZ. P; CHAN PH: "Free, radical pathways in CNS injury", J NEUROTRAUMA., vol. 17, 2000, pages 871 - 890
LI C; HOSSIENY P; WU BJ; QAWASMEH A; BECK K; STOCKER R: "Pharmacologic induction of heme oxygenase-1", ANTIOXID REDOX SIGNAL., vol. 9, 2007, pages 2227 - 2239
LIU F; GRUNDKE-IQBAL I; IQBAL K; ODA Y; TOMIZAWA K; GONG CX.: "Truncation and activation of calcineurin A by calpain I in Alzheimer disease brain", J BIOL CHEM., vol. 280, 2005, pages 37755 - 37762
LUE LF1; KUO YM; ROHER AE; BRACHOVA L; SHEN Y; SUE L; BEACH T; KURTH JH; RYDEL RE; ROGERS J: "Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease", AMJ PATHOL, vol. 155, 1999, pages 853 - 862
LYNCH AM; LYNCH MA: "The age-related increase in IL-1 type I receptor in rat hippocampus is coupled with an increase in caspase-3 activation", EUR J NEUROSCI, vol. 15, 2002, pages 1779 - 1788
MACHOVA, E.; JAKUBIK, J.; MICHAL, P; OKSMAN, M.; I VONEN, H; TANILA, H.; DOLEZAL, V: "Impairment of muscarinic transmission in transgenic APPswe/PSldE9 mice", NEUROBIOL. AGING, vol. 29, 2008, pages 368E378
MAMMIS A; MCINTOSH TK; MANIKER AH: "Erythropoietin as a neuroprotective agent in traumatic brain injury.", SURG NEUROL., vol. 71, 2009, pages 527 - 531
MATTSON MP: "Neuronal life-and-death signaling, apoptosis, and neurodegenerative disorders", ANTIOXID REDOX SIGNAL, vol. 8, 2006, pages 1997 - 2006
C. MCLEAN; R. CHERNY; F. FRASER; S. FULLER; M. SMITH; K.BEYREUTHER; A. BUSH; C. MASTERS: "Soluble pool of A? amyloid as a determinant of severity of neurodegeneration in Alzheimer's Disease", ANN NEUROL, vol. 46, 1999, pages 860 - 866
MERELLI A; CALTANA L; LAZAROWSKI A; BRUSCO A: "Experimental evidence of the potential use of erythropoietin by intranasal administration as a neuroprotective agent in cerebral hypoxia", DRUG METABOL DRUG INTERACT., vol. 26, 2011, pages 65 - 69
MERELLI A; CZORNYJ L; LAZAROWSKI A: "Erythropoietin: a neuroprotective agent in cerebral hypoxia, neurodegeneration, and epilepsy", CURR PHARM DES., vol. 19, 2013, pages 6791 - 801
MITTL PRE; MARCO SD; KREBS JF; KARANEWSKY DS; PRIESTLE JP; TOMASELLI KJ; GRUTTER MG: "Structure of Recombinant Human CPP32 in Complex with the Tetrapeptide Acetyl-Asp-Val-Ala-Asp Fluoromethyl Ketone", J BIOL CHEM., vol. 272, 1997, pages 6539 - 6547
NEWTON SS; FOURNIER NM; DUMAN RS: "Vascular growth factors in neuropsychiatry", CELL MOL LIFE SCI, vol. 70, 2013, pages 1739 - 1752
NGUYEN T; HAMBY A; MASSA SM: "Clioquinol down-regulates mutant huntihgtin expression in vitro and mitigates pathology in a Huntington's disease mouse model", PROC NATL ACAD SCI U S A., vol. 102, 2005, pages 11840 - 11845
NOH MY; CHO KA; KIM H; KIM, SM; KIM SH: "Erythropoietin modulates the immune-inflammatory response of a SOD1(G93A) transgenic mouse model of amyotrophic lateral sclerosis (ALS", NEUROSCI LETT, vol. 574, 2014, pages 53 - 58
ORRENIUS S: "Reactive oxygen species in mitochondria-mediated cell death", DRUG METAB REV, vol. 39, 2007, pages 443 - 455
PALMIERI, G: "A Practical o-Hydroxybenzylamines Promoted Enantioselective Addition of Dialkylzmcs to Aldehydes with Asymmetric Amplification", TETRAHEDRON ASYMMETRY, vol. 11, 2000, pages 3361
J. P. PHILLIPS; R. W. KEOWN; Q. FERNANDO: "The reaction of anils with 8-quinolinol", J. ORG. CHEM., vol. 19, 1954, pages 907
J. P. PHILLIPS; E. M. BARRALL: "Notes - Betti Reactions of Some Phenols.", J. ORG. CHEM., vol. 21, 1956, pages 692
J. P. PHILLIPS: "The Reactions Of 8-Quinolinol", CHEM. REV., vol. 56, 1956, pages 286
REGLAND B; LEHMANN W; ABEDINI I; BLENNOW K; JONSSON M; KARLSSON I; SJOGREN M; WALLIN A; XILINAS M; GOTTFRIES CG: "Treatment of Alzheimer's disease with clioquinol", DEMENT GERIATR COGN DISORD., vol. 12, 2001, pages 408 - 414
SAMA MA; MATHIS DM; FURMAN JL; ABDUL HM; ARTIUSHIN IA; KRANER SD; NORRIS CM.: "Interleukin-lbeta-dependent signaling between astrocytes and neurons depends critically on astrocytic calcineurin/NFAT activity", J BIOL CHEM., vol. 283, 2008, pages 21953 - 21964
SCHAFER S; PAJONK FG; MULTHAUP G; BAYER TA: "Copper and clioquinol treatment in young APP transgenic and wild-type mice: effects on life expectancy, body weight, and metal-ion levels", J MOL MED (BERL)., vol. 85, 2007, pages 405 - 413
SMIRNOVA NA; RAKHMAN I; MOROZ N; BASSO M; PAYAPPILLY J; KAZAKOV S; HERNANDEZ-GUZMAN F; GAISINA IN; KOZ KOWSKI AP; RATAN RR: "Utilization of an in vivo reporter for high throughput identification of branched small molecule regulators of hypoxic adaptation", CHEM BIOL., vol. 17, 2010, pages 380 - 391
STEFANIS L: "Caspase-dependent and -independent I neuronal death: two distinct pathways to neuronal injury", NEUROSCIENTIST, vol. 11, 2005, pages 50 - 62
SZABO C: "Mechanisms of cell necrosis", CRIT CARE MED., vol. 33, 2005, pages 530 - 534
UZ, T.; PESOLD, C.; LONGONE, P.; MANEV, H.: "Agingassociated up-regulation of neuronal 5-lipoxygenase expression: putative role in neuronal vulnerability", FASEB J., vol. 12, 1998, pages 439 - 449
VERKADE JMM; VAN HEMERT LJC; QUAEDFLIEG PJLM; RUTJES FPJT: "Organocatalysed asymmetric Mannich reactions", CHEM. SOC. REV., vol. 37, 2008, pages 29
J. WANG; D.W. DICKSON; J.Q. TROJANOWSKI; V.M. LEE: "The levels of soluble versus,insoluble brain Abeta distinguish Alzheimer's disease from normal and pathologic aging", EXP NEUROL., vol. 158, 1999, pages 328 - 337
XUE YQ; ZHAO LR; GUO WP; DUAN WM.: "Intrastriatal administration of erythropoietin protects dopaminergic neurons and improves neurobehavioral outcome in a rat model of Parkinson's disease", NEUROSCIENCE, vol. 146, 2007, pages 1245 - 1258
ZHANG, C.P.; ZHU, L.L.; ZHAO, T.; ZHAO, H.; HUANG, X.; MA, X.; WANG, H.; FAN, M: "Characteristics of neural stem cells expanded in lowered oxygen and the potential role of hypoxia-inducible factor-1Alpha", NEUROSIGNALS, vol. 15, 2006, pages 259 - 265
ZHOU, Y.; WEI, E. Q.; FANG, S. H.; CHU, L.; WANG, M. L.; ZHANG, W. P.; YU, G. L.; YE, Y. L.; LIN, S. C.; CHEN, Z: "Spatio-temporal properties of 5-lipoxygenase expression and activation in the brain after focal cerebral ischemia in rats", LIFE SCI., vol. 79, 2006, pages 1645 - 1656
CAHALAN MD: "Murine skin transplantation", J VIS , ?X?, vol. 11, 2008, pages 634
Attorney, Agent or Firm:
GIBER, János (Somfai and Partners Industrial Rights Co. Ltd.Pozsonyi út 38, Budapest, HU)
Download PDF:
Claims:
Claims

1. Novel R-enantiomeric derivatives of 8-hydroxyquinoline

derivatives of general formula (I) and novel S-enantiomer ic derivatives of 8-hydroxyquinoline derivatives of general formula (II), and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof,

R-enantiomer S-enantiomer wherein

in general formulas (I) and (II)

Ri represents lower alkyl group; lower cycloalkyl group; aryl group; aralkyl group or 5 or 6 membered heteroaryl group comprising 1; 2 or 3 nitrogen; oxygen or sulfur atoms or combinations thereof wherein the above groups can be optionally substituted with 1; 2; 3; or 4 electron withdrawing groups or electron donating groups in ortho; meta or para positions;

R2 represents a hydrogen atom; aryl group; aralkyl group;

5 or 6 membered heteroaryl group comprising 1; 2 or 3 nitrogen; oxygen or sulfur atoms or combinations thereof wherein the above groups can be optionally substituted with 1; 2; 3; or 4 electron withdrawing groups or electron donating groups in ortho; meta or para positions;

P-1418 R3 represents a hydrogen atom; lower alkyl group; -CH2F; -CHF2; -CF3; -CH2CH2F; -CH2CHF2; -CH2CF3; -CH2OR5; - CH2CH2OR6; or -CH2-NR7R8 group;

R4 represents a hydrogen atom; halogen atom; methylthio group; methylsulfinyl group; methylsulfonyl group; or azido group;

R5 represents a hydrogen atom; or lower alkyl group;

R6 represents a hydrogen atom; or lower alkyl group;

R7 represents a hydrogen atom; or lower alkyl group;

R8 represents a hydrogen atom; or lower alkyl group;

R7 and R8 represents jointly -(CH2)n- group; or - CH2CH2OCH2CH2- group or-CH2CH2SCH2CH2- group or- CH2CH2NR9CH2CH2- group,

wherein

n is 4, 5 or 6;

R9 represents a lower alkyl group; or -COR10 group,

R10 represents a hydrogen atom; lower alkyl group; methoxy group; or ethoxy group;

in the general formula (I)

X represents a hydrogen substituted C atom with "R" configuration;

in the general formula (II)

Y represents a hydrogen substituted C atom with "S" configu ation;

with the proviso that

R1 cannot represent non-substituted phenyl group, in case

R2 represents a non-substituted phenyl group; or non- substituted 2-pyridyl group; or 4-carboxyphenyl group; or 2-carboxyphenylgroup;

R3 represents a hydrogen atom or methyl group;

R4 represents hydrogen atom or chlorine substituent; and 18 R1 cannot represent 3, 4-dimethylphenyl group, in case

R2 represents a non-substituted 2-pyridyl group;

R3 represents methyl group and

R4 a hydrogen atom;

and

R1 cannot represent 2-furyi group, in case

R2 represents a non-substituted 2-pyridyl group;

R3 represents a hydrogen atom and

R4 represents a chlorine substituent,

and

R1 cannot represent a non-substituted 2-pyridyl group, in case

R2 represents 5-methylisoxazol-3-yl group;

R3 represents a hydrogen atom;

R4 represents a hydrogen atom.

Novel R-enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formula (Ι') according to claim 1 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof,

R-enantiomer

wherein

in general formula (Ι')

R1' represents an aryl group substituted with an electron withdrawing group in meta or para position, or an aryl group substituted with an electron donating group in ortho, meta or para position; or a double-substituted aryl group with electron withdrawing groups in meta and para positions; or an aryl group double- substituted with electron withdrawing groups in ortho and para positions; or a substituted or unsubstituted heteroaryl group;

R2' represents an aryl group substituted with an electron withdrawing group in para position, or an aryl group substituted with an electron donating group in ortho, meta or para position; or an unsubstituted

heteroaromatic group or a heteroaromatic or aryl group substituted with alkyl group and/or with electron withdrawing groups in ortho, meta or para positions;

R3' represents a hydrogen atom

R4' represents a hydrogen atom and

in general formula (Ι')

X represents a hydrogen substituted C atom with "R"

configuration .

3. Novel S-enantiomeric derivatives of 8-hydroxyquinoline

derivatives of general formula (II' ) according to claim 1 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof,

S-enantiomer

wherein

in general formula (II' )

R1' represents an aryl group substituted with an electron withdrawing group in meta or para position, or an aryl group substituted with an electron donating group in

P-1418 ortho, meta or para position; or a double-substituted aryl group with electron withdrawing groups in meta and para positions; or an aryl group double- substituted with electron withdrawing groups in ortho and para positions; or a substituted or unsubstituted heteroaryl group;

R2 represents an aryl group substituted with an electron withdrawing group in para position, or an aryl group substituted with an electron donating group in ortho, meta or para position; or an unsubstituted

heteroaromatic group or a heteroaromatic or aryl group substituted with alkyl group and/or with electron withdrawing groups in ortho, meta or para positions;

R3' represents a hydrogen atom;

R4' represents a hydrogen atom and

in general formula (Ι')

Y represents a hydrogen substituted C atom with "S" configuration.

4. Novel R-enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formula (I'') according to any one of claims 1 or 2 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof,

R-enantiomer

wherein

in general formula (I'')

P-1418 R1 represents a phenyl or pyridyl group optionally single or double substituted with a trifluoromethyl group, hydroxy group, fluorine atom or isopropoxy group;

R represents a phenyl group optionally single or double

substituted with a trifluoromethyl group or methoxy- carbonyl group; or a pyridyl, pyrimidyl, pyrrolidinyl, oxazolidinyl group optionally single or double substituted with a methyl group or fluorine atom;

R3" represents hydrogen atom;

R4" represents hydrogen atom; and

in general formula (I'')

X represents a hydrogen substituted C atom with "R"

configuration;

5. Novel S-enantiomeric derivatives of 8-hydroxyquinoline

derivatives of general formula (II'') according to any one of claims 1 or 3 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof,

S-enantiomer

wherein

in general formula (II'')

R1" represents a phenyl or pyridyl group optionally single or double substituted with a trifluoromethyl group, hydroxy group, fluorine atom or isopropoxy group;

R2" represents a phenyl group optionally single or double with a trifluoromethyl group or methoxy-carbonyl group; or a pyridyl, pyrimidyl, pyrrolidinyl, oxazolidinyl group

P-1418 optionally single or double substituted with a methyl group or fluorine atom;

R3" represents hydrogen atom;

R4" represents hydrogen atom and

in general formula (II'')

Y represents a hydrogen substituted C atom with VS"

configuration .

6. Novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 1 to 5 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof characterized in listed as follows : 7- [ (R) - [ (4- ethylpyrimidin-2-yl) amino] [4-

(trifluoromethyl ) phenyl] -methyl] quinolin-8-ol) ,

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4- ( tr ifluoromethyl ) phenyl] -methyl] q inolin-8-ol ,

Potassium 7- [ (R) - [ ( 4-methylpirimidin-2-yl ) amino] [4- (trifluoromethyl) -phenyl ] methyl ] quinolin-8-olate,

Potassium 7- [ (S) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- ( trifluoromethyl ) -phenyl ] methyl ]quinolin-8-olate,

Natrium 7- [ (R) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinolin-8-olate,

7- [ ( R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) - phenyl]methyl] quinolin-8-ol fumarate,

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl ) - phenyl ] methyl ] quinolin-8-ol fumarate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] -methyl] quinolin-8-ol zinc complex, 7- [ (R) - [ (6-Methylpyridin-2-yl) amino] 4-nitrophenyl ) methyl ] - quinolin-8-ol,

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] 4-nitrophenyl ) methyl ] - quinoline-8-ol,

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] 3-hydroxyphenyl ) methyl ] -

P-1418 quinoline-8-ol,

7- [ (S) - [ ( 6-Methylpyridin-2-yl ) amino] 3-hydroxyphenyl ) methyl ] - quinoline-8-ol,

7- [ (R) - [ ( 6-Methylpyridin-2-yl ) amino] ( 4-hydroxy-3- methoxiphenyl ) methyl] -quinoline-8-ol,

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] ( 4-hydroxy-3- methoxiphenyl ) methyl] -quinol.ine-8-ol ,

7- [ ( ) - [ (6-Methylpyridin-2-yl) amino] ( 5-bromopyridin-2- yl) methyl] -quinoline-8-ol ,

7- [ (S) - [ ( 6-Methylpyridin-2-yl ) amino] ( 5-bromopyridin-2- yDmethyl] -quinoline-8-ol ,

7- [ (R) - [ ( 6-Methylpyridin-2-yl ) amino] 2-hydroxyphenylmethyl ] - quinoline-8-ol,

~l - [ ( S ) - [ ( 6- ethylpyridin-2-yl ) amino] 2-hydroxyphenylmethyl ] - quinoline-8-ol,

5-Chloro-7- [ (R) - [ (4-methylpyrimidin-2-yl) mino] [4- (trif luoromethyl) phenyl] methyl] quinolin-8-ol,

5-Chloro-7- [ (S) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol,

5-Chloro-7- [ (R) - [ ( 6-methylpyridin-2-yl ) amino] [4- (trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol ,

2-Methyl-7-[ (R) - [ (4-methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl) phenyl ] methyl] quinoline-8-ol,

2-Methyl-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline-8-ol,

2- [ (Dimethylamino) methyl] -7- [ (R) - [ (4-methylpyrimidin-2- yl) amino] [4- (trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol, 2- [ (Dimethylamino) methyl] -7- [ (S) - [ ( 4-methylpyrimidin-2- yl) amino] [4- (trifluoromethyl) phenyl ] methyl ] quinoline-8-ol, 2- [ (Dimethylamino)methyl] -7- [ (R) - [ ( 4-methylpyridin-2- yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol, 2- [ (Dimethylamino)methyl] -7- [ (S) - [ ( 4-methylpyridin-2- yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol, 5-Nitro-7- [ (R) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline-8-ol,

P-1418 5-Nitro-7- [ (S) - [ ( -methylpyrimidin-2-yl ) amino] [4-

(trifluoromethyl) phenyl] methyl] quinoline-8-ol ,

7- [ (R) - [ (Pyridin-2-yl) [4- ( trifluoromethyl ) phenylamino] methyl ] quinoline-8-ol ,

7-[ (S)-[ (Pyridin-2-yl) [4- (trifluoromethyl ) phenylamino] methyl ] quinoline-8-ol

2- (Hydroxymethyl) -7- [ (R) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline-8-ol,

2- (Hydroxymethyl) -7- [ (S) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline- 8-ol .

7. Novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 1 to 6 and

pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof characterized in that the metal complexes are iron, zinc or copper complexes.

8. Novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 1 to 7 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof characterized in that the metal complex is zinc complex.

9. Novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 6 to 8 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof, characterized in that the enantiomeric derivative is 7- [ ( R) - [ ( 4 -Methylpyrimidin-2- yl) amino] [4- (trifluoromethyl) phenyl] -methyl] quinolin-8-ol .

10. Novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 6 to 8 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof, characterized in that the

P-1418 enantiomeric derivative is 7- [ (S) - [ ( 4 -Methylpyrimidin-2- yl) amino] [ 4 - (trifluoromethyl) phenyl]—methyl] quinolin-8-ol .

.. Novel enantiomeric derivatives of 8-hydroxyquinoline derivatives according to any one of claims 6 to 8 and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof, characterized in that the salts and the complex are listed as follows:

Potassium 7 - [ (R) - [ ( 4-methylpirimidin-2-yl ) ami o ] [4-

( trifluoromethyl) -phenyl ] methyl ] quinolin-8-olate,

Potassium 7- [ (S) - [ ( 4-methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl) -phenyl ] methyl ] quinolin-3-olate,

Natrium 7- [ (R) - [ ( -methylpyrimidin-2-yl ) amino] [4-

(trifluoromethyl) phenyl] methyl] quinolin-8-olate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) - phenyl ] methyl ] quinolin-8-ol fumarate,

7- [ (S) - [ ( 4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) - phenyl ] methyl ] quinolin-8-ol fumarate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl) henyl] -methyl] quinolin-8-ol zinc complex.

'.. Medicinal and/or pharmaceutical compositions comprising the novel enantiomeric derivatives of 8-hydroxyquinoline

derivatives according to any one of claims 1 to 11 and/or pharmaceutically acceptable salts and complexes with divalent or polyvalent metals thereof.

S . Medicinal and/or pharmaceutical compositions according to claim 12, characterized in comprising the novel enantiomeric derivatives of 8-hydroxyquinoline derivatives according to any one of claims 1 to 11 and/or pharmaceutically acceptable salts and metal complexes thereof and inert, pharmaceutically acceptable, solid or liquid carrier and/or excipient.

1418

14. Medicinal and/or pharmaceutical compositions according to claims 12 to 13, characterized in comprising medicinal acceptable carriers and/or excipients as following listed: starch, gelatinized starch, cellulose, microcrystalline cellulose or cellulose-derivatives, lactose, lactose

monohydrate, talcum, mannitol, sodium chloride, sodium carbonate, saccharose, maltose, calcium carbonate, colloidal anhydrous silicon dioxide, stearic acid, magnesium stearate and/or isomalt.

15. Medicinal and/or pharmaceutical compositions according to any one of claims 12 to 14, characterized in that the composition is solid, semi-solid or liquid. 16. Medicinal and/or pharmaceutical compositions according to any one of claims 12 to 15, characterized in that the composition is tablet, inhalation powder, capsule,

suppository or solution for injection. 17. Medicinal and/or pharmaceutical compositions according to any one of claims 12 to 16, characterized in that the composition is tablet.

18. A process for the preparation of medicinal and/or

pharmaceutical compositions according to any one of claims 12 to 17, characterized in that the enantiomeric derivatives according to any one of claims 1 to 11 and/or

pharmaceutically acceptable salts and/or metal complexes thereof are mixed with pharmaceutically applicable inert, solid or liquid carriers and/or excipients according to any one of claims 12 to 17 then formulated to a medicinal and/or pharmaceutical composition according to any one of claims 12 to 17 by using the usual, standard formulation technics.

P-1418 «<

19. A novel, stereoselecti e process for tiie preparation of novel enantiomeric derivatives of general formulas (I) and (II) according to claim 1 and pharmaceutically acceptable salts

and/or complexes thereof according to any one of claims 1 or 7 to 8, characterized in reacting an 8-hydroxyquinoline derivative of general formula (III)

with an amine of general formula (IV) ,

R2 NH2 (IV) and with an oxo-compound of general formula

using quinidine or quinine as catalyst, obta

P-1418

enantiomerically pure R- or S-enantiomeric derivative depending on the used catalyst, where Rl, R2 , R3, R4 , X and Y represent the groups according to claim 1.

20. A process according to claims 1 and 19 for the preparation of novel R-enantiomeric derivatives of general formulas (Ι') and (I'') and specifically named by chemical names according to any one of claims 2, 4, 6, 9 and 11 and pharmaceutically acceptable salts and/or complexes thereof according to any one of claims 1 or 7 to 8, characterized in that the symbols of the general formula are according to the claims, and X represents a hydrogen substituted carbon atom with "R" configuration and quinidine is used as catalyst.

21. A process according to claims 1 and 19 for the preparation of novel S-enantiomeric derivatives of general formulas (II' ) and (II'') and specifically named by chemical names according to any one of claims 3, 5, 6, 10 and 11 and pharmaceutically acceptable salts and/or complexes thereof according to any one of claims 1 or 7 to 8, characterized in that the symbols of the general formula are according to the claims, and Y represents a hydrogen substituted carbon atom with "S" configuration and quinine is used as catalyst.

P-1418

2. A general process according to any one of claims 1, 19 and 20 for the preparation of novel R-enantiomeric derivatives according to the claims characterised by that, to a solution of 50 mmol of.quinidine in the proper solvent (180 ml), formic acid (0.84 equivalent) was added in inert

athmospherethen amine derivative (1 equivalent), aldehyde compound (1 equivalent) and 8-hyd.roxyquinoline derivative (1.2 equivalent) were added then the mixture was stirred at a proper temperature until the R-enantiomer according to any one of claims 1, 19 and 20 was formed in a desired quantity then the reaction mixture was concentrated in vacuum to its third volume, the residue was dissolved in dichloromethane, the solution was washed with 1 M NaOH and extracted further by 1M NaOH 6 times, a non-polar solvent was added to the organic phase then the dichloromethane was evaporated off and the solution obtained was added to 100 ml of 3 M HCl, the phases were separated, the organic layer was extracted with 3 M HCl solution, too the combined HCl phase methyl-t-butyl- ether was added then the pH of the biphasic system was adjusted with 40% NaOH solution to four and then

the precipitated quinidine was filtered off, the biphasic filtrate was separated then the water layer was washed twice by methyl-t-butyl-ether , the combined organic phase was dried on sodium sulfate and filtered, a proper solvent was added to the filtrate, the methyl-t-butyl-ether was evaporated off, the residue was stirred at room temperature for 16 hours, the precipitated racemic crystals were filtered then the mother liquor was concentrated in reduced pressure, and the

remaining pure R-enantiomer according to any one of claims 1, 19 and 20 was dissolved in 40 ml of isopropanol, stirred at room temperature for 48 hours then the precipitated crystals were filtered off, to give pure crystalline R-enantiomer according to any one of claims 1, 19 and 20.

-1418

23. A general process according to claim 22 for the preparation of novel R-enantiomeric derivatives characterized in that the proper solvent of the reaction mixture and added to the filtrate is acetonitrile.

24. A general process according, to any one of claims 1, 19 and 21 for the preparation of novel S-enantiomeric derivatives according to the claims, characterized in that to a solution of quinine (55 mmol) in a proper solvent (300 ml) in inert athmosphere, formic acid (0.8 equivalent), amine (2.5 equivalent), aldehyde derivative (3.7 equivalent), and finally 8-hydroxyquinoline derivative (1.0 equivalent) were added then the mixture was stirred at a proper temperature until the S-enantiomer according to claims 1, 19 and 21 was formed in a desired quantity, the solvent was evaporated off in vacuum, the residue was dissolved in dichloromethane and chromatographed on silica gel then the fractions containing the product were collected and the solvent was evaporated off, the raw product obtained was purified by normal phase

Flash chromatography using hexane-ethyl-acetate gradient then the fractions containing the product were collected and concentrated, the residue was dissolved in 2-propanol and after 2 hours stirring the precipitated racemic crystals were filtered off then the mother liquor was concentrated in vacuum to get the pure S-enantiomer according to claims 1, 19 and 21, and the raw product was isolated and purified by the usual methods. 25. A general process according to claim 24 for the preparation of novel S-enantiomeric derivatives characterized in that the proper solvent of the reaction mixture is acetonitrile and the raw S-enantiomer according to claims 1, 19 and 21 is isolated by filtration, centrifugation and is purified by recrystallization or chromatography.

P-1418

26. A process according to any one of claims 1, 19, 20 and 22, 23, characterized in that the 7- f (R) - [ ( 4-Methylpyrimidin-2- yl) amino] [4- (trifluorome thyl ) phenyl ] ethyl ] quinolin-8-ol enantiomeric derivative according to any one of claims 6 and 9 is prepared.

27. A process according to any one of claims.1, 19, 21 and 24, 25, characterized in that the 7- [ (S ) - [ ( 4-Methylpyrimidin-2- yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinolin-8-ol enantiomeric derivative according to any one of claims 6 and 10 is prepared.

28. A process according to any one of claims 1, 19 to 23,

characterized in that the salts and metal complexes of enantiomeric derivatives according to any one of claims 6 to 8 and 11 are prepared.

29. Novel 8-hydroxyquinoline enantiomeric derivatives according to claims 1 to 11 and the medicinal and/or pharmaceutical compositions according to claim 12 and the applications according to claims 32 to 49, characterized in that oral medicinal and /or pharmaceutical composition is prepared. 30. Novel 8-hydroxyquinoline enantiomeric derivatives according to claims 1 to 11 and the medicinal and/or pharmaceutical compositions according to claim 12 and the applications according to claims 32 to 49, characterized in that buccal, sublingual or rectal medicinal and /or pharmaceutical composition is prepared.

31. Novel 8-hydroxyquinoline enantiomeric derivatives according to claims 1 to 11 and the medicinal and/or pharmaceutical compositions according to claim 12 and the applications according to claims 32 to 49, characterized in that

P-1418 parenteral medicinal and /or pharmaceutical composition is prepared.

32. The use of novel 8-hydroxyquinoline enantiomeric

derivatives according to any one -of claims 1 to 11 or pharmaceutically acceptable salts or metal complexes thereof for manufacture of medicinal and/or pharmaceutical

composition suitable for treatment and/or prevention of neuropsychiatric diseases.

33. The use according to claim 32, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of anxiety disorders,

schizophrenia, depression, bipolar disorder.

34. The use according to claim 33 characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of schizophrenia.

35. The use according to claim 33 characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of depression and/or bipolar disorder .

36. The use of novel 8-hydroxyquinoline enantiomeric

derivatives according to any one of claims 1 to 11 or pharmaceutically acceptable salts or metal complexes thereof for manufacture of medicinal and/or pharmaceutical

composition suitable for treatment and/or prevention of neurologic diseases.

37. The use according to claim 36, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of epilepsy, amnesia, different

P-1418 memory disorders, ' cognitive functional problems, neurodegenerative diseases.

38. The use according to claim 37, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of amnesia, different memory disorders, cognitive functional problems.

39. The use according to claim 37, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of epilepsy and/or different memory disorders.

40. The use according to claim 37, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of neurodegenerative diseases.

41. The use according to claim 40, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of Alzheimer's disease,

Huntington disease, Parkinson disease, Wilson disease, amyotrophic lateral sclerosis (ALS) .

42. The use according to claim 41, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of Alzheimer's disease.

43. The use according to claim 41, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of amyotrophic lateral sclerosis (ALS) .

44. The use of novel 8-hydroxyquinoline enantiomeric

derivatives according to any one of claims 1 to 11 or

P-1418 pharmaceutically acceptable salts or metal complexes thereof for manufacture of medicinal and/or pharmaceutical

composition suitable for treatment and/or prevention of diseases in connection with transplantations and in

connection with ischemia and reperfusion injuries thereof.

45. The use according to claim 44, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of cardiovascular disorders, blood vessel catastrophes, traumatic injuries,

neurodegenerative traumas, diseases in connection with transplantations and in connection with ischemia and

reperfusion injuries thereof. 46. The use according to any one of claims 44 and 45,

characterized in manufacture of medicinal and/or

pharmaceutical composition suitable for treatment and/or prevention of impairments of the brain, heart, liver, kidney or lung.

47. The use according to claim 46, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for treatment and/or prevention of impairments of traumatic brain injuries .

48. The use according to claim 44, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for inhibition of organ graft rejection. 49. The use according to claim 48, characterized in manufacture of medicinal and/or pharmaceutical composition suitable for inhibition of skin graft rejection.

50. The use according to any one of claims 36 to 43,

characterized in use of novel 8-hydroxyquinoline R-

P-1418 enantiomeric derivative according to any one of claims 1, 2, 4, 6, 9 and 11 or pharmaceutically acceptable salt or metal complex according to any one of claims 1, 7 to 8 thereof. 51. The use of novel 8-hydroxyquinoline enantiomeric

derivatives according to any one of claims 1 to 11 or pharmaceutically acceptable salts or metal complexes thereof for manufacture of cytoprotective, neuroprotective,

cardioprotective medicinal and /or pharmaceutical composition according to claim 12, suitable for treatment and/or prevention of diseases according to any one of claims 32 to 49 and showing favourable binding to metal chelate formin target proteins. 52. The use of cytoprotective, neuroprotective,.

cardioprotective medicinal and/or pharmaceutical composition according to claim 12, suitable for treatment and/or prevention of diseases of patients suffering from diseases associated by cell-death and for protection from cytotoxic attacks.

53. The use of medicinal and/or pharmaceutical composition according to claim 12 for treatment and/or prevention of diseases according to any one of claims 32 to 49, which compositions can be administered by a usual method in medicine .

54. The use according to claim 53 characterized in using oral or buccal or sublingual or rectal or parenteral, medicinal and/or pharmaceutical compositions which can be administered also by intravenous or inhalation method.

55. The use according to any one of claims 53 and 54

characterized in using oral, medicinal and/or pharmaceutical compositions.

P-1418

56. The use of medicinal and/or pharmaceutical compositions according to claim 12 for treatment and/or prevention of diseases according to any one of claims 32 to 49.

57. A cytoprotective, neuroprotective, cardioprotective process for treatment and/or prevention of diseases according to any one of claims 32 to 49 characterized in administering the medicinal and/or pharmaceutical composition according to claim 12 by a usual method in medicine, in a medicinal effective dose.

58. A process according to claim 57 characterized in

administering the medicinal and/or pharmaceutical composition according to claim 12 by oral or buccal or sublingual or rectal or parenteral or intravenous or inhalation method.

59. A process according to claim 57 characterized in

administering the medicinal and/or pharmaceutical composition according to claim 12 by oral method.

60. A process according to claim 57 characterized in

administering for treatment and/or prevention of diseases according to any one of claims 36 to 43 a medicinal and/or pharmaceutical composition comprising the novel 8- hydroxyquinoline R-enantiomeric derivative according to any one of claims 1, 2, 4, 6, 9 and 11 or pharmaceutically acceptable salt or metal complex according to any one of claims 1, 7 to 8 thereof.

P-1418

Description:
ENANTIOMERS OF 8-HYDROXY QUINOLINE DERIVATIVES AND THE

SYNTHESIS THEREOF

Our invention relates to novel R-enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formula (I) and novel S-enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formula (II), and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof,

R-enantiomer S-enantiomer wherein

in general formulas (I) and (II)

Ri represents lower alkyl group; lower cycloalkyl group; aryl group; aralkyl group or 5 or 6 membered heteroaryl group comprising 1; 2 or 3 nitrogen; oxygen or sulfur atoms or combinations thereof wherein the above groups can be optionally substituted with 1; 2; 3; or 4 electron

withdrawing groups or electron donating groups in ortho; meta or para positions; represents a hydrogen atom; aryl group; aralkyl group; 5 or 6 membered heteroaryl group comprising 1; 2 or 3 nitrogen; oxygen or sulfur atoms or combinations thereof wherein the above groups can be optionally substituted with 1 ; 2; 3; or 4 electron withdrawing groups or electron donating groups in ortho; meta or para positions; R 3 represents a hydrogen atom; lower alkyl group; -CH 2 F;

-CHF 2 ; -CF 3 ; -CH 2 CH 2 F; -CH 2 CHF 2 ; -CH 2 CF 3 ; -CH 2 OR 5 ; -CH 2 CH 2 OR 6 ; or -CH 2 -NR 7 R 8 group;

R 4 represents a hydrogen atom; halogen atom; methylthio group methylsulfinyl group; methylsulfonyl group; or azido group R 5 represents a hydrogen atom; or lower alkyl group;

R 6 represents a hydrogen atom; or lower alkyl group;

R 7 represents a hydrogen atom; or lower alkyl group;

R 8 represents a hydrogen atom; or lower alkyl group;

R 7 and R 8 represents jointly -(CH 2 ) n - group; or -CH 2 CH 2 OCH 2 CH 2 - group or -CH 2 CH 2 SCH 2 CH 2 - group or-CH 2 CH 2 NR 9 CH 2 CH 2 - group, wherein

n is 4, 5 or 6;

R 9 represents a lower alkyl group; or -COR 10 group,

R 10 represents a hydrogen atom; lower alkyl group; methoxy group; or ethoxy group;

in the general formula (I)

X represents a hydrogen substituted C atom with "R" configuration;

in the general formula (II)

Y represents a hydrogen substituted C atom with "S" configuration;

with the proviso that

R 1 cannot represent non-substituted phenyl group, in case R 2 represents a non-substituted phenyl group; or non- substituted 2-pyridyl group; or 4-carboxyphenyl group; or 2- carboxyphenylgroup;

R 3 represents a hydrogen atom or methyl group;

R 4 represents hydrogen atom or chlorine substituent;

and

R 1 cannot represent 3, -dimethylphenyl group, in case

R 2 represents a non-substituted 2-pyridyl group; R 3 represents methyl group and

R 4 a hydrogen atom;

and

R 1 cannot represent 2-furyl group, in case

R 2 represents a non-substituted 2-pyridyl group;

R 3 represents a hydrogen atom and

R 4 represents a chlorine substituent

and

R 1 cannot represent a non-substituted 2-pyridyl group, in case

R 2 represents 5-methylisoxazol-3-yl group

R 3 represents a hydrogen atom

R4 represents a hydrogen atom

The subject matter of the invention furthermore relates advantageously to novel R-enantiomeric derivatives of 8- hydroxyquinoline derivatives of general formula (Ι') and novel S-enantiomeric derivatives of 8-hydroxyquinoline derivatives o general formula (II') / and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals,

advantageously iron, copper or zinc complexes thereof,

R-enantiomer S-enantiomer wherein

in general formulas (Ι') and (II' )

R 1 ' represents an aryl group substituted with an electron

withdrawing group in meta or para position, or an aryl group substituted with an electron donating group in ortho, meta or para position; or a double-substituted aryl group with electron withdrawing groups in meta and para

positions; or an aryl group double substituted with electron withdrawing groups in ortho and para positions; or a substituted or unsubstituted heteroaryl group;

R 2 represents an aryl group substituted with an electron

withdrawing group in para position, or an aryl group substituted with an electron donating group in ortho, meta or para position; or an unsubstituted heteroaromatic group or a heteroaromatic or aryl group substituted with alkyl group and/or with electron withdrawing groups in ortho, meta or para positions;

R 3' represents advantageously a hydrogen atom

R 4' represents advantageously a hydrogen atom and

in general formula (I 7 )

X represents a hydrogen substituted C atom with "R"

configuration ;

In general formula (II 7 )

Y represents a hydrogen substituted C atom with "S"

configuration.

The subject matter of the invention furthermore relates advantageously to novel R-enantiomeric derivatives of 8- hydroxyquinoline derivatives of general formula (I 7 ') and novel S-enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formula (II'') and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof,

P-1418 R-enantiomer S-enantiomer

wherein

in general formulas (I'') and (II'')

R 1 represents advantageously a phenyl or pyridyl group

optionally single or double substituted with a

trifluoromethyl group, hydroxy group, fluorine atom or isopropoxy group;

R 2 represents advantageously a phenyl group optionally single or double substituted with a trifluoromethyl group or methoxy-carbonyl group; or a pyridyl, pyrimidyl,

pyrrolidinyl , oxazolidinyl group optionally single or double substituted with a methyl group or fluorine atom; R 3" represents advantageously hydrogen atom;

R 4 " represents advantageously hydrogen atom;

In general formula (I

X represents a hydrogen substituted C atom with "R"

configuration; ;

In general formula (II'')

represents a hydrogen substituted C atom with "S

configuration.

The subject matter of the invention furthermore relates advantageously to novel enantiomeric derivatives of 8- hydroxyquinoline derivatives, and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof,

especially advantageously zinc complexes thereof as listed detailed as follows:

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] -methyl] quinolin-8-ol ) ,

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] -methyl] quinolin-8-ol,

Potassium 7- [ (R) - [ ( -methylpirimidin-2-yl ) amino] [4- ( trifluoromethyl) -phenyl] methyl] quinolin-8-olate,

P-1418 Potassium 7- [ (S)-[ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) -phenyl ] methyl ] quinolin-8-olate,

Natrium 7- [ (R) - [ ( -methylpyrimidin-2-yl ) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinolin-8-olate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl ) - phenyl ] methyl] quinolin-8-ol fumarate,

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) - phenyl ] methyl] quinolin-8-ol fumarate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] -methyl] quinolin-8-ol zinc complex, 7- [ (R) - [ ( 6-Methylpyridin-2-yl ) amino] -nitrophenyl ) methyl] - quinolin-8-ol,

7- [ (S ) - [ ( 6-Methylpyridin-2-yl ) amino] -nitrophenyl ) methyl ] - quinoline-8-ol,

7- [ (R) - [ ( 6-Methylpyridin-2-yl) amino] 3-hydroxyphenyl ) methyl] - quinoline-8-ol,

7- [ (S) - [ ( 6-Methylpyridin-2-yl ) amino] 3-hydroxyphenyl ) methyl] - quinoline-8-ol ,

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] ( 4-hydroxy-3- methoxiphenyl ) methyl ] -quinoline-8-ol,

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] ( -hydroxy-3- methoxiphenyl ) methyl ]-quinoline-8-ol,

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] ( 5-bromopyridin-2- yDmethyl] -quinoline-8-ol,

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] ( 5-bromopyridin-2- yl ) methyl ] -quinoline-8-ol ,

7- [ (R) - [ ( 6-Methylpyridin-2-yl ) amino] 2-hydroxyphenylmethyl] - quinoline-8-ol,

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] 2-hydroxyphenylmethyl] - quinoline-8-ol ,

5-Chloro-7- [ (R) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl ] methyl ] quinolin-8-ol,

5-Chloro-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl) phenyl] methyl] quinoline-8-ol, 5-Chloro-7- [ (R) - [ ( 6-methylpyridin-2-yl) amino] [4- (trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol,

2-Methyl-7- [ (R) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl ] methyl ] quinoline-8-ol ,

2-Methyl-7- [ (S) - [ ( 4-methylpyrimidin-2-yl ) amino] [4- ( trifluoromethyl ) phenyl]methyl] quinoline-8-ol ,

2- [ (Dimethylamino) methyl] -7- [ (R) - [ (4-methylpyrimidin-2- yl) amino] [4- (trifluoromethyl ) phenyl] methyl] quinoline-8-ol , 2- [ (Dimethylamino) methyl] -7- [ (S) - [ (4-methylpyrimidin-2- yl)amino] [ 4- (trifluoromethyl ) phenyl] methyl ] quinoline-8-ol , 2- [ (Dimethylamino)methyl] -7- [ (R) - [ ( 4-methylpyridin-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol, 2- [ (Dimethylamino)methyl] -7- [ (S) - [ (4-methylpyridin-2- yl) amino] [4- (trifluoromethyl) phenyl] methyl ] quinoline-8-ol , 5-Nitro-7 - [ (R) - [ ( -methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl ) phenyl] methyl ] quinoline-8-ol ,

5-Nitro-7- [ (S) - [ ( -methylpyrimidin-2-yl ) amino] [4- ( trifluoromethyl) phenyl]methyl] quinoline-8-ol,

7- [ (R) - [ ( Pyridin-2-yl) [4- (trifluoromethyl) phenylamino] methyl] quinoline-8-ol ,

7- [ (S) - [ ( Pyridin-2-yl) [4- (trifluoromethyl) phenylamino] methyl] quinoline-8 -ol

2- (Hydroxymethyl) -7-[ (R) - [ (4-methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyl ] methyl ] quinoline-8 -ol ,

2- (Hydroxymethyl) -7- [ (S) -[ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl ] quinoline-8-ol .

The subject matter of the invention furthermore relates especially advantageously to the novel enantiomeric derivatives of 8-hydroxyquinoline derivatives, and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, especially advantageously zinc complexes thereof as listed detailed as follows: 7- ί (R) - [ ( -Methylpyrimidin-2-yl ) amino] [4- (trifluoromethyl) phenyljmethyl] quinoline-8-ol,

7-[ (S)-[ (4-Methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl ) phenyljmethyl ] quinoline-8-ol .

The subject matter of the invention furthermore relates especially advantageously to the pharmaceutically acceptable salts and zinc complex of novel enantiomeric derivatives of 8- hydroxyquinoline derivatives, as listed detailed as follows:

Potassium 7- [ (R) - [ ( 4-methylpirimidin-2-yl ) amino] [4- ( trifluoromethyl ) -phenyl ] methyl ] quinoline-8-olate,

Potassium 7- [ (S) - [ ( 4-methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl) -phenyl] methyl] quinoline-8-olate,

Natrium 7- [ (R) - [ (4-methylpyrimidin-2-yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl] quinoline-8-olate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) - phenyl] methyl ] quinoline-8-ol fumarate,

1-[ (S) - [ ( 4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl ) - phenyl] methyl ] quinoline-8-ol fumarate,

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] {4-

(trifluoromethyl) phenyl] -methyl] quinoline-8-ol zinc complex.

The subject matter of the invention furthermore relates to medicinal and/or pharmaceutical compositions comprising novel enantiomeric derivatives of 8-hydroxyquinoline derivatives disclosed by general formulas (I) and (II), advantageously (Ι' ) and (II'), advantageously (I'') and (II'') - henceforth General Formulas according to the Invention - and advantageously named specifically as above, and/or pharmaceutically acceptable salts and/or complexes with divalent or polyvalent metals,

advantageously iron, copper or zinc complexes thereof, as active agent, which compositions are containinq inert,

pharmaceutically acceptable, solid or liquid carrier and/or excipient, advantageously starch, gelatinized starch,

P-1418 cellulose, microcrystalline cellulose or cellulose-derivatives, lactose, lactose monohydrate, talcum, mannitol, sodium

chloride, sodium carbonate, saccharose, maltose, calcium carbonate, colloidal anhydrous silicon dioxide, stearic acid, magnesium stearate or isomalt.

The subject matter of the invention furthermore relates to medicinal and/or pharmaceutical composition, advantageously solid composition, especially advantageously tablet, inhalation powder or capsule, advantageously semi-solid composition, especially advantageously suppository, or advantageously liquid composition especially advantageously solution for injection.

The subject matter of the invention furthermore relates to process for the preparation of medicinal and/or pharmaceutical compositions comprising novel enantiomeric derivatives of 8- hydroxyquinoline derivatives described by General Formulas according to the Invention and advantageously named

specifically as above, and pharmaceutically acceptable salts and/or complexes with divalent or polyvalent metals,

advantageously iron, copper or zinc complexes thereof by mixing the enantiomeric derivatives and/or pharmaceutically acceptable salts and/or complexes thereof, according to the invention with pharmaceutically applicable carriers and/or excipients

disclosed as above then by formulating the mixture to a medicinal and/or pharmaceutical composition advantageously to tablet, inhalation powder or capsule, suppository or solution especially advantageously to tablet, using the usual, standard formulation technics.

The subject matter of the invention furthermore relates to novel, stereoselective process for the preparation of novel enantiomeric derivatives of 8-hydroxyquinoline derivatives described by General Formulas according to the Invention and advantageously named specifically as above, and

P-1418 pharmaceutically acceptable salts and/or complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, specifically concerning the

enantiomeric derivatives of general formula (T) and (II) by reacting an 8-hydroxyquin.oline derivative of general formula III

with an amine of general formula (IV) ,

R NH 2 IV and with an oxo-compound of general formula

using quinidine (obtaining R-enantiomer ) or quinine (obtaining S-enan iorner ) as catalyst.

P-1418

By this method pure R-enant iomeric derivative of general formula (I) or pure S-enantiomeric derivative can be obtained, and can be used as active agent or can be converted to

pharmaceutically acceptable salts or to complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes specifically advantageously to zinc complexes thereof or can be released from the salt or complex thereof.

Concerning the further advantageous versions of the compounds of general formulas according to the invention, the process according to the invention is described as above, using the proper R' and R' ' substituents indexed properly in the general formulas (III'), (III"), (IV) (IV") and (V") and (V") of the starting materials and reagents according to the different versions of General Formulas according to the Invention and compounds advantageously specifically named as above.

The subject matter of the invention furthermore relates to novel, stereoselective process for the preparation of novel R- enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formulas (I), (Ι') and (I"), and advantageously named specifically as above, and pharmaceutically acceptable salts and/or complexes with divalent or polyvalent metals,

advantageously iron, copper or zinc complexes thereof,

P-1418 specifically concerning the enantiomeric derivatives of general, formula (I) and (II) by reacting an 8-hydroxyquinoline

derivative of general formula (III), advantageously (III'), further advantageously (III'') with an amine of general

formulas (IV), advantageously (IV), further advantageously (IV ' ) and with an oxo-compound of general formulas (V) , advantageously (V), further advantageously (IV'),

advantageously using quinidine as catalyst and obtaining by this method pure R-enantiomeric derivative.

The subject matter of the invention furthermore relates to novel, stereoselective process for the preparation of novel S- enantiomeric derivatives of 8-hydroxyquinoline derivatives of general formulas (I), (Ι') and (I''), and advantageously named specifically as above, and pharmaceutically acceptable salts and/or complexes with divalent or polyvalent metals,

advantageously iron, copper or zinc complexes thereof,

specifically concerning the enantiomeric derivatives of general formula (I) and (II) by reacting an 8-hydroxyquinoline

derivative of general formula (III), advantageously (III'), further advantageously (III'') with an amine of general

formulas (IV), advantageously (IV), further advantageously (IV') and with an oxo-compound of general formulas (V), advantageously (V), further advantageously (IV'),

advantageously using quinine as catalyst and obtaining by this method pure S-enantiomeric de rivat ive .

For the process according to the invention solvent,

advantageously organic solvent or water, especially

advantageously acetonitrile was used as reaction medium.

The reaction can be implemented by using advantageously acidic catalyst, especially advantageously formic acid.

For the preparation of the compounds according to the invention the following novel, general stereoselective processes have been applied according to the invention, depending on the starting materials:

For the process according to the invention solvent,

advantageously organic solvent or water, especially

advantageously acetonitrile was used as reaction medium.

The reaction can be implemented by using advantageously acidic catalyst, especially advantageously formic acid.

For the preparation of the compounds according to the invention the following novel, general stereoselective processes have been applied according to the invention, depending on the starting materials:

Process "R" for preparation of enantiomers with „R"

con guration :

To a solution of 50 mmol of quinidine in the proper solvent (180 ml), formic acid (0.84 equivalent) was added in inert athmosphere then amine derivative (1 equivalent), aldehyde compound (1 equivalent) and 8-hydroxyquinoline derivative (1.2 equivalent) were added.

The mixture was stirred at a proper temperature until the product was formed in a desired quantity.

The reaction mixture was concentrated in vacuum to its third volume, the residue was dissolved in dichloromethane . The solution was washed with 1 M NaOH and extracted further by 1M NaOH 6 times.

A non-polar solvent was added to the organic phase then the dichloromethane was evaporated off. The solution obtained was added to 100 ml of 3 M HC1. The phases were separated and the organic layer was extracted with 3 M HC1 solution. To the combined HC1 phase methyl-t-butyl-ether was added then the pH of the biphasic system was adjusted with 40% NaOH solution to four. The precipitated quinidine was filtered off, the biphasic filtrate was separated then the water layer was washed twice by methyl-t-butyl-ether, the combined organic phase was dried on sodium sulfate and filtered.

A proper solvent was added to the filtrate, the methyl-t- butyl-ether was evaporated off and the residue was stirred at room temperature for 16 hours. The precipitated racemic crystals were filtered.

The mother liquor was concentrated in reduced pressure and the remaining pure R-enantiomer was dissolved in 40 ml of isopropanol, stirred at room temperature for 48 hours then the precipitated crystals were filtered off, to give pure crystalline R-enantiomer. Process "S" for preparation of enantiomers with S- con guration

To a solution of quinine (55 mmol) in a proper solvent (300 ml) in inert athmosphere , formic acid (0.8 equivalent), amine ( 2 . 5 equivalent), aldehyde derivative ( 3 . 7 equivalent), and finally 8-hydroxyquinoline derivative (1.0 equivalent) were added.

The mixture was stirred at a proper temperature until the product was formed in a desired quantity.

The solvent was evaporated off in vacuum, the residue was dissolved in dichloromethane and chromatographed on silica gel. The fractions containing the product were collected and the solvent was evaporated off.

The raw product obtained was purified by normal phase Flash chromatography using hexane-ethyl-acetate gradient then the fractions containing the product were collected and concentrated.

The residue was dissolved in 2-propanol. After 2 hours stirring the precipitated racemic crystals were filtered off. The mother liquor was concentrated in vacuum to get the S-enantiomer . The raw product was isolated by the usual methods (e.g. filtration,

P-1418 centrifugation) and was purified by known methods if needed (e.g. recrystallization or chromatography).

The subject matter of the invention furthermore relates to the use of novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, for the manufacture of a medicinal and/or pharmaceutical composition suitable for the treatment and/or prevention of neuropsychiatric diseases advantageously anxiety disorders, schizophrenia, depression, bipolar disorder, especially advantageously bipolar disorder, depression.

The subject matter of the invention furthermore relates to the use of novel enantiomeric derivatives, advantageously the R- enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, for the manufacture of a medicinal and/or pharmaceutical composition suitable for the treatment and/or prevention of neurologic diseases, advantageously epilepsy, amnesia, different memory disorders, cognitive functional problems, neurodegenerative diseases especially advantageously memory disorders, epilepsy, amnesia, cognitive functional problems, Alzheimer's disease, Huntington disease, Parkinson disease, Wilson disease,

amyotrophic lateral sclerosis (ALS) .

The subject matter of the invention furthermore relates to the use of novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically

P-1418 acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, for the manufacture of a medicinal and/or pharmaceutical composition suitable for the treatment and/or prevention of ischemia and reperfusion injuries thereof, advantageously of cardiovascular disorders, blood vessel catastrophes, traumatic injuries, neurodegenerative traumas, diseases in connection with transplantations and in connection with ischemia and reperfusion injuries thereof, advantageously of impairments of the brain, heart, liver, kidney or lung especially

advantageously of traumatic brain injuries and for inhibition of organ, advantageously of skin graft rejection, especially advantageously for inhibition of skin graft rejection. The subject matter of the invention furthermore relates to the use of novel enantiomeric derivatives of 8-hydroxyguinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, for the manufacture of metal chelate forming, cyto-protective, neuroprotective and/or cardio-protective medicinal and/or pharmaceutical composition, showing favourable binding to the target protein, suitable for the treatment and/or prevention of neuropsychiatric, neurologic diseases and of diseases in connection with ischemia.

The subject matter of the invention furthermore relates to the use of novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof, for the manufacture of a cyto-protective, neuroprotective and/or cardio-protective medicinal and/or pharmaceutical composition suitable for protecting the cells from the

cytotoxic attacks, and for the treatment of patients suffering in diseases combined with "cell-death" and/or prevention of their diseases.

The subject matter of the invention furthermore relates to the use of medicinal and/or pharmaceutical compositions comprising novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and

advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof for treatment and prevention of the above described diseases, which compositions can be administered by a usual method in medicine, advantageously by oral, buccal, sublingual,

parenteral or intravenous, rectal or inhalation method

especially advantageously administered by oral method.

The subject matter of the invention furthermore relates to a cytoprotective , neuroprotective or cardioprotective process for treatment and/or prevention of cardiovascular diseases, blood vessel catastrophes, traumatic injuries,

neurodegenerative traumas, diseases in connection with

transplantations and in connection with ischemia and

reperfusion injuries thereof, advantageously of impairments of the brain, heart, liver, kidney or lung especially

advantageously of traumatic brain injuries and for inhibition of organ, advantageously for skin graft rejection, especially advantageously for inhibition of skin graft rejection,

by

administering a medicinal and/or pharmaceutical composition by a usual method in medicine, advantageously by oral, buccal, sublingual, parenteral or intravenous, rectal or inhalation method especially advantageously administered by oral method, which composition comprises novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof.

The subject matter of the invention furthermore relates to a cytoprotective, neuroprotective process for treatment and/or prevention of neuropsychiatry diseases advantageously anxiety disorders, schizophrenia, depression, bipolar disorder, especially advantageously bipolar disorder, and depression and further of neurologic diseases, advantageously epilepsy, amnesia, different memory disorders, cognitive functional problems, neurodegenerative diseases especially advantageously memory disorders, epilepsy, amnesia, cognitive functional problems, Alzheimer's disease, Huntington disease, Parkinson disease, Wilson disease, amyotrophic lateral sclerosis (ALS) by administering a medicinal and/or pharmaceutical composition by a usual method in medicine, advantageously by oral, buccal, sublingual, parenteral or intravenous, rectal or inhalation method especially advantageously administered by oral method, which composition comprises novel enantiomeric derivatives of 8-hydroxyquinoline described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable salts and complexes with divalent or polyvalent metals, advantageously iron, copper or zinc complexes thereof.

II. History, the state of the art, mechanism of action

1) The specification of the history, state of the art,

mechanism of action of the novel enantiomeric derivatives of 8- hydroxyquinoline according to the invention, described by General Formulas according to the Invention and advantageously named specifically as above and pharmaceutically acceptable

P-1418 salts and metal complexes thereof and medicines suitable for treatment and/or prevention of different diseases and

comprising the subject compounds. The prior art and patents referred and cited in the present specification hereinafter are all part of the state of the art.

The various etiological cell injuries and cell deaths are the main characteristics of many cardiovascular, neurological and inflammatory disorders. Cell injuries may occur as the results of cellular hypoxia or ischemia, formation of various kinds of oxidants or free radicals and/or overproduction of various biological mediators (cytokines, chemokines, lipid mediators) and overproduction and/or aggregation of different toxic peptides (e.g. β-amyloid peptides) or of proteins (synuclein, huntigtin, prion) in case of neurodegenerative diseases

(comprehensive literature: Orrenius, 2007; Mattson, 2006).

These processes are often interdependent; so those occur as parts of self-amplifying („suicidal") intracellular cycles and form the determining basis of many human diseases.

Though cell death is typically referred as apoptosis or necrosis, these two forms only represent the two ends of the range of the forms of cell injuries.

The intercellular mechanisms taking part in the above cell death processes are complex, but often activate the cell death effector family called caspases and mitochondrial dysfunction, mitochondrial depolarisation, generation of reactive oxygen species and release of mitochondrial components into the cytosol (comprehensive literature: Szabo, 2005; Duprez et al., 2009; Degterev and Yuan, 2008; Wang et al., 2009; Stefanis, 2005) .

In the case of Alzheimer's disease, neuronal cell death results mainly from the direct cytotoxic effects of aggregated β-

P-1418 amyloid peptides which, in part are generated by caspase- triggered apoptosis.

The other reason of the deterioration of the cognitive

functions is the quantitative decreasing of different synaptic proteins playing role in neurotransmission and the reduced reactivity of different receptors like acetylcholine and muscarine (Machova et al . , 2008; Bartus, 2000).

There are more and more evidences, that in the course of β- amyloid peptide aggregation not the fibrillary but the oligomer form has a correlation with neuropathological changes in connection with mortality and dementia, like development of protein tangles and neuritic disorders (e.g. reduced dentrit- spike and reduced numbers of active synapses) (comprehensive literature: Lue et al., 1999; McLean et al., 1999; Wang et al. f 1999).

However besides the oligomer forms of β-amyloid peptide aggregates the fibrillary forms results also neuronal

destruction and reduced functionality partly through LDI receptors (Janciauskiene et al., 1999).

The compounds preventing cell injury and cell death are usually called ( cytoprotective" compounds. Cytoprotection may be achieved by many pharmacological and biochemical methods. The following examples of them are mentioned here: scavengers of oxidants and free radicals, inhibitors of certain „death effector pathways", stabilisation of cell membranes, etc. In the course of ischemia or several related disease processes, iron and copper cations are released from the tissues which catalyse hydroxy-free radical formation in the Haber-Weiss pathway in a known manner causing cell injuries. Inactivation or chelate formation of these metal cations may result in a cytoprotective effect. Thus experiments were conducted to mitigate the catalytic efficiency of iron and copper cations in

P-1418 such a way that iron-chelate forming siderophores (e.g.

deferoxamine) were administered (Lewen et al., 2000; Britton et al. , 2002) .

It is known that glutamate is released along with zinc cations from the synaptosomes of the nervous system cells using glutamate as a chemical messenger. Usually, the zinc released in the nerve synapse is quickly built again in the

synaptosomes. As a result of ischemia, lasting attacks and cerebral lesion, the zinc released from the synaptosomes is accumulated in the extracellular liquid surrounding the neurons. When an excessive amount of zinc enters the cell body, zinc may trigger cell death via apoptosis and necrosis. Zinc- chelate forming through that mechanism may result in

neuroprotection and influence the outcome of various

neuropsychiatric diseases. (Regland et al., 2001; Koh et al., 1996) .

Therefore the zinc-chelating agents may also be useful in treatment of the Alzhemier's disease by binding zinc occurring in the plaques thus weakening the structure of the plaques

(Frederickson et al., 2005; Schafer et al., 2007). The zinc- chelating agents may also be useful in the treatment of

Huntington's disease (Nguyen et al., 2005). According to another way of cytoprotection, the intracellular pathways mediating protective effects are induced. A prototype of this approach is the so-called „ischemic preconditioning" where the cells or organs are subjected to ischemia for a short time in order to induce over-regulation of the cytoprotective genes (e.g. genes of antioxidant enzymes, heat shock proteins and others). Induction of heme oxygenase enzyme (HMOX-1) has demonstrated cytoprotection in several experimental systems (e.g. Li et al., 2001; Idris et al., 2008).

P-1418 Compounds used for inhibition of endoplasmatic reticulum stress have been described in the American patent application

publication no. US 2008/293699.. Part of the state of the art is our own earlier patent

application also, where recently new racemic 8-hydroxquinoline derivatives and indications thereof have been described and where some members of this compound family have been identified by cell-based screening tests for systematic identification of cytoprotective compounds (WO2011148208) .

In this test a certain form of cell injury was simulated and a chemical library was screened in order to identify compounds preventing or retarding cell injury

(e.g. Gero et al., 2007).

By means of the cell-based screening method, we have found and identified novel racemic 8-hydroxyquinoline derivatives.

These compounds protect the cells from injuries induced by oxidative stress therefore these can potentially be used in the treatment of many diseases.

The racemic compounds exert various cellular effects e.g. iron- copper and zinc chelating, inhibition of PARP-activation, inhibition of mitochondrial dysfunction, or activation of heme oxygenase enzyme.

However according to our earlier invention only the racemic compounds have been described, the enantiomerically pure derivatives thereof have not been prepared at all, and neither the chemical characters thereof have been disclosed.

Moreover concerning the earlier cytoprotection tests, no direct evidence has been provided, that neuronal destruction triggered by the different aggregated forms of β-amyloid peptides

(oligomer, and high molecular weight fibrillary complexes) can

P-1418 be prevented by the racemic compounds and whether they can be used for increasing cognitive functions.

Earlier it was also not known, through which molecular

mechanisms the positive effects of each enantiomer can be achieved on neurons, cardiac sells and in neurodegenerative animal models induced by beta oligomers.

It was shown that, the pure R-enantiomeric derivative has effect on several targets that can be related to the

neuroprotective effect and inhibiting effect concerning

neuroinflammation of the clinical candidate.

The R-enantiomer compound of the Example 2 according to the invention inhibits in micromolar concentration the caspase-3 and 5-lipoxygenase enzymes and the transcriptional induction regulated by the activated T-cell nuclear factor (NF-AT) .

The activation of caspase-3 is strongly related to several degenerative processes of the aging brain (Lynch et al . , 2002) and to the pathogenesis of neurodegenerative diseases occurred in old age (Eckert et al., 2003).

The activation of caspase-3 is a common connection point of many toxic stimulations, included the oxidative injury and the toxicosis induced by the aggregated β-amyloid peptides.

The activated caspase-3 protease induces degradation of several key intracellular proteins, resulting apoptosis at the end (Hengartner, 2000) .

However the caspase-3 plays important role not only in the late phase of neurodegeneration inducing neuronal destruction but also encourages the development of the pathological changes in the early stage, because it is responsible also for the proteolytic cleavage of both of amyloid precursor protein (APP) and the GGA3 adaptor which are transforming to toxic amyloid peptide aggregates at the end.

P-1418 The axonal microtubule-associated tau protein forming neurofibrillary tangles is. also substrate of the caspase-3 enzyme, so increased function of the caspase-3 enzyme reduces the binding of the tau with total length to microtubule, leading to a neuritic degeneration at the end.

Moreover it has been shown that the fibrillary tau protein deposits in neurons are not causes but consequences of the degenerative processes within the cell induced by the

activation of caspase (Calignon et al., 2010).

Coming from all these, the neuronal destruction related to the neurodegenerative processes for different reasons (oxidative stress, effects of β-amyloid peptide aggregates) and the developed deterioration of the cognitive processes can be inhibited inter alia by using the pure R-enantiomeric

derivative according to the invention and to the Example 2 for inhibition the caspase enzyme in the brain.

Disturbance in calcium ion homeostasis and the following activation of calcineurin can induce pathological changes related to the Alzheimer's disease (Liu et al., 2005.

The activated T-cell nuclear factor (NFAT) is dephosphorylated by calcineurin, translocated in the cell nucleus by

dephosphorylation, triggering expressions of several genes which are inducing neuronal destruction, activation of

astrocyte and neuroinflammatory processes. Among these the induction of interleukin-ΐβ (IL-Ιβ) is of primary importance, being one of the causes of the increased level of extracellular glutamic acid, and are involved in hyperexcitable synaptic activity, generally in induction and maintenance of

neuroinflammatory processes related to several

neurodegenerative diseases (Sama et al . , 2008).

Recently it was shown that, the neurotoxicity triggered by β- amyloid could be reduced by pharmacological inhibition of NFAT in transgenic mouse (Hudry et al., 2012).

P-1418 Furthermore different levels of each NAFT isoforms have been stated in brain samples of patients of Alzheimer' s disease suffering from dementia with different severity (Abdul et al., 2009) .

It was published by the same authors also, that the protein level of excitatory amino acid transporter (EAAT-2) was NFAT depending reduced by β-amyloid oligomers in astrocytes and the level of glutamic acid resulting neuronal destruction was increased.

Using general NFAT inhibitors all these effects could be reversed proving that NFAT transcriptional activation has important role in pathological processes related to the central nervous system and with the inhibition thereof it was proved that the NFAT system could be an important target concerning the therapy of neurodegenerative diseases.

It is well known that the inhibition of calcineurin/NFAT system reduces the organ graft rejection, so the compounds according to the invention could also be suitable for inhibition of organ graft rejection.

Kozismert, hogy a kalcineurin/NFAT rendszer gatlasa szerv transzplantacio soran a szervkilokodest csokkenti (Lee es Park, 2006) :Lee M, Park J. Regulation of NFAT activation : a

potential therapeutic target for immunosuppression. Mol Cells. 2006 Aug 31 ;22 (1) : 1-7. ) , ezaltal a talalmany szerinti

vegyuletek a transzplantacio utani szervkildkodes

megakadalyozasara is alkalmasak.

NFAT transcriptional induction triggered by A23187 ionophore and PMA inductive agent was inhibited by the compound according to the Example 2 and the invention in micromolar concentration. Therefore it can be concluded that the enantiomerically pure R- enantiomeric derivative according to Example 2 is able to inhibit the increased calcineurin activity induced by β-amyloid and by the increased level of intracellular calcium and the consequent calcineurin activity, and therefore it is able to

P-1418 inhibit the apoptotic and neuroinflammatory processes, and positive effect may have generated in different

neurodegenerative diseases and by transplantations. The calcineurin/NFAT system and the caspase-3 apoptotic activation system are related to each other.

The calcineurin over function induces caspase-3 activation and apoptosis (Asai et al., 1999). As the compound according to Example 2 inhibits both NFAT transcriptional activity and caspase-3 enzyme activity the compound according to the invention reduces the neuronal destruction by double effects, but also has positive effect on the neuroinflammatory processes induced by NFAT.

Therefore it is assumed that pure R-enantiomeric derivative has more efficiency than the solely on NAFT system active or solely the caspase-3 enzyme inhibiting compounds.

The third target of the pure R-enantiomeric derivative

according to the invention and to Example 2 is the enzyme 5- lipoxygenase (5-LOX) having an important role in inflammatory and traumatic (excitotoxic and ischemic) processes related to central nervous system (Zhou et al., 2006) and in

neurodegenerative diseases related to aging (Uz et al., 1998). It was shown in transgenic mice that inhibition of 5-LOX reduced β-amyloid aggregates and improved the cognitive functions (Firuzi et al., 2008).

Increased activity of 5-LOX reduced the level of PSD-95, synaptophysin and MAP2 proteins being important in synaptic integrity (Chu et al., 2013).

It is concluded from all these that, the R-enantiomeric derivative according to the Example 2 having micromole activity validly, reduces the neuroinflammatory processes by inhibition of 5-LOX enzyme thereof and has positive influence on

pathological changes related to β-amyloid peptides and tau proteins

P-1418 The cell injury caused by hypoxia is a common characteristic of different diseases including brain infarct, brain stroke and infarct of heart muscle.

An important and central element of cell's accommodation to hypoxia is the hypoxia-inducible factor (HIF) , a transcription factor activating expressions of several genes inter alia including the genes playing important role in glucose

metabolism, in the antioxidant system, in angiogenesis or in blood cell formation (comprehensive article: Chowdhury et al., 2008) .

It was shown that, impacts causing rise of the level of HIF, have advantageous effect on ischemic diseases and on

reproduction of stem cells (Zhang et al., 2006) .

Using a screening system described earlier, HIF system

activating small molecules, including several 8- hydroxyguinoline type compounds with furcate structure could be identified (Smirnova et al., 2010). However among the molecules listed in the examples there was not any pure enantiomer, only racemic

mixtures have been worked with.

The subject racemic compounds activated the expression of genes with a value between 2-10 μΜ IC50 by 2-7 x maximal value.

According to our invention, it has been found that, the R- enantiomeric derivatives according to Example 2 and S- enantiomeric derivatives according to Example 3 were capable to increase the gene activity of phosphoglycerate kinase 1 (PGK- 1), the vascular endothelial growth factor (VEGF) , the hem- oxygenase 1 (HMOX-1) and the erythropoietin (EPO) in the cells, all regulated by HIF.

According to our invention the highest activation was found in case of EPO gene, in submicromolar concentration with over 20x maximal activation value.

The PGK1, VEGF, EPO activations have been disclosed in the subject document (Smirnova et al., 2010) , but it was not shown any activity of 8-hydroxyquinoline type compounds

P-1418 Furthermore recently it has been shown that, the hem-oxygenase 1 (HMOX-l), an antioxidant enzyme induced by HIF has strong cytoprotective effect in neurons (Chen et al . , 2000) and in endothelial cells of brain capillaries (Bresgen et al., 2003) .

As oxidative stress plays an important role in several

neurodegenerative and neuroinflammatory processes, the

pharmacological induction of HMOX-l is judged as a logic and desirable therapeutic solution (Calabrese et al., 2003; Jazwa and Cuadrado. , 2010).

The induction of HMOX-l influenced also the outcome of heart transplantation advantageously (Bach 2006) .

In one of our earlier publication it has been shown that, a racemic 8-hydroxyquinoline causes increase of HMOX-l gene activity in cardiac sells, explaining partly the

cardioprotective characteristics thereof in cell system and protective effect

thereof in case of heart muscle injury caused by

ischemia/reperfusion in rat (Korkmaz et al., 2013).

On the other hand none of the enantiomerically pure form of the compound group described in the present patent application of general formula has been examined yet.

It was shown that, the R-enantiomeric derivative according to Example 2 and S-enantiomeric derivative according to Example 3 caused the induction of HMOX-l in astrocytes, concluding that, both pure enantiomer described by us can have cytoprotective effect concerning the diseases with oxidative stress.

The highest inductive effect of R-enantiomeric derivative according to Example 2 and S-enantiomeric derivative according to Example 3 on expression of gene was stated in case of EPO gene .

Moreover it has been shown that, by oral administration, the R- enantiomeric derivative according to Example 2 resulted

P-1418 Recently it was shown that, the EPO in brain has outstanding role in neuroprotection and advantageous characteristics thereof revealed itself in several psychiatric diseases like depression, schizophrenia, bipolar disorder, epilepsy (Newton et al., 2013) in neurodegenerative diseases like Alzheimer's disease, Parkinson disease (Arabpoor et al., 2012; Xue et al., 2007), traumatic brain injury, blood vessel catastrophes in brain (Mammis et al., 2009), diseases characterized by

neuroinflamination, like sclerosis multiplex (Hagemeyer et al., 2012), and had positive effect in animal models and several clinical tests concerning other diseases related to central nervous system, like amyotrophic lateral sclerosis (ALS) (Noh at al., 2C14; Merelli et al., 2013; Chong et al., 2013).

Because of this there were several methods examined targeting the ingestion of the EPO directly into the brain.

The intravenous EPO injection could cause system side effects, therefore there were experiments also for ingesting EPO through the nose (Merelli et al., 2011).

However these kind of applications could have several

disadvantages inter alia the costly production, the involved administration, stability, dosage and the side effect on possible blood cell forming organs of EPO.

Therefore according to an advantageous solution an EPO

synthesis could be induced locally in the brain by a small molecule.

This goal has been achieved by the enantiomeric derivatives according to the invention, described in the present patent application and concrete examples has been given concerning the EPO inducing effect of R-enantiomeric derivative according to Example 2 and S-derivative according to Example 3 in cell system.

Concerning the R-enantiomeric derivative according to the Example 2 and the invention it was shown that, it can increase the EPO gene activity in different regions of brain also in living animal.

P-1418 By these characteristics of the compounds according to the invention and disclosed in the present patent application it is obvious to use them in case of different psychiatric diseases, diseases related to ischemia and central nervous system, neurodegenerative disorders, and inflammatory processes related to the brain.

2 ) History of the process according to the invention The process is actually a modification according to the invention of the known Betti-reaction, which makes possible to prepare novel enantiomeric derivatives by novel, stereoselective synthesis.

In the course of the stereoselective process according to our invention the R- or S-enantiomeric derivatives have been prepared by using cinchona stereoisomer (quinidine or quinine) in a novel enantioselective version of Betti reaction (Betti, 1900; Betti, 1903;, Phillips et al., 1954; Phillips et al., 1956; Phillips, 1956) modified and optimised by us.

The advantage of our process is that the proper enantiomers were obtained by a simple recrystallization over 99 % enantiomeric excess, without using any separation technique (e.g. chiral preparative HPLC) using low-cost cinchona organocatalysts : catalysed by quinidine R-enantiomer, catalysed by quinine S-enantiomer could be obtained.

The known processes disclosed in the background literature of stereoselective Betti-reaction mainly enantiomerically pure reactant (e.g. aralkyl amine) was used then the proper enantiomers were isolated by separation of the chiral support (Palmieri, 2000).

Organocatalysed asymmetric reactions using cinchona alkaloids or modified derivatives thereof are known in case of Mannich reactions (Verkade at al., 2008).

P-1418 III . Summary

The essence and advantages of the invention can be summarised as follows.

The enantiomerically pure -enantiomeric derivative prevents the destruction of neurons, the neuroinflammatory processes induced by astrocytes and microglia and furthermore

advantageously reduces the pathological reactions related to amyloid peptides and to tau proteins which are destroying the neurotoxic and synaptic systems,

In consequence the compounds according to the invention, advantageously to Example 2 reduces the deterioration of the cognitive functions related to neurodegenerative processes and therefore they could be therapeutically effective in treatment of several degenerative diseases related to the central nervous system.

As the of enantiomerically pure compounds according to the invention, advantageously to Example 2 influence all the three identified targets (caspase-3, NAFT, 5-LOX) , effectively, they are effective not only in neurodegenerative processes, but also in different ischemic processes like ischemic and inflammatory processes related to heart vascular system, therefore the compound could be also effective in treatment of diseases related to these processes.

The active ingredient of the medicinal and/or pharmaceutical compositions according to our invention is a molecule on one side not complexed with iron, copper or zinc cations, on the other side forms complex with iron, copper or zinc cations. The essence of the subject matter of the invention and base of novelty is that all prior art documents and patents being part of the state of the art disclosed only the racemic products and characteristics, biological effect thereof, the pure

enantiomers according to the invention and characteristics, biological effect thereof are disclosed in our present patent application for the first time.

P-1418 The essence of our invention is furthermore that the novel enantiomers according to the subject matter of invention are prepared by the stereoselective synthesis also according to the subject matter of the invention therefore the novel

pharmaceutical compositions are well applicable for prevention and/or treatment of the diseases listed in present

specification, which applications are also part of the subject matter of the invention.

The essence of the stereoselective process according to the invention is that using quinidine catalyst enantiomerically pure R-enantiomer, using quinine catalyst enantiomerically pure S-enantiomer is formed.

The medicinal and/or pharmaceutical compositions according to our invention can be used for prevention and/or treatment of the diseases listed in present specification as cytoprotective, neuroprotective and cardioprotective agent.

So the subject matter of our invention furthermore relates to a cytoprotective, neuroprotective and cardioprotective process for prevention and/or treatment of the diseases listed in present specification by administering the medicinal and/or pharmaceutical compositions according to the invention by a usual method in medicine, advantageously administered by oral method . IV. Statement of the figures

Figure 1

The calculated configuration of the conformer of the S- enantiomer compound according to the invention (1) with the lowest energy.

Figure 2

The FTIR (Fourier transform infrared spectroscopy ) and VCD (Vibrational Circular Dichroism) spectrum of the sample of R-

P-1418 enantiomer compound according to the invention measured in

CDCI3 (deuterated chloroform) solvent.

Figure 3

The calculated VCD spectrum of the conformers of the

S-enantiomer compound according to the invention with greater population than 1 %.

Figure 4.

The theoretical VCD spectrum of S- and R-enantiomer compounds according to the invention and the conformers thereof measured in CDCI 3 calculated as with the population weighted sum.

Figure 5

The inhibiting effect of the R- and S-enantiomer compounds according to the invention on different matrix metalloprotease enzymes is shown.

Figure 6

The effect of R-enantiomer compound according to the invention on cell death in vitro caused by hydrogen-peroxide on cardiac sells is shown.

Figure 7

The effect of R-enantiomer compound according to the invention on cell death in vitro caused by hydrogen-peroxide on SH5Y neuron cells is shown.

Figure 8

The effect of S-enantiomer compound according to the invention on cell death in vitro caused by hydrogen-peroxide on cardiac sells is shown.

Figure 9

P-1418 The effect of S-enantiomer compound according to the invention on. cell death in vitro caused by hydrogen-peroxide on SH5Y neuron cells is shown.

Figure 10

The effect of sodium salt of R-enantiomer compound according to the invention on cell death in vitro caused by fibrillar aggregates of β-amyloid peptides on primer cortical neuron cells is shown.

Figure 11

The effect of zinc complex of R-enantiomer compound according to the invention on cell death in vitro caused by fibrillar aggregates of β-amyloid peptides on primer cortical neuron cells is shown.

Figure 12

The effect of R-enantiomer and S-enantiomer compounds according to the invention and according to Example 2 and 3 on short-term memory disorders in vivo caused by oligomer aggregate of β- amyloid peptides is shown.

Figure 13

The effect of different concentration of R-enantiomer compound according to the invention and according to Example 2 on short- term memory disorders in vivo caused by oligomer aggregate of β-amyloid peptides is shown.

Figure 14

The effect of R-enantiomer and S-enantiomer compounds according to the invention and according to Example 2 and 3 on short-term memory disorders in vivo caused by scopolamine is shown.

P-1418 Figure 15

The diagram of inhibition effect of R-enantiomeric derivative according to the invention and according to Example 2 on NFAT protein transcriptional activity

Figure 16

The effect of R-enantiomeric derivative according to the invention and according to Example 2 influencing the gene expression in primer astrocytes

Figure 17

The effect of R-enantiomeric derivative according to the invention and according to Example 2 influencing erythropoietin gene expression in hippocampus and cerebral cortex of elderly animals in course of chronical treatment.

Figure 18

The effect of R-enantiomeric derivative according to the invention and according to Example 2 influencing the survival of the skin graft in course of chronical treatment after skin grafting .

V. Explanation of expressions, shortages used in the

specification

Expression " enantiomerically pure":

Characteristic property of a certain enantiomeric form

containing the other enantiomeric form to a specified

percentage: so e.g. in case of 100 % purity there is no other enantiomeric form at all, in case of 98 % purity the other enantiomeric form is in 2 % present etc.

Expression "lower alkyl group":

Straight or branched chained alky groups with 1-4 carbon atoms like e.g. methyl, ethyl, isopropyl groups etc.

P-1418 3.6

Expression "cyclo alkyl group "

Cyclic groups comprising 3-8 carbon atoms like cyclopropyl, cyclobutyl, cyclohexyl group etc.

Expression "aryl group":

Monocyclic or bicyclic aromatic hydrocarbon groups like e.g. phenyl group, naphthyl group Expression "aralkyl group":

Alkyl group described above, substituted by one or two aryl groups described above, like e.g. benzyl, beta phenyl-ethyl group etc. Expression " heteroaryl group":

5 or 6 membered aryl groups containing one or more oxygen, nitrogen and/or sulfur atoms like pyridyl, pyrimidyl, pyrroryl, oxazolyl groups etc. Expression "halogen atom":

Fluorine, chlorine, bromine or iodine atoms

Expression "Electron withdrawing substitutes":

The indicator of electron withdrawing groups, represented as substituents, advantageously halogen atoms, nitro groups, trifluoromethyl , methysulfinyl or methylsul onyl groups.

Expression "Electron donating group":

The indicator of electron donating groups represented as substituents advantageously lower alkyl groups with 1-4 carbon atoms like e.g. methyl group.

Expression " General Formulas according to the Invention" The common meaning of the general formulas (I) and (II), advantageously (Ι') and (II') / especially advantageously (I''

P-1418 and II''), where the general formulas signed by comma are meaning the advantageous properly substituted versions.

Pharmaceutically acceptable salts:

The novel enantiomers of 8-hydroxyquinoline derivatives according to the invention described by the General Formulas according to the Invention as given above and named

specifically can form salts with bases on the hydroxy group and with acids on nitrogen atom in the substituent attached to position 7 of the quinoline ring.

For the salt formation pharmaceutically suitable bases

advantageously alkaline metal hydroxydes like sodium-hydroxyde potassium-hydroxyde or organic acids advantageously hydrogen bromide, acetic acid, fumaric acid, maleic acid, malic acid, succinic acid, tartaric acid, benzenesulfonic acid,

p-toluenesulfonic acid or methane sulfonic acid can be used.

Pharmaceutically acceptable complexes:

The novel enantiomers of 8-hydroxyquinoline derivatives according to the invention described by the General Formulas according to the Invention as given above and named

specifically can coordinatively bind divalent or polyvalent metals (like e.g. iron, zinc, copper etc.) and forming

complexes with them.

In such complexes the free electron pairs of the oxygen atom of hydroxyl group and of nitrogen atom of the quinolone are participating .

Chelate forming effect:

The novel enantiomers of 8-hydroxyquinoline derivatives according to the invention described by the General Formulas according to the Invention as given above and named

specifically are capable to form chelates with the free electron pairs described as above the same way as complexes are formed. Thank to this chelate forming feature the unbeneficial

P-1418 processes caused by metal ions can be inhibited in different diseases, and also the functioning of proteins containing metal ions can be influenced advantageously. Indications, explanations of sets of diseases

The following statement of sets and collective nouns of indications and diseases gives help to interpret and to understand the specification of the application and the use claims containing the indications.

Neuropsychiatric diseases

anxiety disorders, schizophrenia, depression, bipolar disorder

Neurologic diseases:

epilepsy, amnesia, different memory disorders, cognitive functional problems and neurodegenerative diseases including memory disorders, epilepsy, amnesia, cognitive functional problems, Alzheimer's disease, Huntington disease, Parkinson disease, Wilson disease, amyotrophic lateral sclerosis (ALS) .

Ischemia and reperfusion injuries thereof in connection mainly but not solely with cardiovascular disorders, blood vessel catastrophes, traumatic injuries and neurodegenerative traumas, diseases in connection with transplantations: impairments of the brain, including traumatic brain injuries, and impairments of heart, liver, kidney or lung and organ, advantageously skin graft rejections.

P-1418 VI . Chemical examples

The subject matter of our invention is supported by the

following examples without limiting the scope of protection to the examples.

Example 1

The determination of the absolute configuration

The racemic mixture of molecules according to Example 2 and Example 3 comprises one chiral center, but they have

significant conformation freedom so to determine the absolute configuration also detailed analysis of conformation was needed.

The essence of the determination of the absolute conformation based on VCD spectroscopy is the ab initio (DFT) levelled calculation of the VCD spectrum of one or the other

enantiomeric form, and then the comparison of the calculated and the measured spectrum thereof. As the VCD spectrum

comprises high number of bands with alternating signals the comparison is generally extremely reliable.

In case the position and signal-pattern of the bands of

calculated and measured spectrum correspond, the absolute configuration corresponds with the configuration of the chosen enantiomeric form, if mirrored, the configuration is the opposite .

In presence of more conformers the measured spectrum should be compared with the theoretic spectrum obtained as population- weighted average of calculated spectrum of each of enantiomeric compounds .

The FTIR and VCD spectra of the sample were recorded by a

Bruker PMA-37 typed VCD-module connected with a Bruker

Equinox55 FTIR spectrometer with resolution of 4 cm "1 , in a BaF 2 cuvette with 0,05 mm layer thickness, in CDC1 3 solvent, in 100 mg/ml concentration, using MCT detector cooled by liquid N 2 . The spectrometer was optimized on fingerprint-range therefore an optical filter transparent in 18(30-800 cm "1 range was used and the photoelastic modulator of the device was set on 1300 cm "1 wave number.

The calibration of the device was implemented with the help of a Cds standard double refracted crystal (CdS multiple wave- plate, MPW) and a polarisation filter as analyser with metal grid and with KRS-5 carrier.

In case of the sample and the reference approximately 42000 interferograms were averaged that corresponds to a recording time if 12 hours to optimize the bad signal-to-noise ratio caused by the low signal-intensity of VCD spectra.

To correct the baseline of the VCD spectrum, the solvent spectrum recorded under same condition was extracted from the raw spectrum.

The infrared spectrum can be obtained together with the VCG spectrum from the ratio of one channelled DC spectrum of the sample and that of the reference (solvent) (this is a global infrared signal not containing any chiral information) .

The quantum chemical calculations were performed on a Supermicro server (2 x Intel Xeon™ X5680 3.3 GHz 6-corn processor, 72 GB RAM) using a Gaussian 09 software package (Frisch et al., 2010.).

The quantum chemical calculations were performed on the S- enantiomeric form on B3LYP/6-31G** DFT theoretical level, under vacuum condition, the mapping of the conformers was performed along the five torsion angle (Θ1-Θ5) (Figure 1) describing the flexibility of the molecule with partly systematic partly heuristic searching.

The calculation of the IR- and VCD-spectra of the structures with optimized geometry was performed also on B3LYP/6-31G** DFT level . The calculated frequency (wave number.) values were corrected by 0,97 scale factor usable in case of B3LYP/6-31G** theoretical level .

For the generation of the theoretical spectra were assumed a signal-shape of Lorentz diagram and half-width measured at 6 cm -1 mid-height.

For visualization of the vibrational spectrum GaussView 5.0 program package has been used. For estimating the population formed on temperature T=298K, Boltzmann-distribution was assumed. According to the Boltzmann distribution the relative population of the ith conformer compared to the conformer having the lowest free enthalpy

(i = 0) is:

The mole fraction of ith conformer (the population measured to the whole conformer mixture) is:

N>

In that case the calculated IR- or VCD spectrum is the weighted sum of the spectra of each conformers:

S(^) = ^x,5,(^) (JR,VCD)

i

It can be stated after implementation of the population analysis of the 32 calculated conformer results that there are very small energy differences between the individual conformers so no dominant conformer can be determined.

Among of these eleven have higher population than 1 %. Summarizing these eleven conformers they take 99,6 %, so the spectral contribution of conformers with higher energy was neglected .

P-1418 The geometrical data, relative free enthalpies and population of the eleven conformers v/ith higher population than 1 % are summarized in Table 1.

Table 1.

The characterising torsion angle da ta, relative free enthalpy and estimated population of the S-enantiomer at 298 K temperature, on the basis of quahtum chemical calculations carried out on B3LYP/6-31G** levelled vacuum status.

Confor- 0 5 n Δ6 Populati mer (kJ/mol) on (%)

1 142.0 -173.5 -95.5 -74.4 -179.2 0.0 20.3

1 136.3 -14.7 162.1 70.6 179.9 0.6 15.9

3 82.7 -172.8 65.5 87 178.9 1.0 13.6

4 137.3 166.7 160.5 71.0 179.8 1.3 12.2

5 83.3 -172.8 -109.4 87.4 178.9 1.4 11.5

6 88.3 -10.8 13.7 -136.6 179.4 3.1 5.8

7 88.4 -10.8 -166.6 -136.9 179.4 3.1 5.7

8 a 80.9 8.2 62.2 87.0 178.7 3.3 5.4

9 a 81.2 8.2 119.0 87.4 178.6 3.8 4.4

10 143.8 6.7 -93.0 -737 -179.5 4.2 3.7

11 89.6 169.4 12.7 -136.4 179.3 7.5 1.0

Differing only in spacing of CF3 group

It is characteristic for all these cjonformers that they have ti ¬ bound between the hydrogen atom of the hydroxyl group and the nitrogen of the quinolone ring, In some conformers not mentioned here this is missing, respectively the conformers with H-bound with the N-atom of tne aminopyrimidine part are energetically also less favourable The spatial structure of the conformers with the lowest energy is shown on Scheme 1.

P-1418 Infrared and VCD spectra, the determination of absolute con iguration

The FTIR and VCD spectrum of compound according to Example 2, is shown in Figure 2 in the spectral range of 1700-1000 cm "1 .

This approximately corresponds to the spectral region limited by the characteristics of optics of VCD spectrometer and the transmission of the solvent (CDC1 3) used in 0,05 mm cuvette. It can be stated that there are more bands in the VCD spectrum than in the FTIR spectrum referring to the fact that in several cases there is a band overlap in the infrared spectrum so the corresponding bands of VCD spectrum can be identified either only as a "shoulder" or cannot be identified at all.

The extreme values (maximums or minimums) in VCD and in infrared spectrum should appear at identical wavelength in principle; small differences can be explained also by band overlap. . It can generally be stated that the vibrations of aromatic and heteroaromat ic rings couple strongly with the functional groups attached to them hence their assignation is fairly difficult due to their derealization .

The most intensive band of the IR spectrum at 1326 cm "1 derives from the coupling of the symmetric deformation vibration of CF 3 group and the skeletal vibration of benzene ring, but the corresponding VCD band has a very low intensity.

One of the most indicative part of VCD the spectrum is the negative-positive band-pair at 1502 cm "1 and 1512 cm "1 that are not separated in the IR spectrum but they merge into a broader band at 1507 cm "1 . According to the calculations the positive band at 1512 cm "1 has a special diagnostic value as it derives from coupling of the deformation of the C-H bond attached to the chiral center and the in-plane deformation vibration of C- N-H part of 2-aminopyrimidine group. The negative band at 1507- cm "1 derives from the skeletal vibration of quinoline ring and the in-plane OH deformation vibration of its OH substituent, to a certain extent coupled with the deformation of the C-H bond

P-1418 of the chirality center. The intensive band at 1583 cm "1 derives from coupling of the skeletal vibration of pyrimidine ring and the in-plane N-H deformation vibration of the seconder amino group attached to it.

The calculated VCD spectrum of the relevant conformers of the S-enantiomer is shown in Figure 3. It can be seen from the spectrum that the pattern of the VCD bands depends strongly from the conformers, resulting high averaging and due to to coupling of vibrations it is not possible to abstract from the conformation conditions of the achiral aromatic and heterocyclic parts. In the case of the bands at the area of 1500 cm "1 mentioned above having high diagnostic value, there is luckily almost no change at least concerning the conformers with higher population. The analysis of all calculated spectra of the conformers (32 pieces) resulted in that the sign of these bands with diagnostic value turns to the opposite only in case of conformers where the N-H bond and the bond of chiral center have the same direction. These conformers are energetically negligible, very unfavourable forms with a population far below 0.1 %. It is obvious that for the determination of absolute configuration both the experimental spectrum and the calculated spectrum weighted with population are needed (Figure 4), but according to Figure 3 it is also shown that the signs of most of the bands (included also the bands at the area of 1500 cm "1 ) are opposite from the signs of experimental VCD spectrum.

Considering that the VCD spectra of enantiomers are mirrored, based on the wave number and sing-pattern of VCD bands of the measured spectrum and the theoretical, calculated spectrum weighted with the population (Figure 4) it has been concluded that the compound according to Example 2 is an R-enantiomer (Scheme 2) . The measured VCD spectrum is in good agreement with its theoretical spectrum and it has basically opposite spectrum

P-1418 compared to the molecule with S-configuration used in the course of molecular modelling.

Example 2

Preparation of enantiomerically pure

7- t (R) - [ (4-Methylpyrimidin-2-yl) amino] [4-

(trifluoromethy1) pheny1] methy1] quino1in-8-ol

In inert atmosphere 180 ml of acetonitrile, 16.22 g (50 mmol, 0.5 eq) of quinidine, 3.87 g (84 mmol, 0.84 eq) of formic acid, 10.91g (100 mmol, 1.0 eq) of 2-amino-4- methylpyrimidine , 17.41 g (100 mmol, 1.0 eq) of 4- trifluoromethylbenzaldehyde, and finally 17.42 g (120 mmol, 1.2 eq) of 8-hydroxyquinoline were added into a 500 ml round bottom flask. The mixture was stirred for 16 hours at acetonitrile reflux temperature.

The acetonitrile solution was concentrated in reduced pressure to its third volume; the residue was dissolved in 100 ml of dichloromethane . The solution was washed twice with 100 ml of 1M NaOH solution and extracted further six times by 50 ml of 1M NaOH solution. 100 ml of toluene was added to the organic phase then the dichloromethane was evaporated off. The solution obtained was added to 100 ml of 3 M HC1. The phases were separated and the toluene phase was extracted with 30 ml of 3 M HC1 solution. To the combined HC1 phases methyl-t-butyl-ether was added then the pH of the biphasic system was adjusted with 40% NaOH solution to .

The precipitated quinidine was filtered off, the biphasic filtrate was separated then the water layer was washed twice by 20 ml of methyl-t-butyl-ether, the combined ether phase was dried on sodium sulphate and filtered. 30 ml acetonitrile was added to the filtrate, the methyl-t-butyl- ether was evaporated off and the acetonitrile residue was stirred at room temperature for 16 hours. The precipitated

P-1418 crystals were filtered to get 7.61 g (18.5 mmol, HPLC: 95%) of racemic crystalline product.

The mother liquor was concentrated in reduced pressure to give 14.3 g of crude enantiomerically pure R-isomer (HPLC: 80 %) . The crude R-enantiomer was then dissolved in 40 ml of

isopropanol, stirred at room temperature for 48 hours then the precipitated crystals were filtered off, to give 5.67 g (13.8 mmol, HPLC: 98.9%, ee: 99%) of pure crystalline R-enantiomer. 1 H-NMR (ppm) : 10.1, 1H, s; 8.85, 1H, dd; 8.30, 1H, dd; 8.15, 1H, d; 8.07, 1H, d; 7.75, 1H, d; 7.65, 2H, d; 7.60, 2H, d; 7.53, 1H, dd; 7.41, 1H, d; 7.09, 1H, d; 6.51, 1H, d; 2.25, 3H, s

Example 3

Preparation of enantiomerically pure

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-ol

In inert atmosphere 300 ml of acetonitrile, 18 g (55 mmol, 0.5 eq) of quinine, 3.05 g (66 mmol, 0.80 eq) of formic acid, 22.00 g (200 mmol, 2.5 eq) of 2-amino-4- methylpyrimidine, 51.00 g (290 mmol, 3.7 eq) of 4- trifluoromethylbenzaldehyde, and finally 11.5 g (79 mmol, 1.0 eq) of 8-hydroxyquinoline were added in a 1 1, four necked round bottom flask.

The mixture was stirred at 73 °C for six days.

The solvent was evaporated off in reduced pressure. The residue (93.4 g) was dissolved in 200 ml of dichloromethane and chromatographed on 100 g silica gel.

The fractions containing the product were collected and the solvent was evaporated off (60.4 g) . The raw product obtained was purified by normal phase Flash chromatography using hexane- ethyl-acetate gradient then the fractions containing the product were collected and concentrated (19.81 g) .

The residue was dissolved in 190 ml of 2-propanol. After 2 hours stirring the precipitated racemic crystals were filtered

P-1418 off. The mother liquor was concentrated in vacuum obtaining 13.44g of pure product (HPLC:96.2%, ee : +99%) . .

1H-NMR (ppm) : 10.1, 1H, s; 8.85, 1H, dd; 8.30, 1H, dd;

8.15, 1H, d; 8.07, 1H, d; 7.75, 1H, d; 7.65, 2H, d; 7.60, 2H, d; 7.53, 1H, dd; 7.41, 1H, d; 7.09, 1H, d; 6.51, 1H, d; 2.25, 3H, s.

Example 4

Preparation of enantiomerically pure

Potassium-7- [ (R) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-olate

10 g (24.4 mmol) of neutral compound according to Example 2 was dissolved in 60 ml of ethanol then a solution of 2.73 g (24.4 mmol) of potassium tert-butoxide in ethanol (60 ml) was added drop-wise. After 1 hour stirring 360 ml of methylcyclohexane was added and the alcohol was evaporated off in reduced pressure. After 16 hours stirring the precipitated crystals were filtered to get 6.09 g of pure product (13.6 mmol, HPLC: 98.1%, ee: +99%) .

1H-NMR (ppm) : 9.97, 1H, s; 8.43, 1H, dd; 8.07, 1H, d; 7.87, 1H, dd; 7.76, 2H, d; 7.51, 2H, d; 7.24, 1H, d; 7.14, 1H, dd; 6.39, 1H, d; 6.38, 1H, d; 6.15, 1H, d; 3.38, 1H, s; 2.21, 3H, s.

13 C-NMR (ppm) : 161.5, 160.3, 150.0, 145.3, 139.8, 138.0, 129.6, 128.5, 127.3, 124.6, 122.7, 121.0, 109.8, 107.6, 67.0, 56.6, 25.1, 23.7

Example 5

Preparation of enantlomerxcally pure

Potassium-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-olate

6.12 g (13.9 mmol) of neutral compound according to Example 3 was dissolved in 17 ml of ethanol then a solution of 1.56 g (13.9 mmol) of potassium tert-butoxide in ethanol (9 ml) was added drop-wise. After 1 hour stirring 55 ml of

P-1418 methylcyclohexane was added then the alcohol was evaporated off in reduced, pressure and after 16 hours stirring the precipitated crystals were filtered to get 5.08 g of pure product (13.6 mmol, HPLC: 98.1%, ee: +99%) .

1H-NMR (ppm) : 9.97, 1H, s; 8.43, 1H, dd; 8.07, 1H, d; 7.87, 1H, dd; 7.76, 2H, d; 7.51, 2H, d; 7.24, 1H, d; 7.14, 1H, dd; 6.39, 1H, d; 6.38, 1H, d; 6.15, 1H, d; 3.38, 1H, s; 2.21, 3H, s.

1 3 C-NMR (ppm) : 161.5, 160.3, 150.0, 145.3, 139.8, 138.0, 129.6, 128.5, 127.3, 124.6, 122.7, 121.0, 109.8, 107.6, 67.0, 56.6, 25.1, 23.7

Example 6

Preparation of enantxomerically pure

Sodium 7- [ (R) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-olate

0.50 g (1.22 mmol) of neutral compound according to Example 2 was dissolved in 5 ml of ethanol then this solution was added drop-wise to 1 ml (1.22 mmol) of ethanol solution of sodium ethoxide (28 mg Na + 1 ml ethanol) . After 1 hour stirring 360 ml of methylcyclohexane was added then the alcohol was evaporated off in reduced pressure and after 16 hours stirring the precipitated crystals were filtered, and 408 mg (0.94 rmnol) pure product was obtained. Example 7

Preparation of enantiomer-pure

7- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl) phenyl] methyl] quinolin-8-ol fumarate

22.6 g (55.0 mmol) of neutral compound according to Example 2 was dissolved in 30 ml of ethanol then a solution of 3.2 g (27.5 mmol) of fumaric acid in ethanol (90 ml) was added drop-wise. After 16 hours stirring the precipitated

crystals were filtered to get 16.8 g of pure product (35.9 mmol, HPLC: 98.1%, ee: 99.4%) .

P-1418 1 H-NMR (ppm) : 8.86, 1H, d; 8.30, 1H, dd; 8.15, 1H, d; 8.06, 1H, d; 7.74, 1H, d; 7.65, 2H, d; 7.59, 2H, d; 7.55, 1H, dd; 7.41, 1H, d; 7.07, 1H, d; 6.63, 1H, s; 6.51, 1H, d; 2.25, 3H, s.

1 3 C-NMR (ppm) : 167.4, 165.9, 161.5, 149.5, 148.3, 148.1, 138.0, 136.0, 133.9, 127.6, 127.3, 127.1, 126.7, 125.1, 124.5, 121.8, 117.5, 110.3, 51.6

Example 8

Preparation of enantiomerically pure

7- [ (S) - [ (4-Methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl) phenyl] methyl] quinolin-8-ol fumarate

9.21 g (22.4 mmol) of neutral compound according to Example 3 was dissolved in 15 ml of ethanol then a solution of 1.30 g (11.2 mmol) of fumaric acid in ethanol (40 ml) was added drop-wise. After 16 hours stirring the precipitated

crystals were filtered to get 8.34 g of pure product (17.8 mmol, HPLC: 98.5%, ee: 99%) .

X H-NMR (ppm) : 8.86, 1H, d; 8.30, 1H, dd; 8.15, 1H, d; 8.07, 1H, d; 7.74, 1H, d; 7.65, 2H, d; 7.59, 2H, d; 7.55, 1H, dd; 7.41, 1H, d; 7.07, 1H, d; 6.63, . 1H, s; 6.51, 1H, d; 2.25, 3H, s.

13 C-NMR (ppm) : 167.4, 165.9, 161.5, 149.5, 148.3, 148.1, 138.0, 136.0, 133.9, 127.6, 127.3, 127.1, 126.7, 125.1, 124.5, 121.8, 117.5, 110.3, 51.6 Example 9

Preparation of enantiomerically pure

1- [ (R) - [ (4-Methylpyrimidin-2-yl) amino] [4-

( trifluoromethyl) phenyl] methyl] quinolin-8-ol zinc complex

1.098 g (5.0 mmol) of zinc acetate dihydrate was dissolved in 150 ml of tetrahydrofuran then a solution of 4.104 g (10 mmol) of neutral compound according to Example 2 in tetrahydrofuran (150 ml) was added drop-wise. To the mixture obtained 300 ml of hexane was added drop-wise.

After 16 hours stirring the precipitated crystals were filtered to get 2.73 g (3.09 mmol) of pure product.

P-1418 ¾0 '

1 H-NMR (ppm) : 8.80, 1H, d ; S..75, 1H, s; 8.32, 1H, d; 8.16, 1H, d; 7.75, 2H, d; 7.58, 2H, d; 7.50, 2H, d; 6.85, 1H, d; 6.58, 1H, d; 6.48, 1H, d; 3.60, 1H, t; 2.28, 3H, s; 1.75, 1H, s.

1 C-NMR (ppm) : 161.5, 160.3, 150.0, 145.3, 139.8, 138.0, 129.6, 128.5, 127.3, 124.6, 122.7, 121.0, 109.8, 107.6, 67.0, 56.6, 25.1, 23.7.

Example 10

Preparation of enantiomerically pure

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] 4-nitrophenyl) methyl] - quinolin-8-ol

In inert atmosphere into 18 ml of acetonitrile 1.622 g (5.0 mmol, 0.5 eq) of quinidine, 0.387 g (8.4 mmol, 0.84 eq) of formic acid, 1.091g (10 mmol, 1.0 eq) of 2-amino- 6-methyl- pyridine, 4.379 g (29 mmol, 3.7 eq) of -nitrobenzaldehyde , and finally 1.742 g (12 mmol, 1.2 eq) of 8-hydroxyquinoline were added.

The mixture was stirred at room temperature for four days. The reaction mixture was processing up as given above to get the product as pure R-enantiomer (HPLC:98.9%, ee: 99%) .

C22H18 4O3 (MS: 386.1); HPLC (Chiralpak ADn-Hexane /IPA /TEA = 90/10/0.1) : Tr = 10.30 min. 2 H NMR (DMSO-d 6 ) δ 2.2 (3H, s, CH 3 ) , 6.37 (1H, d, J = 7.0 Hz), 6.49 (1H, d, J = 7.9 Hz), 6.98 (1H, d, J = 8.8 Hz, NHCH), 7.28 (1H, t, J = 7.9 Hz), 7.40 (2H, t, J = 7.9 es 8.8 Hz), 7.50-7.55 (1H, m ), 7.59-7.66 (3H, m) , 8.16 (2H, d, J = 8.8 Hz), 8.28 (1H, d, J = 7.9 Hz), 10.1 (1H, wide s, OH); 13 C NMR (D SO-d 6 ) δ 24.2 (CH 3 ) , 51.5 (CH) , 105.6 (CH) ,

111.6 (CH), 117.7 (CH), 121.9 (CH) , 123.5 (2xCH), 124.5 (Cq) ,

126.7 (CH) , 127.7 (Cq) , 128.2 (2*CH), 136.1 (CH) , 137.3 (CH) , 138.2 (Cq) , 146.2 (Cq) , 148.4 (CH) , 149.8 (Cq) , 152.0, 155.7 and 157.3 (Cq)

P-1418 Example 11

Preparation of enantiomerically pure

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] 4-nitrophenyl) methyl] - quinolin-8-ol

In inert atmosphere into 30 ml of acetonitrile 1.8 g (5.5 mmol, 0.7 eq) of quinine, 0,305 g (6.6 mmol, 0.8 eq) of formic acid, 2.20 g (20 mmol, 2.0 eq) of 2-amino-6- methylpyridine, 4.38 g (29 mmol, 3.7 eq) of 4- nitrobenzaldehyde, and finally 1.15 g (7.9 mmol, 1.0 eq) of 8-hydroxyquinoline were added.

The mixture was stirred at room temperature for six days. The reaction mixture was processing up as given above to get the product as pure S-enantiomer (HPLC:98.9%, ee: 99%) .

C 22 Hi8N 4 03 (MS: 386.1); HPLC (Chiralpak ADn-Hexane /IPA /TEA = 90/10/0.1) : Tr = 10.30 min. X H NMR (DMS0-d 6 ) δ 2.2 (3H, s, CH 3 ) , 6.37 (1H, d, J = 7.0 Hz), 6.49 (1H, d, J = 7.9 Hz), 6.98 (1H, d, J = 8.8 Hz, NHCH) , 7.28 (1H, t, J = 7.9 Hz), 7.40 (2H, t, J = 7.9 es 8.8 Hz), 7.50-7.55 (1H, m ), 7.59-7.66 (3H, m) , 8.16 (2H, d, J = 8.8 Hz), 8.28 (1H, d, J = 7.9 Hz), 10.1 (1H, wide s, OH); 13 C NMR (DMSO-d 6 ) δ 24.2 (CH 3 ) , 51.5 (CH) , 105.6 (CH) ,

111.6 (CH) , 117.7 (CH) , 121.9 (CH) , 123.5 (2*CH) , 124.5 (Cq) ,

126.7 (CH) , 127.7 (Cq) , 128.2 (2*CH) , 136.1 (CH) , 137.3 (CH) , 138.2 (Cq), 146.2 (Cq) , 148.4 (CH) , 149.8 (Cq) , 152.0, 155.7 and 157.3 (Cq)

Example 12

Preparation of enantiomerically pure

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] 3-hydroxyphenyl) - methyl] quinolin-8-ol

In inert atmosphere into 18 ml of acetonitrile 1.622 g (50 mmol, 0,5 eq) of quinidine, 0.387 g (8.4 mmol, 0.84 eq) of formic acid, 1.091 g (10 mmol, 1.0 eq) of 2-amino-6-methyl- pyridine, 3.538 g (29 mmol, 3.7 eq) of 3- hydroxybenzaldehyde, and finally 1.742 g (12 mmol, 1.2 eq) of 8-hydroxyquinoline were added.

P-1418 32

The mixture was stirred at reflux temperature of the solvent for 16 hours. The reaction mixture was processing up as given above to get the product as pure R-enantiomer (HPLC:98.9%, ee: 99%) .

C 2 2H a8 4 03 (MS: 357); HPLC (Chiralpak AD n- Hexane/IPA/TEA=90/10/0.1) : Tr = 8.90 min.

Example 13

Preparation of enantiomerically pure

7- [ (S) - [ ( 6-Methylpyridin-2-yl) amino] 3-hydroxyphenyl) - methyl] quinolin-8-ol

In inert atmosphere into 30 ml of acetonitrile 1.8 g (5.5 mmol, 0.7 eq) of quinine, 0.305 g (6.6 mmol, 0.8 eq) of formic acid, 2.2 g (20 mmol, 2.0 eq) of 2-amino-6-methyl- pyridine, 3.538 g (29 mmol, 3.7 eq) of 3- hydroxybenzaldehyde , and finally 1.15 g (7.9 mmol, 1.0 eq) of 8-hydroxyquinoline were added.

The mixture was stirred at reflux temperature of the solvent for six days. The reaction mixture was processing up as given above to get the product as pure S-enantiomer (HPLC: 98.9%, ee: 99%).

C22H18 4O3 (MS: 357); HPLC (Chiralpak AD n-

Hexane/IPA/TEA=90/10/0.1) : Tr = 17.18 min.

Example 14

Preparation of enantiomerically pure

7- [ (R) - [ ( 6-Methylpyridin-2-yl) amino] 3-methoxyphenyl) - methyl] quinolin-8-ol

In inert atmosphere into 18 ml of acetonitrile 1.622 g (50 mmol, 0,5eq) of quinidine, 0.387 g (8.4 mmol, 0.84 eq) of formic acid, 1.091g (10 mmol, 1.0 eq) of 2-amino-6-methyl- pyridine, 1.52 g (10 mmol, 1.0 eq) of 4-hydroxy-3-methoxy-

P-1418 benzaldehyde, and finally 1.742 g (12 mittol, 1.2 eq) of 8- hydroxyquinoline were added.

The mixture was stirred at reflux temperature of the solvent for 16 hours. The reaction mixture was processing up as given above to get the product as pure R-enantiomer (HPLC: 95 %, ee: 99%) .

C 22 H 18 N 4 0 3 (MS = 387); HPLC (Chiralpak AD n- Hexane/IPA/TEA=90/10/0, 1) : Tr = 8.61 min. Example 15

Preparation of enantiomerxcally pure

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] 3-methoxyphenyl) - methyl] quinolin-8-ol

In inert atmosphere into 30 ml of acetonitrile 1.8 g (5.5 mmol, 0.7 eq) of quinine, 0.305 g (6.6 mmol, 0.8 eq) of formic acid, 2.2 g (20 mmol, 2.0 eq) of 2-amino-6~methyl- pyridine, 4.408 g (29 mmol, 3.7 eq) of 4-hydroxy-3-methoxy- benzaldehyde, and finally 1.15 g (7.9 mmol, 1.0 eq) of 8- hydroxyquinoline were added.

The mixture was stirred at reflux temperature of the solvent for six days. The reaction mixture was processing up as given above to get the product as pure S-enantiomer (HPLC: 95 %, ee: 99%) .

C22H18 4O3 (MS = 387); HPLC (Chiralpak AD n- Hexane/IPA/TEA=90/10/0.1) : Tr = 11.14 min.

Example 16

Preparation of enantiomerxcally pure

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] (5-bromopyridin-2- yl) methyl] quinolin-8-ol

Using 8-hydroxyquinoline, 5-bromopyridine-2-carbaldehyde, 2-amino-6-methylpyridine and quinidine, pure R-enantiomer was obtained according to the general R-process (HPLC: +99%, ee: +99%) .

P-1418 C 21 Hi 7 BrN 4 0 (MS = 421); HFLC ^Lux 5u Cellulose-4, 100*4.6 n- Hexane/IPA/TEA=90/10/0.1) : Tr = 5.20 min.

Example 17

Preparation of enantiomerically pure

7-[ (S) -[ (6-Methylpyridin~2-yl) amino] (5-bromopyridin-2- yl) methyl] quinolin-8-ol

Using 8-hydroxyquinoline, 5-bromopyridine-2-carbaldehyde, 2-amino-6-methlypyridine and quinine, pure R-enantiomer was obtained according to the general S-process (HPLC: +99%, ee : + 99%) .

C2. l H 17 BrN 4 0 (MS = 421); HPLC (Lux 5u Cellulose-4, 100*4.6 n- Hexane/IPA/TEA=90/10/0.1) : Tr - 6.56 min. Example 18

Preparation of enantiomerically pure

7- [ (R) - [ (6-Methylpyridin-2-yl) amino] 2-hydroxyphenyl) - methyl] uinolin-8-ol

Using 8-hydroxyquinoline, 2-hydroxybenzaldehyde, 2-amino-6- methylpyridine and quinidine, pure R-enantiomer was obtained according to the general R-process (HPLC: +99%, ee: + 99%) .

C22H19 3O2 (MS = 357); HPLC (Lux 5u Cellulose-4, 100*4.6 n- Hexane/IPA/TEA=9C/10/0.1) : Tr = 2.39 min.

Example 19

Preparation of enantiomerically pure

7- [ (S) - [ (6-Methylpyridin-2-yl) amino] 2-hydroxyphenyl) - methyl] quinolin-8-ol

Using 8-hydroxyquinoline, 2-hydroxybenzaldehyde, 2-amino-6- methlypyridine and quinine, pure S-enantiomer was obtained according to the general S-process (HPLC: +99%, ee : +99%) . C22H19N3O2 (MS = 357); HPLC (Lux 5u Cellulose-4, 100*4.6 n- Hexane/IPA/TEA=90/10/0.1) : Tr =7.04 min.

P-1418 Example 20

Preparation of enantiomerically pure

5-Chloro-7- [ ( ) - [ (4-methylpyrimidin-2-yl) amino] [4-

( rifluoromethyl)phenyl]methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile , 540 mg (1,67 mmol, 0,5eq) of quinidine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4- methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of 4- ( trifluoromethyl) benzaldehyde, and finally 716 mg (4 mmol, 1,2 eq) 5-chloro-8-hydroxyquinoline were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual

to get the pure product.

C22H16C I F3 4O ; mass (ESI positive mode): 445 (444+H + ). HPLC

(Lux4; Hexane: Isopropanol 95:5) Tr = 22,8 minute.

1H NMR (500 MHz, D6MS0) δ 10,43 (wide s, 1H) , 8.95-8.91 (m,

1H), 8.47-8.42 (m, 1H) , 8.20 (d, J = 9.6 Hz, 1H) , 8.15 (d, J =

4.9 Hz, 1H), 7.71-7.66 (m, 1H) , 7.65 (d, J = 8.2 Hz, 2H) , 7.58 (d, J = 8.1 Hz, 2H) , 7,09 (1H, d, J = 9,5 Hz), 6,51 (1H, d, J =

4,95Hz), 2,24 (3H, s) ; 13C-NMR (125MHz, D6MSO) : 25,5, 110,5,

118,8, 123,0, 125,0, 125,3, 125,4, 126,6, 127,8, 132,5, 138,7,

147,5, 149,2, 149,4, 161,5. Example 21

Preparation of enantiomerically pure

5-Chloro-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl)phenyl]methyl] quinolin-8-ol In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4-methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of

4- (trifluoromethyl) benzaldehyde, and finally 716 mg (4 mmol, 1,2 eq) 5-chloro-8-hydroxyquinoline were added into a round

P-1418 bottom flask.

The mixture was stirred for 6 days at 75 °C temperature.

The reaction mixture was processing up as usual to get the pure product .

C22H16C I F3IS O ; mass (ESI positive mode) : 445 (444+H + ) . HPLC (Lux4; Hexane: Isopropanol 95:5) Tr = 22,8 minute.

1H NMR (500 MHz , D6MSO) δ 10,43 (wide s, 1H) , 8.95-8.91 (m, 1H), 8.47-8.42 (m, 1H) , 8.20 (d, J = 9.6 Hz, 1H) , 8.15 (d, J = 4.9 Hz, 1H), 7.71-7.66 (m, 1H) , 7.65 (d, J = 8.2 Hz, 2H), 7.58 (d, J = 8.1 Hz, 2H), 7,09 (1H, d, J = 9,5 Hz), 6,51 (1H, d, J = 4,95Hz), 2,24 (3H, s) ; 13C-NMR (125MHz, D6MSO) : 25,5, 110,5, 118,8, 123,0, 125,0, 125,3, 125,4, 126,6, 127,8, 132,5, 138,7, 147,5, 149,2, 149,4, 161,5. Example 22

Preparation of enantlomerically pure

5-Chloro-7- [ ( ) - [ (6-methylpyridin-2-yl) amino] [4- ( rifluorome hyl) phenyl] methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinidine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-6-picoline then 580 mg (3,33 mmol, 1,0 eq) of

4- ( ri fluoromethyl ) benzaldehyde , and finally 716 mg (4 mmol, 1,2 eq) 5-chloro-8-hydroxyquinoline were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual to get the pure product.

C23H17CI F3N3O ; mass (ESI positive mode) : 444 (443+H + ) .

Example 23

Preparation of enantlomerically pure

2-Me hyl-7- [ (R) - [ (4-methylpyrimidin-2-yl) amino] [4-

(trifluoromethyl) phenyl]methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, P-1418 0,5eq) of quinidine, 129 mg (2,8 raitiol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4- methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of

4- (trifluoromethyl ) enzaldehyde, and finally 636 mg (4 mmol, 1,2 eq) 8-hydroxyquinaldine were added into a round bottom flask.

The mixture was stirred for 6 days at 75 °C temperature.

The reaction mixture was processing up as usual to get the pure product .

C 2 3Hi9F 3 4 0, mass (ESI positive mode) : 425 (424+H + ) Example 24

Prepara'txon of enantxomerlcally pure

2-Methyl-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino] [4- (trifluorome hyl) phenyl] me hyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile , 540 mg (1,67 mmol, 0,5eq) of quinine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4-methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of 4- ( trifluoromethyl ) benzaldehyde , and finally 636 mg (4 mmol, 1,2 eq) 8-hydroxyquinaldine were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual tc get the pure product.

C23H 19F3N4O , mass (ESI positive mode) : 425 (424+H + ) Example 25

Preparation of enantiomerically pure

2- [ (Dimethylamino) methyl] -7- [ (R) - [ (4-methylpyrimidin-2- yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinidine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4- methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of 4- P-1418 ( trifluoromethyl) benzaldehyde, and finally 808 mg (4 mmol, 1,2 eq) 2- ( (dimethylami.no ) methyl ) quinolin-8-ol were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual to get the pure product .

C25H 24 F 3 5 0; mass (ESI positive mode): 468 (467 +H + ) Example 26

Preparation of enantiomerically pure

2- [ (Dime hylamino) methyl] -7- [ (S) - [ (4-methylpyrimidin-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg ( 3,33 mmol, 1,0 eq) of 2-amino-4 -methylpirimidine then 580 mg (3,33 mmol, 1,0 eq) of 4-

(trifluoromethyl) benzaldehyde, and finally 808 mg (4 mmol, 1,2 eq) 2- [ (dimethylamino) methyl] quinolin-8-ol were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual to get the pure product .

25H 24 F 3 5 0; mass (ESI positive mode) : 468 (467+H + ) Example 27

Preparation of enantiomerically pure

2- [ (Dimethylamino) methyl] -7- [ (R) - [ (4-methylpyridin-2- yl) amino] [4- (trifluoromethyl)phenyl]methyl]quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinidine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4-picoline then 580 mg (3,33 mmol, 1,0 eq) of

-( trifluoromethyl ) benzaldehyde, and finally 808 mg (4 mmol, 1,2 eq) 2- [ (dimethylamino)methyl] quinolin-8-ol were added into a round bottom flask.

P-1418 5

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual to get the pure product .

C26H 5F3 4O ; tomeg (ESI positive mode) : 467 (466+H + )

Example 28

Preparation of enantiomerically pure

2- [ (Dimethylamino) methyl] -7- [ (S) - [ (4-methylpyridin-2- yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinolin-8-ol

In inert atmosphere 3 ml of acetonitrile, 540 mg (1,67 mmol, 0,5eq) of quinine, 129 mg (2,8 mmol, 0,84 eq) of formic acid, 364 mg (3,33 mmol, 1,0 eq) of 2-amino-4-picoline then 580 mg (3,33 mmol, 1,0 eq) of

4- ( trifluoromethyl ) benzaldehyde, and finally 808 mg (4 mmol, 1,2 eq) 2- [ (dimethylamino)methyl] quinolin-8-ol were added into a round bottom flask.

The mixture was stirred for 6 days at 75°C temperature.

The reaction mixture was processing up as usual to get the pure product .

C26H25 3N4O ; mass ( ESI positive mode) : 467 (466+H + ) Example 29

Preparation of enantiomerically pure

5-Nitro-7- [ (R) - [ (4-methylpyrimidin-2-yl) amino]

[4- (trifluoromethyl)phenyl]methyl]quinolin-8-ol

Using 5-nit.ro-8-hydroxyquinoline ,

4- ( trifluoromethyl ) benzaldehyde, 2-amino-4-methlypyrimidine and quinidine, pure R-enantiomer was obtained according to the general R-process (HPLC: +99%, ee: +99%).

C22H16 3 5O3 ; mass (ESI positive mode) : 456 (455+H + )

P-1418 Example 30

Preparation of enantiomerically pure

5-Nitro-7- [ (S) - [ (4-methylpyrimidin-2-yl) amino]

[4- (trifluoromethyl) phenyl] methyl] quinolin-8-ol

Using 5-nitro-8-hydroxyquinoline,

4- ( trifluoromethyl ) benzaldehyde, 2-amino- -methlypyr imidine and quinine, pure S-enantiomer was obtained according to the general S-process (HPLC: +99%, ee: +99%) .

C22H16F3N5O3; mass (ESI positive mode) : 456 (455+H + )

Example 31

Preparation of enantiomerically pure

7- [ <R) - [ (Pyridin-2-yl)

[4- (trifluoromethyl) phen lamino] methyl] quinolin-8-ol

Using 8-hydroxyquinoline, 4- (trifluoromethyl) aniline, 2- pyr idine-carbaldehyde and quinidine, pure R-enantiomer was obtained according to the general R-proeess.

C22H16 3N3O ; mass (ESI positive mode) : 396 (395+H + ) Example 32

Preparation of enantiomerically pure

7- [ (S) - [ (Pyridin-2-yl)

[4- (trifluoromethyl) phenylamino] methyl] quinolin-8-ol

Using 8-hydroxyquinoline, 4- ( trifluoromethyl ) aniline , 2- pyridine-carbaldehyde and quinine, pure S-enantiomer was obtained according to the general S-process.

C22 H 1 6 F3N 3O ; mass (ESI positive mode) : 396 (395+H + )

Example 33

Preparation of enantiomerically pure

2- (Hydroxymethyl) -7- [ (R) - [ (4-methylpyrimidin-2-yl) amino]

[4- (trifluoromethyl) phenyl] methyl] quinolin-8-ol

Using 2-hydroxymethyl-8-hydroxyquinoline , 4-

( tri fluoromethyl ) benzaldehyde, 2 -amino- 4 -methylpy imidine

P-1418 and quinidine, pure R-enantiomer was obtained according to the general R-process.

C23H19F3K O2 ; mass (ESI positive mode) 441 (440+H + ) Example 34

Preparation of enantiomerically pure

2- (Hydroxymethyl) -7- [ (R) - [ (4-methylpyrimidin-2-yl) amino]

[4- (trifluoromethyl) phenyl] meth l] quinolin-8-ol Using 2-hydroxymethyl-8-hydroxyquinoline, 4-

( trifluoromethyl ) benzaldehyde, 2 -amino-4 -methylpyrimidine and quinine, pure S-enantiomer was obtained according to the general S-process.

C23H19 F3 O2 ; mass (ESI positive mode) 441 (440+H + )

VII. Biological examples: In vitro procedures Example 35

Inhibition of matrix metalloproteinase 2 (MMP-2 , 72 kDa gelatinase) , matrix metalloproteinase 8 (MMP-8) , matrix metalloproteinase 10 (MMP-10) , matrix metalloproteinase 12 (MMP-12) , matrix metalloproteinase 13 (MMP-13) , and matrix metalloproteinase 14 (MMP-14) activity with compounds 7- [ (R) - [ (4- ethylpirimidine-2-yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol and 7-[(S)-[(4- Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl) phenyl] methyl] quinoline-8-ol according to Examples 2 and 3.

Different matrix metalloproteinase activities were investigated in a set of fluorophotometric, biochemical measurements. Recombinant matrix metalloproteinase enzymes were pre-incubated at 37 °C with different concentrations (25 μΜ-195 n ) of compounds 7- [ (R) - [ ( - ethylpirimidine-2-yl ) amino] [4-

(trifluoromethyl)phenyl]methyl]quinoline-8-ol and 7-[(S)-[(4-

P-1418 MethyIpirimidine-2-yl ) amino J [ 4-

(trifluoromethyl) phenyl ] methyl ] quinoline-8-ol according to Examples 2 and 3.

Cleavage of the applied substrate resulted in a fluorescently active molecule which was measured one hour after reaction start with a Wallac Victor microtiter plate reader by using 355 ran extinction and 460 nm emission filters. Enzyme activities were compared to controls which were not treated with the inhibitor, and are given as percentages. Figure 5 shows the activity of the six investigated MMP enzymes being dependent on concentrations of compounds according to Examples 2 and 3. According to the IC 50 values (half maximal inhibitory concentrations) shown in the figure, the R- and S-enantiomers inhibited activities of the investigated enzymes to a similar extent.

Example 36

Treatment with the compound 7- [ (R) - [ (4-Methylpirimidine-2- yl) amino] [4- (trifluoromethyl)phenyl]methyl] quinoline-8-ol according to Example 2 for inhibition of H 2 0 2 -induced cell death of cardiac muscle cells in vitro.

Cell line H9c2 (ATCC, Rockville, 10MD, USA) , derived from embryonic rat heart, was cultured in Dulbecco's Modified Eagle Medium containing 10% bovine serum, 4 mM L-glutamine (Sigma- Aldrich, Hungary) , 100 U/ml penicillin, and 100 g/ml streptomycin. Cells were cultured in 100 mm TC-treated culture dishes (Orange Scientific, Belgium) in an incubator set at 37 C and 5% C0 2 with humid air. For real-time monitoring of cell viability, a Roche xCELLigence SP and DP (ACEA-Roche, Hungary) was used, which gives us information about cell viability on the basis of changes in cell conductivity. Prior to plating, the special 96-well e-plate was covered with 0.2% type I collagen, and then placed into the incubator for 30 min. Cell- free baseline impedance was measured once in a minute for 10 min. After plating (6000 cells/well), measuring began.

P-1418 Treatment of cells was always carried out the morning after plating. Measurement proceeded for 72 h.

Results are shown in Figure 6. Curve 1 refers to untreated control, curve 2. to control treated with H 2 0 2 , curve 3, 4, 5, 6 refer to different concentrations (3: 0.11 μΜ compound according to Example 2 + 900 μΜ of H 2 0 2 , 4: 0.33 μΜ compound according to Example 2 + 900 μΜ of H 2 0 2 , 5: 1 μΜ compound according to Example 2 + 900 μΜ of H 2 0 2 , 6: 3 μΜ compound according to Example 2 + 900 μΜ of H 2 0 2 ) of the compound according to Example 2 (7- [ (R) - [ (4-Methylpirimidine-2- yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinoline-8 -ol ) . B: refers to percentage of viability at 6 and 24 h after treatment, compared to control cells. Example 37

Treatment with the compound 7- [ (R) - [ (4-MethylpirimidLine-2- yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline-8-ol according to Example 2 for inhibition of H 2 0 2 - nduced cell death of SH-SY5Y human neuroblastoma cells in vitro

SH-SY5Y cells (ATCC, Rockville, 10MD, USA) were cultured in 100 mm TC-treated culture dishes (Orange Scientific, Belgium) in an incubator set at 37 ° C and 5% C0 2 with humid air. For real-time monitoring of cell viability, a Roche xCELLigence SP and DP (ACEA-Roche, Hungary) was used, which gives us information about cell viability on the basis of changes in cell conductivity. For plating, DMEM (Dulbecco's Modified Eagle Medium) supplemented with 10% FBS was used. Prior to plating, the special 96-well e-plate was covered with 0.2% type I collagen, and then placed into the incubator for 30 min. Cell- free baseline impedance was measured once in a minute for 10 min. After plating (20.000 cells/well), measuring began. Treatment of cells was always carried out the morning after plating. Measurement proceeded for 72 h.

Results are shown in Figure 7 Curve 1 refers to untreated control, curve 2 to control treated with H 2 0 2 ,, curve 3, 4, 5,

P-1418 6 refer to different concentrations (3: 0.11 μ compound according to Example 2 + 500 μΜ of H 2 0 2 , 4: 0.33 μΜ compound according to Example 2 + 500 μΜ of H 2 0 2 , 5: 1 μΜ compound according to Example 2 + 500 μΜ of H 2 0 2/ 6: 3 μΜ compound according to Example 2 + 500 μΜ of H 2 0 2 ) of the compound according to Example 2 (7- [ (R) - [ (4-Methylpirimidine-2- yl) amino] [ - (trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol ) . B: refers to percentage of viability at 6 and 24 h after treatment, compared to control cells.

Example 38

Treatment with the compound 7- [ (S) - [ (4-Methylpirimidine-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol according to Example 3 for inhibition of H 2 0 2 -induced cell death of cardiac muscle cells in vitro

Cell line H9c2 (ATCC, Rockville, 10MD, USA) , derived from embryonic rat heart, was cultured in Dulbecco 1 s Modified Eagle Medium containing 10% bovine serum, 4 mM L-glutamine (Sigma- Aldrich, Hungary) , 100 U/ml penicillin, and 100 pg/ml streptomycin. Cells were cultured in 100 mm TC-treated culture dishes (Orange Scientific, Belgium) in an incubator set at 37 C and 5% C0 2 with humid air. For real-time monitoring of cell viability, a Roche xCELLigence SP and DP (ACEA-Roche, Hungary) was used, which gives us information about cell viability on the basis of changes in cell conductivity. Prior to plating, the special 96-well e-plate was covered with 0.2% type I collagen, and then placed into the incubator for 30 min. Cell- free baseline impedance was measured once in a minute for 10 min. After plating (6000 cells/well), measuring began. Treatment of cells was always carried out the morning after plating. Measurement proceeded for 72 h.

Results are shown in Figure 8. Curve 1. refers to untreated control, curve 2. to control treated with ¾(¾,, curve 3, 4, 5, 6 refer to different concentrations (3: 0.11 μΜ compound according to Example 3 + 900 μΜ of H 2 0 2 , 4: 0.33 μΜ compound

P-1418 according to Example 3 -t 900 μΜ of H 2 0 2 , 5 : 1 μΜ compound according to Example 3 + 900 μΜ of H 2 0 2 , 6: 3 μΜ compound according to Example 3 + 900 μΜ of H 2 0 2 ) of the compound according to Example 3. (7- [ (S) - [ (4-Methylpirimidine-2- yl)amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol) . B: refers to percentage of viability at 6 and 24 h after treatment, compared to control cells.

Example 39

Treatment with the compound 7- [ (S) - [ (4-Methylpirimidine-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol according to Example 3 for inhibition of H 2 0 2 -induced cell death of SH-SY5Y human neuroblastoma cells in -vitro

SH-SY5Y cells (ATCC, Rockville, 10MD, USA) were cultured in 100 mm TC-treated culture dishes (Orange Scientific, Belgium) in an incubator set at 37 ° C and 5% C0 2 with humid air. For real-time monitoring of cell viability, a Roche xCELLigence SP and DP (ACEA-Roche, Hungary) was used, which gives us information about cell viability on the basis of changes in cell conductivity. For plating, DMEM (Dulbecco's Modified Eagle Medium) supplemented with 10% FBS was used. Prior to plating, the special 96-well e-plate was covered with 0.2% type I collagen, and then placed into the incubator for 30 min. Cell- free baseline impedance was measured once in a minute for 10 min. After plating (20.000 cells/well), measuring began. Treatment of cells was always carried out the morning after plating. Measurement proceeded for 72 h.

Results are shown in Figure 9 Curve 1. refers to untreated control, curve 2 to control treated with H 2 0 2 , , curve 3, 4, 5, 6 refer to different concentrations (3: 0.11 μΜ compound according to Example 3 + 500 μΜ of H 2 0 2 , 4: 0.33 μΜ compound according to Example 3 + 500 μΜ of H 2 0 2 , 5 : 1 μΜ compound according to Example 3 + 500 μΜ of H 2 0 2 , 6: 3 μΜ compound according to Example 3 + 500 μΜ of H 2 0 2 ) of the compound according to Example 3 (7- [ (S) - [ (4-Methylpirimidine-2-

P-1418 yl) amino] [4- (trif luoromethyl) pneny 1 ] methyl ] quinoline-8-ol) . B: refers to percentage of vi a bi l it y at 5 and 24 h after treatment, compared to control cells. Example 40

Treatment with the compound Potassium-7- [ (R) - [ (4- Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl)phenyl]methyl]quinoline-8-olate according to Example 4 for inhibition of cell death caused by fibrillar aggregates of amyloid peptides in primary cortical neurons in vitro

Cortical neurons from embryonic day 16-17 Wistar rat fetuses were maintained as previously described [1]. Peptide used: Αβΐ- 42 (Bachem, ref. H1368, batch .# 1025459) . Oligomer preparation: the preparation of Αβ oligomers was performed according to SynAging standard operating procedures. The oligomeric preparation contains a mixture of stable dimers, trimers and tetramers of AB1-42 peptide, as well as monomeric forms of the peptide. The same preparation of oligomers was used for all experimental set-ups and has been previously characterized in terms of oligomer composition, neurotoxicity in vitro as well as induction of cognitive impairment.

Primary cortical neurons were incubated with increasing concentrations of the compound according to Example 2 in the presence or absence of 1 DM Αβ1-42 oligomers. Following 24 h incubation, neuronal viability was monitored using the calcein- AM assay as previously described [1, 2] . Data shown in Figure 10 are obtained from 3-4 separate experiments (mean ± SEM) . Student's t-test (**, p < 0.05, p < 0.001) as well as ANOVA followed by a Scheffe's post hoc test were used to test statistical significance.

P-1418 Example 41

Treatment with the compound 7- [ (R) - [ (4- ethylpirimidine-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol zinc complex according to Example 9 for inhibition of cell death caused by fibrillar aggregates of amyloid peptides in primary cortical neurons in vitro

Cortical neurons from embryonic day 16-17 Wistar rat fetuses were maintained as previously described [1]. Peptide used: Αβΐ- 42 (Bachem, ref. H1368, batch # 1025459) . Oligomer preparation: the preparation of Αβ oligomers was performed according to SynAging standard operating procedures. The oligomeric preparation contains a mixture of stable dimers, trimers and tetramers of AB1-42 peptide, as well as monomeric forms of the peptide. The same preparation of oligomers was used for all experimental set-ups and has been previously characterized in terms of oligomer composition, neurotoxicity in vitro as well as induction of cognitive impairment.

Primary cortical neurons were incubated with increasing concentrations of the compound according to Example 2 in the presence or absence of 1 ΠΜ AB1-42 oligomers. Following 24 h incubation, neuronal viability was monitored using the calcein- AM assay as previously described [1, 2] . Data shown in Figure 11. are obtained from 3-4 separate experiments (mean ± SEM) . Student's t-test (**, p < 0.05, p < 0.001) as well as ANOVA followed by a Scheffe's post hoc test were used to test statistical significance.

VIII. Biological examples, in vivo procedures In vivo experiments were carried out according to the National Institute of Health guidelines for the care and use of laboratory animals, approved by the French Ministry for Research and Technology. In the course of experiments, animals were housed at a standard temperature (22±1 °C) and in a light- controlled environment (lights on from 7 a.m. to 8 p.m.) with P-1418 ad libitum access to food and water. Male C57BL/6 mice (C57BL/6J Rj , ref. SC.-CJ- 12w- , Janvier, France) were housed 5 animals per cage. From one week before the start of the experiment until the end of the experiment, mice were housed individually. Animals were monitored twice-a-day by laboratory personnel (8 a.m. and 4 p.m.). In this project, young (14 weeks old) as well as aged (15-16 months old) mice were used. Experiments were carried out using 12 mice per experimental group .

Investigation of short-term working memory - Y maze test

Y maze tests were carried out as previously described [1, 3]. Prior to testing, mice were brought to the experimental room for at least 30 min to acclimatize to experimental room conditions. The Y maze is made of opaque Plexiglas and each arm is 40 cm long, 16 cm high, 9 cm wide and positioned at equal angles. The apparatus was placed in the test room in such a way that it was brightened homogeneously with 12-15 lux in the arms as well as in the central zone. One mouse at a time was placed at the end of one arm and was allowed to move in the maze freely during a 5 min examination. Each arm entry was recorded with those entries being counted where the mouse completely placed its hind paws in the chosen arm. Alternation was defined as successive entry into the 3 arms on overlapping triplet sets. Results were calculated as the percentage of the ratio of actual detected alternations to possible alternations (defined as the number of total arm entries minus 2) .

Example 42.

Effects of treatment with compounds 7-[(R)-[(4- Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl)phenyl]methyl]quinoline-8-ol and 7-[(S)-[(4- Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl) phenyl]me hyl] quinoline-8-ol according to Example 2 and 3 on short-term memory impairment caused by

P-1418 fibrillar aggregates of amyloid peptides in in vivo C57BL6/J mice

For the examination of memory impairment caused by Αβ oligomers, a mouse model developed and validated by SynAging was used [1-3] . Mice receiving icv injection of Αβ oligomers develop memory deficits associated with a decrease of hippocampal synaptic protein levels. Under anesthetization, 1 μΐ of soluble Α β oligomers (50 pmolj or vehicle (saline) was injected into the right ventricle. The stereotaxic coordinates from the bregma are as follows (in mm): AP-0.22, L-1.0 and D- 2.5. For treatments, a 10 ml Hamilton microsyringe fitted with a 26-gauge needle was used. Treatment with molecules described in Example 2. and 3. was carried out once daily for four days with the use of a per os probe (100 μΐ/animal) . Four experimental groups were established: Group A - vehicle/AB oligomer, Group B - compound according to Example 2 (4 mg/kg body weight) and Αβ oligomer, Group C - compound according to Example 3 (4 mg/kg body weight) and Αβ oligomer, Group D - compounds from both Example 2. and 3. (4 mg/kg body weight) and Αβ oligomer. At day 4 post Αβ treatment, short-term memory was assessed with the Y maze test.

Treatment with compound 7- [ (R) - [ ( 4-Methylpirimidine-2- yl) amino] [4- (trifluoromethyl) phenyl ] methyl ] quinoline-8-ol according to Example 2 inhibited while treatment with compound 7- [ (S) - [ (4-Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl) phenyl ] methyl ] quinoline-8-ol ) according to Example 3 did not inhibit short-term memory impairment caused by fibrillar aggregates of amyloid peptides in in vivo C57BL6/J mice .

Example 43

Effects of treatment with different concentrations (related to body weight) of compound 7- [ (R) - [ (4-Methylpirimidine-2- yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol according to Example 2 on short-term memory impairment caused

P-1418 by fibrillar aggregates of amyloid peptides in in vivo C57BL6/J mice

Treatment with Αβ oligomers was carried out as previously described in Example 16. Treatment with the examined compound according to Example 2 was carried out once daily for four days with the use of a per os probe (100 μΐ/animal) . Four experimental groups were established: Group A - vehicle/AB oligomer, Group B - compound according to Example 2. (0.5 mg/kg body weight) and Αβ oligomer, Group C - compound according to Example 2 (2 mg/kg body weight) and Αβ oligomer, Group D - compound according to Example 2 (4 mg/kg body weight) and Αβ oligome .

At day 4 post Αβ treatment, short-term memory was assessed with the Y maze test. Results are summarized in Figure 13.

Treatment with compound 7- [ (R) - [ (4- ethylpirimidine-2- yl) amino] [4- (trif luoromethyl) phenyl] methyl] quinoline-8-ol according to Example 2 inhibited short-term memory impairment caused by fibrillar aggregates of amyloid peptides in in vivo C57BL6/J mice in a dose-dependent manner.

Example 44

Effect of compounds 7- [ (R) - [ (4-Methylpirimidine-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] uinoline-8-ol and 7-[(S)-[(4- ifethylpirimidine-2-yl) amino] [4- (trifluoromethyl) phenyl] methyl] quinoline-8-ol according to Example 2 and 3 on scopolamine-induced short-term memory impairment in in vivo C57BL6/J mice

Scopolamine has been widely used to induce cognitive deficits (typically short-term memory impairment) in animals [FR4] . Test protocol: 30 in prior to behavioral test, scopolamine (0.6 mg/kg) or vehicle was administered i.p. in control and treated mice (12 mice, per experimental groups) . Mice were returned to their cage until the start of the test. Administration of drug- candidates described in Example 2. and 3. was carried out with a per os gavage (administration time based on PK data) . Y-maze P-1418 tests were carried out as previously described. Results are summarized in Figure 14.

Treatment with compound 7- [ (R) - [ ( 4-Methylpirirnidine-2- yl) amino] [4- ( trifluoromethyl ) phenyl ] methyl ] quinoline-8-ol according to Example 2 inhibited, while treatment with compound 7- [ (S) - [ (4-Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl)phenyl]methyl]quinoline-8-ol according to Example 3 did not inhibit scopolamine-induced short-term memory impairment in in vivo C57BL6/J mice.

Example 45

Inhibition of the activity of caspase 3 peptidase with compound 7- [ ( ) - [ (4-Methylpirimidine-2-yl) amino] [4-

( r fluorome hyl) phenyl]me hyl] quinoline-8-ol according to Example 2

Throughout the experiments, human caspase 3 enzyme produced in E. coli was used as previously described (Mittl PRE, Marco SD, Krebs JF, Karanewsky DS, Priestle JP, Tomaselli KJ and Grutter MG (1997) Structure of Recombinant Human CPP32 in Complex with the Tetrapeptide Acetyl-Asp-Val-Ala-Asp Fluoromethyl Ketone. J Biol Chem. 272: 6539-6547). In brief: the enzyme (150 U/ml) was pre-incubated together with the compound according to Example 2 (10 μΜ) in modified HEPES buffer (50 mM HEPES, pH=7.4, 100 mM NaCl, 0 . 1 % CHAPS, 1 mM EDTA, 10% glycerin, 10 mM DTT) for 15 min at 37 ° C. After the addition of 50 mM Ac-DEVD-AMC substrate, the reaction ran for 60 min. The amount of generated AMC ( 7-amino-4-methylcoumarine) , which gives information about the enzyme activity, was detected by fluorescence at 360/465 nm. At the applied concentration of 10 mM, the compound according to Example 2 reduced the activity of caspase 3 enzyme by 74%.

P-1418 Example 46

Transcriptional inhibition of NF-AT (nuclear factor of activated T cells) protein with the compound 7-[(R)-[(4- Methylpirimidine-2-yl) amino] [4- (trifluoromethyl) phenyl]methyl] quinoline-8-ol according to Example 2

Experiments were carried out on human T Jurkat cells transfected with , a reporter construct expressing β- galactosidase and containing the binding site of NFAT1 transcription factor based on the previously described method (Emmel EA, Verweij CL, Durand DB, Higgins KM, Lacy E and Crabtree GR (1989) Cyclosporin A specifically inhibits function of the nuclear proteins involved in T cell activation. Science. 246:1617-1620.; Karttunen J and Shastri N (1991) Measurement of ligand-induced activation in single viable T cells using the lacZ reporter gene. Proc Natl Acad Sci USA. 88:3972). Cells (3xl0 6 cells/ml) were incubated with 10 μΜ compound according to Example 2 in RPMI-1640 medium (pH=7.4) for 20 min at 37 ' c . Following this, incubation lasted for an additional 4 h in the presence of 0.5 μΜ A23187 and 50 ng/ml P A (12-0- tetradecanoylphorbol-13-acetate) . Induced effects can be determined by the β-galactosidase activity of cells treated with the compound according to Example 2 and untreated cells as it catalyzes the transformation of FDG (fluorescein di-p-D- galactopyranoside) into fluorescein. Reading of results was carried out with a SpectraFluor Plus plate reader. Decrease of β -galactosidase activity was compared to the effect of 1 μΜ cyclosporine A which was served as a positive control. The compound according to Example 2 Reduced the transcriptional activity of NF-AT protein by 2.2 μΜ half-value concentration. Results are summarized in Figure 15.

P-1418 Example 47

Inhibition of 5-lipoxygenase enzyme activity with the compound 7- [ (R) - [ (4-Methylpirimidine-2-yl) amino] [4-

(trifluoromethyl)phenyl]methyl]quinoline-8-ol according to Example 2

Human 5-lipoxygenase produced by Sf9 cells was used throughout the experiments. The enzyme was pre-incubated with 10 μΜ compound according to Example 2 at room temperaturethen - after the addition of 25 μΜ arachidonic acid substrate - the amount of generated rhodamine 123 was determined by fluorescence measurement. Percentage of inhibition rate was calculated by comparison to untreated control reactions. In 10 μΜ concentration the compound according to Example 2 inhibited 5- lipoxygenase activity by 54%.

Example 48

Gene expression influencing effect of the R-enantiomeric derivative according to Example 2 on primary astrocytes

Pure astrocyte culture was obtained from neonatal rats according to the protocol found in literature (Protocols for Neural Cell Culture 2001, pp 117-127. ch 9. Ruth Cole, Jean de Vellis. Preparation of Astrocyte, Oligodendrocyte, and Microglia Cultures from Primary Rat Cerebral Cultures) . Astrocytes (1 million) were cultured in 100 mm TC-treated culture dishes (Orange Scientific, Belgium) in an incubator set at 37 ° C and 5% C0 2 with humid air. The R-enantiomeric derivative according to Example 2 was added to cells in two final concentrations: 330 nM and 1000 nM. Cells were also treated with 1000 nM PBT2 which bears the basic structure of the R-enantiomer according to Example 2 ( 8-hydroxy-quinoline ring) but contains conformational differences and was developed for the treatment of neurodegenerative disorders (Pub. No.: WO/2004/007461) . Treatments were carried out with a 100 μΜ DMSO-based stock solution. Control cells were treated with the same amount of DMSO without the agent. Each treatment was

P-1418 conducted in three separate culture dishes. After treatment, cells were incubated at 37 'c for 3 h then the medium was removed, cells were washed with PBS and total RNA was extracted from cells with the Accuzol™ Total RNA Extraction Solution (Bioneer, Daejeon, South Korea), . according to the manufacturer's protocol. cDNA transcription was carried out with the High Capacity cDNA Reverse Transcription Kit (Life Technologies, Foster City, CA, USA) . During the test, the expression of four genes (PGK1, EPO, HMOX-1, VEGF) controlled by HIFla was determined by qRT-PCR, with HPRT gene serving as a control. Relative expression of genes in treated samples was compared to untreated controls. The PCR was conducted with a LightCycler® Nano Instrument (Roche, Budapest, Hungary) in the presence of UPL (Universal Probe Library) probes (HPRT: 95, HMOX1: 4, EPO: 16, VEGF: 1,PGK1: 66), specific primers (HPRTl: 5'- gaccggttctgtcatgtcg-3' , HPRT2: 5' - acctggttcatcatcactaatcac—3' ; HMOXl-1: 5' -gtcaagcacagggtgacaga- 3', HMOX1-2: 5' -ctgcagctcctcaaacagc-3 ' ; EPOl: 5'- agtcgcgttctggagaggta-3' , EP02: 5'- ccttctgcacagcccatt-3' ; VEGF1: 5'- aaaaacgaaagcgcaagaaa-3' , VEGF2: 5'- tttctccgctctgaacaagg-3' ; PGKl-1: 5'- ccagataacgaataaccaaagga- 3', PGK1-2: 5'- gacttggctccattgtcca-3 ' ) in 20 μΐ PCR reaction volume with 20 ng cDNA template and 10 μΐ Lightcycler DNA Probes Master (5x) Reagent Kit (Roche) according to the following protocol: activation of enzyme at 95 C for 10 min, 50 cycles: denaturation at 95 ° C for 15 sec, hybridization and polymerizationthen detection at 60 ° C for 30 sec. Results are shown in Figure 16. While the compound according to Example 2 caused dose-dependent and robust activation of EPO and HMOX-1 genes, even 1000 nM of PBT2 did not induced differences in gene activity.

P-1418 Example 49

Effect of chronic treatment with the R-enantiomeric derivative according to Example 2 on erythropoietin gene expression in the hippocampus and cortex of aged animals

Twenty-four 18-month-old C57BL/6 female mice (Innovo Ltd., Budapest, Hungary) were divided into two groups randomly: Group 1 - untreated control, Group 2 - treated with the R-enantiomer according to Example 2, administered in drinking water (20 mg/1) for four months. From both groups, 10-10 mice were sacrificed by C0 2 inhalation. After the withdrawal reflex ceased, the whole animal was subjected to perfusion with lx phosphate buffer according to the described protocol (Whole Animal Perfusion Fixation for Rodents; Gregory J. Gage, Daryl R. Kipke, William Shain; J. Vis. Exp. (65), e3564, doi:10.3791/3564 (2012). After perfusion, brain tissue was removed from the animals and hippocampal and cortical regions were separated. Half of the brain was processed per animal. Half of the samples was used for gene expression studies, the other half was used for protein expression (Example 35.) studies.

To study gene expression, total RNA was extracted from the brain tissues with Accuzol™ Total RNA Extraction Solution (Bioneer, Daejeon, South Korea), according to the manufacturer's protocol. cDNA transcription was carried out with the High Capacity cDNA Reverse Transcription Kit (Life Technologies, Foster City, CA, USA) . Expression of the gene coding for erythropoietin, the EPO gene was determined by qRT- PCR (with HPRT gene serving as a control) with the method and instrument described in Example 33. but here, mouse-specific UPL probes (HPRT: 95, EPO: 16) and primer pairs were applied (mouse HPRT1: 5'- tcctcctcagaccgctttt-3' , mouse HPRT2: 5'- cctggttcatcatcgctaatc-3' ; mouse EPOl : 5'- tctgcgacagtcgagttctg- 3', mouse EP02 : 5'- cttctgcacaacccatcgt-3' ) . One hippocampal control, one cortical control and one treated cortical sample did not give appreciable results. Results are shown in Figure

P-1418 17. Chronic treatment with the R-enantiomeric derivative according to Example 2 induced both hippocampal and cortical EPO expression compared to samples of untreated control mice. Example 50

Effect of chronic treatment with the R-enantiomeric derivative according to Example 2 on erythropoietin protein expression in the hippocampus and cortex of aged animals

Twenty-four 18-month-old female C57BL/6 mice (Innovo Ltd., Budapest, Hungary) were treated according to the experimental setup described in Example 34. in two groups: Group 1 - control group given normal drinking water, Group 2 - treated with the R-enantiomer according to Example 2, administered in drinking water (20 mg/1) for four months. Brain preparation was carried out under conditions described in Example 34. Brain samples were washed with lx PBS to remove bloodthen tissues were homogenized in lx PBS and for 3 days, they were held at -20 ° C overnight and 25 C at daytime every day. After the third round of thawing, samples were centrifuged (5 min, 5000 rpm) and the supernatant was collected for further protein expression studies. Determination of erythropoietin levels of samples was done with Quantikine® ELISA Mouse Erythropoetin (R&D Systems, cat. no.: MEP00B) according to the manufacturer's protocol. At the end of the test, the developoed colour was measured with a THERMO-LABSYSTEMS Multiskan FC Absorbance Plate Reader at 450 nm. Results obtained through ELISA and measured at 450 nm were compared to standard dilution series of purified mouse EPO. Values (pg/ml) obtained based on the dilution series were compared to actual protein content of tissues. Protein content of tissue homogenates was measured with Nanodrop-1000 adjusted for BSA protein, at 280 nm. Two hippocampal and two cortical controls as well as one treated hippocampal and cortical sample did not give appreciable results. Results are shown in Figure 18. Chronic treatment with the R-enantiomeric derivative according to Example 2 induced increased EPO protein levels in

P-1418 both hippocampal and cortical samples compared to samples of untreated control mice.

Example 51.

Inhibition of skin graft rejection with the compound 7-[(R)- [ (4-me hylpyrimidin-2-yl) amino] [4

(trifluoromethyl) henyl]methyl] quinoline-8-ol according to Example 2

Mouse skin transplantation is a standard method to assay host T cell responses to MHC-disparate donor antigens, which protocol was followed in the present example (Garrod and Cahalan; 2008). Briefly, WT C57BL/6 recipient mice (8-12 weeks old) and WT BALB/c donor mice (8-12 weeks old) were anesthetized with a combination of 10 mg/kg Xylazine and 100 mg/kg Ketamine administered by i.p. injection and 0.05 mg/kg Buprenorphine was administered by subcutaneous injection for analgesia. Ear skin (1.0 cm.2) from the WT BALB/c donor mouse was grafted onto the flank of the WT C57BL/6 recipient. The implanted skin (graft) was covered with a sterile bandage, which was removed on day 7 post-transplant.

Recipients received either no treatment (controls) , or treated with compound according to Example 2 orally at 5 mg/kg dose, once every day until the end of the experiment from the day of transplantation. As positive control we administered tacrolimus at 5 mg/kg dose, once every day until the end of the experiment from the day of transplantation via intraperitoneal administration .

Different scores were used to define the quality of the grafts: intact grafts (Score 0), early stages of rejection (Score 1-4, corresponding to first clear signals of graft rejection: 1; >25% rejection rate: 2; >50% rejection rate: 3; >75% rejection rate: 4) up to complete graft rejection (Score 5). The different scores describe rejection by the area that was destroyed by the immune system of the host. Scoring was done at day 9 and day 10 post-transplant. Each group contained 6

P-1418 animals. Average scores with SEM were calculated for each group.

We can see from the results (Figure 18) that both the positive control, tacrolimus compound as well as the compound according to Example 2 resulted in smaller score values both on day 9 and on day 10, from which we can conclude that both compounds prevented skin graft rejection.

I . Formulation examples Example 52

20 mg of active ingredient, enantiomerically pure

7- [ ( ) - [ ( -methylpyrimidin-2-yl ) amino] [ 4-

(trifluoromethyl) phenyl] methyl] quinolin-8-ol was mixed with microcrystalline cellulose, mannitol, colloidal anhydrous silicon dioxide and magnesium stearate and the mixture was formulated under pressure, with the usual formulation technic to tablet.

Example 53

20 mg of active ingredient, enantiomerically pure

7- [ (R) - [ (4-methylpyrimidin-2-yi) amino] [4-

(trifluoromethyl) phenyl] methyl] quinolin-8-ol was mixed with microcrystalline cellulose, mannitol, colloidal anhydrous silicon dioxide and magnesium stearate and the homogenised mixture was filled into gelatin capsules as solid powder.

P-1418 References :

Abdul et al., 2009: Abdul HM, Sama MA, Furman JL, Mathis DM, Beckett TL, eidner AM, Patel ES, Baig I, Murphy MP, LeVine H 3rd, Kraner SD, Norris CM. Cognitive decline in Alzheimer's disease is associated with selective changes in

calcineurin/NFAT signaling. J Neurosci. 2009, 29:12957- 12969.;

Araboor et al., 2012: Arabpoor Z, Hamidi G, Rashidi B, Shabrang M, Alaei H, Sharifi MR, Salami M, Dolatabadi HR, Reisi P.

Erythropoietin improves neuronal proliferation in dentate gyrus of hippocampal formation in an animal model of

Alzheimer's disease. Adv Biomed Res. 2012, 1:50.;

Asai et al., 1999: Asai A, Qiu Jh, Narita Y, Chi S, Saito N, Shinoura N, Hamada H, Kuchino Y, Kirino T. High level

calcineurin activity predisposes neuronal cells to apoptosis. J Biol Chem. 1999, 274:34450-34458.;

Bartus, 2000: Bartus, R.T. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp. Neurol. 2000, 163:495e529.

Bach, 2006: Bach FH. Heme Oxygenase-1 and Transplantation

Tolerance Human Immunology 2006, 67:430-432.;

Betti, 1900: Betti, M. Gazz. Chim. Ital. 1900, 30 II, 301;

Betti, 1903: Betti, M. Gazz. Chim. Ital. 1903, 33 II, 2;

Bresgen et al., 2003: Bresgen N, Karlhuber G, Krizbai I, Bauer H, et al. Oxidative stress in cultured cerebral endothelial cells induces chromosomal aberrations, micronuclei, and apoptosis, J. Neurosci. Res. 2003 72:327-333.;

Britton et al., 2002: Britton RS, Leicester KL, Bacon BR. Iron toxicity and chelation therapy. Int J Hematol. 2002, 76:219- 228;

Calabrese et al., 2010: Calabrese V, Butterfield DA, Stella AM, Nutritional antioxidants and the heme oxygenase pathway of stress tolerance: novel targets for neuroprotection in

P-1418 Alzheimer's disease, Ital. J. Biochem. 2003, 52:177-181.; Targeting heme oxygenase-1 for neuroprotec ion and

neuroinflammation in neurodegenerative diseases. Jaz a A, Cuadrado A. Curr Drug Targets. 2010, 11:1517-1531.;

Chen et al., 2003: Chen K, Gunter K, Maines MD. Neurons

overexpressing heme oxygenase-1 resist oxidative stress- mediated cell death, J. Neurochem. 2000, 75:304-313.)

Chong et al., 2013: Chong ZZ, Shang YC, u Y , Cui S, Yao Q, Maiese K. Targeting erythropoietin for chronic

neurodegenerative diseases. Expert Opin Ther Targets. 2013, 17:707-720.;

Chowdhury et al . , 2008: The human oxygen sensing machinery and its manipulation. Chowdhury R, Hardy A, Schofield CJ. Chem Soc Rev. 2008, 37:1308-1319.;

Chu et al., 2013: Chu J, Li JG, Ceballos-Diaz C, Golde T,

Pratico D.The influence of 5-lipoxygenase on Alzheimer's disease-related tau pathology: in vivo and. in vitro evidence. Biol Psychiatry. 2013, 74:321-328.;

Cole and de Vellis, 2001: Ruth Cole, Jean de Vellis.

Preparation of Astrocyte, Oligodendrocyte, and Microglia

Cultures from Primary Rat Cerebral Cultures , Protocols for Neural Cell Culture konyv 2001, 9. fejezet, pp 117-127.;

De Calignon et al., 2010: de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, Hyman BT. Caspase activation precedes and leads to tangles. Nature. 2010, 464: 1201-1204;

Degterev and Yuan, 2008: Degterev A, Yuan J. Expansion and evolution of cell death programmes. Nat Rev ol Cell Biol. 2008, 9:378-390;

Eckert et al., 2003: Eckert A, Marques CA, Keil U, Schussel K, Muller WE. Increased apoptotic cell death in sporadic and genetic Alzheimer's disease. Ann N Y Acad Sci. 2003,

1010: 604-609. ;

Emmel et al., 1991: Emmel EA, Verweij CL, Durand DB, Higgins KM, Lacy E and Crabtree GR ( 1989) Cyclosporin A specifically

P-1418 inhibits function of the nuclear proteins involved in T cell activation. Science. 246:1617-1620.;

Firuzi et al. 2008: Firuzi 0, Zhuo J, Chinnici CM, Wisniewski

T, Pratico D.5-Lipoxygenase gene disruption reduces amyloid- beta pathology in a mouse model of Alzheimer's disease. FAS ' EB

J. 2008, 22:1169-1178.;

Frederickson et al., 2005: Frederickson CJ, Koh JY, Bush AI .

The neurobiology of zinc in health and disease. Nat Rev

Neurosci. 2005, 6:449-462.;

Gage et al., 1999: Gage GJ, Kipke DR, Shain W. Whole Animal

Perfusion Fixation for Rodents J. Vis. Exp. 2012, (65: e3564; Frisch MJ, et al., 2010: Frisch MJ, Trucks GW, Schlegel HB,

Scuseria GE et al., Gaussian 09 rev. B01, Gaussian, Inc.,

Wallingford CT, 2010.

Hagemeyer et al., 2012: Hagemeyer N, Boretius S, Ott C, Von

Streitberg A, Welpinghus H, Sperling S, Frahm J, Simons M,

Ghezzi P, Ehrenreich H. Erythropoietin attenuates

neurological and histological consequences of toxic

demyelination in mice. Mol Med. 2012, 18:628-635.;

Hengartner, 2000: Hengartner MO. The biochemistry of apoptosis.

Nature 2000, 407:770-776.;

Hudri et al., 2012: Hudry E, Wu HY, Arbel-Ornath M, Hashimoto

T, Matsouaka R, Fan Z, Spires-Jones TL, Betensky RA, Bacskai

BJ, Hyman BT. Inhibition of the NFAT pathway alleviates amyloid β neurotoxicity in a mouse model of Alzheimer's disease. J Neurosci . 2012, 32:3176-3192.) .

Idris et al., 2008: Idriss NK, Blann AD, Lip GY . Hemoxygenase-1 in cardiovascular disease. J Am Coll Cardiol. 2008, 52:971-

978.

Janciauskiene et al., 1999: Janciauskiene S, Wright HT,

Lindgren S. Fibrillar Alzheimer's amyloid peptide Abeta(l-42) stimulates low density lipoprotein binding and cell

association, free radical production and cell cytotoxicity in PC12 cells. Neuropeptides. 1999, 33:510-516.

P-1418 Jazwa and Cuadrado, 2010: Jazwa A, Cuadrado A. Targeting heme oxygenase-1 for neuroprotection and neuroinflamination in neurodegenerative diseases. Curr Drug Targets. 2010, 11:1517- 1531.

Karttunen et al. 1991: Karttunen J and Shastri N. Measurement of ligand-induced activation in single viable T cells using the lacZ reporter gene. Proc Natl Acad Sci USA. 1991,

88:3972. ;

Koh et al., 1996:. Koh JY, Suh SW, Gwag BJ, He YY, Hsu CY, Choi D . The role of zinc in selective neuronal death after transient global cerebral ischemia. Science. 1996, 272:1013- 1016;

Korkmaz et al., 2013: Korkmaz S, Barnucz E, Loganathan S, Li S, Radovits T, Hegedus P, Zubarevich A, Hirschberg K, eymann A, Puskas LG, Ozsvari B, Farago N, Kanizsai I, Fabian G, Gyuris M, Merkely B, Karck M, Szabo C, Szabo G. Q50, an iron- chelating and zinc-complexing agent, improves cardiac

function in rat models of ischemia/reperfusion-induced myocardial injury. Circ J. 2013;77:1817-1826.;

Lewen et al., 2000: Lewen A, Matz P, Chan PH. Free radical pathways in CNS injury. J Neurotrauma. 2000, 17:871-890;

Li et al., 2007: Li C, Hossieny P, u BJ, Qawasmeh A, Beck K, Stocker R. Pharmacologic induction of heme oxygenase-1.

Antioxid Redox Signal. 2007, 9:2227-2239;

Liu et al., 2005: Liu F, Grundke-Iqbal I, Iqbal K, Oda Y,

Tomizawa K, Gong CX. Truncation and activation of calcineurin A by calpain I in Alzheimer disease brain. J Biol Chem. 2005, 280:37755-37762.;

Lue, et al., 1999: Lue LF1 , uo YM, Roher AE, Brachova L, Shen Y, Sue L, Beach T, Kurth JH, Rydel RE, Rogers J. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease, AmJ Pathol 1999, 155:853-862.; Lynch et al., 2002: Lynch AM, Lynch MA. The age-related

increase in IL-1 type I receptor in rat hippocampus is

P-1418 coupled with an increase in caspase-3 activation. Eur J

Neurosci 2002, 15:1779-1788.;

Machova et al., 2008: Machova, E., Jakubik, J., Michal, P.,

Oksman, M . , Iivonen, H. , Tanila, H., Dolezal, V. Impairment of muscarinic transmission in transgenic APPswe/PSldE9 mice.

Neurobiol. Aging 2008, 29 : 368e378. ;

Mammis et al., 2009: Mammis A, Mcintosh TK, Maniker AH.

Erythropoietin as a neuroprotective agent in traumatic brain injury. Surg Neurol. 2009, 71:527-531.;

Mattson, 2006: Mattson MP. Neuronal life-and-death signaling, apoptosis, and neurodegenerative disorders. Antioxid Redox

Signal. 2006, 8:1997-2006.;

McLean et al., 1999: C. McLean, R. Cherny, F. Fraser, S.

Fuller, M. Smith, K . Beyreuther, A. Bush and C. Masters, Soluble pool of Αβ amyloid as a determinant of severity of neurodegeneration in Alzheimer's Disease, Ann Neurol 1999,

46: 860-866;

Merelli et al., 2011 : Merelli A, Caltana L, Lazaro ski A, Brusco A. Experimental evidence of the potential use of erythropoietin by intranasal administration as a

neuroprotective agent in cerebral hypoxia. Drug Metabol Drug Interact. 2011, 26:65-69.;

Merelli et al . , 2013 : Merelli A, Czornyj L, Lazarowski A. Erythropoietin: a neuroprotective agent in cerebral hypoxia, neurodegeneration, and epilepsy. Curr Pharm Des. 2013,

19:6791-801.;

Mittl et al., 1997 : Mittl PRE, Marco SD, Krebs JF, Karanewsky DS, Priestle JP, Tomaselli KJ and Grutter MG Structure of Recombinant Human CPP32 in Complex with the Tetrapeptide Acetyl-Asp-Val-Ala-Asp Fluoromethyl Ketone. J Biol Chem. 1997, 272: 6539-6547.;

Newton et al. 2013: Newton SS, Fournier NM, Duman RS. Vascular growth factors in neuropsychiatry. Cell Mol Life Sci . 2013, 70: 1739-1752. ;

P-1418 Nguyen et al., 2005: Nguyen T, Hamby A, Massa SM . Clioquinol down-regulates mutant huntingtin expression in vitro and mitigates pathology in a Huntington's disease mouse model. Proc Natl Acad Sci U S A. 2005, 102:11840-11845.;

Noh et al., 2014: Noh MY, Cho KA, Kim H, Kim SM, Kim SH.

Erythropoietin modulates the immune-inflammatory response of a SOD1 (G93A) transgenic mouse model of amyotrophic lateral sclerosis (ALS) . Neurosci Lett. 2014, 574:53-58.;

Orrenius, 2007: Orrenius S. Reactive oxygen species in

mitochondria-mediated cell death. Drug Metab Rev. 2007, 39: 443-455.

Palmieri, 2000: Palmieri, G. A Practical o-Hydroxybenzylamines Promoted Enantioselective Addition of Dialkylzincs to

Aldehydes with Asymmetric Amplification Tetrahedron Asymmetry 2000, 11:3361;

Phillips et al., 1954: J. P. Phillips, R. W. Keown, Q.

Fernando; The reaction of anils with 8-quinolinol . , J. Org. Che . 1954, 19:907;

Phillips and Barrall, 1956: J. P. Phillips, E. M. Barrall.

Notes - Betti Reactions of Some Phenols. J. Org. Chem. 1956, 21:692; .

Phillips, 1956: J. P. Phillips, The Reactions Of 8-Quinolinol,

Chem. Rev. 1956, 56:286;

Regland et al., 2001: Regland B, Lehmann , Abedini I, Blennow K, Jonsson M, Karlsson I, Sjogren M, Wallin A, Xilinas M,

Gottfries CG. Treatment of Alzheimer's disease with

clioquinol. Dement Geriatr Cogn Disord. 2001, 12:408-414; Sama et al., 2008: Sama MA, Mathis DM, Furman JL, Abdul HM,

Artiushin IA, Kraner SD, Norris CM. Interleukin-lbeta- dependent signaling between astrocytes and neurons depends critically on astrocytic calcineurin/NFAT activity. J Biol

Chem. 2008, 283:21953-21964.;

Schafer et al., 2007: Schafer S, Pajonk FG, Multhaup G, Bayer

TA. Copper and clioquinol treatment in young APP transgenic

P-1418 and wild-type mice: effects on life expectancy, body weight, and metal-ion levels. J Mol Med (Berl) . 2007, 85:405-413.; Smirnova et al . , 2010: Smirnova NA, Rakhman I, oroz N, Basso

M, Payappilly J, Kazakov S, Hernandez-Guzman F, Gaisina IN, Kozikowski AP, Ratan RR, Gazaryan IG. Utilization of an in vivo reporter for high throughput identification of branched small molecule regulators of hypoxic adaptation. Chem Biol.

2010, 17:380-391.;

Stefanis, 2005: Stefanis L. Caspase-dependent and -independent neuronal death: two distinct pathways to neuronal injury.

Neuroscientist . 2005, 11:50-62.

Szabo, 2005: Szabo C. Mechanisms of cell necrosis. Crit Care

Med. 2005, 33:S530-534;

Uz et al., 1998: Uz, T., Pesold, C., Longone, P., and Manev, H. Agingassociated up-regulation of neuronal 5-lipoxygenase expression: putative role in neuronal vulnerability. FASEB J.

1998, 12:439-449.;

Verkade et al., 2008: Verkade JMM, van Hemert LJC, Quaedflieg

PJLM, Rutjes FPJT . Organocatalysed asymmetric Mannich

reactions; Chem. Soc. Rev., 2008, 37:29;

Wang et al., 1999: J. Wang, D.W. Dickson, J.Q. Trojanowski and

V.M. Lee, The levels of soluble versus insoluble brain Abeta distinguish Alzheimer's disease from normal and pathologic aging, Exp Neurol. 1999, 158:328-337.

Xue et al., 2007 : Xue YQ, Zhao LR, Guo WP, Duan WM.

Intrastriatal administration of erythropoietin protects dopaminergic neurons and improves neurobehavioral outcome in a rat model of Parkinson's disease. Neuroscience . 2007,

146:1245-1258.;

Zhang et al., 2006 : Zhang, CP., Zhu, L.L., Zhao, T., Zhao,

H., Huang, X., Ma, X., Wang, H . , and Fan, M. Characteristics of neural stem cells expanded in lowered oxygen and the potential role of hypoxia-inducible factor-lAlpha .

Neurosignals 2006, 15:259-265.;

Zhou et al., 2006: Zhou, Y . , Wei, E. Q. , Fang, S. H., Chu, L.

P-1418 S., Wang, M. L., Zhang, W. P., Yu, G. L., Ye, Y. L., Lin, S. C. , and Chen, Z. Spatio-temporal properties of 5-lipoxygenase expression and activation in the brain after focal cerebral ischemia in rats. Life Sci . 2006, 79:1645-1656;

Garrod KR, Cahalan MD (2008) Murine skin transplantation. J Vis Exp 11:634.

P-1418