Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HEXAHYDRODIAZEPINOQUINOLINES CARRYING A SUBSTITUTED ALKYL RADICAL
Document Type and Number:
WIPO Patent Application WO/2015/136091
Kind Code:
A1
Abstract:
The present invention relates to tricyclic hexahydrodiazepinoquino lines carrying a substituted alkyl radical, to a method for producing them, to a pharmaceutical composition containing such compounds, to their use as modulators, especially agonists or partial agonists, of the 5-HT2C receptor, their use for preparing a medicament for the prevention or treatment of conditions and disorders which respond to the modulation of 5- HT2C receptor, to a method for preventing or treating conditions and disorders which respond to the modulation of 5-HT2C receptor, and processes for preparing such compounds and compositions.

Inventors:
BACKFISCH GISELA (DE)
BAKKER MARGARETHA HENRICA MARIA (DE)
BLAICH GÜNTER (DE)
BRAJE WILFRIED (DE)
DRESCHER KARLA (DE)
ERHARD THOMAS (DE)
HAUPT ANDREAS (DE)
LAKICS VIKTOR (DE)
LINSENMEIER ANNA (DE)
MACK HELMUT (DE)
PETER RAIMUND (GB)
RELO ANA LUCIA (DE)
SMYTH LYNETTE (DE)
Application Number:
PCT/EP2015/055336
Publication Date:
September 17, 2015
Filing Date:
March 13, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ABBVIE DEUTSCHLAND (DE)
International Classes:
C07D471/04; A61K31/5513; A61P25/00
Domestic Patent References:
WO2003091250A12003-11-06
WO2014041131A12014-03-20
WO2014041131A12014-03-20
WO2003091250A12003-11-06
Foreign References:
US20070225277A12007-09-27
US20080146583A12008-06-19
US20070225274A12007-09-27
Other References:
DAVIS KL; KAHN RS; KO G; DAVIDSON M: "Dopamine in schizophrenia: a review and re-conceptualization", AM J PSYCHIATRY, vol. 148, 1991, pages 1474 - 86
WEINBERGER DR; BERMAN KF: "Prefrontal function in schizophrenia: confounds and controversies", PHILOS TRANS R SOC LOND B BIOL SCI, vol. 351, 1996, pages 1495 - 503
REMINGTON G; KAPUR S: "Atypical antipsychotics: are some more atypical than others?", PSYCHOPHARMACOL, vol. 148, 2000, pages 3 - 15, XP002483914, DOI: doi:10.1007/s002130050017
KAUFMAN MJ; HIRATA F: "Cyclic GMP inhibits phosphoinositide turnover in choroid plexus: evidence for interactions between second messengers concurrently triggered by 5-HT receptors", NEUROSCI LETT, vol. 206, 1996, pages 153 - 156
POMPEIANO M; PALACIOS JM; MENGOD G: "Distribution of the serotonin 5-HT2 receptor family mRNAs: comparison between 5-HT and 5-HT receptors", BRAIN RES MOL BRAIN RES, vol. 23, 1994, pages 163 - 178, XP024352210, DOI: doi:10.1016/0169-328X(94)90223-2
LOPEZ-GIMENEZ JF; MENGOD G; PALACIOS JM; VILARO MT: "Regional distribution and cellular localization of 5-HT receptor mRNA in monkey brain: comparison with [3H]mesulergine binding sites and choline acetyltransferase mRNA", SYNAPSE, vol. 42, 2001, pages 12 - 26
G. J. DIAZ ET AL., JOURNAL OF PHARMACOLOGICAL AND TOXICOLOGICAL METHODS, vol. 50, 2004, pages 187 - 199
"Advances in drug research", vol. 10, 1966, BIRKHAUSER VERLAG, pages: 224
TETRAHEDRON, vol. 57, 2001, pages 9199 - 9225
"Microwaves in Organic Synthesis", 2002, WILEY-VCH
T. GREENE; P. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
TECOTT LH; SUN LM; AKANA SF; STRACK AM; LOWENSTEIN DH; DALLMAN MF; JULIUS D: "Eating disorder and epilepsy in mice lacking 5-HT serotonin receptors", NATURE, vol. 374, 1995, pages 542 - 546, XP001193395, DOI: doi:10.1038/374542a0
CHOU-GREEN JM; HOLSCHER TD; DALLMAN MF; AKANA SF: "Compulsive behavior in the 5-HT receptor knockout mouse", PHYS. BEHAV., vol. 78, 2003, pages 641 - 649
CHOU-GREEN JM; HOLSCHER TD; DALLMAN MF; AKANA SF: "Repeated stress in young and old 5-HT c receptor knockout mouse", PHYS. BEHAV., vol. 79, 2003, pages 217 - 226
FRANK MG; STRYKER MP; TECOTT LH: "Sleep and sleep homeostasis in mice lacking the 5-HT receptor", NEUROPSYCHOPHARMACOLOGY, vol. 27, 2002, pages 869 - 873
ROCHA BA; GOULDING EH; O'DELL LE; MEAD AN; COUFAL NG; PARSONS LH; TECOTT LH: "Enhanced locomotor, reinforcing and neurochemical effects of cocaine in serotonin 5-hydroxytryptamine 2C receptor mutant mice", J. NEUROSCI., vol. 22, 2002, pages 1 0039 - 1 0045
WERRY, TD; LOIACONO R; SEXTON PA; CHRISTOPOULOS A: "RNA editing of the serotonin 5-HT receptor and its effects on cell signaling, pharmacology and brain function", PHARMAC. THERAP., vol. 119, 2008, pages 7 - 23, XP022820231, DOI: doi:10.1016/j.pharmthera.2008.03.012
SCHMAUSS C: "Serotonin 2C receptors: suicide, serotonin, and runaway RNA editing", NEUROSCIENTIST, vol. 9, 2003, pages 237 - 242, XP009033018, DOI: doi:10.1177/1073858403253669
IWAMOTO K; KATO T: "RNA editing of serotonin 2C receptor in human postmortem brains of major mental disorders", NEUROSCI. LETT., vol. 346, 2003, pages 169 - 172, XP001182238, DOI: doi:10.1016/S0304-3940(03)00608-6
IWAMOTOA K; NAKATANIB N; BUNDOA M; YOSHIKAWAB T; KATOA T: "Altered RNA editing of serotonin 2C receptor in a rat model of depression", NEUROSCI. RES, vol. 53, 2005, pages 69 - 76
DU Y; STASKO M; COSTA AC; DAVISSONE MT; GARDINER KJ: "Editing of the serotonin 2C receptor pre-mRNA Effects of the Morris Water Maze", GENE, vol. 391, 2007, pages 186 - 197, XP005917928, DOI: doi:10.1016/j.gene.2006.12.023
NISWENDER CM; HERRICK-DAVIS K; DILLEY GE; MELTZER HY; OVERHOLSER JC; STOCKMEIER CA; EMESON RB; SANDERS-BUSH E: "RNA Editing of the Human Serotonin 5-HT Receptor: Alterations in Suicide and Implications for Serotonergic Pharmacotherapy", NEUROPSYCHOPHARM, vol. 24, 2001, pages 478 - 491, XP002288022, DOI: doi:10.1016/S0893-133X(00)00223-2
SMITH BM ET AL.: "Discovery and structure-activity relationship of (IR)-8-chloro-2,3,4,5-tetrahydro-1-methyl-1H-3-benzazepine (Lorcaserin), a selective serotonin 5-HT receptor agonist for the treatment of obesity", JMED CHEM, vol. 51, 2008, pages 305 - 313, XP009138515, DOI: doi:10.1021/jm0709034
THOMSEN WJ; GROTTICK AJ; MENZAGHI F; REYES-SALDANA H; ESPITIA S; YUSKIN D; WHELAN K; MARTIN M; MORGAN M; CHEN W: "Lorcaserin, A Novel Selective Human 5-HT Agonist: In Vitro and In Vivo Pharmacological Characterization", J PHARMACOL EXP THER, vol. 325, 2008, pages 577 - 587, XP009138374, DOI: doi:10.1124/jpet.107.133348
ROSENZWEIG-LIPSON S; ZHANG J; MAZANDARANI H; HARRISON BL; SABB A; SABALSKI J; STACK G; WELMAKER G; BARRETT JE; DUNLOP J: "Antiobesity-like effects of the 5-HT receptor agonist WAY-161503", BRAIN RES., vol. 1073-107, 2006, pages 240 - 251, XP025064592, DOI: doi:10.1016/j.brainres.2005.12.052
DUNLOP J; SABB AL; MAZANDARANI H; ZHANG J; KAL-GAONKER S; SHUKHINA E; SUKOFFS, VOGEL RL; STACK G; SCHECHTER L; HARRISON BL: "WAY-163909 [97bR, lOaR)-1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta-[b][1,4]diazepino[6,7,lhi]indole], a novel 5-hydroxytryptamine 2C receptor -selective agonist with anorectic activity", JPHARMACØ/ EXP THER, vol. 313, 2005, pages 862 - 869
ROSENZWEIG-LIPSON S; SABB A; STACK G; MITCHELL P; LUCKI I; MALBERG JE; GRAUER S; BRENNAN J; CRYAN JF; SUKOFF RIZZO SJ: "Antidepressant-like effects of the novel, selective, 5-HT receptor agonist WAY-163909 in rodents", PSYCHOPHARMACOLOGY (BERLIN, vol. 192, 2007, pages 159 - 170, XP002450646
ROSENZWEIG-LIPSON S; DUNLOP J; MARQUIS KL: "5-HT receptor agonists as an innovative approach for psychiatric disorders", DRUG NEWS PERSPECT, vol. 20, 2007, pages 565 - 571
CRYAN, JF; LUCKI I: "Antidepressant-like behavioral effects mediated by 5-Hydroxytryptamine 2C receptors", J. PHARM. EXP. THER., vol. 295, 2000, pages 1120 - 1126, XP002450645
DI MATTEO, V.; DI GIOVANNI, G.; DI MASCIO, M.; ESPOSITO, E.: "SB 242084, a selective serotonin 2C receptor antagonist, increases dopaminergic transmission in the mesolimbic system", NEUROPHARMACOLOGY, vol. 38, 1999, pages 1195 - 1205
DI GIOVANNI, G.; DI MATTEO, V.; DI MASCIO, M.; ESPOSITO, E.: "Preferential modulation of mesolimbic vs. nigrostriatal dopaminergic function by serotonin2C/2B receptor agonists: a combined in vivo electrophysiological and microdialysis study", SYNAPSE, vol. 35, 2000, pages 53 - 61
MARQUIS KL; SABB AL; LOGUE SF; BRENNAN JA; PIESLA MJ; COMERY TA; GRAUER SM; ASHBY CR, JR.; NGUYEN HQ; DAWSON LA: "WAY-163909 [(7bR,lOaR)-1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta-[b][1,4]diazepino[ 6,7,1hi]indole]: A novel 5-hydroxytryptamine 2C receptor-selective agonist with preclinical antipsychotic-like activity", J PHARMACOL EXP THER, vol. 320, 2007, pages 486 - 496
MARQUIS KL; SABB AL; LOGUE SF; BRENNAN JA; PIESLA MJ; COMERY TA; GRAUER SM; ASHBY CR, JR.; NGUYEN HQ; DAWSON LA: "WAY-163909 [(7bR,10aR)-1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta-[b][1,4]diazepino[ 6,7,1hi]indole]: A novel 5-hydroxytryptamine 2C receptor-selective agonist with pre-clinical antipsychotic-like activity", J PHARMACOL EXP THER, vol. 320, 2007, pages 486 - 496
SIUCIAK JA; CHAPIN DS; MCCARTHY SA; GUANOWSKY V; BROWN J; CHIANG P; MARALA R; PATTERSON T; SEYMOUR PA; SWICK A: "CP-809,101, a selective 5-HT agonist, shows activity in animal models of antipsychotic activity", NEUROPHARMACOLOGY, vol. 52, 2007, pages 279 - 290, XP005856696, DOI: doi:10.1016/j.neuropharm.2006.07.024
DUNLOP J; MARQUIS KL; LIM HK; LEUNG L; KAO J; CHEESMAN C; ROSENZWEIG-LIPSON S: "Pharmacological profile of the 5-HT receptor agonist WAY-163909; therapeutic potential in multiple indications", CNS DUG REV, vol. 12, 2006, pages 167 - 177, XP009089393
ISAAC M: "Serotonergic 5-HT receptors as a potential therapeutic target for the antiepileptic drugs", CURR. TOPICS MED. CHEM., vol. 5, 2005, pages 59 - 67
THORSLUND K; NORDLIND K: "Serotonergic drugs-a possible role in the treatment of psoriasis?", DRUG NEWS PERSPECT, vol. 20, 2007, pages 521 - 525
ESPOSITO E; DI MATTEO V; PIERUCCI M; BENIGNO A; DI GIAVANNI, G: "Role of central 5-HT receptor in the control of basal ganglia functions", THE BASAL GANGLIA PATHOPHYSIOLOGY: RECENT ADVANCES, 2007, pages 97 - 127
BARR AM; LAHMANN-MASTEN V; PAULUS M; GAINETDINOV RP; CARON MG; GEYER MA: "The selective serotonin-2A receptor antagonist M 1 00907 reverses behavioral deficits in dopamine transporter knockout mice", NEUROPSYCHOPHARMACOLOGY, vol. 29, 2004, pages 221 - 228, XP003000531, DOI: doi:10.1038/sj.npp.1300343
DEKEYNE A; MANNOURY LA COUR C; GOBERT A; BROCCO M; LEJUENE F; SERRES F; SHARP T; DASZUTA A; SOUMIER A; PAPP M: "S32006, a novel 5-HT receptor antagonists displaying broad-based antidepressant and anxiolytic properties in rodent models", PSYCHOPHARMACOLOGY, vol. 199, pages 549 - 568, XP019621119
NUNES-DE-SOUZA V; NUNES-DE-SOUZA RL; RODGERS RJ; CANTO-DE-SOUZA A: "5-HT2 receptor activation in the midbrain periaqueductal grey (PAG) reduces anxiety-like behavior in mice", BEHAV. BRAIN RES., vol. 187, 2008, pages 72 - 79, XP022382026, DOI: doi:10.1016/j.bbr.2007.08.030
LEONE M; RIGAMONTI A; D'AMICO D; GRAZZI L; USAI S; BUSSONE G: "The serotonergic system in migraine", JOURNAL OFHEADACHE AND PAIN, vol. 2, no. 1, 2001, pages S43 - S46, XP021242176, DOI: doi:10.1007/s101940170008
ARJONA AA; POOLER AM; LEE RK; WURTMAN RJ: "Effect of a 5-HT serotonin agonist, dexnorfenfluramine, on am-yloid precursor protein metabolism in guinea pigs", BRAIN RES., vol. 951, 2002, pages 135 - 140, XP001149841, DOI: doi:10.1016/S0006-8993(02)03153-0
NAKAE A; NAKAI K; TANAKA T; HAGIHIRA S; SHIBATA M; UEDA K; MASIMO T: "The role of RNA editing of the serotonin 2C receptor in a rat model of oro-facial neuropathic pain", THE EUROPEAN JOURNAL FNEUROSCIENCE, vol. 27, 2008, pages 2373 - 2379
NAKAE A; NAKAI K; TANAKA T; TAKASHINA M; HAGIHIRA S; SHIBATA M; UEDA K; MASHIMO T: "Serotonin 2C receptor mRNA editing in neuropathic pain model", NEUROSCI. RES, vol. 60, 2008, pages 228 - 231, XP022439568, DOI: doi:10.1016/j.neures.2007.10.004
KAO T; SHUMSKY JS; JACOB-VADAKOT S; TIMOTHY HB; MURRAY M; MOXON, KA: "Role of the 5-HT receptor in improving weight-supported stepping in adult rats spinalized as neonates", BRAIN RES.L, vol. 112, 2006, pages 159 - 168, XP025101348, DOI: doi:10.1016/j.brainres.2006.07.020
MOTOFEI IG: "A dual physiological character for sexual function: the role of serotonergic receptors", B U INTERNATIONAL, vol. 101, 2008, pages 531 - 5 4
SHIMADA I; MAENO K; KONDOH Y; KAKU H; SUGASAWA K; KIMURA Y; HATANAKA K; NAITOU Y; WANIBUCHI F; SAKAMOTO S: "Synthesis and structure-activity relationships of a series of benzazepine derivatives as 5-HT receptor agonists", BIOORG. MED. CHEM., vol. 16, 2008, pages 3309 - 3320, XP022558565, DOI: doi:10.1016/j.bmc.2007.12.009
FLETCHER PJ; LE AD; HIGGINS GA: "Serotonin receptors as potential targets for modulation of nicotine use and dependence", PROGRESS BRAIN RES., vol. 172, 2008, pages 361 - 83, XP009183073, DOI: doi:10.1016/S0079-6123(08)00918-7
BUBAR MJ; CUNNINGHAM KA: "Prospects for serotonin 5-HT2R pharmacotherapy in psychostimulant abuse", PROGRESS BRAIN RES., vol. 172, 2008, pages 319 - 46, XP009183072, DOI: doi:10.1016/S0079-6123(08)00916-3
SHARIF NA; MCLAUGHLIN MA; KELLY CR: "AL-34662: a potent, selective, and efficacious ocular hypotensive serotonin-2 receptor agonist", J OCUL PHARMACOL THER, vol. 23, 2006, pages 1 - 13, XP008109787, DOI: doi:10.1089/jop.2006.0093
OBATA, HIDEAKI; ITO, NAOMI; SASAKI, MASAYUKI; SAITO, SHIGERU; GOTO, FUMIO: "Possible involvement of spinal noradrenergic mechanisms in the antiallodynic effect of intrathecally administered 5 - HT2C receptor agonists in the rats with peripheral nerve injury", EUROPEAN JOURNAL OFPHARMACOLOGY, vol. 567, no. 1-2, 2007, pages 89 - 94, XP022095316, DOI: doi:10.1016/j.ejphar.2007.03.029
NAKAE, AYA; NAKAI, KUNIHIRO; TANAKA, TATSUYA; TAKASHINA, MASAKI; HAGIHIRA, SATOSHI; SHIBATA, MASAHIKO; UEDA, KOICHI; MASHIMO, TAKA: "Department of Anesthesiology & Intensive Care Medicine, Graduate School of Medicine, Osaka University", NEUROSCIENCE RESEARCH (AMSTERDAM, NETHERLANDS, vol. 60, no. 2, 2008, pages 228 - 231
OBATA, HIDEAKI; SAITO, SHIGERU; SAKURAZAWA, SHINOBU; SASAKI, MASAYUKI; USUI, TADASHI; GOTO, FUMIO: "Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.", PAIN, vol. 108, no. 1-2, 2004, pages 163 - 169
BRUS, RYSZARD; KASPERSKA, ALICJA; OSWIECIMSKA, JOANNA; SZKILNIK, RYSZARD: "Department of Pharmacology, Silesian Medical University, Zabrze, Pol.", MEDICAL SCIENCE MONITOR, vol. 3, no. 5, 1997, pages 654 - 656
BRENNAN, PAUL E.; WHITLOCK, GAVIN A.; HO, DANNY K. H.; CONLON, KELLY; MCMURRAY, GORDON, BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 19, no. 17, 2009, pages 4999 - 5003
MBAKI, Y.; RAMAGE, A. G.: "Department of Pharmacology, University College London, London, UK", BRITISH JOURNAL OFPHARMACOLOGY, vol. 155, no. 3, 2008, pages 343 - 356
SHEN JHQ ET AL.: "A 6-week randomized, double-blind, placebo-controlled, comparator referenced trial of vabicaserin in acute schizophrenia", JOURNAL OF PSYCHIATRIC RESEARCH, vol. 53, 2014, pages 14 - 22, XP028839400, DOI: doi:10.1016/j.jpsychires.2014.02.012
LIU J ET AL.: "Prediction of Efficacy of Vabicaserin, a 5-HT Agonist, for the Treatment of Schizophrenia Using a Quantitative Systems Pharmacology Model", CPT PHARMACOMETRICS SYST. PHARMACOL., vol. 3, 2014, pages 11
DUNLOP J ET AL.: "Characterization of Vabicaserin (SCA-136), a Selective 5-Hydroxytryptamine 2C Receptor Agonist", J PHARMACOL EXP THER, vol. 337, 2011, pages 673 - 80
SIUCIAK J ET AL.: "CP-809,101, a selective 5-HT agonist, shows activity in animal models of antipsychotic activity", NEUROPHARMACOLOGY, vol. 52, 2007, pages 279 - 290, XP005856696, DOI: doi:10.1016/j.neuropharm.2006.07.024
MOSIENKO V ET AL.: "Exaggerated aggression and decreased anxiety in mice deficient in brain serotonin", TRANSL PSYCHIATRY, vol. 2, 2012, pages E122
DEL GUIDICE T ET AL.: "Stimulation of 5-HT c Receptors Improves Cognitive Deficits Induced by Human Tryptophan Hydroxylase2 Loss of Function Mutation", NEUROPSYCHOPHARMACOLOGY, vol. 39, 2014, pages 1125 - 1134
ROSENZWEIG-LIPSON ET AL.: "Antidepressant-like effects of the novel, selective, 5-HT receptor agonist WAY-163909 in rodents", PSYCHOPHARMACOLOGY, vol. 192, 2007, pages 159 - 170, XP002450646
DEL GUIDICE T ET AL.: "Stimulation of 5-HT Receptors Improves Cognitive Deficits Induced by Human Tryptophan Hydroxylase2 Loss of Function Mutation", NEUROPSYCHOPHARMACOLOGY, vol. 39, 2014, pages 1125 - 1134
"Methods in Cell Biology", vol. XIV, 1976, ACADEMIC PRESS, pages: 33
S. M. BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1
Attorney, Agent or Firm:
REITSTÖTTER - KINZEBACH (Ludwigshafen, DE)
Download PDF:
Claims:
We claim:

A compound of the formula I

wherein

R is selected from the group consisting of hydrogen, cyano, Ci-C6-alkyl, fluor- inated Ci-C6-alkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, -C(=0)R9, phenyl, phenyl-Ci-C2- alkyl and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated heterocyclic ring containing 1, 2, 3 or 4 heteroa- toms or heteroatom groups independently selected from N, O, S, NO, SO and SO2 and optionally also 1 or 2 C=0 and/or C=S groups as ring members, where the cyclic moieties in the three last-mentioned radicals may be substituted with one or more substituents R10; each R is independently selected from the group consisting of cyano, nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6-alkynyl, C3-C8- cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6-alkoxy-Ci-C4-alkyl, -CH2NRl laRl lb, -C(=0)R9,

phenyl, phenyl-Ci-C2-alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, SO2, C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substit- uents R10; or

two radicals R2 bound to the same carbon atom, together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, par- tially unsaturated or maximally unsaturated ring, where the ring may contain

1, 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S02, C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R10;

R3a and R3b, independently of each other, are selected from the group consisting of hydrogen, cyano, nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6- hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6-alkoxy-Ci-C4-alkyl,

-CH2NRl laRl lb, -C(=0)R9, phenyl, phenyl-Ci-C2-alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, SO2, C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R10;

R4a and R4b, independently of each other, are selected from the group consisting of hydrogen, cyano, nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6- hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl,

Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6-alkoxy-Ci-C4-alkyl,

-CH2NRl laRl lb, -C(=0)R9, phenyl, phenyl-Ci-C2-alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, SO2, C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R10; or R4a and R4b form together a group =0 or =S; or

R4a and R4b, together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring, where the ring may contain 1, 2, 3 or 4 heteroatoms or het- eroatom-containing groups selected from O, S, N, SO, S02, C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R10; and R , independently of each other, are selected from the group consisting of hydrogen, halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6- alkyl, Ci-C6-hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6- alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated C3-C8- cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-alkoxy-Ci-C4- alkyl, fluorinated Ci-C6-alkoxy-Ci-C4-alkyl, Ci-C6-hydroxyalkoxy, Ci-C6- alkoxy-Ci-C4-alkoxy, Ci-C6-alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6- alkylsulfinyl, fluorinated Ci-C6-alkylsulfinyl, Ci-C6-alkylsulfonyl, fluorinated Ci-Ce-alkylsulfonyl, -NRl laRl lb, -CH2NRl laRl lb, -NRl laC(0)R9, -C(=0)R9, S02NRl laRl lb, Ci-Ce-alkylcarbonyloxy, fluorinated Ci -C6- alkylcarbonyloxy, phenyl, phenyl-Ci-C2-alkyl, phenoxy, phenylsulfonyl, benzyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S02, C=0 and C=S as ring members, where the cyclic moieties in the six last- mentioned radicals may be substituted with one or more substituents R10; where R5a and R5b are not simultaneously hydroxyl; or

R5a and R5b, together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring, where the ring may contain 1, 2, 3 or 4 heteroatoms or het- eroatom-containing groups selected from O, S, N, SO, S02, C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R10; R6 is Ci-C6-alkyl which carries one or more substituents R8;

R7 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6- alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, C3-C8- cycloalkenyl, fluorinated Cs-Cs-cycloalkenyl, Ci-C6-alkoxy, fluorinated Ci- C6-alkoxy, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6-alkoxy-Ci-C4- alkyl, Ci-C6-hydroxyalkoxy, Ci-C6-alkoxy-Ci-C4-alkoxy, Ci-C6-alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6-alkylsulfinyl, fluorinated Ci-C6- alkylsulfmyl, Ci-C6-alkylsulfonyl, fluorinated Ci-C6-alkylsulfonyl,

-NRl laRl lb, -CH2NRl laRl lb, -NRl laC(0)R9, -C(=0)R9, S02NRl laRl lb, Ci- C6-alkylcarbonyloxy, fluorinated Ci-C6-alkylcarbonyloxy, phenyl, phenyl- Ci-C2-alkyl, phenoxy, phenylsulfonyl, benzyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S02, C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R10; or

two radicals R7 bound on neighboring carbon atoms, together with the carbon atoms they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered partially unsaturated or maximally unsaturated ring, where the ring may contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S02, C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R10: each R is independently selected from the group consisting of hydroxyl, halogen, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-hydroxyalkoxy, Ci-C6- alkoxy-Ci-C4-alkoxy, Ci-C6-alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6- alkylsulfmyl, fluorinated Ci-C6-alkylsulfinyl, Ci-C6-alkylsulfonyl, fluorinated Ci-Cg-alkylsulfonyl and -NRl laRl lb; each R9 is independently selected from the group consisting of hydrogen, cy- ano, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6- alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-Ce-alkoxy, -NR1 laR! lb, -CH2NRl laR1 lb, phenyl, phenyl-Ci-

C2-alkyl, phenoxy, benzyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S02, C=0 and C=S as ring members, where the cyclic moieties in the five last-mentioned radicals may be substituted with one or more sub- stituents R10; each R10 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6- alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6- alkoxy-Ci-C4-alkyl, Ci-C6-hydroxyalkoxy, Ci-C6-alkoxy-Ci-C4-alkoxy, Ci- C6-alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6-alkylsulfinyl, fluorinated Ci-C6-alkylsulfmyl, Ci-C6-alkylsulfonyl, fluorinated Ci-C6-alkylsulfonyl, -

COOH, -NRl laRl lb, -CH2NRl laRl lb, Ci-C6-alkylcarbonyl, fluorinated Ci- C6-alkylcarbonyl, Ci-C6-alkoxycarbonyl, fluorinated Ci-C6-alkoxycarbonyl, S02NRl laRl lb, Ci-Ce-alkylcarbonyloxy and fluorinated Ci-C6- alky lcarbony lo xy ;

or two radicals R10, together with the atom(s) they are bound to, form a saturated, partially unsaturated or maximally unsaturated 3-, 4-, 5-, 6- or 7- membered carbocyclic or heterocyclic ring, where the heterocyclic ring contains 1 , 2 or 3 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S02, C=0 and C=S as ring members;

Rl la and Rl lb, independently of each other and independently of each occurrence, are selected from the group consisting of hydrogen, cyano, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6- alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated C3-C8- cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-alkylcarbonyl, fluorinated Ci-C6-alkylcarbonyl, Ci-C6-alkoxycarbonyl, fluorinated Ci-C6- alkoxycarbonyl, phenyl and benzyl, where the phenyl moieties in the two last-mentioned radicals may carry 1 , 2 or 3 substituents selected from halogen, cyano nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-alkoxy and fluorinated Ci-C6-alkoxy; or,

if Rl la and Rl lb are bound to the same nitrogen atom, together with this nitrogen atom may form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated heterocyclic ring, where the ring may further contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S02, C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents selected from halogen, cyano nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-alkoxy and fluorinated Ci-C6-alkoxy; a is 0, 1 or 2; and b is 0, 1 , 2 or 3; or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

The compound as claimed in claim 1 , where R1 is selected from hydrogen and Ci- Ce-alkyl.

The compound as claimed in claim 2, where R1 is hydrogen.

The compound as claimed in any of the preceding claims, where R2 is selected from Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-alkoxy-Ci-C4-alkyl and fluori- nated Ci-C6-alkoxy-Ci-C4-alkyl.

5. The compound as claimed in claim 4, where R2 is selected from Ci-C6-alkyl and fluorinated Ci-C6-alkyl.

The compound as claimed in claim 5, where R2 is selected from methyl and CF3.

The compound as claimed in any of the preceding claims, where R3a and R3b, independently of each other, are selected from hydrogen, cyano, nitro, Ci-C6-alkyl and fluorinated Ci-C6-alkyl.

8. The compound as claimed in claim 7, where R3a is selected from hydrogen, cyano, nitro, Ci-C6-alkyl and fluorinated Ci-C6-alkyl; and R3b is hydrogen. 9. The compound as claimed in claim 8, where R3a is selected from hydrogen and methyl; and R3b is hydrogen.

10. The compound as claimed in claim 9, where R3a and R3b are hydrogen.

The compound as claimed in any of the preceding claims, where R a and R , independently of each other, are selected from hydrogen, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C4-alkoxy-Ci-C4-alkyl and fluorinated Ci-C4-alkoxy-Ci-C4-alkyl, or form together a group =0, or form together a group -(CH2)m-, where m is 2, 3 or 4.

The compound as claimed in claim 1 1 , where R4a and R4b are hydrogen.

The compound as claimed in any of the preceding claims, where R5a is selected from hydrogen, halogen, Ci-C6-alkyl, fluorinated Ci-C6-alkyl and Ci-C6-alkoxy-

5b

Ci-C4-alkyl and R is hydrogen

14. The compound as claimed in claim 13, where R5a is hydrogen or methyl and R5b is hydrogen.

15. The compound as claimed in any of the preceding claims, where each R is inde- pendently selected from the group consisting of halogen, Ci-C6-alkoxy and fluori- nated Ci-C6-alkoxy.

16. The compound as claimed in claim 15, where R8 is halogen. 17. The compound as claimed in claim 16, where R8 is fluorine.

18. The compound as claimed in claim 17, where R6 is fluorinated Ci-C4-alkyl.

19. The compound as claimed in claim 18, where R6 is fluorinated Ci-C2-alkyl.

20. The compound as claimed in claim 19, where R6 is fluorinated methyl.

21. The compound as claimed in claim 20, where R6 is CHF2 or CF3. 22. The compound as claimed in claim 15, where R6 is selected from Ci-C6-alkoxy- Ci-C4-alkyl and fluorinated Ci-C6-alkoxy-Ci-C4-alkyl.

23. The compound as claimed in claim 22, where R6 is fluorinated Ci-C6-alkoxy-Ci- C4-alkyl.

24. The compound as claimed in claim 22, where R6 is Ci-C4-alkoxy-methyl.

25. The compound as claimed in any of claims 1 to 14, where R6 is Ci-C4-alkyl which carries a hydroxyl substituent.

26. The compound as claimed in any of the preceding claims, where each R7 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, C2-C6-alkenyl, fluorinated C2-C6-alkenyl, C2-C6-alkynyl, fluorinated C2-C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated C3-C8- cycloalkyl, Ci-C6-alkoxy and fluorinated Ci-C6-alkoxy.

27. The compound as claimed in claim 26, where each R7 is independently selected from halogen, Ci-C4-alkyl, fluorinated Ci-C4-alkyl, Ci-C4-alkoxy and fluorinated Ci-C4-alkoxy.

28. The compound as claimed in claim 27, where R7 is fluorine.

29. The compound as claimed in any of the preceding claims, where a is 0 or 1.

30. The compound as claimed in claim 29, where in case that a is 1 , R2 is bound position to the nitrogen ring atom carrying R1.

31. The compound as claimed in any of the preceding claims, where b is 0 or 1.

32. The compound as claimed in any of the preceding claims, of formula 1.1

wherein R2, R6, R7 and b are as defined in any of claims 1 , 4 to 6, 15 to 28 and 31 and a is 0 or 1.

33. The compound as claimed in claim 32, of formula 1.1.1

(1.1 .1 ) wherein

R7a is H, CI, F or methyl;

a is 0 or 1 ; and

R2, R6, R7 and b are as defined in any of claims 1, 4 to 6, 15 to 28 and 31.

34. A compound selected from the group consisting of

8-(Trifluoromethyl)-2, 3,4,6,7, 8-hexahydro- lH-[ 1 ,4]diazepino[6,7, 1 -ij]quinoline;

(S)-8-(Trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

(R)-8-(Trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

8-(2,2,2-Trifluoroethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

(S)- 8-(2,2,2-Trifluoroethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

(R)-8-(2,2,2-Trifluoroethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

8-(Difluoromethyl)-2, 3,4,6,7, 8-hexahydro-lH-[l,4]diazepino[6, 7, l-ij]quino line;

(S)-8-(Difiuoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

(R)-8-(Difiuoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

4-Methyl-8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline;

8-(Difluoromethyl)-4-methyl-2, 3,4,6,7, 8-hexahydro-lH-[l, 4]diazepino[6,7, 1- ij]quinoline;

(4-Methyl-2,3,4,6,7,8-hexahydro-lH 1,4]diazepino[6,7,l-ij]quinolin-8- yl)methanol;

8-(Fluoromethyl)-4-methyl-2, 3,4,6,7, 8-hexahydro-lH-[l, 4]diazepino[6,7, 1- ij]quinoline;

8-(Ethoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline; 8-(Methoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinoline; and the stereoisomers and pharmaceutically acceptable salts thereof.

A method for producing a compound of formula I according to claim 1 wherein R1, R4a, R4b and R5b are hydrogen, and R2, if present, is bound in β-position to the nitrogen ring atom carrying R1 (compound I"), comprising following steps:

- reacting the boronic acid 11 and the α,β-unsaturated ester 12 in the presence of a Rhodium catalyst to the dihydroquinolone 13;

- reducing the dihydroquinolone 13 to the 1,2,3,4-tetrahydroquinoline 14;

- reacting the 1,2,3,4-tetrahydroquinoline 14 with the 2-halogenoacetamide 6, wherein Y is CI, Br or I, to the 1-acetamide-substituted 1,2,3,4- tetrahydroquinoline compound 15;

- reducing the carbonyl group of the 1-acetamide-substituted 1,2,3,4- tetrahydroquinoline compound 15 to the ethylamine-substituted tetrahydro- quinoline compound 16; and

- reacting 16 with the aldehyde or ketone 9, wherein R3a and R3b are as defined in claim 1, but for nitro or cyano, in the presence of a strong acid to I":

A pharmaceutical composition comprising a therapeutically effective amount of at least one compound as claimed in any of claims 1 to 34 or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof, in combination with at least one pharmaceutically acceptable carrier and/or auxiliary substance.

The compound as claimed in any of claims 1 to 34 or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for use as a medicament.

The compound as claimed in any of claims 1 to 34 or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the treatment of disorders which respond to the modulation of the 5-HT2c receptor.

The use of a compound as claimed in any of claims 1 to 34 or of an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of disorders which respond to the modulation of the 5-HT2c receptor.

40. A method for treating disorders which respond to the modulation of the 5-HT2c receptor, which method comprises administering to a subject in need thereof at least one compound as defined in any of claims 1 to 34 or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

41. The compound as claimed in claim 38 or the use as claimed in claim 39 or the method as claimed in claim 40, where the disorders are selected from the group consisting of damage of the central nervous system, disorders of the central nervous system, eating disorders, ocular hypertension, cardiovascular disorders, gas- trointestinal disorders and diabetes.

The compound or the use or the method as claimed in claim 41, where the disorders are selected from the group consisting of bipolar disorder, depression, atypical depression, mood episodes, adjustment disorders, anxiety, panic disorders, post-traumatic syndrome, psychoses, schizophrenia, cognitive deficits of schizophrenia, memory loss, dementia of aging, Alzheimer's disease, neuropsychiatric symptoms in Alzheimer's disease, behavioral disorders associated with dementia, social phobia, mental disorders in childhood, attention deficit hyperactivity disorder, organic mental disorders, autism, mutism, disruptive behavior disorder, impulse control disorder, borderline personality disorder, obsessive compulsive disorder, migraine and other conditions associated with cephalic pain or other pain, raised intracranial pressure, seizure disorders, epilepsy, substance use disorders, alcohol abuse, cocaine abuse, tobacco abuse, smoking cessation, sexual dysfunction/erectile dysfunction in males, sexual dysfunction in females, premenstrual syndrome, late luteal phase syndrome, chronic fatigue syndrome, sleep disorders, sleep apnoea, chronic fatigue syndrome, psoriasis, Parkinson's disease, psychosis in Parkinson's disease, neuropsychiatric symptoms in Parkinson's disease, Lewy Body dementia, neuropsychiatric symptoms in Lewy Body dementia, spinal cord injury, trauma, stroke, pain, bladder dysfunction/urinary incontinence, encephalitis, meningitis, eating disorders, obesity, bulimia, weight loss, anorexia nervosa, ocular hypertension, cardiovascular disorders, gastrointestinal disorders, diabetes insipidus, diabetes mellitus, type I diabetes, type II diabetes, type III diabetes, di- abetes secondary to pancreatic diseases, diabetes related to steroid use, diabetes complications, hyperglycemia and insulin resistance.

The compound or the use or the method as claimed in claim 42, where the disorders are selected from schizophrenia, depression, bipolar disorders, obesity, substance use disorders, neuropsychiatric symptoms in Alzheimer's disease and neu- ropsychiatric symptoms in Parkinson's disease.

Description:
HEXAHYDRODI AZEPINOQUINOLINE S CARRYING A SUBSTITUTED

ALKYL RADICAL

FIELD OF THE INVENTION

The present invention relates to tricyclic hexahydrodiazepinoquino lines carrying a substituted alkyl radical, to a method for producing them, to a pharmaceutical composition containing such compounds, to their use as modulators, especially agonists or partial agonists, of the 5-HT 2 c receptor, their use for preparing a medicament for the prevention or treatment of conditions and disorders which respond to the modulation of 5-HT 2 c receptor, to a method for preventing or treating conditions and disorders which respond to the modulation of 5-HT 2C receptor, and processes for preparing such compounds and compositions.

BACKGROUND OF THE INVENTION

Diseases, disorders and conditions where 5-HT 2C modulation is desired are for example depression, anxiety, schizophrenia, bipolar disorder, obsessive compulsive disorder, migraine, pain, epilepsy, substance abuse, eating disorders, obesity, diabetes, erectile dysfunction and others.

Serotonin (5-hydroxytryptamine, 5-HT), a monoamine neurotransmitter and local hormone, is formed by the hydroxylation and decarboxylation of tryptophan. The greatest concentration is found in the enterochromaffin cells of the gastrointestinal tract, the remainder being predominantly present in platelets and in the Central Nervous System (CNS). 5-HT is implicated in a vast array of physiological and pathophysiological pathways. In the periphery, it contracts a number of smooth muscles and induces endo- thelium-dependent vasodilation. In the CNS, it is believed to be involved in a wide range of functions, including the control of appetite, mood, anxiety, hallucinations, sleep, vomiting and pain perception.

Neurons that secrete 5-HT are termed serotonergic. The function of 5-HT is exerted upon its interaction with specific (serotonergic) neurons. Seven types of 5-HT receptors have been identified: 5-HTi (with subtypes 5-HTi A , 5-HTi B , 5-HTi D , 5-HTi E and 5-HTIF), 5-HT 2 (with subtypes 5-HT 2A , 5-HT 2B and 5-HT 2C ), 5-HT 3 , 5-HT 4 , 5-HT 5 (with subtypes 5-HT 5A and 5-HT 5B ), 5-HT 6 and 5-HT 7 . Most of these receptors are coupled to G-proteins that affect the activities of adenylate cyclase or phospholipase Cy.

Alterations in the activity of multiple neurotransmitter receptor systems (dopamine, serotonin, glutamate, GAB A, acetylcholine) have been implicated in the manifes- tation of the symptoms of schizophrenia. The most widely accepted "Dopamine Hypothesis of Schizophrenia" in its simplest form states that the positive symptoms of this pathology relate to a functional hyperactivity of the meso limbic dopaminergic system, while the negative and cognitive aspects can be traced to a functional hypoactivity of the mesocortical dopaminergic projections. Atypical antipsychotics block the meso- limbic dopaminergic neurotransmission, thereby controlling positive symptoms, with little or no effect on the nigrostriatal system, leading to less induction of extrapyramidal side effects (EPS).

Primary negative and cognitive symptoms of schizophrenia reflect a dysfunction of the frontal cortex ("hypofrontality"), which is thought to be induced by a decreased tone in the mesocortical dopaminergic projection field [Davis KL, Kahn RS, Ko G and Davidson M (1991). Dopamine in schizophrenia: a review and re-conceptualization. Am J Psychiatry 148: 1474 - 86. Weinberger DR and Berman KF (1996). Prefrontal function in schizophrenia: confounds and controversies. Philos Trans R Soc Lond B Biol Sci 351 : 1495 - 503]. Agents that selectively enhance dopamine levels in the cortex have the potential to address the negative symptoms of this disorder. Atypical antipsychotics lack robust efficacy against negative and cognitive components of the schizophrenic syndrome.

The schizophrenic symptomatology is further complicated by the occurrence of drug-induced so-called secondary negative symptoms and cognitive impairment, which are difficult to distinguish from primary negative and cognitive symptoms [Remington G and Kapur S (2000). Atypical antipsychotics: are some more atypical than others? Psychopharmacol 148: 3 - 15]. The occurrence of secondary negative symptoms not only limits therapeutic efficacy but also, together with these side effects, negatively affects patient compliance.

It may thus be hypothesized that a novel mechanistic approach that blocks dopaminergic neurotransmission in the limbic system but does not affect the striatal and pituitary projection fields, and stimulates frontocortical projection fields, would provide an efficacious treatment for all parts of the schizophrenic pathology, including its positive, negative and cognitive symptoms. Moreover, a selective compound that is substantially free of the ancillary pharmacology that characterizes current agents would be expected to avoid a variety of off-target side effects that plague current treatments such as ex- trapyramidal side effects (EPS) and weight gain.

The 5-HT 2 c receptor, previously named 5-HTlC, is a G-protein-coupled receptor, which couples to multiple cellular effector systems including the phospholipase C, A and D pathways. It is found primarily in the brain and its distribution is particularly high in the plexus choroideus, where it is assumed to control cerebrospinal fluid produc- tion [Kaufman MJ, Hirata F (1996) Cyclic GMP inhibits phosphoinositide turnover in choroid plexus: evidence for interactions between second messengers concurrently triggered by 5-HT 2C receptors. Neurosci Lett 206: 153-156]. Very high levels were also found in the retrosplenial, piriform and entorhinal cortex, anterior olfactory nucleus, lateral septal nucleus, subthalamic nucleus, amygdala, subiculum and ventral part of CA3, lateral habenula, substantia nigra pars compacta, several brainstem nuclei and the whole grey matter of the spinal cord [Pompeiano M, Palacios JM, Mengod G (1994). Distribution of the serotonin 5-HT2 receptor family mRNAs: comparison between 5- HT 2A and 5-HT 2C receptors. Brain Res Mol Brain Res 23: 163-178]. A comparison of the distribution of 5-HT 2 c mRNA with that of 5-HT 2 c protein in monkey and human brains has revealed both pre- and postsynaptic localization [Lopez-Gimenez JF, Men- god G, Palacios JM, Vilaro MT (2001) Regional distribution and cellular localization of 5-HT 2 c receptor mRNA in monkey brain: comparison with [ 3 H]mesulergine binding sites and choline acetyltransferase mRNA. Synapse ΙΛΙ-Ιβ ' .

It is anticipated that modulation of the 5-HT 2 c receptor will improve disorders such as depression, anxiety, schizophrenia, cognitive deficits of schizophrenia, obsessive compulsive disorder, bipolar disorder, neuropsychiatric symptoms in Parkinson' disease, in Alzheimer's disease or Lewy Body dementia, migraine, epilepsy, substance abuse, eating disorders, obesity, diabetes, sexual dysfunction/erectile dysfunction, sleep disorders, psoriasis, Parkinson's disease, pain conditions and disorders, and spinal cord injury, smoking cessation, ocular hypertension and Alzheimer's disease. Modulators of the 5-HT 2 c receptor are also shown to be useful in the modulation of bladder function, including the prevention or treatment of urinary incontinence. Compounds with a structure similar to the compounds of the present invention have been described in WO 2014/041131 and WO 03/091250.

There is an ongoing need for providing compounds having high affinity and preferably also selectivity for the 5-HT 2 c receptor. In particular the compounds should have low affinity to adrenergic receptors, such as the α,ι -adrenergic receptor, histamine receptors, such as the Hi -receptor, and dopaminergic receptors, such as the D 2 -receptor, in order to avoid or reduce side effects associated with modulation of these receptors, such as postural hypotension, reflex tachycardia, potentiation of the antihypertensive effect of prazosin, terazosin, doxazosin and labetalol or dizziness associated with the blockade of the (Xi -adrenergic receptor, weight gain, sedation, drowsiness or potentiation of central depressant drugs associated with the blockade of the Hi -receptor, or extrapyramidal movement disorder, such as dystonia, parkinsonism, akathisia, tardive dyskinesia or rabbit syndrome, or endocrine effects, such as prolactin elevation (galactorrhea, gynecomastia, mentstrual changes, sexual dysfunction in males), associated with the block- ade of the D 2 -receptor, and even more important no induction of weight gain in combination with severe metabolic dysfunction found for marketed antipsychotic drugs.

It is moreover desirable that the compounds have low affinity or alternatively an antagonistic effect to/on other serotonergic receptors, especially the 5-HT 2A and/or 5- HT 2B receptors, in order to avoid or reduce side effects associated with modulation of these receptors, such as changes (thickening) of the heart tissue associated with agonism at the 5-HT 2B receptor, and psychotomimetic effect induced by agonism at the 5-HT 2A receptor. Ideally they should show an agonistic action on the 5-HT 2 c receptor, an antagonistic action on the 5-HT 2A receptor or alternatively no affinity to the 5-HT 2A receptor and no affinity to the 5-HT 2B receptor or alternatively an antagonistic action on the 5- HT 2B receptor. Even more ideally the compounds should display an agonistic action on the 5-HT 2 c receptor in combination with an antagonistic action on the 5-HT 2A receptor and no affinity to the 5-HT 2B receptor.

Besides the affinity and selectivity for the 5-HT 2 c receptor, further properties may be advantageous for the treatment and/or prophylaxis of 5-HT 2C -related disorders, such as, for example:

1.) the metabolic stability, for example determined from the half-lives, measured in vitro, in liver microsomes from various species (e.g. rat or human); 2. ) no or only low inhibition of cytochrome P450 (CYP) enzymes: cytochrome P450 (CYP) is the name for a superfamily of heme proteins having enzymatic activity (oxidase). They are also particularly important for the degradation (metabolism) of foreign substances such as drugs or xenobiotics in mammalian organisms. The principal representatives of the types and subtypes of CYP in the human body are: CYP 1A2, CYP 2C9, CYP 2D6 and CYP 3A4. If CYP 3A4 inhibitors (e.g. grapefruit juice, cimet- idine, erythromycin) are used at the same time as medicinal substances which are degraded by this enzyme system and thus compete for the same binding site on the enzyme, the degradation thereof may be slowed down and thus effects and side effects of the administered medicinal substance may be undesirably enhanced;

3. ) a suitable solubility in water (in mg/mL);

4. ) suitable pharmacokinetics (time course of the concentration of the compound of the invention in plasma or in tissue, for example brain). The pharmacokinetics can be described by the following parameters: half-life (in h), volume of distribution (in l » kg- 1), plasma clearance (in l » h-l » kg-l), AUC (area under the curve, area under the concentration-time curve, in ng » h » l-l), oral bioavailability (the dose-normalized ratio of AUC after oral administration and AUC after intravenous administration), the so-called brain- plasma ratio (the ratio of AUC in brain tissue and AUC in plasma);

5. ) no or only low blockade of the hERG channel: compounds which block the hERG channel may cause a prolongation of the QT interval and thus lead to serious disturbances of cardiac rhythm (for example so-called "torsade de pointes"). The potential of compounds to block the hERG channel can be determined by means of the displacement assay with radio labelled dofetilide which is described in the literature (G. J. Diaz et al, Journal of Pharmacological and Toxicological Methods, 50 (2004), 187 199). A smaller IC50 in this dofetilide assay means a greater probability of potent hERG blockade. In addition, the blockade of the hERG channel can be measured by electrophysiological experiments on cells which have been transfected with the hERG channel, by so-called whole-cell patch clamping (G. J. Diaz et al, Journal of Pharmacological and Toxicological Methods, 50 (2004), 187-199).

It was an object of the present invention to provide compounds for the treatment or prophylaxis of various 5-HT 2 c-related diseases. The compounds were intended to have a high affinity on the 5-HT 2 c receptor and be potent and efficacious 5-HT 2 c ago- nists. In addition, the compounds of the invention were intended to have one or more of the aforementioned advantages, namely low affinity to other serotonergic receptors, and especially the lack of potent agonistic effect (antagonism preferred) on the 5-HT 2 A and/or 5-HT 2 B receptors, and additionally one or more of those advantages mentioned under 1.) to 5.).

The present invention provides compounds which have an affinity for the 5-HT 2 c, thus allowing the treatment of disorders related to or affected by the 5-HT 2 c receptor.

SUMMARY OF THE INVENTION

The present invention relates to tricyclic hexahydrodiazepinoquino lines carrying a substituted alkyl radical, to a method for producing them, to compositions comprising such compounds, their use as modulators, especially agonists or partial agonists, of the 5-HT 2 c receptor, their use for preparing a medicament for the prevention or treatment of conditions and disorders which respond to the modulation of 5-HT 2C receptor, to a method for preventing or treating conditions and disorders which respond to the modulation of 5-HT 2C receptor, and processes for preparing such compounds and compositions.

In one aspect, the present invention relates to compounds of the formula (I):

wherein

R 1 is selected from the group consisting of hydrogen, cyano, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, C 2 -C 6 -alkenyl, fluorinated C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, fluorinated C 2 -C 6 -alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C 6 -alkoxy, fluorinated Ci-C 6 -alkoxy, -C(=0)R 9 , phenyl, phenyl-Ci-C 2 -alkyl and a 3-, 4-, 5-,

6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated heterocyclic ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups inde- pendently selected from N, O, S, NO, SO and S0 2 and optionally also 1 or 2 C=0 and/or C=S groups as ring members, where the cyclic moieties in the three last- mentioned radicals may be substituted with one or more substituents R 10 ; each R 2 is independently selected from the group consisting of cyano, nitro, Ci-C 6 - alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C 2 -C6-alkenyl, fluorinated C 2 - C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C6-alkynyl, C3-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C6-alkoxy-Ci-C 4 - alkyl, -CH 2 NR l la R l lb , -C(=0)R 9 , phenyl, phenyl-Ci-C 2 -alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R 10 ; or

two radicals R 2 bound to the same carbon atom, together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring (i.e. a spiro -bound ring), where the ring may contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S0 2 , C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R 10 ;

R 3a and R 3b , independently of each other, are selected from the group consisting of hydrogen, cyano, nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C 2 -C6-alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C 6 - alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy-Ci-C 4 - alkyl, fluorinated Ci-C 6 -alkoxy-Ci-C 4 -alkyl, -CH 2 NR l la R l lb , -C(=0)R 9 , phenyl, phenyl-Ci-C 2 -alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R 10 ; R 4a and R 4b , independently of each other, are selected from the group consisting of hydrogen, cyano, nitro, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C 2 -C6-alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C 6 - alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C6-alkoxy-Ci-C4- alkyl, fluorinated Ci-C 6 -alkoxy-Ci-C 4 -alkyl, -CH 2 NR l la R 1 lb , -C(=0)R 9 , phenyl, phenyl-Ci-C 2 -alkyl, and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R 10 ; or

R 4a and R 4b form together a group =0 or =S; or

R 4a and R 4b , together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring (i.e. a spiro-bound ring), where the ring may contain 1, 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S0 2 , C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R 10 ; and R , independently of each other, are selected from the group consisting of hydrogen, halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci- C6-hydroxyalkyl, C 2 -C6-alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C6-alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C 6 - alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-alkoxy-Ci-C4-alkyl, fluorinated Ci-C 6 - alkoxy-Ci-C4-alkyl, Ci-C6-hydroxyalkoxy, Ci-C6-alkoxy-Ci-C4-alkoxy, Ci-C 6 - alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6-alkylsulfmyl, fluorinated Ci-C 6 - alkylsulfinyl, Ci-C6-alkylsulfonyl, fluorinated Ci-C6-alkylsulfonyl,

-NR l la R l lb , -CH 2 NR l la R l lb , -NR l la C(0)R 9 , -C(=0)R 9 , S0 2 NR l la R l lb , Ci-C 6 - alkylcarbonyloxy, fluorinated Ci-C6-alkylcarbonyloxy, phenyl, phenyl-Ci-C 2 - alkyl, phenoxy, phenylsulfonyl, benzyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the six last- mentioned radicals may be substituted with one or more substituents R 10 ; where R 5a and R 5b are not simultaneously hydro xyl; or

R 5a and R 5b , together with the carbon atom they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring (i.e. a spiro-bound ring), where the ring may contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S0 2 , C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R 10 ; R 6 Ci-Ce-alkyl which carries one or more substituents R ; each R 7 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, Ci-C 6 -hydroxyalkyl, C 2 -C 6 - alkenyl, fluorinated C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, fluorinated C 2 -C 6 -alkynyl, C 3 - Cs-cycloalkyl, fluorinated C 3 -C8-cycloalkyl, C 3 -Cs-cycloalkenyl, fluorinated C 3 -

Cs-cycloalkenyl, Ci-C 6 -alkoxy, fluorinated Ci-C 6 -alkoxy, Ci-C 6 -alkoxy-Ci-C 4 - alkyl, fluorinated Ci-C 6 -alkoxy-Ci-C 4 -alkyl, Ci-C 6 -hydroxyalkoxy, Ci-C 6 -alkoxy- Ci-C 4 -alkoxy, Ci-C 6 -alkylthio, fluorinated Ci-C 6 -alkylthio, Ci-C 6 -alkylsulfinyl, fluorinated Ci-C 6 -alkylsulfinyl, Ci-C 6 -alkylsulfonyl, fluorinated Ci-C 6 - alkylsulfonyl, -NR 1 la R ! lb , -CH 2 NR l la R 1 lb , -NR 1 la C(0)R 9 ,

-C(=0)R 9 , S0 2 NR l la R l lb , Ci-Ce-alkylcarbonyloxy, fluorinated Ci -C 6 - alkylcarbonyloxy, phenyl, phenyl-Ci-C 2 -alkyl, phenoxy, phenylsulfonyl, ben- zyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the six last-mentioned radicals may be substituted with one or more substituents R 10 ; or

two radicals R 7 bound on neighboring carbon atoms, together with the carbon atoms they are bound to, form a 3-, 4-, 5-, 6-, 7- or 8-membered partially unsaturat- ed or maximally unsaturated ring, where the ring may contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S0 2 , C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents R 10 ; each R 8 is independently selected from the group consisting of hydroxyl, halogen, Ci-C6-alkoxy, fluorinated Ci-C6-alkoxy, Ci-C6-hydroxyalkoxy, Ci-C6-alkoxy-Ci-

C 4 -alkoxy, Ci-C6-alkylthio, fluorinated Ci-C6-alkylthio, Ci-C6-alkylsulfinyl, fluorinated Ci-Ce-alkylsulfmyl, Ci-C6-alkylsulfonyl, fluorinated Ci-C 6 - alkylsulfonyl and -NR l la R l lb ; each R 9 is independently selected from the group consisting of hydrogen, cyano, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C 2 -C 6 - alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C6-alkynyl, C 3 - Cs-cycloalkyl, fluorinated C 3 -C8-cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C 6 - alkoxy, -NR l la R l lb , -CH 2 NR l la R l lb , phenyl, phenyl-Ci-C 2 -alkyl, phenoxy, ben- zyloxy and a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1 , 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members, where the cyclic moieties in the five last-mentioned radicals may be substituted with one or more substituents R 10 ; each R 10 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C6-hydroxyalkyl, C 2 -C 6 - alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C6-alkynyl, C 3 - Cs-cycloalkyl, fluorinated C 3 -C8-cycloalkyl, Ci-C6-alkoxy, fluorinated Ci-C 6 - alkoxy, Ci-C6-alkoxy-Ci-C 4 -alkyl, fluorinated Ci-C6-alkoxy-Ci-C 4 -alkyl, Ci-C 6 - hydroxyalkoxy, Ci-C6-alkoxy-Ci-C 4 -alkoxy, Ci-C6-alkylthio, fluorinated Ci-C 6 - alkylthio, Ci-C6-alkylsulfmyl, fluorinated Ci-C6-alkylsulfinyl, Ci-C 6 - alkylsulfonyl, fluorinated Ci-Cg-alkylsulfonyl, -COOH, -NR l la R l lb , - CH 2 NR l la R l lb , Ci-Cg-alkylcarbonyl, fluorinated Ci-Cg-alkylcarbonyl, Ci-C 6 - alkoxycarbonyl, fluorinated Ci-C 6 -alkoxycarbonyl, SO^R 1 la R ! lb , Ci-C 6 - alkylcarbonyloxy and fluorinated Ci-C6-alkylcarbonyloxy; or two radicals R 10 , together with the atom(s) they are bound to, form a saturated, partially unsaturated or maximally unsaturated 3-, 4-, 5-, 6- or 7-membered carbo- cyclic or heterocyclic ring, where the heterocyclic ring contains 1 , 2 or 3 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members;

R l la and R l lb , independently of each other and independently of each occurrence, are selected from the group consisting of hydrogen, cyano, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, C 2 -C 6 -alkenyl, fluorinated C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, fluorinated C 2 -C 6 -alkynyl, Cs-Cs-cycloalkyl, fluorinated Cs-Cs-cycloalkyl, Ci-C 6 -alkoxy, fluorinated Ci-C 6 -alkoxy, Ci-C 6 -alkylcarbonyl, fluorinated Ci-C 6 -alkylcarbonyl, Ci-C 6 -alkoxycarbonyl, fluorinated Ci-C 6 -alkoxycarbonyl, phenyl and benzyl, where the phenyl moieties in the two last-mentioned radicals may carry 1 , 2 or 3 substituents selected from halogen, cyano nitro, Ci-C 6 -alkyl, fluorinated Ci-C 6 - alkyl, Ci-C 6 -alkoxy and fluorinated Ci-C 6 -alkoxy; or,

if R l la and R l lb are bound to the same nitrogen atom, together with this nitrogen atom may form a 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated heterocyclic ring, where the ring may further contain 1 , 2, 3 or 4 heteroatoms or heteroatom-containing groups selected from O, S, N, SO, S0 2 , C=0 and C=S as ring members, and where the ring may be substituted with one or more substituents selected from halogen, cyano nitro, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, Ci-C 6 -alkoxy and fluorinated Ci-C 6 -alkoxy; a is 0, 1 or 2; and b is 0, 1 , 2 or 3; and the N-oxides, tautomeric forms, stereoisomers and pharmaceutically acceptable salts thereof.

In another aspect, the invention relates to a pharmaceutical composition comprising a therapeutically effective amount of at least one compound of formula I or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof, in combination with at least one pharmaceutically acceptable carrier and/or auxiliary substance.

In yet another aspect, the invention relates to a compound of formula I or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for use as a medicament.

In yet another aspect, the invention relates to a compound of formula I or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the treatment of disorders which responds to the modulation of the 5-HT 2 c receptor.

In yet another aspect, the invention relates to the use of a compound of formula I or of an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of disorders which respond to the modulation of the 5-HT 2 c receptor.

In yet another aspect, the invention relates to the use of a compound of formula I or of an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of disorders selected from the group consisting of damage of the central nervous system, disorders of the central nervous system, eating disorders, ocular hypertension, cardiovascular disorders, gastrointestinal disorders and diabetes, and especially from the group consisting of bi- polar disorder, depression, atypical depression, mood episodes, adjustment disorders, anxiety, panic disorders, post-traumatic syndrome, psychoses, schizophrenia, cognitive deficits of schizophrenia, memory loss, dementia of aging, Alzheimer's disease, neuro- psychiatric symptoms in Alzheimer's disease (e.g. aggression), behavioral disorders associated with dementia, social phobia, mental disorders in childhood, attention deficit hyperactivity disorder, organic mental disorders, autism, mutism, disruptive behavior disorder, impulse control disorder, borderline personality disorder, obsessive compulsive disorder, migraine and other conditions associated with cephalic pain or other pain, raised intracranial pressure, seizure disorders, epilepsy, substance use disorders, alcohol abuse, cocaine abuse, tobacco abuse, smoking cessation, sexual dysfunction/erectile dysfunction in males, sexual dysfunction in females, premenstrual syndrome, late luteal phase syndrome, chronic fatigue syndrome, sleep disorders, sleep apnoea, chronic fatigue syndrome, psoriasis, Parkinson's disease, neuropsychiatric symptoms in Parkin- son's disease (e.g. aggression), Lewy Body dementia, neuropsychiatric symptoms in Lewy Body dementia (e.g. aggression), spinal cord injury, trauma, stroke, pain, bladder dysfunction/urinary incontinence, encephalitis, meningitis, eating disorders, obesity, bulimia, weight loss, anorexia nervosa, ocular hypertension, cardiovascular disorders, gastrointestinal disorders, diabetes insipidus, diabetes mellitus, type I diabetes, type II diabetes, type III diabetes, diabetes secondary to pancreatic diseases, diabetes related to steroid use, diabetes complications, hyperglycemia and insulin resistance.

In yet another aspect, the invention relates to a method for treating disorders which respond to the modulation of the 5-HT 2 c receptor, which method comprises ad- ministering to a subject in need thereof at least one compound of formula I or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

In yet another aspect, the invention relates to a method for treating disorders selected from the group consisting of damage of the central nervous system, disorders of the central nervous system, eating disorders, ocular hypertension, cardiovascular dis- orders, gastrointestinal disorders and diabetes, and especially from the group consisting of bipolar disorder, depression, atypical depression, mood episodes, adjustment disorders, anxiety, panic disorders, post-traumatic syndrome, psychoses, schizophrenia, cognitive deficits of schizophrenia, memory loss, dementia of aging, Alzheimer's disease, neuropsychiatric symptoms in Alzheimer's disease (e.g. aggression), behavioral disor- ders associated with dementia, social phobia, mental disorders in childhood, attention deficit hyperactivity disorder, organic mental disorders, autism, mutism, disruptive behavior disorder, impulse control disorder, borderline personality disorder, obsessive compulsive disorder, migraine and other conditions associated with cephalic pain or other pain, raised intracranial pressure, seizure disorders, epilepsy, substance use disor- ders, alcohol abuse, cocaine abuse, tobacco abuse, smoking cessation, sexual dysfunction/erectile dysfunction in males, sexual dysfunction in females, premenstrual syndrome, late luteal phase syndrome, chronic fatigue syndrome, sleep disorders, sleep apnoea, chronic fatigue syndrome, psoriasis, Parkinson's disease, neuropsychiatric symptoms in Parkinson's disease (e.g. aggression), Lewy Body dementia, neuropsychi- atric symptoms in Lewy Body dementia (e.g. aggression), spinal cord injury, trauma, stroke, pain, bladder dysfunction/urinary incontinence, encephalitis, meningitis, eating disorders, obesity, bulimia, weight loss, anorexia nervosa, ocular hypertension, cardio- vascular disorders, gastrointestinal disorders, diabetes insipidus, diabetes mellitus, type I diabetes, type II diabetes, type III diabetes, diabetes secondary to pancreatic diseases, diabetes related to steroid use, diabetes complications, hyperglycemia and insulin resistance, which method comprises administering to a subject in need thereof at least one compound of formula I or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

DETAILED DESCRIPTION The compounds of the formula I may exist in different spatial arrangements.

For example, if the compounds possess one or more centers of asymmetry, polysubsti- tuted rings or double bonds, or as different tautomers, the present invention contemplates the possible use of enantiomeric mixtures, in particular racemates, diastereomeric mixtures and tautomeric mixtures, such as the respective essentially pure enantiomers, diastereomers and tautomers of the compounds of formula I and/or their salts.

It is likewise possible to use physiologically tolerated salts of the compounds of the formula I, especially acid addition salts with physiologically tolerated acids. Examples of suitable physiologically tolerated organic and inorganic acids are hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, acetic acid, trifluoroacetic acid, Ci-C4-alkylsulfonic acids, such as methanesulfonic acid, aromatic sulfonic acids, such as benzenesulfonic acid and toluenesulfonic acid, oxalic acid, maleic acid, fumaric acid, lactic acid, tartaric acid, adipic acid and benzoic acid. Other utilizable acids are described in Fortschritte der Arzneimittelforschung [Advances in drug research], Volume 10, pages 224 et seq., Birkhauser Verlag, Basel and Stuttgart, 1966.

The compounds of formula I may also be present in the form of tautomers. In one aspect, tautomery may be present in compounds I wherein R 4a and R 4b form together a group =0 and R 5a or R 5b is H. For example, the compounds of formula I may have the following tautomeric formulae:

In another aspect, tautomery may be present in compounds I containing rings which have one or more C=0 groups as ring members which are neighboured to a CH 2 group.

The organic moieties mentioned in the above definitions of the variables are, like the term halogen, collective terms for individual listings of the individual group members. The prefix C n -C m indicates in each case the possible number of carbon atoms in the group.

The term "halogen" denotes in each case fluorine, bromine, chlorine or iodine. In one aspect, the halogen may be fluorine, chlorine or bromine.

The term "alkyl" as used herein and in the alkyl moieties of alkoxy, alkylthio, alkylsulfmyl, alkylsulfonyl, alkylcarbonyl, alkoxycarbonyl and the like refers to saturat- ed straight-chain or branched hydrocarbon radicals having 1 to 2 ("Ci-C2-alkyl"), 1 to 3 ("Ci-C 3 -alkyl"), l to 4 ("Ci-C 4 -alkyl") or 1 to 6 ("Ci-C 6 -alkyl") carbon atoms. Ci-C 2 - Alkyl is methyl or ethyl. Ci-C3-Alkyl is additionally propyl and isopropyl. Ci-C4-Alkyl is additionally butyl, 1-methylpropyl (sec-butyl), 2-methylpropyl (isobutyl) or 1 , 1- dimethylethyl (tert-butyl). Ci-C6-Alkyl is additionally also, for example, pentyl, 1- methylbutyl, 2-methylbutyl, 3-methylbutyl, 2,2-dimethylpropyl, 1-ethylpropyl, 1 ,1- dimethylpropyl, 1 ,2-dimethylpropyl, hexyl, 1 -methylpentyl, 2-methylpentyl, 3- methylpentyl, 4-methylpentyl, 1 ,1-dimethylbutyl, 1 ,2-dimethylbutyl, 1 ,3-dimethylbutyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1 , 1 ,2-trimethylpropyl, 1 ,2,2-trimethylpropyl, 1 -ethyl- 1-methylpropyl, or l-ethyl-2- methylpropyl.

The term " fluorinated alkyl" as used herein refers to straight-chain or branched alkyl groups having 1 ("fluorinated methyl), 1 to 2 ("fluorinated Ci-C 2 -alkyl"), 1 to 3 ("fluorinated Ci-C 3 -alkyl"), 1 to 4 ("fluorinated Ci-C 4 -alkyl") or 1 to 6 ("fluorinated Ci- C6-alkyl") carbon atoms (as mentioned above), where some or all of the hydrogen atoms in these groups are replaced by fluorine atoms. Fluorinated methyl is fluoromethyl, difluoromethyl or trifluoromethyl. Fluorinated Ci-C2-alkyl is an alkyl group having 1 or 2 carbon atoms (as mentioned above), where at least one of the hydrogen atoms, e.g. 1 , 2, 3, 4 or 5 hydrogen atoms in these groups are replaced by fluorine atoms, such as difluoromethyl, trifluoromethyl, 1-fluoroethyl, (R)-l-fluoroethyl, (S)-l-fluoroethyl, 2- fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, or pentafluoroethyl. Fluorinated Ci- C4-alkyl is a straight-chain or branched alkyl group having 1 to 4 carbon atoms (as men- tioned above), where at least one of the hydrogen atoms, e.g. 1 , 2, 3, 4 or 5 hydrogen atoms in these groups are replaced by fluorine atoms. Examples are, apart those listed above for Ci-C2-fluoroalkyl, 1-fluoropropyl, (R)-l-fluoropropyl, (S)-l-fluoropropyl, 2- fluoropropyl, (R)-2-fluoropropyl, (S)-2-fluoropropyl, 3-fluoropropyl, 1 , 1- difluoropropyl, 2,2-difluoropropyl, 1 ,2-difluoropropyl, 2,3-difluoropropyl, 1 ,3- difluoropropyl, 3,3-difluoropropyl, 1 ,1 ,2-trifluoropropyl, 1 ,2,2-trifluoropropyl, 1 ,2,3- trifluoropropyl, 2,2,3-trifluoropropyl, 3,3,3-trifluoropropyl, l , l , l-trifluoroprop-2-yl, 2- fluoro-1 -methylethyl, (R)-2-fluoro-l -methylethyl, (S)-2-fluoro-l -methylethyl, 2,2- difluoro- 1 -methylethyl, (R)-2,2-difluoro- 1 -methylethyl, (S)-2,2-difluoro- 1 -methylethyl, 1 ,2-difluoro- 1 -methylethyl, (R)- 1 ,2-difluoro- 1 -methylethyl, (S)- 1 ,2-difluoro- 1 - methylethyl, 2,2,2-trifiuoro- 1 -methylethyl, (R)-2,2,2-trifiuoro- 1 -methylethyl, (S)-2,2,2- trifluoro- 1 -methylethyl, 2-fluoro- 1 -(fluoromethyl)ethyl, 1 -(difluoromethyl)-2,2- difluoroethyl, 1 -(trifluoromethyl)-2,2,2-trifluoroethyl, 1 -(trifluoromethyl)- 1 ,2,2,2- tetrafluoroethyl, 1-fluorobutyl, (R)-l-fluorobutyl, (S)-l-fluorobutyl, 2-fluorobutyl, (R)- 2-fluorobutyl, (S)-2-fluorobutyl, 3-fluorobutyl, (R)-3-fluorobutyl, (S)-3-fluorobutyl, 4- fluorobutyl, 1 ,1-difluorobutyl, 2,2-difluorobutyl, 3,3-difluorobutyl, 4,4-difluorobutyl, 4,4,4-trifluorobutyl and the like. Fluorinated Ci-C6-alkyl is a straight-chain or branched alkyl group having 1 to 6 carbon atoms (as mentioned above), where at least one of the hydrogen atoms, e.g. 1 , 2, 3, 4 or 5 hydrogen atoms in these groups are replaced by fluorine atoms. Additionally examples include for Ci-C4-fluoroalkyl, 1-fluoropentyl, (R)- 1-fluoropentyl, (S)- 1-fluoropentyl, 2-fluoropentyl, (R)-2-fluoropentyl, (S)-2- fluoropentyl, 3-fluoropentyl, (R)-3-fluoropentyl, (S)-3-fluoropentyl, 4-fluoropentyl, (R)-4-fluoropentyl, (S)-4-fluoropentyl, 5-fluoropentyl, (R)-5-fluoropentyl, (S)-5- fluoropentyl, 1-fluorohexyl, (R)-l-fluorohexyl, (S)-l-fluorohexyl, 2-fluorohexyl, (R)-2- fluorohexyl, (S)-2-fluorohexyl, 3-fluorohexyl, (R)-3-fluorohexyl, (S)-3-fluorohexyl, 4- fluorohexyl, (R)-4-fluorohexyl, (S)-4-fluorohexyl, 5-fluorohexyl, (R)-5-fluorohexyl, (S)-5-fluorohexyl, 6-fluorohexyl, (R)-6-fluorohexyl, (S)-6-fluorohexyl, and the like.

The term "haloalkyl" as used herein refers to straight-chain or branched alkyl groups having 1 to 2 ("Ci-C 2 -haloalkyl"), 1 to 3 ("Ci-C 3 -haloalkyl"), 1 to 4 ("Ci-C 4 - haloalkyl") or 1 to 6 ("Ci-C6-haloalkyl") carbon atoms (as mentioned above), where some or all of the hydrogen atoms in these groups are replaced by halogen atoms as mentioned above. Examples are, apart from those mentioned above for fluorinated al- kyl, for Ci-C 2 -haloalkyl chloromethyl, bro mo methyl, dichloromethyl, trichloromethyl, chlorofluoromethyl, dichlorofluoromethyl, chlorodifluoromethyl, 1-chloroethyl, 1- bromoethyl, 2-chloro-2-fluoroethyl, 2-chloro-2,2-difluoroethyl, 2,2-dichloro-2- fluoroethyl or 2,2,2-trichloroethyl; for Ci-C3-haloalkyl, apart those mentioned for Ci- C 2 -haloalkyl, 3-chloropropyl; and for Ci-C4-haloalkyl, apart those mentioned for C 1 -C3- haloalkyl, 4-chlorobutyl and the like.

The term "alkenyl" as used herein refers to monounsaturated straight-chain or branched hydrocarbon radicals having 2 to 3 ("C 2 -C3-alkenyl"), 2 to 4 ("C 2 -C4-alkenyl") or 2 to 6 ("C 2 -C6-alkenyl") carbon atoms and a double bond in any position, such as C 2 - C3-alkenyl, such as ethenyl, 1-propenyl, 2-propenyl or 1-methylethenyl; C 2 -C4-alkenyl, such as ethenyl, 1-propenyl, 2-propenyl, 1-methylethenyl, 1-butenyl, 2-butenyl, 3- butenyl, 1 -methyl- 1-propenyl, 2-methyl- 1-propenyl, l-methyl-2-propenyl or 2-methyl- 2-propenyl; and C 2 -C6-alkenyl, such as ethenyl, 1-propenyl, 2-propenyl, 1- methylethenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1 -methyl- 1-propenyl, 2-methyl- 1- propenyl, l-methyl-2-propenyl, 2-methyl-2-propenyl, 1-pentenyl, 2-pentenyl, 3- pentenyl, 4-pentenyl, 1 -methyl- 1-butenyl, 2-methyl- 1-butenyl, 3 -methyl- 1-butenyl, 1- methyl-2-butenyl, 2-methyl-2-butenyl, 3-methyl-2-butenyl, l-methyl-3-butenyl, 2- methyl-3 -butenyl, 3 -methyl-3 -butenyl, 1 , 1 -dimethyl-2-propenyl, 1 ,2-dimethyl- 1 - propenyl, 1 ,2-dimethyl-2-propenyl, 1 -ethyl- 1-propenyl, l-ethyl-2-propenyl, 1-hexenyl,

2- hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1 -methyl- 1-pentenyl, 2-methyl-l-pentenyl, 3 -methyl- 1 -pentenyl, 4-methyl- 1 -pentenyl, 1 -methyl-2-pentenyl, 2-methyl-2-pentenyl,

3- methyl-2-pentenyl, 4-methyl-2-pentenyl, 1 -methyl-3 -pentenyl, 2-methyl-3 -pentenyl, 3 -methyl-3 -pentenyl, 4-methyl-3 -pentenyl, l-methyl-4-pentenyl, 2-methyl-4-pentenyl, 3-methyl-4-pentenyl, 4-methyl-4-pentenyl, l,l-dimethyl-2-butenyl, l,l-dimethyl-3- butenyl, 1,2-dimethyl-l-butenyl, 1 ,2-dimethyl-2-butenyl, l,2-dimethyl-3-butenyl, 1 ,3-dimethyl- 1 -butenyl, 1 ,3-dimethyl-2-butenyl, 1 ,3-dimethyl-3-butenyl,

2,2-dimethyl-3-butenyl, 2,3-dimethyl- 1 -butenyl, 2,3-dimethyl-2-butenyl,

2,3-dimethyl-3-butenyl, 3,3-dimethyl-l-butenyl, 3,3-dimethyl-2-butenyl,

1 -ethyl- 1 -butenyl, 1 -ethyl-2-butenyl, 1 -ethyl-3 -butenyl, 2-ethyl- 1 -butenyl,

2- ethyl-2-butenyl, 2-ethyl-3 -butenyl, 1 , 1 ,2-trimethyl-2-propenyl,

1 -ethyl- 1 -methyl-2-propenyl, 1 -ethyl-2-methyl- 1 -propenyl, 1 -ethyl-2-methyl-2- propenyl and the like.

The term "fluorinated alkenyl" as used herein refers to unsaturated straight-chain or branched hydrocarbon radicals having 2 to 3 ("fluorinated C2-C3-alkenyl"), 2 to 4 ("fluorinated C2-C4-alkenyl") or 2 to 6 ("fluorinated C2-C6-alkenyl") carbon atoms and a double bond in any position (as mentioned above), where some or all of the hydrogen atoms in these groups are replaced by fluorine atoms, such as, fluorovinyl, fluoroallyl and the like.

The term "alkynyl" as used herein refers to straight-chain or branched hydrocarbon groups having 2 to 3 ("C 2 -C 3 -alkynyl"), 2 to 4 ("C 2 -C 4 -alkynyl") or 2 to 6 ("C 2 -C 6 - alkynyl") carbon atoms and one or two triple bonds in any position, such as C2-C3- alkynyl, such as ethynyl, 1-propynyl or 2-propynyl; C2-C 4 -alkynyl, such as ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, l-methyl-2-propynyl and the like, and C2-C6-alkynyl, such as ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl,

3- butynyl, l-methyl-2-propynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1- methyl-2-butynyl, l-methyl-3-butynyl, 2-methyl-3-butynyl, 3 -methyl- 1-butynyl, 1,1- dimethyl-2-propynyl, l-ethyl-2-propynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl, l-methyl-2-pentynyl, l-methyl-3-pentynyl, l-methyl-4-pentynyl, 2-methyl- 3-pentynyl, 2-methyl-4-pentynyl, 3-methyl-l-pentynyl, 3-methyl-4-pentynyl, 4-methyl- 1-pentynyl, 4-methyl-2-pentynyl, l,l-dimethyl-2-butynyl, l,l-dimethyl-3-butynyl, 1,2- dimethyl-3-butynyl, 2,2-dimethyl-3-butynyl, 3,3-dimethyl-l-butynyl, l-ethyl-2-butynyl, 1 -ethyl-3 -butynyl, 2-ethyl-3-butynyl, 1 -ethyl- l-methyl-2-propynyl and the like.

The term "fluorinated alkynyl" as used herein refers to unsaturated straight-chain or branched hydrocarbon radicals having 2 to 3 ("fluorinated C2-C 3 -alkynyr'), 3 to 4 ("fluorinated C3-C 4 -alkynyl") or 2 to 6 ("fluorinated C2-C6-alkynyl") carbon atoms and one or two triple bonds in any position (as mentioned above), where some or all of the hydrogen atoms in these groups are replaced by fluorine atoms.

The term "cycloalkyl" as used herein refers to monocyclic saturated hydrocarbon radicals having 3 to 8 ("Cs-Cs-cycloalkyl"), in particular 3 to 6 carbon atoms ("C 3 -C6- cycloalkyl") or 3 to 5 carbon atoms ("Cs-Cs-cycloalkyl") or 3 or 4 carbon atoms ("C 3 - C4-cycloalkyl"). Examples for C3-C4-cycloalkyl are cyclopropyl and cyclobutyl. Examples of C3-C 5 -cycloalkyl are cyclopropyl, cyclobutyl and cyclopentyl. Examples of C 3 - C6-cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. Examples of C3- Cs-cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.

The term " fluorinated cycloalkyl" as used herein refers to monocyclic saturated hydrocarbon groups having 3 to 8 ("C 3 -C8-halocycloalkyl" ) or preferably 3 to 6 ("C 3 - C6-halocycloalkyl") or 3 to 5 ("C 3 -C5-halocycloalkyl") carbon ring members (as mentioned above) in which some or all of the hydrogen atoms are replaced by fluorine at- oms. Examples include 1-fluorocyclopropyl, 2-fluorocyclopropyl, (S)- and (R)-2,2- difluorocyclopropyl, 1 ,2-difluorocyclopropyl, 2,3-difluorocyclopropyl, pentafluorocy- clopropyl, 1-fluorocyclo butyl, 2-fluorocyclo butyl, 3-fluorocyclobutyl, 2,2-difluoro- cyclobutyl, 3,3-difluorocyclobutyl, 1 ,2-difluorocyclo butyl, 1 ,3-difluorocyclobutyl, 2,3- difluorocyclo butyl, 2,4-difluorocyclo butyl, 1 ,2,2-trifluorocyclo butyl, 1-fluorocyclo- heptyl, 2-fluorocyclo heptyl, 3-fluorocycloheptyl, 4-fluorocycloheptyl, 1 ,2-difluoro- cycloheptyl, 1 ,3-difluorocycloheptyl, 1 ,4-difluorocycloheptyl, 2,2-difluorocycloheptyl, 2,3-difluorocycloheptyl, 2,4-difluorocycloheptyl, 2,5-difluorocycloheptyl, 2,6-difluoro- cycloheptyl, 2,7-difluorocycloheptyl, 3,3-difluorocycloheptyl, 3,4-difluorocycloheptyl, 3,5-difluorocycloheptyl, 3,6-difluorocycloheptyl, 4,4-difluorocycloheptyl, 4,5-difluoro- cycloheptyl, and the like.

The term "cycloalkenyl" as used herein refers to monocyclic partially unsaturated, non-aromatic hydrocarbon radicals having 3 to 8 ('^-Cs-cycloalkenyl"), in particular 5 to 7 carbon atoms ("Cs-Cy-cycloalkenyl") or 5 or 6 carbon atoms ("C5-C6- cycloalkenyl") and one or more non-cumulative, preferably one, C-C double bonds in the ring. Examples for Cs-Ce-cycloalkenyl are cyclopent-l-en-l-yl, cyclopent-l-en-3-yl, cyclopent-l-en-4-yl, cyclopenta-l ,3-dien-l-yl, cyclopenta-l ,3-dien-2-yl, cyclopenta- l ,3-dien-5-yl, cyclohex-l-en-l-yl, cyclohex-l-en-3-yl, cyclohex-l-en-4-yl, cyclohexa- 1.3- dien-l-yl, cyclohexa-l ,3-dien-2-yl, cyclohexa-l ,3-dien-5-yl, cyclo hexa-l ,4-dien-l- yl and cyclohexa-l ,4-dien-3-yl. Examples of Cs-Cy-cycloalkenyl are, apart those mentioned above, include for Cs-Ce-cycloalkenyl, cyclohept-l-en-l-yl, cyclohept-l-en-3-yl, cyclohept-l-en-4-yl, cyclohept-l-en-5-yl, cyclohepta- 1 ,3-dien-l-yl, cyclohepta- 1 ,3- dien-2-yl, cyclohepta- 1 ,3-dien-5-yl, cyclohepta- 1 ,3-dien-6-yl, cyclohepta- 1 ,4-dien- 1 -yl, cyclohepta-l ,4-dien-2-yl, cyclohepta- l ,4-dien-3-yl and cyclohepta- 1 ,4-dien-6-yl. Examples of C3-C8-cycloalkenyl are, apart those mentioned above for Cs-Ce-cycloalkenyl, cycloprop-l-en-l-yl, cycloprop-l-en-3-yl, cyclobut-l-en-l-yl, cyclobut-l-en-3-yl, cy- clooct-l-en-l-yl, cyclooct-l-en-3-yl, cyclooct-l-en-4-yl, cyclooct-l-en-5-yl, cycloocta- 1 ,3-dien-l-yl, cycloocta-l ,3-dien-2-yl, cycloocta-l ,3-dien-5-yl, cycloocta-l ,3-dien-6-yl, cycloocta-l ,4-dien-l-yl, cycloocta-l ,4-dien-2-yl, cycloocta-l ,4-dien-3-yl, cycloocta-

1.4- dien-6-yl, cycloocta-l ,4-dien-7-yl, cycloocta-l ,5-dien-l-yl, and cycloocta-l ,5-dien- 3-yl.

The term "fluorinated cycloalkenyl" as used herein refers to monocyclic partially unsaturated, non-aromatic hydrocarbon radicals having 3 to 8 ("fluorinated C3-C8- cycloalkenyl"), in particular 5 to 7 carbon atoms ("fluorinated Cs-Cy-cycloalkenyl") or 5 or 6 carbon atoms ("fluorinated Cs-Ce-cycloalkenyl") and one or more non-cumulative, preferably one, C-C double bonds in the ring and in which some or all of the hydrogen atoms are replaced by fluorine atoms.

The term "cycloalkyl-Ci-C 4 -alkyl" refers to a Cs-Cs-cycloalkyl group ("C 3 -C 8 - cycloalkyl-Ci-C 4 -alkyl"), preferably a C3-C 6 -cycloalkyl group ("C 3 -C 6 -cycloalkyl-Ci- C 4 -alkyl") as defined above which is bound to the remainder of the molecule via a Ci- C 4 -alkyl group, as defined above. The term "cycloalkyl-Ci-C 2 -alkyl" refers to a C 3 -C 8 - cycloalkyl group ("C 3 -C 8 -cycloalkyl-Ci-C 2 -alkyl"), preferably a C 3 -C 6 -cycloalkyl group ("C 3 -C 6 -cycloalkyl-Ci-C 2 -alkyl") as defined above which is bound to the remainder of the molecule via a Ci-C 2 -alkyl group, as defined above. Examples for C 3 -C 6 -cycloalkyl- Ci-C 2 -alkyl are cyclopropylmethyl, cyclopropylethyl, cyclobutylmethyl, cyclo butyl- ethyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl and cyclohexylethyl. Examples for C 3 -C 6 -cycloalkyl-Ci-C4-alkyl, apart those mentioned for C 3 -C 6 -cycloalkyl- Ci-C 2 -alkyl, are cyclopropylpropyl, cyclopropylbutyl, cyclo butylpropyl, cyclo butyl- butyl, cyclopentylpropyl, cyclopentylbutyl, cyclohexylpropyl and cyclohexylbutyl. Examples for C 3 -C8-cycloalkyl-Ci-C 2 -alkyl, apart those mentioned for C 3 -C6-cycloalkyl- Ci-C2-alkyl, are cycloheptylmethyl, cycloheptylethyl, cyclooctylmethyl and cy- clooctylethyl. Examples for C3-C8-cycloalkyl-Ci-C4-alkyl, apart those mentioned for C3-C8-cycloalkyl-Ci-C2-alkyl, are cycloheptylpropyl, cycloheptylbutyl, cyclooctylprop- yland cyclooctylbutyl.

The term "fluorinated cycloalkyl-Ci-C4-alkyl" refers to a fluorinated C3-C8- cycloalkyl group ("fluorinated C3-C8-cycloalkyl-Ci-C4-alkyl"), preferably a fluorinated C3-C6-cycloalkyl group ("fluorinated C3-C6-cycloalkyl-Ci-C4-alkyl") as defined above which is bound to the remainder of the molecule via a Ci-C4-alkyl group, as defined above. The term " fluorinated cycloalkyl-Ci-C2-alkyl" refers to a fluorinated C3-C8- cycloalkyl group ("fluorinated C3-C8-cycloalkyl-Ci-C2-alkyl"), preferably a fluorinated C3-C6-cycloalkyl group ("fluorinated C3-C6-cycloalkyl-Ci-C2-alkyl") as defined above which is bound to the remainder of the molecule via a Ci-C2-alkyl group, as defined above.

The term "Ci-C2-alkoxy" is a Ci-C2-alkyl group, as defined above, attached via an oxygen atom. The term "Ci-C3-alkoxy" is a Ci-C3-alkyl group, as defined above, attached via an oxygen atom. The term "Ci-C4-alkoxy" is a Ci-C4-alkyl group, as defined above, attached via an oxygen atom. The term "Ci-C6-alkoxy" is a Ci-C6-alkyl group, as defined above, attached via an oxygen atom. Ci-C2-Alkoxy is methoxy or ethoxy. Ci- C 3 -Alkoxy is additionally, for example, n-propoxy and 1-methylethoxy (isopropoxy). Ci-C4-Alkoxy is additionally, for example, butoxy, 1-methylpropoxy (sec-butoxy), 2- methylpropoxy (isobutoxy) or 1 , 1-dimethylethoxy (tert-butoxy). Ci-C6-Alkoxy is additionally, for example, pentoxy, 1-methylbutoxy, 2-methylbutoxy, 3-methylbutoxy, 1 , 1- dimethylpropoxy, 1 ,2-dimethylpropoxy, 2,2-dimethylpropoxy, 1-ethylpropoxy, hexoxy, 1-methylpentoxy, 2-methylpentoxy, 3-methylpentoxy, 4-methylpentoxy, 1 , 1- dimethylbutoxy, 1 ,2-dimethylbutoxy, 1 ,3-dimethylbutoxy, 2,2-dimethylbutoxy, 2,3- dimethylbutoxy, 3,3-dimethylbutoxy, 1-ethylbutoxy, 2-ethylbutoxy, 1 , 1 ,2- trimethylpropoxy, 1 ,2,2-trimethylpropoxy, 1 -ethyl- 1-methylpropoxy or l-ethyl-2- methylpropoxy. Ci-Cs-Alkoxy is additionally, for example, heptyloxy, octyloxy, 2- ethylhexyloxy and positional isomers thereof.

The term "fluorinated Ci-C2-alkoxy" is a fluorinated Ci-C2-alkyl group, as defined above, attached via an oxygen atom. The term "fluorinated Ci-C3-alkoxy" is a fluorinated Ci-C3-alkyl group, as defined above, attached via an oxygen atom. The term "fluori- nated Ci-C 6 -haloalkoxy" is a fluorinated Ci-C 6 -alkyl group, as defined above, attached via an oxygen atom. Fluorinated Ci-C 2 -alkoxy is, for example, OCH 2 F, OCHF 2 , OCF 3 , 1-fluoroethoxy, (R)-l-fluoroethoxy, (S)-l-fluoroethoxy, 2-fluoroethoxy, 1,1- difluoroethoxy, 1 ,2-difluoroethoxy, 2,2-difluoroethoxy, 1 , 1 ,2-trifluoroethoxy, 1,2,2- trifluoroethoxy, 2,2,2-trifluoroethoxy, 1,1,2,2-tetrafluoroethoxy or OC 2 Fs. Fluorinated Ci-C3-alkoxy is additionally, for example, 1-fluoropropoxy, (R)-l-fluoropropoxy, (S)-l- fluoropropoxy, 2-fluoropropoxy, (R)-2-fluoropropoxy, (S)-2-fluoropropoxy, 3- fluoropropoxy, 1,1-difluoropropoxy, 2,2-difluoropropoxy, 2,3-difiuoropropoxy, 3,3- difluoropropoxy, 3,3,3-trifluoropropoxy, (R)-2-fluoro-l-methylethoxy, (S)-2-fluoro-l- methylethoxy, (R)-2,2-difluoro-l-methylethoxy, (S)-2,2-difiuoro-l-methylethoxy, (R)- 1 ,2-difiuoro- 1 -methylethoxy, (S)- 1 ,2-difiuoro- 1 -methylethoxy, (R)-2,2,2-trifluoro- 1 - methylethoxy, (S)-2,2,2-trifluoro-l -methylethoxy, 2-fluoro-l-(fiuoromethyl)ethoxy, 1- (difluoromethyl)-2,2-difluoroethoxy, OCH 2 -C 2 F 5 , OCF 2 -C 2 F 5 or l-(CH 2 F)-2- fluoroethoxy. Fluorinated Ci-C4-alkoxy is additionally, for example, 1-fluorobutoxy, (R)- 1-fluorobutoxy, (S)- 1-fluorobutoxy, 2-fluorobutoxy, 3-fluorobutoxy,

4-fluorobutoxy, 1,1-difiuorobutoxy, 2,2-difluorobutoxy, 3,3-difiuorobutoxy, 4,4- difluorobutoxy, 4,4,4-trifiuorobutoxy or nonafluorobutoxy. Fluorinated Ci-C6-alkoxy is additionally, for example, 5-fluoropentoxy, undecafluoropentoxy, 6-fluorohexoxy or tridecafluorohexoxy.

The term "Ci-C 4 -alkoxy-Ci-C 4 -alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 4 carbon atoms, as defined above, where one hydrogen atom is replaced by a Ci-C 4 -alkoxy group, as defined above. The term "C 1 -C6- alkoxy-Ci-C4-alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 4 carbon atoms, as defined above, where one hydrogen atom is replaced by a Ci-C6-alkoxy group, as defined above. Examples are methoxymethyl, ethoxymethyl, propoxymethyl, isopropoxymethyl, n-butoxymethyl, sec-butoxymethyl, isobutoxyme- thyl, tert-butoxymethyl, 1-methoxyethyl, 1-ethoxyethyl, 1-propoxyethyl,

1-isopropoxyethyl, 1-n-butoxyethyl, 1-sec-butoxyethyl, 1-isobutoxyethyl, 1-tert- butoxyethyl, 2-methoxyethyl, 2-ethoxyethyl, 2-propoxyethyl, 2-isopropoxyethyl, 2-n- butoxyethyl, 2-sec-butoxyethyl, 2-isobutoxyethyl, 2-tert-butoxyethyl, 1-methoxypropyl, 1-ethoxypropyl, 1-propoxypropyl, 1-isopropoxypropyl, 1-n-butoxypropyl, 1-sec- butoxypropyl, 1-isobutoxypropyl, 1-tert-butoxypropyl, 2-methoxypropyl, 2- ethoxypropyl, 2-propoxypropyl, 2-isopropoxypropyl, 2-n-butoxypropyl, 2-sec- butoxypropyl, 2-isobutoxypropyl, 2-tert-butoxypropyl, 3-methoxypropyl, 3- ethoxypropyl, 3-propoxypropyl, 3-isopropoxypropyl, 3-n-butoxypropyl, 3-sec- butoxypropyl, 3-isobutoxypropyl, 3-tert-butoxypropyl and the like.

The term "Ci-C 2 -alkoxy-methyl" as used herein, refers to methyl where one hydrogen atom is replaced by a Ci-C 2 -alkoxy group, as defined above. Examples are methoxymethyl and ethoxymethyl. The term "Ci-C 4 -alkoxy-methyl" as used herein, refers to methyl where one hydrogen atom is replaced by a Ci-C 4 -alkoxy group, as defined above. Examples are methoxymethyl, ethoxymethyl, propoxymethyl, iso- propoxymethyl, n-butoxymethyl, sec-butoxymethyl, isobutoxymethyl and tert- butoxymethyl.

The term "fluorinated Ci-C 4 -alkoxy-Ci-C 4 -alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 4 carbon atoms, as defined above, where one hydrogen atom is replaced by a Ci-C4-alkoxy group, as defined above, and wherein at least one, e.g. 1, 2, 3, 4 or all of the remaining hydrogen atoms (either in the alkoxy moiety or in the alkyl moiety or in both) are replaced by fluorine atoms. The term "fluorinated Ci-C 6 -alkoxy-Ci-C4-alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 4 carbon atoms, as defined above, where one hydrogen atom is replaced by a Ci-C 6 -alkoxy group, as defined above, and wherein at least one, e.g. 1, 2, 3, 4 or all of the remaining hydrogen atoms (either in the alkoxy moiety or in the alkyl moiety or in both) are replaced by fluorine atoms. Examples are difluo- romethoxymethyl (CHF 2 OCH 2 ), trif uoromethoxymethyl (CF 3 OCH 2 ), 1- dif uoromethoxyethyl (CHF 2 OCH(CH 3 )), 1-trif uoromethoxyethyl (CF 3 OCH(CH 3 )), 2- dif uoromethoxyethyl (CHF 2 OCH 2 CH 2 ), 2-trif uoromethoxyethyl (CF 3 OCH 2 CH 2 ), methoxy-dif uoromethyl (CH 3 OCF 2 ), 2-methoxy- 1 , 1 -difiuoroethyl (CH 3 OCH 2 CF 2 ), 2- methoxy-2,2-difluoroethyl (CH 3 OCF 2 CH 2 ), and the like.

The term "hydroxy-Ci-C4-alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 4 carbon atoms, as defined above, where one hydrogen atom is replaced by a hydroxyl group. The term "hydroxy-Ci-C6-alkyl" as used herein, refers to a straight-chain or branched alkyl group having 1 to 6 carbon atoms, as defined above, where one hydrogen atom is replaced by a hydroxyl group. Examples for hydroxy-Ci-C4-alkyl include hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, 1- hydroxypropyl, 2-hydroxypropyl, 3-hydroxypropyl, l-hydroxyprop-2-yl, 2- hydroxyprop-2-yl,l-hydroxybutyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 1- hydroxybut-2-yl, 2-hydroxybut-2-yl, 3-hydroxybut-2-yl, 4-hydroxybut-2-yl, hydroxy- tert-butyl and the like. Examples for hydroxy-Ci-C6-alkyl are, apart those mentioned for hydroxy-Ci-C4-alkyl, 1-hydroxypentyl, 2-hydroxypentyl, 3-hydroxypentyl, 4- hydroxypentyl, 5-hydroxypentyl, 1-hydroxyhexyl, 2-hydroxyhexyl, 3-hydroxyhexyl, 4- hydroxyhexyl, 5-hydroxyhexyl, 6-hydroxyhexyl and the like.

The term "hydroxy-Ci-C4-alkoxy" as used herein, refers to a Ci-C4-alkoxy group, as defined above, where one hydrogen atom is replaced by a hydroxyl group. The term "hydroxy-Ci-C6-alkoxy" as used herein, refers to a Ci-C6-alkoxy group, as defined above, where one hydrogen atom is replaced by a hydroxyl group. Examples for hydro xy-Ci-C4-alkoxy include hydroxymethoxy, 1 -hydroxy ethoxy, 2-hydroxyethoxy, 1- hydroxypropoxy, 2-hydroxypropoxy, 3-hydroxypropoxy, l-hydroxy-2-propoxy, 2- hydroxy-2-propoxy, l-hydroxybutoxy, 2-hydroxybutoxy, 3-hydroxybutoxy, 4- hydroxybutoxy, l-hydroxy-2-butoxy, 2-hydroxy-2-butoxy, 3-hydroxy-2-butoxy, 4- hydroxy-2-butoxy, hydroxy-tert-butoxy and the like. Examples for hydroxy-Ci-C6- alkoxy include, apart those mentioned for hydroxy-Ci-C4-alkoxy, 1-hydroxypentoxy, 2- hydroxypentoxy, 3-hydroxypentoxy, 4-hydroxypentoxy, 5-hydroxypentoxy, 1- hydroxyhexoxy, 2-hydroxyhexoxy, 3-hydroxyhexoxy, 4-hydroxyhexoxy, 5- hydroxyhexoxy, 6-hydroxyhexoxy and the like.

The term "Ci-C4-alkoxy-Ci-C4-alkoxy" as used herein, refers to a Ci-C4-alkoxy group, as defined above, where one hydrogen atom is replaced by a Ci-C4-alkoxy group, as defined above. The term "Ci-C6-alkoxy-Ci-C4-alkoxy" as used herein, refers to a Ci-C4-alkoxy group, as defined above, where one hydrogen atom is replaced by a Ci-C6-alkoxy group, as defined above. Examples are methoxymethoxy, ethoxymethoxy, propoxymethoxy, isopropoxymethoxy, butoxymethoxy, sec-butoxymethoxy, isobu- toxymethoxy, tert-butoxymethoxy, 1-methoxy ethoxy, 1 -ethoxy ethoxy, 1- propoxyethoxy, 1-isopropoxyethoxy, 1-butoxyethoxy, 1-sec-butoxyethoxy, 1- isobutoxyethoxy, 1-tert-butoxyethoxy, 2-methoxyethoxy, 2-ethoxyethoxy, 2- propoxyethoxy, 2-isopropoxyethoxy, 2-butoxyethoxy, 2-sec-butoxyethoxy, 2- isobutoxyethoxy, 2-tert-butoxyethoxy, 1-methoxypropoxy, 1-ethoxypropoxy, 1- propoxypropoxy, 1-isopropoxypropoxy, 1-butoxypropoxy, 1-sec-butoxypropoxy, 1- isobutoxypropoxy, 1-tert-butoxypropoxy, 2-methoxypropoxy, 2-ethoxypropoxy, 2- propoxypropoxy, 2-isopropoxypropoxy, 2-butoxypropoxy, 2-sec-butoxypropoxy, 2- isobutoxypropoxy, 2-tert-butoxypropoxy, 3-methoxypropoxy, 3-ethoxypropoxy, 3- propoxypropoxy, 3-isopropoxypropoxy, 3-butoxypropoxy, 3-sec-butoxypropoxy, 3- isobutoxypropoxy, 3-tert-butoxypropoxy and the like.

The term "Ci-C2-alkylthio" is a Ci-C2-alkyl group, as defined above, attached via a sulfur atom. The term "Ci-C3-alkylthio" refers to a Ci-C3-alkyl group, as defined above, attached via a sulfur atom. The term "Ci-C4-alkylthio" is a Ci-C4-alkyl group, as defined above, attached via a sulfur atom. The term "Ci-C6-alkylthio" refers to a Ci-C 6 - alkyl group, as defined above, attached via a sulfur atom. The term "Ci-Cio-alkylthio" refers to a Ci-Cio-alkyl group, as defined above, attached via a sulfur atom. C1-C2- Alkylthio is methylthio or ethylthio. Ci-C3-Alkylthio is additionally, for example, n- propylthio or 1-methylethylthio (isopropylthio). Ci-C4-Alkylthio is additionally, for example, butylthio, 1-methylpropylthio (sec-butylthio), 2-methylpropylthio (isobu- tylthio) or 1,1-dimethylethylthio (tert-butylthio). Ci-C6-Alkylthio is additionally, for example, pentylthio, 1-methylbutylthio, 2-methylbutylthio, 3-methylbutylthio, 1,1- dimethylpropylthio, 1,2-dimethylpropylthio, 2,2-dimethylpropylthio, 1-ethylpropylthio, hexylthio, 1-methylpentylthio, 2-methylpentylthio, 3-methylpentylthio, 4- methylpentylthio, 1,1-dimethylbutylthio, 1,2-dimethylbutylthio, 1,3-dimethylbutylthio, 2,2-dimethylbutylthio, 2,3-dimethylbutylthio, 3,3-dimethylbutylthio, 1-ethylbutylthio, 2-ethylbutylthio, 1 ,1,2-trimethylpropylthio, 1,2,2-trimethylpropylthio, 1 -ethyl- 1- methylpropylthio or l-ethyl-2-methylpropylthio.

The term "fluorinated Ci-C2-alkylthio" refers to a fluorinated Ci-C2-alkyl group, as defined above, attached via a sulfur atom. The term "fluorinated Ci-C3-alkylthio" refers to a fluorinated Ci-C3-alkyl group, as defined above, attached via a sulfur atom. The term "fluorinated Ci-C4-alkylthio" refers to a fluorinated Ci-C4-alkyl group, as defined above, attached via a sulfur atom. The term "fluorinated Ci-C6-alkylthio" refers to a fluorinated Ci-C6-alkyl group, as defined above, attached via a sulfur atom. Fluorinated Ci-C2-alkylthio refers to, for example, SCH 2 F, SCHF 2 , SCF 3 , 2-fluoroethylthio, 2,2-difluoroethylthio, 2,2,2-trif uoroethylthio, or SC2F5. Fluorinated Ci-C3-alkylthio may additionally, for example, include 2-fluoropropylthio, 3-f uoropropylthio,

2,2-difluoropropylthio, 2,3-difluoropropylthio, 3,3,3-trifluoropropylthio, SCH2-C2F5, SCF 2 -C 2 F 5 or l-(CH 2 F)-2-fluoroethylthio. Fluorinated Ci-C 4 -alkylthio may additionally, for example, include 4-fluorobutylthio or nonafluorobutylthio. Fluorinated Ci-C 6 - alkylthio is additionally, for example, 5-fluoropentylthio, undecafluoropentylthio, 6- fluorohexylthio or dodecafluorohexylthio.

The term "Ci-C 2 -alkylsulfinyl" refers to a Ci-C 2 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. The term "Ci-C4-alkylsulfmyl" is a Ci-C 4 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. The term "C 1 -C 6 - alkylsulfinyl" is a Ci-C 6 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. Ci-C 2 -Alkylsulfinyl is methylsulfinyl or ethylsulfmyl. Ci-C 4 -Alkylsulfinyl is additionally, for example, n-propylsulfinyl, 1-methylethylsulfinyl (isopropylsulfinyl), butylsulfinyl, 1 -methylpropylsulfinyl (sec-butylsulfinyl), 2-methylpropylsulfinyl (isobu- tylsulfinyl) or 1,1-dimethylethylsulfinyl (tert-butylsulfinyl). Ci-Ce-Alkylsulfmyl is additionally, for example, pentylsulfmyl, 1-methylbutylsulfinyl, 2-methylbutylsulfinyl, 3-methylbutylsulfinyl, 1 , 1 -dimethylpropylsulfinyl, 1 ,2-dimethylpropylsulfmyl,

2,2-dimethylpropylsulfinyl, 1-ethylpropylsulfinyl, hexylsulfinyl, 1- methylpentylsulfinyl, 2-methylpentylsulfinyl, 3-methylpentylsulfinyl, 4- methylpentylsulfmyl, 1 , 1 -dimethylbutylsulfinyl, 1 ,2-dimethylbutylsulfinyl,

1 ,3-dimethylbutylsulfinyl, 2,2-dimethylbutylsulfinyl, 2,3-dimethylbutylsulfinyl, 3,3-dimethylbutylsulfinyl, 1-ethylbutylsulfinyl, 2-ethylbutylsulfinyl, 1,1,2- trimethylpropylsulfinyl, 1 ,2 ,2-trimethylpropylsulfinyl, 1 -ethyl- 1 -methylpropylsulfinyl or 1 -ethyl-2-methylpropylsulfmyl.

The term "fluorinated Ci-C 2 -alkylsulfinyl" refers to a fluorinated Ci-C 2 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. The term "fluorinated Ci- C 3 -alkylsulfinyl" refers to a fluorinated Ci-C 3 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. The term "fluorinated Ci-C 4 -alkylsulfinyl" refers to a fluorinated Ci-C 4 -alkyl group, as defined above, attached via a sulfinyl [S(O)] group. The term "fluorinated Ci-Ce-alkylsulfmyl" refers to a fluorinated Ci-C6-alkyl group, as defined above, attached via a sulfinyl [S(O)] group. Fluorinated Ci-C 2 -alkylsulfmyl is, for example, S(0)CH 2 F, S(0)CHF 2 , S(0)CF 3 , 2-fluoroethylsulfmyl,

2,2-difluoroethylsulfinyl, 2,2,2-trifluoroethylsulfinyl, or S(0)C 2 F 5 . Fluorinated C 1 -C3- alkylsulfinyl may additionally, for example, include 2-fluoropropylsulfinyl, 3- fluoropropylsulfinyl, 2,2-difluoropropylsulfinyl, 2,3-difluoropropylsulfinyl, 3,3,3- trifluoropropylsulfmyl, S(0)CH 2 -C 2 F 5 , S(0)CF 2 -C 2 F 5 or l-(CH 2 F)-2- fluoroethylsulfinyl. Fluorinated Ci-C4-alkylsulfinyl may additionally, for example, include 4-fluorobutylsulfinyl or nonafluorobutylsulfinyl. Fluorinated Ci-Ce-alkylsulfmyl may additionally, for example, include 5-fluoropentylsulfinyl, undecafluoropentylsulfi- nyl, 6-fluorohexylsulfinyl or dodecafluorohexylsulfinyl.

The term "Ci-C 2 -alkylsulfonyl" refers to a Ci-C 2 -alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. The term "Ci-C4-alkylsulfonyl" refers to a C1-C4- alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. The term "C1-C6- alkylsulfonyl" is a Ci-C6-alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. Ci-C 2 -Alkylsulfonyl refers to a methylsulfonyl or ethylsulfonyl. C1-C4- Alkylsulfonyl is additionally, for example, n-propylsulfonyl, 1-methylethylsulfonyl (isopropylsulfonyl), butylsulfonyl, 1-methylpropylsulfonyl (sec-butylsulfonyl), 2- methylpropylsulfonyl (isobutylsulfonyl) or 1,1-dimethylethylsulfonyl (tert- butylsulfonyl). Ci-C6-Alkylsulfonyl is additionally, for example, pentylsulfonyl, 1- methylbutylsulfonyl, 2-methylbutylsulfonyl, 3-methylbutylsulfonyl, 1,1- dimethylpropylsulfonyl, 1 ,2-dimethylpropylsulfonyl, 2,2-dimethylpropylsulfonyl, 1- ethylpropylsulfonyl, hexylsulfonyl, 1-methylpentylsulfonyl, 2-methylpentylsulfonyl, 3-methylpentylsulfonyl, 4-methylpentylsulfonyl, 1,1-dimethylbutylsulfonyl, 1,2- dimethylbutylsulfonyl, 1,3-dimethylbutylsulfonyl, 2,2-dimethylbutylsulfonyl, 2,3- dimethylbutylsulfonyl, 3,3-dimethylbutylsulfonyl, 1-ethylbutylsulfonyl, 2- ethylbutylsulfonyl, 1,1,2-trimethylpropylsulfonyl, 1 ,2,2-trimethylpropylsulfonyl, 1- ethyl- 1-methylpropylsulfonyl or l-ethyl-2-methylpropylsulfonyl.

The term "fluorinated Ci-C 2 -alkylsulfonyl" refers to a fluorinated Ci-C 2 -alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. The term "fluorinated Ci-C3-alkylsulfonyl" refers to a fluorinated Ci-C3-alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. The term "fluorinated Ci-C4-alkylsulfonyl" refers to a fluorinated Ci-C4-alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. The term "fluorinated Ci-Ce-alkylsulfonyl" refers to a fluorinated Ci-C6-alkyl group, as defined above, attached via a sulfonyl [S(0) 2 ] group. Fluorinated Ci-C 2 -alkylsulfonyl is, for example, S(0) 2 CH 2 F, S(0) 2 CHF 2 , S(0) 2 CF 3 , 2-fluoroethylsulfonyl,

2,2-difluoroethylsulfonyl, 2,2,2-trifluoroethylsulfonyl, or S(0) 2 C 2 F 5 . Fluorinated C1-C3- alkylsulfonyl is additionally, for example, 2-fluoropropylsulfonyl, 3- fluoropropylsulfonyl, 2,2-difluoropropylsulfonyl, 2,3-difluoropropylsulfonyl, 3,3,3- trifluoropropylsulfonyl, S(0) 2 CH 2 -C 2 F 5 , S(0) 2 CF 2 -C 2 F 5 or l-(CH 2 F)-2- fluoroethylsulfonyl. Fluorinated Ci-C4-alkylsulfonyl is additionally, for example, 4-fluorobutylsulfonyl or nonafluorobutylsulfonyl. Fluorinated Ci-C6-alkylsulfonyl is additionally, for example, 5-fluoropentylsulfonyl, undecafluoropentylsulfonyl, 6- fluorohexylsulfonyl or dodecafluorohexylsulfonyl.

Ci-C4-Alkylcarbonyl refers to a straight-chain or branched alkyl group having from 1 to 4 carbon atoms), which is bound to the remainder of the molecule via a carbonyl group (CO), such as in acetyl, propionyl, isopropylcarbonyl, butylcarbonyl, sec- butylcarbonyl, isobutylcarbonyl, and tert-butylcarbonyl. Ci-C6-Alkylcarbonyl is a straight-chain or branched alkyl group having from 1 to 6 carbon atoms, which is bound to the remainder of the molecule via a carbonyl group (CO). Examples include, apart those listed above for Ci-C4-alkylcarbonylpentylcarbonyl, hexylcarbonyl and the constitutional isomers thereof.

Fluorinated Ci-C4-alkylcarbonyl refers to a straight-chain or branched fluorinated alkyl group having from 1 to 4 carbon atoms as defined above, which is bound to the remainder of the molecule via a carbonyl group (CO). Fluorinated Ci-C6-alkylcarbonyl is a straight-chain or branched fluorinated alkyl group having from 1 to 6 carbon atoms as defined above, which is bound to the remainder of the molecule via a carbonyl group (CO). Examples include trifluoromethylcarbonyl, 2,2,2-trifluoroethylcarbonyl and the like.

C 3 -C 6 -cycloalkylcarbonyl relates to a C 3 -C 6 -cycloalkyl group as defined above which is bound to the remainder of the molecule via a carbonyl group (CO), such as in cyclopropylcarbonyl, cyclobutylcarbonyl, cyclopentylcarbonyl and cyclo hexylcarbonyl.

Ci-C6-Alkoxycarbonyl refers to a straight-chain or branched alkoxy group having from 1 to 6, especially 1 to 4 carbon atoms (= Ci-C4-alkoxycarbonyl), in particular 1 to 3 carbon atoms (= Ci-C3-alkoxycarbonyl), which is bound to the remainder of the molecule via a carbonyl group (CO), such as in methoxycarbonyl, ethoxycarbonyl, propy- loxycarbonyl, and isopropyloxy carbonyl.

Fluorinated Ci-C6-alkoxycarbonyl refers to a straight-chain or branched fluorinated alkoxy group having from 1 to 6, especially 1 to 4 carbon atoms (=fluorinated C 1 -C4- alkoxy carbonyl), in particular 1 to 3 carbon atoms (=fluorinated Ci-C3-alkoxycarbonyl) as defined above, which is bound to the remainder of the molecule via a carbonyl group (CO). Examples include trifluoromethoxycarbonyl, 2,2,2-trifluoroethoxycarbonyl and the like.

Ci-C4-Alkylcarbonyloxy refers to a straight-chain or branched alkyl group having from 1 to 4 carbon atoms, which is bound to the remainder of the molecule via a car- bonyloxy group [C(0)-0-], such as in acet(yl)oxy, propionyloxy, isopropylcarbonyloxy, butylcarbonyloxy, sec-butylcarbonyloxy, isobutylcarbonyloxy, and tert- butylcarbonyloxy. Ci-C6-Alkylcarbonyloxy is a straight-chain or branched alkyl group having from 1 to 6 carbon atoms, which is bound to the remainder of the molecule via a carbonyloxy group [C(0-0-]. Examples include, apart those listed above for C1-C4- alkylcarbonyloxy, pentylcarbonyloxy, hexylcarbonyloxy and the constitutional isomers thereof.

Fluorinated Ci-C4-alkylcarbonyloxy refers to a straight-chain or branched fluori- nated alkyl group having from 1 to 4 carbon atoms as defined above, which is bound to the remainder of the molecule via a carbonyloxy group [C(0)-0-]. Fluorinated Ci-C 6 - alkylcarbonyloxy is a straight-chain or branched fluorinated alkyl group having from 1 to 6 carbon atoms as defined above, which is bound to the remainder of the molecule via a carbonyloxy group [C(0)-0-]. Examples include trifluoromethylcarbonyloxy, 2,2,2-trifluoroethylcarbonyloxy and the like.

Phenyl-Ci-C2-alkyl is a phenyl group bound to the remainder of the molecule via a Ci-C2-alkyl group. Examples are benzyl, 1-phenylethyl and 2-phenylethyl

(phenethyl).

The term "3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated heterocyclic ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO and S0 2 and optionally also 1 or 2 C=0 and/or C=S groups as ring members" denotes a 3-, 4-, 5-, 6-, 7- or 8-membered, preferably a 3-, 4-, 5-, 6- or 7-membered saturated, partially unsaturated or maximum unsaturated heteromonocyclic ring containing 1, 2, 3 or 4 (preferably 1, 2 or 3) heteroatoms or heteroatom groups selected from N, O, S, SO and S0 2 and optionally also 1 or 2 C=0 and/or C=S groups as ring members.

Unsaturated rings contain at least one C-C and/or C-N and/or N-N double bond(s). Maximally unsaturated rings contain as many conjugated C-C and/or C-N and/or N-N double bonds as allowed by the ring size. Maximally unsaturated 5- or 6- membered heterocyclic rings are aromatic. 7- and 8-membered rings cannot be aromatic. They are homoaromatic (7-membered ring, 3 double bonds) or have 4 double bonds (8-membered ring). Partially unsaturated rings contain less than the maximum number of C-C and/or C-N and/or N-N double bond(s) allowed by the ring size. The heterocyclic ring may be attached to the remainder of the molecule via a carbon ring member or via a nitrogen ring member. As a matter of course, the heterocyclic ring contains at least one carbon ring atom. If the ring contains more than one O ring atom, these are not adjacent.

In heterocyclic rings containing N as a ring member this nitrogen atom may either be present as tertiary N formally bound by a double and a single bond (like in pyridyl) or may be present as secondary NH (if N is not part of a ring double bond). In case that N is actually present as NH, and the ring is substituted, the substituent may either be bound to a carbon ring atom or to such a secondary nitrogen ring atom. In case that N is actually present as NH, the ring may either be bound to the remainder of the molecule via a carbon ring atom or such a secondary nitrogen ring atom.

Examples of a 3-, 4-, 5-, 6- or 7-membered saturated heterocyclic ring include: Oxiranyl, thiiranyl, aziridinyl, oxetanyl, thietanyl, azetidinyl, tetrahydrofuran-2-yl, tet- rahydrofuran-3-yl, tetrahydrofuran-2-on-3-yl, tetrahydrofuran-2-on-4-yl, tetrahydrofu- ran-2-on-5-yl, tetrahydrofuran-2-thion-3-yl, tetrahydrofuran-2-thion-4-yl, tetrahydrofu- ran-2-thion-5-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, tetrahydrothien-2-on-3-yl, tetrahydrothien-2-on-4-yl, tetrahydrothien-2-on-5-yl, tetrahydrothien-2-thion-3-yl, tet- rahydrothien-2-thion-4-yl, tetrahydrothien-2-thion-5-yl, pyrrolidin-l-yl, pyrrolidine-2- on-l-yl, pyrrolidine-2,5-dion-l-yl, pyrrolidine-2-thion-l-yl, pyrrolidin-2-yl, pyrrolidin- 3-yl, pyrrolidine-2-on-3-yl, pyrrolidine-2-on-4-yl, pyrrolidine-2-on-5-yl, pyrrolidine- 2,5-dion-3-yl, pyrrolidine-2-thion-3-yl, pyrrolidine-2-thion-4-yl, pyrrolidine-2-thion-5- yl, pyrazolidin-l-yl, pyrazolidin-3-yl, pyrazolidin-4-yl, pyrazolidin-5-yl, imidazolidin- 1-yl, imidazolidin-2-on-l-yl, imidazolidin-2-thion-l-yl, imidazolidin-2-yl, imidazolidin- 4-yl, imidazolidin-2-on-4-yl, imidazolidin-2-thion-4-yl, oxazolidin-2-yl, oxazolidin-3- yl, oxazolidin-4-yl, oxazolidin-5-yl, isoxazolidin-2-yl, isoxazolidin-3-yl, isoxazolidin-4- yl, isoxazolidin-5-yl, thiazolidin-2-yl, thiazolidin-3-yl, thiazolidin-4-yl, thiazolidin-5-yl, isothiazolidin-2-yl, isothiazolidin-3-yl, isothiazolidin-4-yl, isothiazolidin-5-yl, 1 ,2,4-oxadiazolidin-3-yl, 1 ,2,4-oxadiazolidin-5-yl, 1 ,2,4-thiadiazolidin-3-yl,

1 ,2,4-thiadiazolidin-5-yl, 1 ,2,4-triazolidin-3-yl, 1 ,3,4-oxadiazolidin-2-yl,

1.3.4- thiadiazolidin-2-yl, 1,3,4-triazolidin-l-yl, l ,3,4-triazolidin-2-yl, 2- tetrahydropyranyl, 4-tetrahydropyranyl, l,3-dioxan-5-yl, 1 ,4-dioxan-2-yl, piperidin-1- yl, piperidin-2-on- 1 -yl, piperidin-2,5-dion- 1 -yl, piperidine-2-thion- 1 -yl, piperidin-2-yl, piperidin-3-yl, piperidin-2-on-3-yl, piperidin-2,5-dion-3-yl, piperidin-2-thion-3-yl, pi- peridin-4-yl, hexahydropyridazin-3-yl, hexahydropyridazin-4-yl, hexahydropyrimidin-

2- yl, hexahydropyrimidin-4-yl, hexahydropyrimidin-5-yl, piperazin-l-yl, piperazin-2-yl,

1.3.5- hexahydrotriazin-l-yl, l,3,5-hexahydrotriazin-2-yl and l,2,4-hexahydrotriazin-3- yl, morpholin-2-yl, morpholin-3-yl, morpholin-4-yl, thiomorpholin-2-yl, thiomorpholin-

3- yl, thiomorpholin-4-yl, l-oxothiomorpholin-2-yl, l-oxothiomorpholin-3-yl, 1- oxothiomorpholin-4-yl, l,l-dioxothiomorpholin-2-yl, l,l-dioxothiomorpholin-3-yl, 1,1- dioxothiomorpholin-4-yl, azepan-1-, -2-, -3- or -4-yl, oxepan-2-, -3-, -4- or -5-yl, hexa- hydro-l,3-diazepinyl, hexahydro-l,4-diazepinyl, hexahydro-l,3-oxazepinyl, hexahydro- 1 ,4-oxazepinyl, hexahydro- 1 ,3-dioxepinyl, hexahydro- 1 ,4-dioxepinyl and the like.

Examples of a 3-, 4-, 5-, 6-, 7- or 8-membered saturated heterocyclic ring further include oxocane, thiocane, azocane, [l,3]diazocane, [l,4]diazocane, [l,5]diazocane, [l,5]oxazocane and the like.

Examples of a 3-, 4-, 5-, 6- or 7-membered partially unsaturated heterocyclic ring include: 2,3-dihydrofur-2-yl, 2,3-dihydrofur-3-yl, 2,4-dihydrofur-2-yl, 2,4-dihydrofur-3- yl, 2,3-dihydrothien-2-yl, 2,3-dihydrothien-3-yl, 2,4-dihydrothien-2-yl, 2,4- dihydrothien-3-yl, 2-pyrrolin-2-yl, 2-pyrrolin-3-yl, 3-pyrrolin-2-yl, 3-pyrrolin-3-yl, 2- isoxazolin-3-yl, 3-isoxazolin-3-yl, 4-isoxazolin-3-yl, 2-isoxazolin-4-yl, 3-isoxazolin-4- yl, 4-isoxazolin-4-yl, 2-isoxazolin-5-yl, 3-isoxazolin-5-yl, 4-isoxazolin-5-yl, 2- isothiazolin-3-yl, 3-isothiazolin-3-yl, 4-isothiazolin-3-yl, 2-isothiazolin-4-yl, 3- isothiazolin-4-yl, 4-isothiazolin-4-yl, 2-isothiazolin-5-yl, 3-isothiazolin-5-yl, 4- isothiazolin-5-yl, 2,3-dihydropyrazol-l-yl, 2,3-dihydropyrazol-2-yl, 2,3-dihydropyrazol- 3-yl, 2,3-dihydropyrazol-4-yl, 2,3-dihydropyrazol-5-yl, 3,4-dihydropyrazol-l-yl, 3,4- dihydropyrazol-3-yl, 3,4-dihydropyrazol-4-yl, 3,4-dihydropyrazol-5-yl, 4,5- dihydropyrazol-l-yl, 4,5-dihydropyrazol-3-yl, 4,5-dihydropyrazol-4-yl, 4,5- dihydropyrazol-5-yl, 2,3-dihydrooxazol-2-yl, 2,3-dihydrooxazol-3-yl, 2,3- dihydrooxazol-4-yl, 2,3-dihydrooxazol-5-yl, 3,4-dihydrooxazol-2-yl, 3,4- dihydrooxazol-3-yl, 3,4-dihydrooxazol-4-yl, 3,4-dihydrooxazol-5-yl, 3,4- dihydrooxazol-2-yl, 3,4-dihydrooxazol-3-yl, 3,4-dihydrooxazol-4-yl, 2-, 3-, 4-, 5- or 6- di- or tetrahydropyridinyl, 3-di- or tetrahydropyridazinyl, 4-di- or tetrahydropyridazinyl, 2-di- or tetrahydropyrimidinyl, 4-di- or tetrahydropyrimidinyl, 5-di- or tetrahydropyrim- idinyl, di- or tetrahydropyrazinyl, 1,3, 5-di- or tetrahydrotriazin-2-yl, 1,2,4-di- or tetra- hydrotriazin-3-yl, 2,3,4,5-tetrahydro[lH]azepin-l-, -2-, -3-, -4-, -5-, -6- or -7-yl, 3,4,5, 6-tetrahydro[2H]azepin-2-, -3-, -4-, -5-, -6- or -7-yl, 2,3,4,7-tetrahydro[lH]azepin- 1-, -2-, -3-, -4-, -5-, -6- or -7-yl, 2,3,6,7-tetrahydro[lH]azepin-l-, -2-, -3-, -4-, -5-, -6- or -7-yl, tetrahydrooxepinyl, such as 2,3,4, 5-tetrahydro[lH]oxepin-2-, -3-, -4-, -5-, -6- or -7-yl, 2,3,4,7-tetrahydro[lH]oxepin-2-, -3-, -4-, -5-, -6- or -7-yl, 2,3,6,7- tetrahydro[lH]oxepin-2-, -3-, -4-, -5-, -6- or -7-yl, tetrahydro-l,3-diazepinyl, tetrahy- dro-l,4-diazepinyl, tetrahydro-l,3-oxazepinyl, tetrahydro-l,4-oxazepinyl, tetrahydro- 1,3-dioxepinyl and tetrahydro-l,4-dioxepinyl.

Examples of a 3-, 4-, 5-, 6-, 7- or 8-membered partially unsaturated heterocyclic ring further include 1,2,3, 4,5, 6-hexahydroazocine, 2,3,4,5,6,7-hexahydroazocine,

1, 2,3,4,5, 8-hexahydroazocine, 1,2,3,4,7,8-hexahydroazocine, 1,2,3,4,5,6-hexahydro- [l,5]diazocine,l,2, 3,4,7, 8-hexahydro-[l,5]diazocine and the like.

Examples of a 3-, 4-, 5-, 6- or 7-membered maximally unsaturated (including aromatic) heterocyclic ring include 5- or 6-membered heteroaromatic rings, such as 2- furyl, 3-furyl, 2-thienyl, 3-thienyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 1-pyrazolyl, 3- pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-thiazolyl, 4- thiazolyl, 5-thiazolyl, 1-imidazolyl, 2-imidazolyl, 4-imidazolyl, 1,3,4-triazol-l-yl, 1,3,4- triazol-2-yl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, l-oxopyridin-2-yl, l-oxopyridin-3-yl, l-oxopyridin-4-yl, 3-pyridazinyl, 4-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5- pyrimidinyl and 2-pyrazinyl, and also homoaromatic radicals, such as lH-azepine, 1H- [l,3]-diazepine and lH-[l,4]-diazepine.

Examples of a 3-, 4-, 5-, 6-, 7- or 8-membered maximally unsaturated heterocyclic ring further include [l,3]diazocine, [l,5]diazocine and [l,5]diazocine.

A 3-, 4-, 5-, 6-, 7- or 8-membered saturated, partially unsaturated or maximally unsaturated ring containing 1, 2, 3 or 4 heteroatoms or heteroatom groups independently selected from N, O, S, NO, SO, S0 2 , C=0 and C=S as ring members is either saturated, partially unsaturated and carbocyclic (if it contains only C=0 and/or C=S as heteroatom group and no further heteroatoms or heteroatom groups) or saturated, partially unsaturated or maximally unsaturated heterocyclic. Examples are, in addition to the heterocyclic rings mentioned above, carbocyclic rings, such as cyclopropanonyl, cyclobuta- nonyl, cyclopentanonyl, cyclohexanonyl, cyclohexandionyl, cycloheptanonyl, cyclooc- tanonyl, cyclopropanthionyl, cyclobutanthionyl, cyclopentanthionyl, cyclohexanthionyl, cyclohexandithionyl, cycloheptanthionyl, cyclooctanthionyl, cyclopropenonyl, cyclo- pentenonyl, cyclohexenonyl and the like.

The remarks made above and in the following with respect to preferred aspects of the invention, e.g. to preferred meanings of the variables R 1 , R 2 , R 3a , R 3b , R 4a , R 4b , R 5a , R 5b , R 6 , R 7 , R 8 , R 9 , R 10 , R l la , R l lb , a and b of compounds I, to preferred compounds I and to preferred embodiments of the method or the use according to the invention, apply in each case on their own or in particular to combinations thereof.

In a preferred embodiment, R 1 is selected from hydrogen and Ci-C6-alkyl, in par- ticular from hydrogen and methyl, and is specifically hydrogen.

In a preferred embodiment, R 2 is selected from cyano, nitro, Ci-C 6 -alkyl, fluori- nated Ci-C 6 -alkyl, Ci-C 6 -alkoxy-Ci-C 4 -alkyl and fluorinated Ci-C 6 -alkoxy-Ci-C 4 -alkyl, more preferably from Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, Ci-C 4 -alkoxy-Ci-C 4 -alkyl and fluorinated Ci-C 4 -alkoxy-Ci-C 4 -alkyl, even more preferably from Ci-C 6 -alkyl and fluorinated Ci-C 6 -alkyl, in particular from methyl, ethyl, propyl, isopropyl and CF 3 , more particularly from methyl, ethyl and CF 3 , and is specifically methyl or CF 3 .

In a preferred embodiment, R 3a and R 3b , independently of each other, are selected from hydrogen, cyano, nitro, Ci-C 6 -alkyl and fluorinated Ci-C 6 -alkyl. More preferably, R 3a is selected from hydrogen, cyano, nitro, Ci-C 6 -alkyl and fluorinated Ci-C 6 -alkyl, and R 3b is hydrogen. Even more preferably, R 3a is selected from hydrogen and methyl and R 3b is hydrogen. In particular, both R 3a and R 3b are hydrogen.

In a preferred embodiment, R 4a and R 4b , independently of each other, are selected from hydrogen, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, Ci-C4-alkoxy-Ci-C4-alkyl and fluorinated Ci-C4-alkoxy-Ci-C4-alkyl, or form together a group =0, or form together a group -(CH 2 ) m -, where m is 2, 3 or 4, in particular 2 or 3, especially 2, thus forming together a spiro-bound ring. More preferably, R 4a is selected from hydrogen, Ci-C 6 - alkyl, fluorinated Ci-C6-alkyl, Ci-C4-alkoxy-Ci-C4-alkyl and fluorinated Ci-C4-alkoxy- Ci-C4-alkyl and R 4b is hydrogen, or R 4a and R 4b are methyl, or R 4a and R 4b form together a group -(CH 2 ) m -, where m is 2, 3 or 4, in particular 2 or 3, especially 2, thus forming together a spiro-bound ring. In a particular embodiment, they are hydrogen.

In a preferred embodiment, R 5a is selected from hydrogen, halogen, Ci-C6-alkyl, fluorinated Ci-C6-alkyl and Ci-C6-alkoxy-Ci-C4-alkyl and R 5b is hydrogen. In particular, R 5a is hydrogen or methyl, specifically hydrogen, and R 5b is hydrogen.

Preferably, each R 8 as a substituent on R 6 is independently selected from the group consisting of halogen, Ci-C 6 -alkoxy and fluorinated Ci-C 6 -alkoxy. In particular, R 8 is halogen, especially fluorine.

R 6 is thus preferably Ci-C 4 -haloalkyl, more preferably fluorinated Ci-C 4 -alkyl, in particular fluorinated Ci-C 2 -alkyl, such as CH 2 F, CHF 2 , CF 3 and CH 2 CF 3 , and specifically fluorinated methyl; very specifically CHF 2 or CF 3 .

Alternatively, R 6 is preferably selected from Ci-C6-alkoxy-Ci-C4-alkyl and fluorinated Ci-C6-alkoxy-Ci-C4-alkyl, and is in particular fluorinated Ci-C6-alkoxy-Ci-C4- alkyl. In another particular embodiment, R 6 is Ci-C6-alkoxy-Ci-C4-alkyl, specifically Ci-C4-alkoxy-methyl and very specifically Ci-C 2 -alkoxy-methyl.

In another embodiment, R 6 is Ci-C4-alkyl which carries a hydroxy 1 substituent.

Among Ci-C6-alkoxy-Ci-C4-alkyl, Ci-C4-alkoxy-Ci-C4-alkyl, fluorinated Ci-C 6 - alkoxy-Ci-C 4 -alkyl and fluorinated Ci-C 4 -alkoxy-Ci-C 4 -alkyl radicals R 2 , R 4a , R 4b , R 5a and R 6 preference is given to CH 2 OCH 3 , CH 2 CH 2 OCH 3 , CH 2 OCH 2 CH 3 ,

CH 2 CH 2 OCH 2 CH 3 , CH 2 OCF 3 , CH 2 CH 2 OCF 3 , CH 2 OCHF 2 and CH 2 CH 2 OCHF 2 .

In a preferred embodiment, each R 7 is independently selected from the group consisting of halogen, cyano, nitro, hydroxyl, Ci-C6-alkyl, fluorinated Ci-C6-alkyl, C 2 -C 6 - alkenyl, fluorinated C 2 -C6-alkenyl, C 2 -C6-alkynyl, fluorinated C 2 -C6-alkynyl, C 3 -Cs- cycloalkyl, fluorinated C 3 -Cs-cycloalkyl, Ci-C6-alkoxy and fluorinated Ci-C6-alkoxy. More preferably, each R 7 is independently selected from halogen, Ci-C4-alkyl, fluorinated Ci-C4-alkyl, Ci-C4-alkoxy and fluorinated Ci-C4-alkoxy, and is in particular fluorine.

In a preferred embodiment, a is 0 or 1. If a is 1 , R 2 is preferably bound in β- position to the nitrogen ring atom carrying R 1 .

In a preferred embodiment, b is 0 or 1 and specifically 0. If not specified otherwise in a specific context, R 9 , R 10 , R l la and R l lb have following preferred meanings:

Preferably each R 9 is independently selected from the group consisting of hydrogen, hydroxyl, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, Ci-C 6 -hydroxyalkyl, C3-C 6 - cycloalkyl, fluorinated C3-C 6 -cycloalkyl, Ci-C 6 -alkoxy, fluorinated Ci-C 6 -alkoxy, and - NR l la R l lb , where R l la and R l lb have one of the above general or, in particular, one of the below preferred meanings. In particular, each R 9 is independently selected from the group consisting of hydrogen, Ci-C 4 -alkyl, fluorinated Ci-C 2 -alkyl, C 1 -C 4 - hydroxyalkyl, Ci-C 4 -alkoxy, fluorinated Ci-C 2 -alkoxy, and -NR l la R l lb , where R l la and R l lb have one of the above general or, in particular, one of the below preferred meanings.

Preferably, each R 10 is independently selected from the group consisting of halogen, cyano, Ci-C 4 -alkyl, fluorinated Ci-C 4 -alkyl, Ci-C 4 -alkoxy and fluorinated C 1 -C4- alkoxy, more preferably from F, CI, cyano, CH 3 , CF 3 , OCH 3 and OCF 3 , and in particu- lar from F, CH 3 , CF 3 , OCH 3 and OCF 3 .

Preferably, R l la and R l lb , independently of each other and independently of each occurrence, are selected from the group consisting of hydrogen, Ci-C 6 -alkyl, fluorinated Ci-C 6 -alkyl, Ci-C 6 -alkylcarbonyl and fluorinated Ci-C 6 -alkylcarbonyl.

In a particular embodiment, the compounds of the invention are compounds c formula 1.1

wherein R 2 , R 6 , R 7 and b have one of the above general or, in particular, one of the above preferred meanings, and a is 0 or 1.

In a more particular embodiment, the compounds of the invention are compounds of formula 1.1.1

wherein

R 7a is H, CI, F or methyl, in particular H or F, specifically H;

a is 0 or 1 ; and

R 2 , R 6 , R 7 and b have one of the above general or, in particular, one of the above preferred meanings.

In compounds 1.1.1 (b-1) is preferably 0.

In compounds 1.1 and 1.1.1 R 2 is specifically methyl.

Examples of preferred compounds are compounds of the following formulae Ia. l to Ia.36, where the variables have one of the general or preferred meanings given above Examples of preferred compounds are the individual compounds compiled in the tables 1 to 9360 below. Moreover, the meanings mentioned below for the individual variables in the tables are per se, independently of the combination in which they are mentioned, a particularly preferred embodiment of the substituents in question.

Ia.1 la.2 la.3

la.13 la.14 la.15

la.25 la.26 la.27

la.34 la.35 la.36

Table 1

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 F and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 2

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 3

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Table 4

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CH 2 CHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 5

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CH 2 CF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 6

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CF 2 CH 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 7

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CF 2 CHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 8

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CF 2 CF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Ta- ble A.

Table 9

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 OH and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 10

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CF 2 OH and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 11

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CH 2 OCH 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Table 12

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 OCH 2 CH 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 13

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCH 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 14

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCH 2 CH 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 15

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 OCHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 16

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 OCF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 17

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 18

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Table 19

Compounds of the formula la.1 in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCH 2 CHF 2 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Table 20

Compounds of the formula Ia.l in which R 4a is H, R 4b is H, R 6 is CH 2 CH 2 OCH 2 CF 3 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 21 to 40

Compounds of the formula la.1 in which R 4a is methyl, R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 41 to 60

Compounds of the formula la.1 in which R 4a is ethyl, R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 61 to 80

Compounds of the formula Ia. l in which R 4a is CH 2 OCH 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 81 to 100

Compounds of the formula Ia. l in which R 4a is CH 2 OCH 2 CH 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 101 to 120

Compounds of the formula Ia. l in which R 4a is CH 2 CH 2 OCH 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 121 to 140

Compounds of the formula Ia. l in which R 4a is CH 2 CH 2 OCH 2 CH 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 141 to 160

Compounds of the formula la.1 in which R 4a is CH 2 OCHF 2 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Tables 161 to 180

Compounds of the formula Ia. l in which R 4a is CH 2 OCF 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 181 to 200

Compounds of the formula Ia. l in which R 4a is CH 2 CH 2 OCHF 2 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 201 to 220

Compounds of the formula la.1 in which R 4a is CH 2 CH 2 OCF 3 , R 4b is H, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 221 to 240

Compounds of the formula la.1 in which R 4a is methyl, R 4b is methyl, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 241 to 260

Compounds of the formula la.1 in which R 4a and R 4b form together -CH 2 -CH 2 -, R 6 is as defined in tables 1 to 20 and the combination of R 5a , R 5b and R 7a for a compound corre- sponds in each case to one row of Table A.

Tables 261 to 520

Compounds of the formula Ia.2 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 521 to 780

Compounds of the formula la.3 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 781 to 1040

Compounds of the formula Ia.4 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Tables 1041 to 1300

Compounds of the formula la.5 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 1301 to 1560

Compounds of the formula la.6 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 1561 to 1820

Compounds of the formula la.7 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 1821 to 2080

Compounds of the formula la.8 in which the combination of R 4a , R 4b and R 6 is as de- fined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 2081 to 2340

Compounds of the formula la.9 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corre- sponds in each case to one row of Table A.

Tables 2341 to 2600

Compounds of the formula la.10 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 2601 to 2860

Compounds of the formula la.11 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 2861 to 3120

Compounds of the formula la.12 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Tables 3121 to 3380

Compounds of the formula la.13 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 3381 to 3641

Compounds of the formula la.14 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 3641 to 3900

Compounds of the formula la.15 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 3901 to 4160

Compounds of the formula la.16 in which the combination of R 4a , R 4b and R 6 is as de- fined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 4161 to 4420

Compounds of the formula la.17 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corre- sponds in each case to one row of Table A.

Tables 4421 to 4680

Compounds of the formula la.18 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 4681 to 4940

Compounds of the formula la.19 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 4941 to 5200

Compounds of the formula Ia.20 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Tables 5201 to 5460

Compounds of the formula Ia.21 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 5461 to 5720

Compounds of the formula Ia.22 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 5721 to 5980

Compounds of the formula Ia.23 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 5981 to 6240

Compounds of the formula Ia.24 in which the combination of R 4a , R 4b and R 6 is as de- fined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 6241 to 6500

Compounds of the formula Ia.25 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corre- sponds in each case to one row of Table A.

Tables 6501 to 6760

Compounds of the formula Ia.26 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 6761 to 7020

Compounds of the formula Ia.27 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 7021 to 7280

Compounds of the formula Ia.28 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Tables 7281 to 7540

Compounds of the formula Ia.29 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 7541 to 7800

Compounds of the formula la.30 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 7801 to 8060

Compounds of the formula la.31 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 8061 to 8320

Compounds of the formula Ia.32 in which the combination of R 4a , R 4b and R 6 is as de- fined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 8321 to 8580

Compounds of the formula la.33 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corre- sponds in each case to one row of Table A.

Tables 8581 to 8840

Compounds of the formula Ia.34 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 8841 to 9100

Compounds of the formula la.35 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A.

Tables 9101 to 9360

Compounds of the formula la.36 in which the combination of R 4a , R 4b and R 6 is as defined in tables 1 to 260 and the combination of R 5a , R 5b and R 7a for a compound corresponds in each case to one row of Table A. Table A

Among the above compounds, preference is given to compounds Ia. l and la.10.

In a specific embodiment, the invention relates to compounds I selected from the com- pounds of the examples, either in form of free bases or of any pharmaceutically acceptable salt thereof or a steroisomer, the racemate or any mixture of stereoisomers thereof.

The compounds of the present invention can be prepared by using routine tech- niques familiar to a skilled person. In particular, the compounds of the formula I can be prepared according to the following schemes, wherein the variables, if not stated otherwise, are as defined above.

Compounds of formula I wherein R 4b and R 5b are H (= compounds Γ) can be synthe- sized as described in scheme 1 below. The readily available quinoline 1 in which X is a leaving group, such as CI, Br I or triflate, is reacted with a suitable boron compound of R 6 , such as R 6 -boronic acid (R 6 -B(OH) 2 ) or the potassium (trifluoro)borate of R 6 in a Suzuki coupling reaction to 2. The reaction is carried out in the presence of a Pd catalyst, such as Pd(OAc) 2 , in general in the presence of a phosphine ligand (e.g. tricyclo- hexylphosphine; di(adamantan-l-yl)(butyl)-phosphine etc.), or tetrakis-

(triphenylphosphine)palladium(O) and the like. Quinoline 2 is then N-alkylated with a suitable protective group, such as benzyl, e.g. by reaction with benzyl bromide, to 3. Reduction of the quinolinium compound 3 with a suitable reduction agent, such as sodium boro hydride and H 2 /Raney nickel, yields the 1, 2,3, 4-tetrahydroquino line 4, which is then deprotected to 5. Reaction with the 2-halogenoacetamide 6, wherein Y is CI, Br or I, affords the acetamide 7, the keto group of which is reduced with common reduction agents, like borane, borane-tetrahydrofurane-complex, borane-dimethylsulfide-complex or borohydrides such as sodium borohydride or LAH (lithium aluminium hydride) or DIBAL-H (diisobutyl aluminium hydride), to 8. Cyclization with formalde- hyde/aldehyde/ketone 9 (C(0)R 3a R 3b = formaldehyde if R 3a and R 3b are hydrogen; another aldehyde if R 3a is hydrogen and R 3b is (fluorinated) alkyl, hydroxyalkyl, (fluori- nated) alkenyl, (fluorinated) alkynyl, (fluorinated) cycloalkyl, phenyl, phenyl-alkyl or a heterocyclic ring; a ketone if R 3a and R 3b are (fluorinated) alkyl, hydroxyalkyl, (fluori- nated) alkenyl, (fluorinated) alkynyl, (fluorinated) cycloalkyl, phenyl, phenyl-alkyl or a heterocyclic ring), generally in the presence of a strong acid, such as trifluoroacetic acid, yields Γ.

Alternatively, 2 can be reacted with the 2-halogenoacetamide 6 to 10, which is then reduced to 7.

Scheme 1

tively be synthesized as described in scheme 2 below. The boronic acid 11 and the α,β- unsaturated ester 12 are reacted in a 1,4-addition in the presence of a Rhodium catalyst, e.g. [RhOH(COD)] 2 or Rh(acac)(CO) 2 , if desired in the presence of a phosphine ligand, such as l,4-bis(diphenylphosphino)butane (dppb), followed by lactam formation. The lactam 13 is then reduced with common reduction agents, like borane, borane- tetrahydrofurane-complex, borane-dimethylsulfide-complex or borohydrides such as sodium borohydride, to the tetrahydroquinoline 14. Like in the reaction sequence of scheme 1, reaction with the 2-halogenoacetamide 6, wherein Y is CI, Br or I, affords the acetamide 15, the keto group of which is reduced with common reduction agents, like borane, borane-tetrahydrofurane-complex, borane-dimethylsulfide-complex or borohy- drides such as sodium borohydride or LAH (lithium aluminium hydride) or DIBAL-H (diisobutyl aluminium hydride), to 16. Cyclization with formaldehyde / aldehyde / ketone 9, generally in the presence of a strong acid, such as trifluoroacetic acid, yields I".

Scheme 2

For obtaining compounds I wherein R a and R form together =0, the lactam 13 is not reduced, but is directly reacted with the 2-halogenoacetamide 6 to 17, followed by reduction of the CO group in the amide to 18 and finally cyclisation, as shown in scheme 3 below. Alternatively, compounds Γ" can be obtained as described below scheme 4.

Scheme 3

Compounds of formula I wherein R a , R and R are H (= compounds I") can alternatively be synthesized as described in scheme 4 below. The protected tetrahydro- benzodiazepine 19, wherein Z is a hydrogen or a halogen atom, such as CI, Br or I and PG is a common protective group, such as a carbamate, especially boc, is acylated with the acrylic acid derivative 20, wherein LG is an appropriate leaving group, such as CI or an anhydride or a chloroformate, in the presence of a base, such as triethylamine or Hunig's base, in an organic solvent, such as ether or methylene chloride. Reaction of 21 with a Lewis acid or a Bronstedt acid HA or irradiation with a suitable wavelength commonly derived from a mercury lamp in an adequate solvent, such as acetone or toluene, in a common photoreactor yields cyclization to 22. Reduction of the carbonyl group with common reduction agents like borohydrides such as sodium borohydride or borane-tetrahydrofurane-complex yields 23, which is deprotected using suitable rea- gents such as strong bases or acids to I", wherein R 4a and R 4b and R 5b are H. Compounds I" " wherein R 4a and R 4b form together =0 can be obtained by skipping the reduction step to 23 and deprotecting 22.

Alternatively, compound 22 can be directly synthesized by reacting the tetrahy- drobenzodiazepine 19, wherein Z is -B(OH) 2 with the acrylic acid derivative 20 in a 1,4-addition in the presence of a Rhodium catalyst, e.g. [RhOH(COD)] 2 or

Rh(acac)(CO) 2 , if desired in the presence of a phosphine ligand, such as 1,4- bis(diphenylphosphino)butane (dppb), followed by lactam formation to 22. This is then further reacted as described above. Scheme 4

If compound 22 is deprotected, this yields compound Γ " as well as other compounds I in which (R 2 ) a is not necessarily bound to the same position as in compounds Γ".

If desired, substituents R 1 different from hydrogen can be introduced for example via alkylation under typical conditions such as stirring in an appropriate solvent in the presence of an alkylhalide and a base or via other common substitution reactions, or via reductive amination using a suitable aldehyde or ketone in the presence of reduction agent, such as borohydrides, e.g. triacetoxyborohydride, sodium cyanoborohydride or sodium borohydride. -C(=0)R 9 as radical R 1 can be introduced via amidation under typical amidation conditions, e.g. via reaction with a compound X-C(=0)R 9 , where X is OH or a halogen atom, under heating and removal of reaction water or using a coupling reagent, such as DCC (dicyclohexylcarbodiimide), DIC (diisopropylcarbodiimide), HATU (0-(7-azabenzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), HBTU ((0-benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), HCTU (lH-benzotriazolium-l-[bis(dimethylamino)methylene]-5-chloro tetra- fluoroborate), BOP ((benzotriazol-l-yloxy)-tris(dimethylamino)phosphonium hexafluorophosphate), Py-BOP ((benzotriazol-l-yloxy)-tripyrrolidinphosphonium hex- afluorophosphate) or Py-BrOP (bromotripyrrolidinphosphonium hexafluorophosphate).. Compounds I wherein R 5b is different from H can be prepared, for example, by reacting compound 22 with a compound LG-R 5b in the presence of a base, wherein LG is an appropriate leaving group, such as CI or Br.

Alternatively to the method depicted in scheme 4, compounds I wherein R a , R and R 5b are H (= compounds I") can be synthesized as described in scheme 5 below. Readily available anilines 24 are derivatized with carbonyl moieties 25 by acylation procedures employing appropriate leaving groups LG, such as chlorides or anhydrides, in the presence of a base such as triethylamine or Hunig's base, in an organic solvent, such as diethyl ether or methylene chloride to yield 26. Cyclization products 13 are received by irradiation with a suitable wavelength commonly derived from a mercury lamp in an adequate solvent such as acetone or toluene in a common photoreactor known to those skilled in the art. These are further reacted as depicted in schemes 2 and

Scheme 5

Compounds I wherein R a and R are not H or do not form together a group =0 can be prepared by standard derivatization methods of compounds wherein R 4a and R 4b form together a group =0. For instance, compounds wherein R 4a and R 4b form together a group =S may be prepared by reaction with a sulfurization agent, such as Lawesson's reagent or P2S5. Alkyl and related groups as radicals R 4a and R 4b may be introduced via Grignard reduction. Amino and related groups may be introduced via reductive animation. Hydroxyl group R 4a or R 4b may be introduced by reducing the carbonyl group. This may be alkylated to yield alkoxy and related groups R 4a and R 4b or substituted by diverse groups. If not otherwise indicated, the above-described reactions are generally carried out in a solvent at temperatures between room temperature and the boiling temperature of the solvent employed. Alternatively, the activation energy which is required for the reaction can be introduced into the reaction mixture using microwaves, something which has proved to be of value, in particular, in the case of the reactions catalyzed by transition metals (with regard to reactions using microwaves, see Tetrahedron 2001, 57, p. 9199 ff. p. 9225 ff. and also, in a general manner, "Microwaves in Organic Synthesis", Andre Loupy (Ed.), Wiley- VCH 2002.

The acid addition salts of compounds I are prepared in a customary manner by mixing the free base with a corresponding acid, where appropriate in solution in an organic solvent, for example a lower alcohol, such as methanol, ethanol or propanol, an ether, such as methyl tert-butyl ether or diisopropyl ether, a ketone, such as acetone or methyl ethyl ketone, or an ester, such as ethyl acetate.

Routine experimentations, including appropriate manipulation of the reaction conditions, reagents and sequence of the synthetic route, protection of any chemical functionality that may not be compatible with the reaction conditions, and deprotection at a suitable point in the reaction sequence of the preparation methods are within routine techniques.

Suitable protecting groups and the methods for protecting and deprotecting dif- ferent substituents using such suitable protecting groups are well known to those skilled in the art; examples of which may be found in T. Greene and P. Wuts, Protective Groups in Organic Synthesis (3 rd ed.), John Wiley & Sons, NY (1999), which is herein incorporated by reference in its entirety. Synthesis of the compounds of the invention may be accomplished by methods analogous to those described in the synthetic schemes described hereinabove and in specific examples.

Starting materials, if not commercially available, may be prepared by procedures selected from standard organic chemical techniques, techniques that are analogous to the synthesis of known, structurally similar compounds, or techniques that are analogous to the above described schemes or the procedures described in the synthetic exam- pies section.

When an optically active form of a compound of the invention is required, it may be obtained by carrying out one of the procedures described herein using an opti- cally active starting material (prepared, for example, by asymmetric induction of a suitable reaction step), or by resolution of a mixture of the stereoisomers of the compound or intermediates using a standard procedure (such as chromatographic separation, re- crystallization or enzymatic resolution).

Similarly, when a pure geometric isomer of a compound of the invention is required, it may be obtained by carrying out one of the above procedures using a pure geometric isomer as a starting material, or by resolution of a mixture of the geometric isomers of the compound or intermediates using a standard procedure such as chromatographic separation.

The present invention further relates to a pharmaceutical composition comprising a therapeutically effective amount of at least one compound I as defined above or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof, in combination with at least one pharmaceutically acceptable carrier and/or auxiliary substance.

The present invention further relates to a compound I as defined above or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for use as a medicament.

The present invention also relates to a compound I as defined above or an N- oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the treatment of disorders which respond to the modulation of the 5-HT 2 c receptor.

The present invention also relates to the use of a compound I as defined above or of an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of disorders which respond to the modulation of the 5-HT 2 c receptor, and to a method for treating disorders which respond to the modulation of the 5-HT 2C receptor, which method comprises administering to a subject in need thereof at least one compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

The compounds of the present invention are modulators of the 5-HT 2 c receptor. Specifically, the compounds of formula I are agonists or partial agonists of the 5-HT 2C receptor. Thus, in a specific embodiment, the invention relates to a compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically ac- ceptable salt thereof for the treatment of disorders which respond to 5-HT 2 c receptor agonists, further to the use of a compound I as defined above or of an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of disorders which respond to 5-HT 2 c recep- tor agonists, and to a method for treating disorders which respond to 5-HT 2C receptor agonists, which method comprises administering to a subject in need thereof at least one compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof.

Within the meaning of the invention, the term "disorder" denotes disturbances and/or anomalies which are as a rule regarded as being pathological conditions or functions and which can manifest themselves in the form of particular signs, symptoms and/or malfunctions. While the treatment according to the invention can be directed toward individual disorders, i.e. anomalies or pathological conditions, it is also possible for several anomalies, which may be causatively linked to each other, to be combined into patterns, i.e. syndromes, which can be treated in accordance with the invention.

In one aspect of the invention, the diseases to be treated are disorders are damage of the central nervous system, disorders of the central nervous system, eating disorders, ocular hypertension, cardiovascular disorders, gastrointestinal disorders and diabetes.

Disorders or diseases of the central nervous system are understood as meaning disorders which affect the spinal cord and, in particular, the brain. These are, for example, cognitive dysfunction, attention deficit disorder/hyperactivity syndrome and cognitive deficits related with schizophrenia, attention deficit/hyperactivity syndrome, personality disorders, affective disorders, motion or motor disorders, pain, migraine, sleep disorders (including disturbances of the Circadian rhythm), feeding disorders, diseases associated with neurodegeneration, addiction diseases, obesity or psoriasis.

Examples of cognitive dysfunction are deficits in memory, cognition, and learning, Alzheimer's disease, age-related cognitive decline, and mild cognitive impairment, or any combinations thereof. Examples of personality disorders are schizophrenia and cognitive deficits related to schizophrenia. Examples of affective disorders are depres- sion, anxiety, bipolar disorder and obsessive compulsive disorders, or any combination thereof. Examples of motion or motor disorders are Parkinson's disease and epilepsy. Examples of feeding disorders are obesity, bulimia, weight loss and anorexia, especially anorexia nervosa. Examples of diseases associated with neurodegeneration are stroke, spinal or head trauma, and head injuries, such as hydrocephalus.

Pain condition includes nociceptive pain, neuropathic pain or a combination thereof. Such pain conditions or disorders can include, but are not limited to, post- operative pain, osteoarthritis pain, pain due to inflammation, rheumatoid arthritis pain, musculoskeletal pain, burn pain (including sunburn), ocular pain, the pain associated with dental conditions (such as dental caries and gingivitis), post-partum pain, bone fracture, herpes, HIV, traumatic nerve injury, stroke, post-ischemia, fibromyalgia, reflex sympathetic dystrophy, complex regional pain syndrome, spinal cord injury, sciatica, phantom limb pain, diabetic neuropathy, hyperalgesia and cancer.

In certain other embodiments, the disease condition is bladder dysfunction, including urinary incontinence.

Diabetes includes diabetes insipidus, diabetes mellitus, type I diabetes, type II diabetes, type III diabetes, diabetes secondary to pancreatic diseases, diabetes related to steroid use, diabetes complications, hyperglycemia and insulin resistance.

The addiction diseases include psychiatric disorders and behavioral disturbances which are caused by the abuse of psychotropic substances, such as pharmaceuticals or narcotics, and also other addiction diseases, such as addiction to gaming (impulse control disorders not elsewhere classified). Examples of addictive substances are: opioids (e.g. morphine, heroin and codeine), cocaine; nicotine; alcohol; substances which interact with the GABA chloride channel complex, sedatives, hypnotics and tranquilizers, for example benzodiazepines; LSD; cannabinoids; psychomotor stimulants, such as 3,4- methylenedioxy-N-methylamphetamine (ecstasy); amphetamine and amphetamine- like substances such as methylphenidate, other stimulants including caffeine and nicotine. Addictive substances which come particularly into consideration are opioids, cocaine, amphetamine or amphetamine- like substances, nicotine and alcohol. Especially, addiction disorders include alcohol abuse, cocaine abuse, tobacco abuse and smoking cessation.

With regard to the treatment of addiction diseases, particular preference is given to those compounds according to the invention of the formula (I) which themselves do not possess any psychotropic effect. This can also be observed in a test using rats, which, after having been administered compounds which can be used in accordance with the invention, reduce their self administration of psychotropic substances, for example cocaine.

Examples of gastrointestinal disorders are irritable bowel syndrome.

Preferably, the disorders are selected from the group consisting of bipolar disor- der, depression, atypical depression, mood episodes, adjustment disorders, anxiety, panic disorders, post-traumatic syndrome, psychoses, schizophrenia, cognitive deficits of schizophrenia, memory loss, dementia of aging, Alzheimer's disease, neuropsychiatric symptoms in Alzheimer's disease (e.g. aggression), behavioral disorders associated with dementia, social phobia, mental disorders in childhood, attention deficit hyperactivity disorder, organic mental disorders, autism, mutism, disruptive behavior disorder, impulse control disorder, borderline personality disorder, obsessive compulsive disorder, migraine and other conditions associated with cephalic pain or other pain, raised intracranial pressure, seizure disorders, epilepsy, substance use disorders, alcohol abuse, cocaine abuse, tobacco abuse, smoking cessation, sexual dysfunction/erectile dysfunc- tion in males, sexual dysfunction in females, premenstrual syndrome, late luteal phase syndrome, chronic fatigue syndrome, sleep disorders, sleep apnoea, chronic fatigue syndrome, psoriasis, Parkinson's disease, psychosis in Parkinson's disease, neuropsychiatric symptoms in Parkinson's disease (e.g. aggression), Lewy Body dementia, neuropsychiatric symptoms in Lewy Body dementia (e.g. aggression), spinal cord injury, trauma, stroke, pain, bladder dysfunction/urinary incontinence, encephalitis, meningitis, eating disorders, obesity, bulimia, weight loss, anorexia nervosa, ocular hypertension, cardiovascular disorders, gastrointestinal disorders, diabetes insipidus, diabetes mellitus, type I diabetes, type II diabetes, type III diabetes, diabetes secondary to pancreatic diseases, diabetes related to steroid use, diabetes complications, hyperglycemia and insulin resistance, and are specifically schizophrenia, depression, bipolar disorders, obesity, substance use disorders, neuropsychiatric symptoms in Alzheimer's disease (e.g. aggression) or neuropsychiatric symptoms in Parkinson's disease (e.g. aggression).

The compounds of the invention may be used for a preventive treatment

(prophylaxis), in particular as relapse prophylaxis or phase prophylaxis, but are prefera- bly used for a treatment in its proper sense (i.e. non-prophylactic), i.e. for the treatment of acute or chronic signs, symptoms and/or malfunctions. The treatment can be orientated symptomatically, for example as the suppression of symptoms. It can be effected over a short period, be orientated over the medium term or can be a long-term treatment, for example within the context of a maintenance therapy.

In another embodiment, the present invention relates to the use of a compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for preparing a medicament for preventing (the development of) a disease condition as described above and to a method for preventing (the development of) a disease condition as described above comprises administering to the subject in need of treatment thereof (e.g., a mammal, such as a human) a therapeutically effective amount of a compound I as defined above or an N-oxide, a tautomeric form, a stereoi- somer or a pharmaceutically acceptable salt thereof. As used herein, the term "prevent" a disease condition by administration of any of the compounds described herein means that the detectable physical characteristics or symptoms of the disease or condition do not develop following the administration of the compound described herein. Alternatively, the method comprises administering to the subject a therapeutically effective amount of a compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof, in combination with a therapeutically effective amount of at least one cognitive enhancing drug.

In yet another embodiment, the present invention relates to the use a compound I as defined above or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutically acceptable salt thereof for preparing a medicament for preventing the progression (e.g., worsening) of a disease condition and to a method for preventing the progression (e.g., worsening) of a disease condition, which method comprises administering to the subject in need of treatment thereof (e.g., a mammal, such as a human) a therapeutically effective amount of a compound I as defined above or an N-oxide, a tautomeric form, a ste- reoisomer or a pharmaceutically acceptable salt thereof.

There are several lines of evidence suggesting that 5-HT 2 c agonists or partial agonists would have therapeutic use in a variety of diseases, disorders and conditions.

Knockout mice models lacking the 5-HT 2 c receptor exhibit hyperphagia, obesity and are more prone to seizures and sudden death [Tecott LH, Sun LM, Akana SF, Strack AM, Lowenstein DH, Dallman MF, Julius D (1995) Eating disorder and epilepsy in mice lacking 5-HT 2 c serotonin receptors. Nature 374:542-546]. They also exhibit compulsive- like behavior [Chou-Green JM, Holscher TD, Dallman MF, Akana SF (2003). Compulsive behavior in the 5-HT 2 c receptor knockout mouse. Phys. Behav. 78:641-649], hyperresponsiveness to repeated stress [Chou-Green JM, Holscher TD, Dallman MF, Akana SF (2003). Repeated stress in young and old 5-HT 2 c receptor knockout mouse. Phys. Behav. 79:217-226], wakefulness [Frank MG, Stryker MP, Tecott LH (2002). Sleep and sleep homeostasis in mice lacking the 5-HT 2 c receptor. Neuropsychopharmacology 27:869-873], hyperactivity and drug dependence [Rocha BA, Goulding EH, O'Dell LE, Mead AN, Coufal NG, Parsons LH, Tecott LH (2002). Enhanced locomotor, reinforcing and neurochemical effects of cocaine in serotonin 5- hydroxytryptamine 2C receptor mutant mice. J. Neurosci. 22: 10039- 10045].

5-HT 2C is unique among other G-protein-coupled receptors (GPCRs) in that its pre-mRNA is a substrate for base modification via hydro lytic deamination of adenosines to yield inosines. Five adenosines, located within a sequence encoding the putative second intracellular domain can be converted to inosines. This editing can alter the coding potential of the triplet codons and allows for the generation of multiple different receptor iso forms. The edited receptor isoforms were shown to have reduced ability to interact with G-proteins in the absence of agonist stimulation [Werry, TD, Loiacono R, Sexton PA, Christopoulos A (2008). RNA editing of the serotonin 5-H >< receptor and its effects on cell signaling, pharmacology and brain function. Pharmac. Therap.

119:7-23].

Edited 5-HT 2C isoforms with reduced function are significantly expressed in the brains of depressed suicide victims [Schmauss C (2003) Serotonin 2C receptors: suicide, serotonin, and runaway RNA editing. Neuroscientist 9:237-242. Iwamoto K, Kato T (2003). RNA editing of serotonin 2C receptor in human postmortem brains of major mental disorders. Neurosci. Lett. 346: 169-172] and in the learned helplessness rats (a well established animal model of depression) [Iwamotoa K, Nakatanib N, Bundoa M, Yoshikawab T, Katoa T (2005). Altered RNA editing of serotonin 2C receptor in a rat model of depression. Neurosci. Res.53: 69-76] suggesting a link between 5-HT 2C function and depression. There are also implications of edited 5-HT 2 c isoforms and spatial memory [Du Y, Stasko M, Costa AC, Davissone MT, Gardiner KJ (2007). Editing of the serotonin 2C receptor pre-mRNA Effects of the Morris Water Maze. Gene 391 : 186- 197]. In addition, fully edited isoforms of the human 5-HT 2C receptor display a striking reduction in sensitivity to lysergic acid diethylamide (LSD) and to atypical antipsychot- ic drugs clozapine and loxapine, suggesting a possible role of the receptor in the etiology and pharmacology of schizophrenia [Niswender CM, Herrick-Davis K,. Dilley GE, Meltzer HY, Overholser JC, Stockmeier CA, Emeson RB, Sanders-Bush E (2001). RNA Editing of the Human Serotonin 5-HT 2 c Receptor: Alterations in Suicide and Im- plications for Serotonergic Pharmacotherapy. Neuropsychopharm. 24:478-491].

Recently, the availability of potent and selective 5-HT 2 c receptor agonists made it possible to directly investigate the effects of 5-HT 2C agonists and their therapeutic potential. Thus recent studies demonstrated that selective 5-HT 2C agonists resulted in decreased food intake and body weight gain in normal and obese rats [Smith BM, et al. (2008). Discovery and structure-activity relationship of (li?)-8-chloro-2,3,4,5- tetrahydro-1 -methyl- lH-3-benzazepine (Lorcaserin), a selective serotonin 5-HT 2 c receptor agonist for the treatment of obesity. J Med Chem 51 :305-313. Thomsen WJ, Grottick AJ, Menzaghi F, Reyes-Saldana H, Espitia S, Yuskin D, Whelan K, Martin M, Morgan M, Chen W, Al-Shama H, Smith B, Chalmers D, Behan D (2008) Lorcaserin, A Novel Selective Human 5-HT 2 c Agonist: In Vitro and In Vivo Pharmacological

Characterization. J Pharmacol Exp Ther. 325:577-587. Rosenzweig-Lipson S, Zhang J, Mazandarani H, Harrison BL, Sabb A, Sabalski J, Stack G, Welmaker G, Barrett JE, Dunlop J (2006) Antiobesity-like effects of the 5-HT 2C receptor agonist WAY-161503. Brain Res. 1073- 1074:240-25 1. Dunlop J, Sabb AL, Mazandarani H, Zhang J, Kal- gaonker S, Shukhina E, Sukoff S, Vogel RL, Stack G, Schechter L, Harrison BL, Rosenzweig-Lipson S (2005). WAY-163909 [97bi?, 10ai?)-l,2,3,4,8,9,10,10a- octahydro-7bH-cyclopenta-[¾][l,4]diazepino[6,7,lhi]indole], a novel 5- hydroxytryptamine 2C receptor -selective agonist with anorectic activity. J Pharmacol Exp Ther. 313:862-869.].

Furthermore, selective 5-HT 2C receptor agonists produce antidepressant effects in animal models of depression comparable to those of SSRIs but with a much faster onset of action and a therapeutic window that avoids antidepressant-induced sexual dysfunction. These agonists were also effective in animal models of compulsive behavior such as scheduled induced polydipsia and they also exhibited decreased hyperactivity and aggression in rodents [Rosenzweig-Lipson S, Sabb A, Stack G, Mitchell P, Lucki I, Malberg JE, Grauer S, Brennan J, Cryan JF, Sukoff Rizzo SJ, Dunlop J, Barrett JE, Marquis KL (2007) Antidepressant-like effects of the novel, selective, 5-HT 2C receptor agonist WAY-163909 in rodents. Psychopharmacology (Berlin) 192: 159-170.

Rosenzweig-Lipson S, Dunlop J, Marquis KL (2007) 5-HT 2 c receptor agonists as an innovative approach for psychiatric disorders. Drug news Perspect, 20: 565-571.

Cryan, JF, Lucki I (2000). Antidepressant-like behavioral effects mediated by 5- Hydroxytryptamine 2C receptors. J. Pharm. Exp. Ther. 295: 1120-1126.].

Acute or chronic administration of 5-HT 2 c agonists decreases the firing rate of ventral tegmental area dopamine neurons but not that of substantia nigra. In addition 5- HT 2C agonists reduce dopamine levels in the nucleus accumbens but not in the striatum (the region of the brain mostly associated with extrapyramidal side effects) [Di Matteo, V., Di Giovanni, G., Di Mascio, M., & Esposito, E. (1999). SB 242084, a selective serotonin 2C receptor antagonist, increases dopaminergic transmission in the meso limbic system. Neuropharmacology 38, 1 195 - 1205. Di Giovanni, G., Di Matteo, V., Di Mascio, M., & Esposito, E. (2000). Preferential modulation of meso limbic vs. nigrostri- atal dopaminergic function by serotonin2C/2B receptor agonists: a combined in vivo electrophysiological and microdialysis study. Synapse 35, 53 - 61. Marquis KL, Sabb AL, Logue SF, Brennan JA, Piesla MJ, Comery TA, Grauer SM, Ashby CR, Jr., Nguyen HQ, Dawson LA, Barrett JE, Stack G, Meltzer HY, Harrison BL, Rosenzweig- Lipson S (2007) WAY-163909 [(7bi?,10ai?)-l,2,3,4,8,9,10,10a-octahydro-7bH- cyclopenta-[£][l,4]diazepino[ 6,7,lhi]indole]: A novel 5 -hydroxytryptamine 2C recep- tor-selective agonist with preclinical antipsychotic-like activity. J Pharmacol Exp Ther 320:486-496.]. Therefore it is expected that 5-HT 2C receptor agonists will selectively decrease mesolimibic dopamine levels without affecting the nigrostriatal pathway thus avoiding the EPS side effects of typical antipsychotics. Several 5-HT 2C receptor agonists have shown antipsychotic activity in animal models of schizophrenia without EPS based on the lack of effect in catalepsy [Marquis KL, Sabb AL, Logue SF, Brennan JA, Piesla MJ, Comery TA, Grauer SM, Ashby CR, Jr., Nguyen HQ, Dawson LA, Barrett JE, Stack G, Meltzer HY, Harrison BL, Rosenzweig-Lipson S (2007) WAY-163909 [(IbR, 1 Oai?)- 1,2,3,4,8,9,10,10a-octahydro-7bH-cyclopenta-[6] [ 1 ,4]diazepino[

6,7,lhi]indole]: A novel 5 -hydroxytryptamine 2C receptor-selective agonist with pre- clinical antipsychotic-like activity. J Pharmacol Exp Ther 320:486-496. Siuciak JA, Chapin DS, McCarthy SA, Guanowsky V, Brown J, Chiang P, Marala R, Patterson T, Seymour PA, Swick A, Iredale PA (2007) CP-809,101, a selective 5-HT 2C agonist, shows activity in animal models of antipsychotic activity. Neuropharmacology 52:279- 290]. The antipsychotic activity of 5-HT 2 c receptor agonists without EPS coupled with their beneficial effects in mood disorders and cognition and their antiobesity like effects render 5-HT 2 c receptor agonists as unique agents to treat schizophrenia [Rosenzweig- Lipson S, Dunlop J, Marquis KL (2007) 5-HT 2 c receptor agonists as an innovative approach for psychiatric disorders. Drug news Perspect, 20: 565-571. Dunlop J, Marquis KL, Lim HK, Leung L, Kao J, Cheesman C, Rosenzweig-Lipson S (2006). Pharmacological profile of the 5-HT 2 c receptor agonist WAY- 163909; therapeutic potential in multiple indications. C S Dug Rev. 12: 167-177.].

In addition 5-HT 2 c modulation has been implicated in epilepsy [Isaac M (2005).

Serotonergic 5-HT > ( receptors as a potential therapeutic target for the antiepileptic drugs. Curr. Topics Med. Chem. 5:59:67], psoriasis [Thorslund K, Nordlind K (2007). Serotonergic drugs-a possible role in the treatment of psoriasis? Drug News Perspect 20:521-525], Parkinson's disease and related motor disorders [Esposito E, Di Matteo V, Pierucci M, Benigno A, Di Giavanni, G (2007). Role of central 5-HT 2 c receptor in the control of basal ganglia functions. The Basal Ganglia Pathophysiology: Recent Advances 97- 127], behavioral deficits [Barr AM, Lahmann-Masten V, Paul us M,

Gainetdinov RP, Caron MG, Geyer A (2004). The selective serotonin-2A receptor antagonist M 100907 reverses behavioral deficits in dopamine transporter knockout mice. Neuropsychopharmacology 29:221-228], anxiety [Dekeyne A, Mannoury la Cour C, Gobert A, Brocco M, Lejuene F, Serres F, Sharp T, Daszuta A, Soumier A, Papp M, Rivet JM, Flik G, Cremers TI, Muller O, Lavielle G, Millan MJ (2208). S32006, a novel 5-HT.x receptor antagonists displaying broad-based antidepressant and anxiolytic properties in rodent models. Psychopharmacology 199:549-568. Nunes-de-Souza V, Nunes-de-Souza RL, Rodgers RJ, Canto-de-Souza A (2008). 5-HT2 receptor activation in the midbrain periaqueductal grey (PAG ) reduces anxiety-like behavior in mice. Be- hav. Brain Res. 187:72-79.], migraine [Leone M, Rigamonti A, D'Amico D, Grazzi L, Usai S, Bussone G (2001). The serotonergic system in migraine. Journal of Headache and Pain 2(Suppl. ./J:S43-S46], Alzheimer ' s disease [A jona A A, Pooler AM, Lee RK, W urt man RJ (2002). Effect of a 5-HT 2 c serotonin agonist, dexnorfen fl uram i ne, on amyloid precursor protein metabolism in guinea pigs. Brain Res. 95 1 : 135- 140], pain and spinal cord injury [Nakae A, akai K, Tanaka T, Uagihira S, Shibata M, Ueda K, asi mo T (2008). The role of RNA editing of the serotonin 2C receptor in a rat model of oro-facial neuropathic pain. The European Journal ofNeuroscience 27:2373-2379. Nakae A, Nakai K, Tanaka T, Takashina M, Hagihira S, Shibata M, Ueda K, Mashimo T (2008). Serotonin 2C receptor mRNA edit ing in neuropathic pain model. Neurosci. Res. 60 :228-23 1 . Kao T, Shumsky JS, Jacob- Vadakot S, Timothy HB, Murray M, Moxon, KA (2006). Role of the 5-HT 2 c receptor in improving weight-supported stepping in adult rats spinalized as neonates. Brain Res.1 1 12: 159-168.], sexual dysfunct ion [Motofei IG (2008). A dual physiological character for sexual function: the role of serotonergic receptors. BJU International 101 :53 1 -534. Shimada I, Maeno K, Kondoh Y, Kaku H, Sugasawa K, Kimura Y, Hatanaka K,; Naitou Y, Wanibuchi F, Sakamoto S,; Tsukamoto S (2008). Synthesis and structure-activ ity relationships of a series of benzazepine derivatives as 5-ΗΤ χ receptor agonists. Bioorg. Med. Chem. 16:3309- 3320.], smoking cessation [Fletcher PJ, Le AD, Higgins GA (2008). Serotonin receptors as potential targets for modulation of nicotine use and dependence. Progress Brain Res. 1 72 :361 -83 ], substance dependence [Bubar MJ, Cunningham KA (2008 ). Prospects for serotonin 5-HT2R pharmacotherapy in psychostimulant abuse. Progress Brain Res. 1 72 :3 1 9-46], and ocular hypertension [Sharif NA, McLaughlin MA, Kel ly CR (2006 ). A L-34662: a potent, select ive, and efficacious ocular hypotensive serotonin-2 receptor agonist. J Ocul Pharmacol Ther. 23 : 1-13].

Further, 5HT modulation can be useful in the treatment of pain, both neuropathic and nocicept ive pain, see for example U.S. Patent applicat ion publ icat ion

US2007/0225277. Obata, Hideaki: Ito, Naomi; Sasaki, Masayuki; Saito, Shigeru; Goto, Fumio. Possible involvement of spinal noradrenergic mechanisms in the antiallodynic effect of intrathecally administered 5 - HT2C receptor agonists in the rats with periph- eral nerve injury. European Journal of Pharmacology (2007), 567(1-2), 89-94. Ser- otonin2C receptor mRNA editing in neuropathic pain model. Nakae, Aya; Nakai, Kunihiro; Tanaka, Tatsuya; Takashina, Masaki; Hagihira, Satoshi; Shibata, Masahiko; Ueda, Koichi; Mashimo, Takashi. Department of Anesthesiology & Intensive Care Medicine, Graduate School of Medicine, Osaka University, Neuroscience Research (Amsterdam, Netherlands) (2008), 60(2), 228-231. Antiallodynic effects of intrathecally administered 5 - HT2C receptor agonists in rats with nerve injury. Obata, Hideaki; Saito, Shigeru; Sakurazawa, Shinobu; Sasaki, Masayuki; Usui, Tadashi; Goto, Fumio. Department of Anesthesiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan. Pain (2004), 108(1-2), 163-169. Influence of 5 ,7- dihydroxytryptamine ( 5 ,7-DHT) on the antinociceptive effect of serotonin ( 5 -HT) 5 - HT2C receptor agonist in male and female rats. Brus, Ryszard; Kasperska, Alicja; Oswiecimska, Joanna; Szkilnik, Ryszard. Department of Pharmacology, Silesian Medical University, Zabrze, Pol. Medical Science Monitor (1997), 3(5), 654-656.

Modulation of 5HT2 receptors may be beneficial in the treatment of conditions related to bladder function, in particular, urinary incontinence. [Discovery of a novel azepine series of potent and selective 5 - HT2C agonists as potential treatments for uri- nary incontinence. Brennan, Paul E.; Whitlock, Gavin A.; Ho, Danny K. H.; Conlon, Kelly; McMurray, Gordon. Bioorganic & Medicinal Chemistry Letters (2009), 19(17), 4999-5003. Investigation of the role of 5 -HT2 receptor subtypes in the control of the bladder and the urethra in the anesthetized female rat. Mbaki, Y.; Ramage, A. G. Department of Pharmacology, University College London, London, UK. Brit- ish Journal of Pharmacology (2008), 155(3), 343-356.] In particular, compounds with agonist activity at 5-HT 2 c have been shown to be useful in treating urinary incontinence, see for example U.S. Patent application publications US2008 0146583 and US 2007/0225274.

Further pre-clinical data suggest that 5-HT 2C agonists could be useful for the treat- ment of a number of psychiatric diseases, including schizophrenia, bipolar disorders, depression/anxiety, substance use disorders and especially disorders like neuropsychiat- ric symptoms in Alzheimer's disease: Aggression, psychosis/ agitation represent key unmet medical needs. Clinical (Shen JHQ et al, A 6-week randomized, double-blind, placebo-controlled, comparator referenced trial of vabicaserin in acute schizophrenia. Journal of Psychiatric Research 53 (2014) 14-22; Liu J et al., Prediction of Efficacy of Vabicaserin, a 5-HT 2 c Agonist, for the Treatment of Schizophrenia Using a Quantitative Systems Pharmacology Model. CPT Pharmacometrics Syst. Pharmacol. (2014) 3, el 11;) and preclinical data (Dunlop J et al, Characterization of Vabicaserin (SCA-136), a Selective 5-Hydroxytryptamine 2C Receptor Agonist. J Pharmacol Exp Ther (2011) 337, 673-80; Siuciak J et al, CP-809,101, a selective 5-HT 2C agonist, shows activity in animal models of antipsychotic activity. Neuropharmacology 52 (2007) 279-290;

Mosienko V et al, Exaggerated aggression and decreased anxiety in mice deficient in brain serotonin. Transl Psychiatry (2012) 2, el 22; Del Guidice T et al, Stimulation of 5-HT 2 c Receptors Improves Cognitive Deficits Induced by Human Tryptophan Hy- droxylase2 Loss of Function Mutation. Neuropsychopharmacology (2014) 39, 1125— 1134; Rosenzweig-Lipson et al, Antidepressant-like effects of the novel, selective, 5- HT 2C receptor agonist WAY-163909 in rodents. Psychopharmacology (2007) 192: 159— 170) suggest 5-HT 2C receptor stimulation to result in therapeutic efficacy in aggression, psychosis agitation and moderate pro-cognitive effects (Del Guidice T et al, Stimulation of 5-HT 2 c Receptors Improves Cognitive Deficits Induced by Human Tryptophan Hydroxylase2 Loss of Function Mutation. Neuropsychopharmacology (2014) 39, 1125— 1134; Siuciak J et al, CP-809,101, a selective 5-HT 2C agonist, shows activity in animal models of antipsychotic activity. Neuropharmacology 52 (2007) 279-290).

In the use and the method of the invention, an effective quantity of one or more compounds, as a rule formulated in accordance with pharmaceutical and veterinary practice, is administered to the individual to be treated, preferably a mammal, in par- ticular a human being, productive animal or domestic animal. Whether such a treatment is indicated, and in which form it is to take place, depends on the individual case and is subject to medical assessment (diagnosis) which takes into consideration signs, symptoms and/or malfunctions which are present, the risks of developing particular signs, symptoms and/or malfunctions, and other factors.

Actual dosage levels of active ingredients in the pharmaceutical compositions of the present invention can be varied so as to obtain an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular subject (e.g., a mammal, preferably, a human (patient)), compositions and mode of administration. The selected dosage level will depend upon the activity of the particular compound, the route of administration, the severity of the condition being treated and the condition and prior medical history of the patient being treated. However, it is within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.

Compounds of the present invention can also be administered to a subject as a pharmaceutical composition comprising the compounds of interest in combination with at least one pharmaceutically acceptable carriers. The phrase "therapeutically effective amount" of the compound of the present invention means a sufficient amount of the compound to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well-known in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.

The total daily dose of the compounds of this invention administered to a subject

(namely, a mammal, such as a human) ranges from about 0.01 mg/kg body weight to about 100 mg/kg body weight. More preferable doses can be in the range of from about 0.01 mg/kg body weight to about 30 mg/kg body weight. If desired, the effective daily dose can be divided into multiple doses for purposes of administration. Consequently, single dose compositions may contain such amounts or submultiples thereof to make up the daily dose.

In one aspect, the present invention provides pharmaceutical compositions. The pharmaceutical compositions of the present invention comprise the compounds of the present invention or an N-oxide, a tautomeric form, a stereoisomer or a pharmaceutical- ly acceptable salt or solvate thereof. The pharmaceutical compositions of the present invention comprise compounds of the present invention that can be formulated together with at least one non-toxic pharmaceutically acceptable carrier.

In yet another embodiment, the present invention provides a pharmaceutical composition comprising compounds of the present invention or an N-oxide, a tautomer- ic form, a stereoisomer or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, alone or in combination with one or more compounds that are not the compounds of the present invention. Examples of one or more compounds that can be combined with the compounds of the present invention in pharmaceutical compositions, include, but are not limited to, one or more cognitive enhancing drugs.

The pharmaceutical compositions of this present invention can be administered to a subject (e.g., a mammal, such as a human) orally, rectally, parenterally, intracister- nally, intravaginally, intraperitoneally, topically (as by powders, ointments or drops), bucally or as an oral or nasal spray. The term "parenterally" as used herein, refers to modes of administration which include intravenous, intramuscular, intraperitoneal, in- trasternal, subcutaneous and intraarticular injection and infusion.

The term "pharmaceutically acceptable carrier" as used herein, means a nontoxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as, but not limited to, lactose, glucose and sucrose; starches such as, but not limited to, corn starch and potato starch; cellulose and its derivatives such as, but not limited to, sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as, but not limited to, cocoa butter and suppository waxes; oils such as, but not limited to, peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols; such a propylene glycol; esters such as, but not limited to, ethyl oleate and ethyl laurate; agar; buffering agents such as, but not limited to, magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as, but not limited to, sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.

Pharmaceutical compositions of the present invention for parenteral injection comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), vegetable oils (such as olive oil), injectable organic esters (such as ethyl oleate) and suitable mixtures thereof. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.

These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride and the like. Pro- longed absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

In some cases, in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.

The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.

Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound may be mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium cit- rate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants such as glycerol; d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents such as paraffin; f) absorption accelerators such as quaternary ammonium compounds; g) wetting agents such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such carriers as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

The solid dosage forms of tablets, dragees, capsules, pills and granules can be prepared with coatings and shells such as enteric coatings and other coatings well- known in the pharmaceutical formulating art. They may optionally contain opacifying agents and may also be of a composition such that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.

The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned carriers.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active com- pounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tet- rahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan and mixtures thereof. Besides inert diluents, the oral compositions may also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar, tragacanth and mixtures thereof.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating carriers or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

Compounds of the present invention can also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or mult i- lamellar hydrated liquid crystals which are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients and the like. The preferred lipids are natural and synthetic phospholipids and phosphatidyl cholines (lecithins) used separately or together.

Methods to form liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p. 33 et seq.

Dosage forms for topical administration of a compound of the present invention include powders, sprays, ointments and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives, buffers or propellants which may be required. Ophthalmic formulations, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.

The compounds of the present invention can be used in the form of pharmaceutically acceptable salts derived from inorganic or organic acids. The phrase "pharma- ceutically acceptable salt" means those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.

Pharmaceutically acceptable salts are well known in the art. For example, S. M.

Berge et al. describe pharmaceutically acceptable salts in detail in (J. Pharmaceutical Sciences, 1977, 66: 1 et seq.). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting a free base function with a suitable organic acid. Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerophosphate, hem- isulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethansulfonate (isothionate), lactate, malate, maleate, methanesulfonate, nico- tinate, 2-naphthalenesulfonate, oxalate, palmitoate, pectinate, persulfate, 3- phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides such as, but not limited to, methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as, but not limited to, decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained. Examples of acids which can be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, hydrobromic acid, sulfuric acid, and phos- phoric acid and such organic acids as acetic acid, fumaric acid, maleic acid, 4- methylbenzenesulfonic acid, succinic acid and citric acid.

Basic addition salts can be prepared in situ during the final isolation and purification of compounds of this invention by reacting a carboxylic acid-containing moiety with a suitable base such as, but not limited to, the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary or tertiary amine. Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as, but not limited to, lithium, sodium, potassium, calcium, magnesium and aluminum salts and the like and nontoxic quaternary ammonia and amine cations including ammonium, tetrame- thylammonium, tetraethylammonium, methylammonium, dimethylammonium, trime- thylammonium, triethylammonium, diethylammonium, ethylammonium and the like. Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine and the like.

The compounds of the present invention can exist in unsolvated as well as solv- ated forms, including hydrated forms, such as hemi-hydrates. In general, the solvated forms, with pharmaceutically acceptable solvents such as water and ethanol among others are equivalent to the unsolvated forms for the purposes of the invention.

The following examples serve to explain the invention without limiting it.

Examples

The compounds were either characterized via proton-NMR in d6-dimethylsulfoxide, d- chloroform or cLi-methanol on a 400 MHz, 500 MHz or 600 MHz NMR instrument (Bruker AVANCE), or by 13 C-NMR at 125 MHz, or by 19 F-NMR at 470 MHz, or by mass spectrometry, generally recorded via HPLC-MS in a fast gradient on C18-material (electrospray-ionisation (ESI) mode).

The magnetic nuclear resonance spectral properties (NMR) refer to the chemical shifts (δ) expressed in parts per million (ppm). The relative area of the shifts in the 1H-NMR spectrum corresponds to the number of hydrogen atoms for a particular functional type in the molecule. The nature of the shift, as regards multiplicity, is indicated as singlet (s), broad singlet (s. br.), doublet (d), broad doublet (d br.), triplet (t), broad triplet (t br.), quartet (q), quintet (quint.), multiplet (m), doublet of doublets (dd), doublet of doublets of doublets (ddd), triplet of doublets (td), doublet of triplets of doublets (dtd), quartet of doublets of doublets (qdd) etc..

Enantiomers were separated / purified by chiral supercritical fluid chromatography (SFC) (method A). Method A

A.l Analytical SFC

Analytical samples were run on an Agilent 1260 Infinity Hybrid SFC System, controlled by Agilent OpenLab CDS ChemStation Edition. The system consists of an injec- tor, a heated column compartment including a switch for 6 columns, a C0 2 -booster pump, a binary pump module for C0 2 and modifier flow and an UV-detector. The backpressure regulator was set to 160 bars and heated to 60 °C. If not stated otherwise, the columns were 100 mm in length, 4.6 mm in diameter and packed with 5 μιη material. They were kept at room temperature during analysis. As mobile phase, a mixture of liquefied C0 2 and organic modifier with additive was used as indicated for each sample. The flow rate was kept at 3.5 mL/min.

A.2 Preparative SFC

Preparative separations were carried out on a Waters Prep lOOq SFC System, controlled by Waters MassLynx Software. The system consists of an open bed injector/collector, a heated column compartment including a switch for 6 columns, a C0 2 -booster pump, a pump module for modifier flow and an UV-detector. To enable quantitative collection, the gas liquid separator was driven with a make-up flow of 30 mL/ min Methanol. The backpressure regulator was set to 120 bar and heated to 60 °C. If not stated otherwise, the columns were 250 mm in length, 20 mm in diameter and packed with 5 μιη material. They were kept at 30 °C during the separation. As mobile phase, a mixture of liquefied C0 2 and organic modifier with additive was used as indicated for each sample. The flow rate was kept at 100 g/min. Abbreviations:

h hour(s)

min minute(s)

d day(s)

r.t. room temperature (20-25°C)

PE petroleum ether

EtOH ethanol

MeOH methanol DCM dichloromethane

DCE dichloroethane

THF tetrahydrofuran

DMF N,N-dimethylformamide

MeCN acetonitrile

EtOAc ethyl acetate

TFA trifluoroacetic acid

DIPEA diisopropylethyl amine

EDC 1 -ethyl-3 -(3 -dimethylaminopropyl)carbodiimide

HOBt hydroxybenzotriazo le

I. Preparation Examples

Example 1

8-(Trifluoromethyl)-2, 3,4,6,7, 8-hexahydro- lH-[ 1 ,4]diazepino[6,7, 1 -ij]quinoline

(compound of formula la.1 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CF 3 )

1.1 4-(Trifluoromethyl)-3 ,4-dihydroquino lin-2( 1 H)-one

In a 5 mL microwave vial containing 100 mg of ethyl (E)-4,4,4-trifluorobut-2-enoate (0.595 mmol, 1.00 eq), 163 mg of 2-aminophenyl boronic ester (1.19 mmol, 2.00 eq), 164 mg of potassium carbonate (1.19 mmol, 2.00 eq) and 13.6 mg of [RhOH(COD)] 2 (0.030 mmol, 0.05 eq) was added 2%wt. TPGS-750-M solution in water (3 mL) (TPGS- 750-M: a surfactant composed of a lipophilic a-tocopherol moiety and a hydrophilic PEG-750-M chain, joined by a succinic acid linker; forms spontaneously micelles upon dissolution in water. From Sigma-Aldrich.). The mixture was stirred vigorously at ambient temperature for 48 h. The reaction mixture was then extracted with ethyl acetate. Then the organic phase was dried over MgSC^, filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (116 mg, 82% yield). 1H NMR (500 MHz, CDC1 3 ): δ 9.05 (s, 1H), 7.32 (td, J= 7.7, 1.5 Hz, 1H), 7.29 (d, J = 7.5 Hz, 1H), 7.08 (td, J= 7.6, 1.2 Hz, 1H), 6.90 (dd, J= 8.0, 1.2 Hz, 1H), 3.64 (qdd, J = 9.6, 7.1, 2.9 Hz, 1H), 3.00 - 2.89 (m, 2H).

13 C NMR (125 MHz, CDC1 3 ): δ 168.39, 137.83, 130.38, 130.08, 126.12 (q, J= 281.0 Hz), 123.43, 116.30, 115.57, 41.15 (q, J = 28.6 Hz), 30.07 (q, J = 2.8 Hz).

1 9 F NMR (470 MHz, CDC1 3 ): δ -72.51.

ESI-MS: m/z (%): 216.20 (100, [M+H] + ).

1.2 4-(Trifluoromethyl)-l,2,3,4-tetrahydroquinoline

197 mg of 4-(trifluoromethyl)-3,4-dihydroquinolin-2(lH)-one from step 1.1 (0.916 mmol, 1.00 eq) was dissolved in 10 mL of THF (anhydrous) and 2.29 mL borane dimethyl sulfide complex (2 M in THF, 4.58 mmol, 5.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 8 h. Then the reaction mixture was quenched with HCl (1 M) and the aqueous phase was extracted with ethyl acetate. Subsequently the organic phase was extracted three times with HCl (1M) and to the aqueous phase was added NaOH (1M) until pH 10. The mixture was then extracted with dichloro- methane three times. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0- 10% methanol in dichloro methane) to yield the title compound (114 mg, 62% yield). ESI-MS: m/z (%): 202.20 (100, [M+H] + ).

1.3 2-(4-(Trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)acetamid e

A mixture of 114 mg of 4-(trifluoromethyl)-l,2,3,4-tetrahydroquinoline from step 1.2 (0.567 mmol, 1.00 eq), 210 mg of 2-iodoacetamide (1.13mmol, 2.00 eq) and 0.385 ml of DIPEA (2.27 mmol, 4.00 eq) in DMF (3 mL) was heated in a microwave at 130 °C for 8 h. Then NaOH (1M) was added and the mixture was extracted with dichloro- methane. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (86 mg, 59%> yield).

ESI-MS: m/z (%): 259.20 (100, [M+H] + ).

1.4 2-(4-(Trifluoromethyl)-3 ,4-dihydroquinolin- 1 (2H)-yl)ethanamine 86 mg of 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)acetamid e from step 1.3 (0.333 mmol, 1.00 eq) was dissolved in 3 mL THF (anhydrous) and 0.833 mL of borane dimethyl sulfide complex (2 M in THF, 1.67 mmol, 5.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 5 h. Then the reaction mixture was quenched with HC1 (1 M) and the aqueous phase was extracted with ethyl acetate. Subsequently the organic phase was extracted three times with HC1 (1M) and to the aqueous phase was added NaOH (1M) until pH 10. The mixture was then extracted with dichloromethane three times. The organic phase was dried over MgSC^ and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (69 mg, 85% yield).

ESI-MS: m/z (%): 245.15 (100, [M+H] + ).

1.5 8-(Trifluoromethyl)-2, 3,4,6,7, 8-hexahydro- lH-[ 1 ,4]diazepino[6,7, 1 -ij]quinoline A mixture of 69 mg of 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)ethanami ne from step 1.4 (0.282 mmol, 1.00 eq), 0.022 mL of formaldehyde (0.282 mmol, 1.00 eq), and 0.022 mL of TFA (0.282 mmol, 1.00 eq) in ethanol (2 mL) was stirred for 3 d at ambient temperature. Then NaOH (1M) was added and the mixture was extracted with dichloromethane. The organic phase was dried over MgSC^ and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (47 mg, 65% yield).

1H NMR (500 MHz, CDC1 3 ) δ 7.17 (d, J= 7.4 Hz, 1H), 7.09 (dd, J= 7.5, 1.6 Hz, 1H), 6.81 (t, J= 7.5 Hz, 1H), 3.98 (d, J= 14.4 Hz, 1H), 3.85 (d, J= 14.5 Hz, 1H), 3.49 (qdd, J= 9.8, 6.1, 3.8 Hz, 1H), 3.31 (ddd, J= 10.1, 3.6, 1.4 Hz, 2H), 3.17 - 3.01 (m, 4H), 2.20 (dq, J= 14.2, 4.0 Hz, 1H), 2.10 - 1.97 (m, 2H).

ESI-MS: m/z (%): 257.20 (100, [M+H] + ).

1.6 Analytical separation of the two enantiomers of 8-(trifluoromethyl)-2, 3,4,6,7, 8- hexahydro-lH-[l,4]diazepino[6,7,l-ij]quinoline obtained in step 1.5

The separation was carried out in two steps via method A.

Two peaks were separated on a Daicel Chiralpak® AS-H column (150x4.6mm, 5 μιη) (t R [min] = 1.4, 1.7). The gradient used holds at 95%> C0 2 and 5%> modifier for 1 min. During 7 mins, it raises up to 50% modifier, which is then held for 1 min. Afterwards, the modifier is ramped back to 5% over 1 min (10 min total). As modifier, MeOH with 0.1 Vol%> of diethylamine was used.

Enantiomer l .A of 8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[ 6,7,l- ij]quinolone (single enantiomer of compound 1.5)

ESI-MS: m/z (%): 257.20 (100, [M+H] + ).

The retention time according to the method described above is 1.4 min.

Enantiomer l .B of 8-(trifluoromethyl)-2, 3,4,6,7, 8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinolone (single enantiomer of compound 1.5)

ESI-MS: m/z (%): 257.20 (100, [M+H] + ).

The retention time according to the method described above is 1.7 min. Example 2

8-(2,2,2-Trifluoroethyl)-2, 3,4,6,7, 8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quino line (compound of formula la.1 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CH 2 CF 3 )

2.1 4-(2,2,2-Trifluoroethyl)-3,4-dihydroquinolin-2(lH)-one

In a 5 mL microwave vial containing ethyl (E)-5,5,5-trifluoropent-2-enoate (100 mg, 0.549 mmol, 1.00 eq), 2-aminophenyl boronic ester (150 mg, 1.10 mmol, 2.00 eq), potassium carbonate (152 mg, 1.10 mmol), 2.00 eq) and [RhOH(COD)] 2 (12.52 mg, 0.027 mmol, 0.05 eq) was added 2%>wt. TPGS-750-M solution in water (3 mL). The mixture was stirred vigorously at ambient temperature for 24 h. The reaction mixture was then extracted with ethyl acetate. Then the organic phase was dried over MgSC^, filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (94 mg, 75%) yield).

1H NMR (500 MHz, CDC1 3 ): δ 9.28 (s, 1H), 7.28 - 7.17 (m, 2H), 7.05 (td, J= 7.5, 1.2 Hz, 1H), 6.92 - 6.86 (m, 1H), 3.40 (dq, J= 9.1, 5.0 Hz, 1H), 2.84 (dd, J= 16.4, 5.9 Hz, 1H), 2.71 (dd, J= 16.4, 3.6 Hz, 1H), 2.47 - 2.32 (m, 2H). 13 C NMR (125 MHz, CDC1 3 ): δ 170.43, 136.39, 128.62, 127.67, 126.20 (q, J= 277.0 Hz), 124.94, 123.67, 116.23, 37.82 (q, J= 27.6 Hz), 35.80, 30.96.

1 9 F NMR (470 MHz, CDC1 3 ): δ -63.33 (t, J= 10.6 Hz).

ESI-MS: m/z (%): 230.20 (100, [M+H] + ).

2.2 4-(2,2,2-Trifluoroethyl)-l,2,3,4-tetrahydroquinoline

229 mg of 4-(2,2,2-trifluoroethyl)-3,4-dihydroquinolin-2(lH)-one from step 2.1 (0.999 mmol, 1.00 eq) was dissolved in 10 mL of THF (anhydrous) and 2.50 mL of borane dimethyl sulfide complex (2 M in THF, 5.00 mmol, 5.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 8 h. Then the reaction mixture was quenched with HC1 (1 M) and the aqueous phase was extracted with ethyl acetate. Subsequently the organic phase was extracted three times with HC1 (1M) and to the aqueous phase was added NaOH (1M) until pH 10. This was then extracted with dichloromethane three times. The organic phase was dried over MgS0 4 and reduced under vac- uum. The crude product was purified by column chromatography on silica (eluent: 0- 10% methanol in dichloromethane) to yield the title compound (170 mg, 79% yield). ESI-MS: m/z (%): 216.20 (100, [M+H] + ).

2.2 2-(4-(2,2,2-Trifluoroethyl)-3,4-dihydroquinolin-l(2H)-yl)ace tamide

A mixture of 170 mg of 4-(2,2,2-trifluoroethyl)-l,2,3,4-tetrahydroquinoline from step 2.2 (0.790 mmol, 1.00 eq), 292 mg of 2-iodoacetamide (1.58 mmol, 2.00 eq) and 0.537 mL of DIPEA (3.16 mmol, 4.00 eq) in DMF (3 mL) was heated in a microwave at 110 °C for 8 h and further 3 h at 130 °C. Then NaOH (1M) was added and the mixture was extracted with dichloromethane. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica

(eluent: 0-10% methanol in dichloromethane) to yield the title compound (168 mg, 78% yield).

ESI-MS: m/z (%): 273.20 (100, [M+H] + ). 2.3 2-(4-(2,2,2-Trifluoroethyl)-3,4-dihydroquinolin- 1 (2H)-yl)ethanamine

165 mg of 2-(4-(2,2,2-trifluoroethyl)-3,4-dihydroquinolin-l(2H)-yl)ace tamide from step 2.2 (0.606 mmol, 1.00 eq) was dissolved in 4 mL of THF (anhydrous) and 0.61 mL of borane dimethyl sulfide complex (2 M in THF, 1.212 mmol, 2.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 2 h. Then the reaction mixture was quenched with HCl (1 M) and the aqueous phase was extracted with ethyl acetate. Subsequently the organic phase was extracted three times with HCl (1M) and to the aque- ous phase was added NaOH (1M) until pH 10. This was then extracted with dichloro- methane three times. The organic phase was dried over MgSC^ and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0- 10% methanol in dichloromethane) to yield the title compound (90 mg, 52% yield). ESI-MS: m/z (%): 259.20 (100, [M+H] + ).

2.4 8-(2,2,2-Trifluoroethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]diaze pino[6,7,l- ij]quinoline

A mixture of 90 mg of 2-(4-(2,2,2-trifluoroethyl)-3,4-dihydroquinolin-l(2H)- yl)ethanamine from step 2.3 (0.314 mmol, 1.00 eq), 23 μΐ ^ of formaldehyde (0.314 mmol, 1.00 eq) and 27 μΐ ^ of TFA (0.314 mmol, 1.00 eq) was stirred at ambient temperature for 72 h. Then NaOH (1M) was added and the mixture was extracted with dichloromethane. The organic phase was dried over MgSC^ and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (62 mg, 72% yield).

1H NMR (600 MHz, DMSO-< 6 ) 6 7.01 (dd, J= 7.7, 1.5 Hz, 1H), 6.95 (dd, J= 7.3, 1.6 Hz, 1H), 6.72 (t, J= 7.4 Hz, 1H), 3.72 (d, J= 14.1 Hz, 1H), 3.64 (d, J= 14.1 Hz, 1H), 3.21 - 3.17 (m, 2H), 3.13 - 3.07 (m, 1H), 3.03 - 2.96 (m, 2H), 2.93 - 2.83 (m, 2H), 1.81 - 1.73 (m, 1H), 3.47 - 3.43 (m, 1H), 2.67 - 2.51 (m, 2H), 1.92 - 1.83 (m, 1H).

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

Example 3

8-(Difluoromethyl)-2, 3,4,6,7, 8-hexahydro- lH-[ 1 ,4]diazepino[6,7, 1 -ij]quinoline

(compound of formula la.1 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CHF 2 )

3.1 4-(Difluoromethyl)-3 ,4-dihydroquino lin-2( 1 H)-one In a 5 mL microwave vial containing ethyl (E)-4,4-difluorobut-2-enoate (110 mg, 0.67 mmol, 1.00 eq), 2-aminophenyl boronic ester (183 mg, 1.33 mmol, 2.00 eq), potassium carbonate (184 mg, 1.33 mmol), 2.00 eq) and [RhOH(COD)] 2 (15 mg, 0.033 mmol, 0.05 eq) was added 2%wt. TPGS-750-M solution in water (3 mL). The mixture was stirred vigorously at ambient temperature for the 24 h. The reaction mixture was then extracted with ethyl acetate. Then the organic phase was dried over MgS0 4 , filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (70 mg, 50% yield).

1H NMR (500 MHz, CDCls): 5 9.05 (s, 1H), 7.27 (dtd, J= 15.4, 7.7, 1.4 Hz, 2H), 7.06 (td, J= 7.5, 1.1 Hz, 1H), 6.88 (dd, J= 7.9, 1.2 Hz, 1H), 5.85 (td, J= 56.0, 4.3 Hz, 1H), 5.74 (d, J= 4.3 Hz, 1H), 3.44 - 3.33 (m, 1H), 2.94 - 2.82 (m, 2H).

1 3 C NMR (125 MHz, CDC1 3 ): δ 169.41, 137.65, 129.55, 129.46, 123.48, 117.92 (t, J = 3.8 Hz), 116.26 (t, J= 245.4 Hz), 116.12, 40.95 (t, J= 22.1 Hz), 29.64.

1 9 F NMR (470 MHz, CDC1 3 ): δ -120.87 (ddd, J= 280.0, 55.9, 13.4 Hz), -123.60 (ddd, J = 280.1, 56.2, 16.5 Hz).

ESI-MS: m/z (%): 198.10 (100, [M+H] + ).

3.2 4-(Difluoromethyl)-l,2,3,4-tetrahydroquinoline

390 mg of 4-(difluoromethyl)-3,4-dihydroquinolin-2(lH)-one from step 3.1 (1.98 mmol, 1.00 eq) was dissolved in 4 mL of THF (anhydrous) and 3.96 mL of borane dimethyl sulfide complex (2 M in THF, 7.92 mmol, 4.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 10 h. Then the reaction mixture was quenched with HCl (1 M) and the aqueous phase was extracted with ethyl acetate. Sub- sequently the organic phase was extracted three times with HCl (1M) and to the aqueous phase was added NaOH (1M) until pH 10. This was then extracted with dichloromethane three times. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0- 10% methanol in dichloromethane) to yield the title compound (220 mg, 55% yield). ESI-MS: m/z (%): 184.10 (100, [M+H] + ).

3.3 2-(4-(Difluoromethyl)-3 ,4-dihydroquino lin- 1 (2H)-yl)acetamide A mixture of 220 mg of 4-(difluoromethyl)-l,2,3,4-tetrahydroquinoline from step 3.2 (1.20 mmol, 1.00 eq), 666 mg of 2-iodoacetamide (3.60mmol, 3.00 eq) and 1 mL of DIPEA (6.00 mmol, 5.00 eq) in DMF (3 mL) was heated in a microwave at 100 °C for 11 h. Then NaOH (1M) was added and the mixture was extracted with dichloromethane. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound (260 mg, 90%> yield).

ESI-MS: m/z (%): 241.20 (100, [M+H] + ). 3.4 2-(4-(Difluoromethyl)-3 ,4-dihydroquinolin- 1 (2H)-yl)ethanamine

260 mg of 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)acetamide from step 3.3 (1.08 mmol, 1.00 eq) was dissolved in 3 mL of THF (anhydrous) and 2.71 mL of borane dimethyl sulfide complex (2 M in THF, 5.41 mmol, 5.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 5 h. Then the reaction mixture was quenched with HC1 (1 M) and the aqueous phase was extracted with ethyl acetate. Subsequently the organic phase was extracted three times with HC1 (1M) and to the aqueous phase was added NaOH (1M) until pH 10. This was then extracted with dichloromethane three times. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0- 10% methanol in dichloromethane) to yield the title compound (155 mg, 57% yield). ESI-MS: m/z (%): 227.20 (100, [M+H] + ).

3.5 8-(Difluoromethyl)-2, 3,4,6,7, 8-hexahydro- lH-[ 1 ,4]diazepino[6,7, 1 -ij]quinoline

A mixture of 155 mg of 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)- yl)ethanamine from step 3.4 (0.685 mmol, 1.00 eq), 0.055 mL of formaldehyde (0.719 mmol, 1.05 eq), and 0.058 mL of TFA (0.754 mmol, 1.10 eq) in ethanol (2 mL) was stirred for 7 h at ambient temperature. Then NaOH (1M) was added and the mixture was extracted with dichloromethane. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-20% methanol in dichloromethane + 0.1% N¾ in ethanol) to yield the title compound (51 mg, 26% yield). 1H NMR (600 MHz, DMSO-d 6 ) δ 8.83 (s, 1H), 7.34 - 7.21 (m, 2H), 6.91 (t, J= 7.5 Hz, 1H), 6.35 (td, J= 56.1, 4.2 Hz, 1H), 4.18 (dt, J= 24.4, 4.4 Hz, 2H), 3.31 - 3.20 (m, 7H), 2.02 - 1.83 (m, 2H).

ESI-MS: m/z (%): 239.20 (100, [M+H] + ).

Example 4

4-Methyl-8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]d iazepino[6,7,l- ij]quinoline (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CF 3 )

4.1 Ethyl 2-(4-(trifluoromethyl)-3 ,4-dihydroquino lin- 1 (2H)-yl)propanoate

A mixture of 1.57 g of 4-(trifluoromethyl)-l,2,3,4-tetrahydroquinoline (7.80 mmol, 1.00 eq) from step 1.2, 14.1 g of ethyl 2-bromopropanoate (78.0 mmol, 10.0 eq and 15.1 mL of DIPEA (117 mmol, 15.0 eq) in DMF (38 mL) was heated in a microwave for 1 h at 175 °C. Then the mixture was diluted with ethyl acetate and washed with aqueous sodium bicarbonate solution. The aqueous phase was extracted three times with ethyl acetate and the combined organic phases were dried over Na 2 SC"4, filtrated and concentrated in vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in dichloromethane) to yield the title compound.

ESI-MS: m/z (%): 302.20 (100, [M+H] + ).

4.2 2-(4-(Trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propanoi c acid

A mixture of 2.59 g of ethyl 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)- yl)propanoate (8.60 mmol, 1.00 eq) from step 4.1 and 43.0 mL of NaOH (1M in H 2 0, 43.0 mmol, 5.00 eq) in MeOH (40 mL) was stirred at room temperature for 16 h. Then saturated aqueous sodium bicarbonate solution was added and the mixture was washed with ethyl acetate. 1M HCl was added to the aqueous phase until pH 3 was reached. The aqueous phase was extracted 5 times with DCM and the organic phase was dried over Na 2 SC"4, filtrated and concentrated in vacuum to yield the title compound (47%, 4.02 mmol).

ESI-MS: m/z (%): 274.20 (100, [M+H] + ). 4.3 2-(4-(Trifluoromethyl)-3 ,4-dihydroquinolin- 1 (2H)-yl)propanamide

To a solution of 1.10 g of 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)- yl)propanoic acid (4.02 mmol, 1.00 eq) from step 4.2, 430 mg of ammonium chloride (8.04 mmol, 2.00 eq) and 0.702 mL of DIPEA (4.02 mmol, 1.00 eq) in DMF (20 mL) was added 1.54 g of EDC (8.04 mmol, 2.00 eq) and 616 mg HOBt (4.02 mmol, 1.00 eq) and the mixture was stirred for 16 h at room temperature. Then DCM was added and the mixture was washed with saturated aqueous sodium bicarbonate solution. The organic layer was dried over Na 2 S0 4 , filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in di- chloromethane) to yield the title compound (86%, 3.46 mmol).

ESI-MS: m/z (%): 273.20 (100, [M+H] + ).

4.4 2-(4-(Trifluoromethyl)-3 ,4-dihydroquinolin- 1 (2H)-yl)propan- 1 -amine To 942 mg of 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propanam ide (3.46 mmol, 1.00 eq) from step 4.3 and 8.65 mL of borane dimethyl sulfide complex (1 M in THF, 17.3 mmol, 5.00 eq) was slowly added. The mixture was stirred for 72 h at room temperature and was then heated at 90 °C for 2 h. Then the reaction mixture was quenched with HCl (1 M) and the aqueous phase was extracted with ethyl acetate. The organic phase was extracted three times with HCl (1M) and to the aqueous phase was added NaOH (1M) until pH 10 and was then extracted with dichloro methane three times. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in dichloromethane) to yield the title compound (1.68 mmol, 49%).

ESI-MS: m/z (%): 259.20 (100, [M+H] + ).

4.5 4-Methyl-8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH-[l,4]d iazepino[6,7,l- ij]quinoline

A mixture of 444 mg of 2-(4-(trifluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propan-l - amine (1.72 mmol, 1.00 eq) from step 4.4, 0.128 mL of formaldehyde (1.72 mmol, 1.00 eq) and 0.146 mL of TFA (1.89 mmol, 1.10 eq) was stirred at ambient temperature for 72 h. Then the solvent was removed under vacuum. The residue was dissolved with DCM and washed with saturated aqueous sodium bicarbonate solution. The organic layer was dried over Na 2 S0 4 , filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in di- chloromethane) to yield the title compound (40%, 0.688 mmol).

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

4.6 Analytical separation of the four isomers of 4-methyl-8-(trifluoromethyl)- 2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quinoline obtained in step 4.5

Four peaks were separated on a Daicel Chiralcel® OD-H column (tR [min] = 1.5, 1.7, 1.9, 2.2). The mobile phase consisted of 93% C0 2 and 7% modifier. As modifier, MeOH with 0.2 Vol% of aqueous ammonia solution (25%) was used.

4.7 Preparative separation of the four isomers of 4-methyl-8-(trifluoromethyl)- 2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quinoline obtained in step 4.5

All 4 Isomers were separated in one step by UV-detection at 254 nm:

Four peaks were separated on a Daicel Chiralcel® OD-H column (250x20mm, 5 μιη) (t R [min] = 4.6, 5.1, 6.1, 6.6). The mobile phase consisted of 95% C0 2 and 5% modifier. As modifier, MeOH with 0.2 Vol% of aqueous ammonia solution (25%) was used.

Isomer 4.A of 4-methyl-8-(trifluoromethyl)-2, 3,4,6,7, 8-hexahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 1 of the compound of step 4.5)

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

The retention time according to the analytical method described above is 1.5 min and according to the preparative method described above is 4.6 min.

1H NMR (600 MHz, CDC1 3 ): δ 7.20 (d, J= 8.0, 1.8 Hz, 1H), 7.10 - 7.06 (m, 1H), 6.82 (t, J= 7.5 Hz, 1H), 3.98 (d, J= 13.6 Hz, 1H), 3.74 (d, J= 13.7 Hz, 1H), 3.53 - 3.42 (m, 1H), 3.42 - 3.34 (m, 1H), 3.34 - 3.24 (m, 2H), 3.12 (dd, J= 13.5, 2.6 Hz, 1H), 2.81 (dd, J= 13.5, 3.8 Hz, 1H), 2.19 - 2.02 (m, 2H), 1.07 (d, J= 6.8 Hz, 3H). (NH not detected) Isomer 4.B of 4-methyl-8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 1 of the compound of step 4.5)

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

The retention time according to the analytical method described above is 1.7 min and according to the preparative method described above is 5.1 min.

1H NMR (600 MHz, CDC1 3 ): δ 7.21 (d, J= 7.8 Hz, 1H), 7.11 - 7.07 (m, 1H), 6.82 (t, J = 7.5 Hz, 1H), 4.00 (d, J= 13.6 Hz, 1H), 3.76 (d, J= 13.6 Hz, 1H), 3.53 - 3.45 (m, 1H), 3.40 - 3.34 (m, 1H), 3.34 - 3.25 (m, 2H), 3.13 (dd, J= 13.6, 2.6 Hz, 1H), 2.83 (dd, J = 13.5, 3.8 Hz, 1H), 2.18 - 2.02 (m, 2H), 1.10 (d, J= 6.8 Hz, 3H). (NH not detected)

Isomer 4.C of 4-methyl-8-(trifluoromethyl)-2,3,4,6,7,8-hexahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 2 of the compound of step 4.5)

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

The retention time according to the analytical method described above is 1.9 min and according to the preparative method described above is 6.1 min.

1H NMR (600 MHz, CDC1 3 ): δ 7.19 (d, J= 7.7 Hz, 1H), 7.13 (dd, J= 7.5, 1.6 Hz, 1H), 6.82 (t, J= 7.5 Hz, 1H), 4.11 (d, J= 14.0 Hz, 1H), 3.70 (d, J= 14.0 Hz, 1H), 3.45 (dt, J = 9.8, 5.3 Hz, 1H), 3.36 (td, J= 6.7, 3.4 Hz, 2H), 3.31 (ddd, J= 12.5, 3.7, 1.4 Hz, 1H), 3.08 (dd, J= 13.6, 2.7 Hz, 1H), 3.02 (dd, J= 13.5, 3.2 Hz, 1H), 2.33 - 2.14 (m, 1H), 2.06 - 1.92 (m, 1H), 1.00 (d, J= 6.8 Hz, 3H). (NH not detected)

Isomer 4.D of 4-methyl-8-(trifluoromethyl)-2, 3,4,6,7, 8-hexahydro-lH- [ 1 ,4]diazepino[6,7, 1 -ij]quinoline (single enantiomer of diastereomer 2 of the compound of step 4.5)

ESI-MS: m/z (%): 271.20 (100, [M+H] + ).

The retention time according to the analytical method described above is 2.2 min and according to the preparative method described above is 6.6 min.

1H NMR (600 MHz, CDC1 3 ): 5 7.15 (d, J= 7.8 Hz, 1H), 7.09 (dd, J= 7.4, 1.6 Hz, 1H), 6.79 (t, J= 7.5 Hz, 1H), 4.03 (d, J= 14.1 Hz, 1H), 3.62 (d, J= 14.2 Hz, 1H), 3.49 - 3.40 (m, 1H), 3.39 - 3.25 (m, 3H), 3.05 (dd, J= 13.7, 2.7 Hz, 1H), 2.93 (dd, J= 13.6, 3.1, 0.9 Hz, 1H), 2.26 (dq, J= 14.3, 3.6 Hz, 1H), 2.06 - 1.92 (m, 1H), 0.92 (d, J= 6.9 Hz, 3H). (NH not detected)

Example 5

8-(Difiuoromethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4 ]diazepino[6,7,l- ij]quinoline (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CHF 2 )

5.1 Ethyl 2-(4-(difluoromethyl)-3 ,4-dihydroquino lin- 1 (2H)-yl)propanoate A mixture of 416 mg of 4-(difluoromethyl)-l,2,3,4-tetrahydroquinoline (2.27 mmol,

1.00 eq) from step 3.2, 4.11 g of ethyl 2-bromopropanoate (22.7 mmol, 10.0 eq and 5.95 mL DIPEA (34.1 mmol, 15. 0 eq) in DMF (10 mL) was heated in a microwave for 1 h at 150 °C. Then the mixture was diluted with ethyl acetate and washed with aqueous sodium bicarbonate solution. The aqueous phase was extracted three times with ethyl acetate and the combined organic phases were dried over Na 2 S0 4 , filtrated and concentrated in vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in dichloromethane) to yield the title compound.

ESI-MS: m/z (%): 284.20 (100, [M+H] + ). 5.2 2-(4-(Difluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propanoic acid

A mixture of 1.07 g of ethyl 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)- yl)propanoate (3.78 mmol, 1.00 eq) from step 5.1 and 18.9 mL of NaOH (1M in H 2 0, 18.9 mmol, 5.00 eq) in MeOH (20 mL) was stirred at room temperature for 16 h. Then saturated aqueous sodium bicarbonate solution was added and the mixture was washed with ethyl acetate. 1M HCl was added to the aqueous phase until pH 3 was reached. The aqueous phase was extracted 5 times with DCM and the organic phase was dried over Na 2 S0 4 , filtrated and concentrated in vacuum to yield the title compound (55%, 2.06 mmol).

ESI-MS: m/z (%): 256.30 (100, [M+H] + ).

5.3 2-(4-(Difluoromethyl)-3 ,4-dihydroquino lin- 1 (2H)-yl)propanamide To a solution of 526 mg of 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)- yl)propanoic acid (2.06 mmol, 1.00 eq) from step 5.2, 220 mg of ammonium chloride (4.12 mmol, 2.00 eq) and 0.360 mL of DIPEA (2.06 mmol, 1.00 eq) in DMF (10 mL) was added 789 mg of EDC (4.12 mmol, 2.00 eq) and 315 mg of HOBt (2.06 mmol, 1.00 eq) and the mixture was stirred for 16 h at room temperature. Then DCM was added and the mixture was washed with saturated aqueous sodium bicarbonate solution. The organic layer was dried over Na 2 S0 4 , filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in dichloromethane) to yield the title compound (64%, 1.31 mmol).

ESI-MS: m/z (%): 255.20 (100, [M+H] + ).

5.4 2-(4-(Difluoromethyl)-3 ,4-dihydroquinolin- 1 (2H)-yl)propan- 1 -amine

To 333 mg of 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propanami de (1.31 mmol, 1.00 eq) from step 5.3 and 6.54 mL of borane dimethyl sulfide complex (1 M in THF, 6.54 mmol, 5.00 eq) was slowly added. The mixture was stirred for 72 h at room temperature and was then heated in a microwave at 90 °C for 30 min. The reaction mixture was quenched with HC1 (1 M) and the aqueous phase was extracted with ethyl acetate. The organic phase was extracted three times with HC1 (1M) and to the aqueous phase was added NaOH (1M) until pH 10 and was then extracted with dichloromethane three times. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30%> methanol in dichloromethane) to yield the title compound (0.720 mmol, 55%).

ESI-MS: m/z (%): 241.25 (100, [M+H] + ). 5.5 8-(Difiuoromethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]di azepino[6,7,l- ij]quinoline

A mixture of 173 mg of 2-(4-(difluoromethyl)-3,4-dihydroquinolin-l(2H)-yl)propan-l- amine (0.720 mmol, 1.00 eq) from step 5.4, formaldehyde (0.720 mmol, 1.00 eq) and TFA (0.792 mmol, 1.10 eq) was stirred at ambient temperature for 72 h. Then the sol- vent was removed under vacuum. The residue was dissolved with DCM and washed with saturated aqueous sodium bicarbonate solution. The organic layer was dried over Na 2 S0 4 , filtrated and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-30% methanol in dichloromethane) to yield the title compound (45%, 0.326 mmol).

ESI-MS: m/z (%): 253.25 (100, [M+H] + ).

1H NMR (600 MHz, DMSO- g): 6 7.11 (d, J= 7.6 Hz, 1H), 6.99 (dd, J= 7.3, 1.5 Hz, 1H), 6.74 (t, J= 7.5 Hz, 1H), 6.29 (td, J= 56.2, 4.1 Hz, 1H), 3.80 (d, J= 13.4 Hz, 1H), 3.47 (d, J= 13.4 Hz, 1H), 3.40 - 3.31 (m, 1H), 3.30 - 3.21 (m, 2H), 3.21 - 3.16 (m, 1H), 2.85 (dd, J= 13.2, 2.6 Hz, 1H), 2.68 (dd, J= 13.1, 3.5 Hz, 1H), 1.98 - 1.89 (m, 1H), 1.89 - 1.80 (m, 1H), 0.91 (d, J= 6.7 Hz, 3H). (NH not detected)

5.6 Analytical separation of the four isomers of 8-(difluoromethyl)-4-methyl- 2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quinoline obtained in step 5.5

Three peaks were separated on a Daicel Chiralcel® OD-H column (tR [min] = 1.9, 2.1 , 2.5). The mobile phase consisted of 93% C0 2 and 7% modifier. As modifier, MeOH with 0.2 Vol% of aqueous ammonia solution (25%) was used.

The second peak could be further separated on a Daicel Chiralpak® AD-H column (tR [min] = 1.8, 2.1). The mobile phase consisted of 93%> C0 2 and 7%> modifier. As modifier, MeOH with 0.2 Vol%> of aqueous ammonia solution (25%>) was used. 5.7 Preparative separation of the four isomers of 8-(difluoromethyl)-4-methyl- 2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quinoline obtained in step 5.5

The separation of the 4 isomers was carried out in two steps by UV-detection at 254 nm:

Three peaks were separated on a Daicel Chiralcel® OD-H column (250x20mm, 5 μιη) (t R [min] = 2.7, 3.2, 3.5). The mobile phase consisted of 90%> C0 2 and 10%> modifier. As modifier, MeOH with 0.1 Vol% of triethylamine was used.

The second peak consisted of two isomers, that were subsequently separated on a YMC CHIRAL Amylose-C column (250x20mm, 5 μπι) (t R [min] = 3.9, 4.3). The mobile phase consisted of 93%> C02 and 7%> modifier. As modifier, MeOH with 0.2 Vol%> of aqueous ammonia solution (25%>) was used. Isomer 5.A of 8-(difluoromethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 1 of the compound of step 5.5)

ESI-MS: m/z (%): 253.25 (100, [M+H] + ).

The retention time according to the analytical method described above is 1.9 min and according to the preparative method described above is 2.7 min.

1H NMR (600 MHz, DMSO-d 6 ): δ 7.11 (d, J= 7.6 Hz, 1H), 6.99 (dd, J= 7.3, 1.5 Hz, 1H), 6.74 (t, J= 7.5 Hz, 1H), 6.29 (td, J= 56.2, 4.1 Hz, 1H), 3.80 (d, J= 13.4 Hz, 1H), 3.47 (d, J= 13.4 Hz, 1H), 3.40 - 3.31 (m, 1H), 3.30 - 3.21 (m, 2H), 3.21 - 3.16 (m, 1H), 2.85 (dd, J= 13.2, 2.6 Hz, 1H), 2.68 (dd, J= 13.1, 3.5 Hz, 1H), 1.98 - 1.89 (m, 1H), 1.89 - 1.80 (m, 1H), 0.91 (d, J= 6.7 Hz, 3H). (NH not detected)

Isomer 5.B of 8-(difluoromethyl)-4-methyl-2, 3,4,6,7, 8-hexahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 1 of the compound of step 5.5)

ESI-MS: m/z (%): 253.25 (100, [M+H] + ).

The retention time according to the analytical method described above is 1.8 min and according to the preparative method described above is 3.9 min.

1H NMR (600 MHz, DMSO-d 6 ) δ 7.04 (dd, J= 7.7, 1.6 Hz, 1H), 6.98 (dd, J= 7.3, 1.6 Hz, 1H), 6.70 (t, J= 7.4 Hz, 1H), 6.27 (td, J= 56.3, 4.8 Hz, 1H), 3.84 (d, J= 13.5 Hz, 1H), 3.39 (d, J= 13.6 Hz, 1H), 3.28 - 3.12 (m, 4H), 2.80 (dd, J= 13.1, 2.5 Hz, 1H), 2.73 (dd, J= 13.1, 3.1 Hz, 1H), 2.07 - 1.96 (m, 1H), 1.88 - 1.75 (m, 1H), 0.87 (d, J = 6.7 Hz, 3H). (NH not detected) Isomer 5.C of 8-(difluoromethyl)-4-methyl-2, 3,4,6,7, 8-hexahydro-lH-

[l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 2 of the compound of step 5.5)

ESI-MS: m/z (%): 253.25 (100, [M+H] + ).

The retention time according to the analytical method described above is 2.1 min and according to the preparative method described above is 4.3 min.

1H NMR (600 MHz, DMSO-d 6 ) δ 7.12 (d, J= 7.6 Hz, 1H), 7.00 (dd, J= 7.3, 1.6 Hz, 1H), 6.75 (t, J= 7.5 Hz, 1H), 6.29 (td, J= 56.2, 4.1 Hz, 1H), 3.81 (d, J= 13.4 Hz, 1H), 3.50 (d, J= 13.4 Hz, 1H), 3.41 - 3.28 (m, 1H), 3.30 - 3.22 (m, 2H), 3.22 - 3.14 (m, 1H), 2.86 (dd, J= 13.1, 2.6 Hz, 1H), 2.70 (dd, J= 13.1, 3.5 Hz, 1H), 1.98 - 1.89 (m, 1H), 1.89 - 1.81 (m, 1H), 0.92 (d, J= 6.7 Hz, 3H). (NH not detected) Isomer 5.D of 8-(difluoromethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-

[l,4]diazepino[6,7,l-ij]quinoline (single enantiomer of diastereomer 2 of the compound of step 5.5)

ESI-MS: m/z (%): 253.25 (100, [M+H] + ).

The retention time according to the analytical method described above is 2.5 min and according to the preparative method described above is 3.5 min.

1H NMR (600 MHz, DMSO-d 6 ) δ 7.05 (dd, J= 7.5, 1.6 Hz, 1H), 6.99 (dd, J= 7.3, 1.6 Hz, 1H), 6.71 (t, J= 7.4 Hz, 1H), 6.27 (td, J= 56.4, 4.8 Hz, 1H), 3.85 (d, J= 13.6 Hz, 1H), 3.41 (d, J= 13.6 Hz, 1H), 3.28 - 3.09 (m, 4H), 2.81 (dd, J= 13.1, 2.5 Hz, 1H), 2.75 (dd, J= 13.1, 3.2 Hz, 1H), 2.07 - 1.96 (m, 1H), 1.86 - 1.75 (m, 1H), 0.88 (d, J = 6.7 Hz, 3H). (NH not detected)

Example 6

(4-Methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]q uinolin-8-yl)methanol (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CH 2 OH)

6.1 ( 1 -( 1 - Aminopropan-2-yl)- 1 ,2,3 ,4-tetrahydroquino lin-4-yl)methano 1

500 mg of l-(2-amino-l-methyl-2-oxo-ethyl)-3,4-dihydro-2H-quinoline-4- carboxylic acid (2.014 mmol, 1.00 eq) was dissolved in 1 mL of THF (anhydrous) and 5.03 mL of borane dimethyl sulfide complex (2 M in THF, 10.07 mmol, 5.00 eq) was slowly added. The mixture was heated in a microwave at 90 °C for 2 h. Then the reaction mixture was quenched with HCl (1 M) and the aqueous phase was extracted with ethyl acetate. The organic phase was extracted three times with HCl (1M) and to the aqueous phase was added NaOH (1M) until pH 10 and was then extracted with dichloromethane three times. The organic phase was dried over MgSC^ and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound. ESI-MS: m/z (%): 221.20 (100, [M+H] + ).

6.2 (4-Methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]q uinolin-8- yl)methanol A mixture of 61 mg of ( 1 -( 1 -aminopropan-2-yl)- 1 ,2,3 ,4-tetrahydroquinolin-4- yl)methanol (0.277 mmol, 1.00 eq) obtained in step 6.1, formaldehyde (0.277 mmol,

1.00 eq) and TFA (0.305 mmol, 1.10 eq) was stirred at ambient temperature for 72 h. Then the solvent was removed under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-10% methanol in dichloromethane) to yield the title compound as TFA-salt (62%, 0.171 mmol).

ESI-MS: m/z (%): 233.30 (100, [M+H] + ).

Example 7

8-(Fluoromethyl)-4-methyl-2, 3,4,6,7, 8-hexahydro-lH-[l,4]diazepino[6,7,l-ij]quino line (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is CH 2 F)

7.1 tert-Butyl 8-(hydroxymethyl)-4-methyl-3,4,7,8-tetrahydro-lH- [ 1 ,4]diazepino[6,7, 1 -ij]quinoline-2(6H)-carboxylate 487 mg of (4-methyl-2, 3,4,6,7, 8-hexahydro-lH-[l,4]diazepino[6, 7, l-ij]quinolin-8- yl)methanol (2.096 mmol, 1.00 eq) from step 6.2 was dissolved in DCM (15 mL) and 0.585 mL of triethylamine (4.19 mmol, 2.00 eq) was added. Then 457 mg (2.096 mmol, 1.00 eq) of di-tert-butyldicarbonat was added and the mixture was stirred for 18 h at room temperature. Citric acid was added and the solution was stirred for 15 min. The organic phase was separated from the aqueous phase and the aqueous phase was extracted with DCM. The organic phase was dried over MgS0 4 and reduced under vacuum to yield the title compound.

ESI-MS: m/z (%): 333.30 (100, [M+H] + ). 7.2 tert-Butyl 4-methyl-8-(((methylsulfonyl)oxy)methyl)-3,4,7,8-tetrahydro- lH- [ 1 ,4]diazepino[6,7, 1 -ij]quinoline-2(6H)-carboxylate 269 mg oftert-butyl 8-(hydroxymethyl)-4-methyl-3,4,7,8-tetrahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline-2(6H)-carboxylate (0.810 mmol, 1.00 eq) from step 7.1 dissolved in DCM (5 mL) was cooled to 0 °C and 0.226 mL of triethylamine (1.62 mmol, 2.00 eq) was added. Then 0.066 mL of methanesulfonyl chloride (0.850 mmol, 1.05 eq) was added and the mixture was stirred for 1 h at 0 °C. Then it was warmed up to room temperature and stirred for further 16 h. After quenching with water the organic phase was separated from the aqueous phase and the aqueous phase was extracted with DCM. The organic phase was dried over MgS0 4 and reduced under vacuum. The crude product was purified by column chromatography on silica (eluent: 0-100% methanol in DCM) to yield the title compound (43%, 0.064 mmol).

ESI-MS: m/z (%): 411.20 (100, [M+H] + ).

7.3 tert-Butyl 8-(fluoromethyl)-4-methyl-3,4,7,8-tetrahydro- 1Η-[ 1 ,4]diazepino[6,7, 1 - ij]quinoline-2(6H)-carboxylate To 54 mg of tert-butyl 4-methyl-8-(((methylsulfonyl)oxy)methyl)-3,4,7,8-tetrahydro- lH-[l,4]diazepino[6,7,l-ij]quinoline-2(6H)-carboxylate (0.132 mmol, 1.00 eq) from step 7.2 dissolved in tert-butanol (2 mL) was added 80 mg of caesium fluoride (0.526, 4.00 eq). The mixture was heated for 2 h in a microwave at 80 °C. Then saturated aqueous sodium bicarbonate solution was added. The organic phase was separated from the aqueous phase and the aqueous phase was extracted with DCM. The organic phase was dried over MgS0 4 and reduced under vacuum to yield the title compound.

ESI-MS: m/z (%): 335.30 (100, [M+H] + ).

7.4 8-(Fluoromethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diaz epino[6,7,l- ij]quinoline

To a solution of 78 mg of tert-butyl 8-(fluoromethyl)-4-methyl-3,4,7,8-tetrahydro-lH- [l,4]diazepino[6,7,l-ij]quinoline-2(6H)-carboxylate (0.233 mmol, 1.00 eq) from step 7.3 in DCM (3 mL) was added 0.180 mL of trifluoroacetic acid (2.33 mmol, 10.00 eq) and the mixture was stirred for 3 d. Then the solvent was evaporated and the crude product was purified via column chromatography on silica (eluent: 0-100% ethyl acetate in heptane) which yielded the title compound (4.5%, 0.010 mmol). ESI-MS: m/z (%): 235.20 (100, [M+H] + ).

1H NMR (500 MHz, Methanol-^): δ 7.37 - 7.25 (m, 1H), 7.19 (dd, J= 7.4, 1.5 Hz, 1H), 6.94 (td, J= 7.6, 2.5 Hz, 1H), 4.84 - 4.44 (m, 3H), 4.28 (dd, J= 13.2, 9.3 Hz, 1H), 4.08 (dd, J= 15.8, 13.1 Hz, 1H), 3.70 - 3.44 (m, 1H), 3.43 - 3.31 (m, 1H), 3.31 - 3.15 (m, 3H), 2.07 - 1.85 (m, 1H), 1.96 - 1.92 (m, 1H), 1.31 - 1.20 (m, 4H).

Example 8

8-(Ethoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diaz epino[6,7,l-ij]quinoline (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is ethoxymethyl (CH 2 OCH 2 CH 3 )

8.1 tert-Butyl 8-(ethoxymethyl)-4-methyl-3,4,7,8-tetrahydro-lH-[l,4]diazepi no[6,7,l ij]quinoline-2(6H)-carboxylate

A solution of 50 mg of (4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinolin-8-yl)methanol (0.150 mmol, 1.00 eq) from step 6.2 in DMF (5 mL) was cooled to 0 °C. Then 9.02 mg of sodium hydride (0.226 mmol, 1.50 eq) was added and the mixture was stirred for 1 h at 0 °C. 28.1 mg iodoethane (0.180 mmol, 1.20 eq) was added and the mixture was stirred for 72 h at room temperature. Then the solvent was evaporated and the crude product was purified via preparative HPLC to yield the title compound.

ESI-MS: m/z (%): 361.50 (100, [M+H] + ).

8.2 8-(Ethoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diaz epino[6,7,l- ij]quinoline To a solution of 23.2 mg of tert-butyl 8-(ethoxymethyl)-4-methyl-3,4,7,8-tetrahydro- lH-[l,4]diazepino[6,7,l-ij]quinoline-2(6H)-carboxylate (0.064 mmol, 1.00 eq) from step 8.1 in DCM (5 mL) was added 99.0 μΕ of TFA (1.29 mmol, 20 eq) and the mixture was stirred for 24 h at room temperature. Then the solvent was evaporated and the crude product was purified via prep-HPLC to yield the title compound (10%, 0.006 mmol). ESI-MS: m/z (%): 261.20 (100, [M+H] + ). 1H NMR (500 MHz, Methanol-^): δ 7.36 - 7.20 (m, 1H), 7.16 (dd, J= 7.4, 1.6 Hz, 1H), 6.91 (t, J= 7.6 Hz, 1H), 4.28 (d, J= 13.1 Hz, 1H), 4.11 - 3.99 (m, 1H), 3.72 - 3.60 (m, 1H), 3.63 - 3.44 (m, 4H), 3.33 - 3.16 (m, 4H), 3.08 - 2.99 (m, 1H), 2.05 - 1.92 (m, 1H), 1.92 - 1.80 (m, 1H), 1.27 - 1.13 (m, 6H).

Example 9

8-(Methoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]dia zepino[6,7,l- ij]quinoline (compound of formula la.10 wherein R 4a , R 4b , R 5a , R 5b and R 7a are hydrogen and R 6 is methoxymethyl (CH 2 OCH 3 )

9.1 tert-Butyl 8-(methoxymethyl)-4-methyl-3,4,7,8-tetrahydro-lH- [ 1 ,4]diazepino[6,7, 1 -ij]quinoline-2(6H)-carboxylate

A solution of 50 mg of (4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]diazepino[6,7,l- ij]quinolin-8-yl)methanol (0.150 mmol, 1.00 eq) from step 6.2 in DMF (5 mL) was cooled to 0 °C. Then 9.02 mg of sodium hydride (0.226 mmol, 1.50 eq) was added and the mixture was stirred for 1 h at 0 °C. 32.3 mg of iodomethane (0.226 mmol, 1.50 eq) was added and the mixture was stirred for 72 h at room temperature. Then the solvent was evaporated and the crude product was purified via column chromatography on silica (eluent: 0-40% methanol in DCM) which yielded the title compound (80%, 0.120 mmol).

ESI-MS: m/z (%): 347.30 (100, [M+H] + ).

9.2 8-(Methoxymethyl)-4-methyl-2,3,4,6,7,8-hexahydro-lH-[l,4]dia zepino[6,7,l- ij]quinoline

To a solution of 41.6 mg of tert-butyl 8-(methoxymethyl)-4-methyl-3,4,7,8-tetrahydro- lH-[l,4]diazepino[6,7,l-ij]quinoline-2(6H)-carboxylate (0.120 mmol, 1.00 eq) from step 9.1 in DCM (5 mL) was added 185 μΐ. of TFA (2.40 mmol, 20.0 eq) and the mixture was stirred for 24 h at room temperature. Then the solvent was evaporated and the crude product was purified via preparative HPLC to yield the title compound (19%>, 0.023 mmol).

ESI-MS: m/z (%): 247.30 (100, [M+H] + ). 1H NMR (500 MHz, Methanol-^): δ 1H NMR (500 MHz, Methanol-^) δ 7.31 - 7.25 (m, 1H), 7.16 (dd, J= 7.6, 1.6 Hz, 1H), 6.91 (t, J= 7.6 Hz, 1H), 4.28 (d, J= 13.2 Hz, 1H), 4.04 (d, J= 13.2 Hz, 1H), 3.61 - 3.54 (m, 1H), 3.57 - 3.49 (m, 2H), 3.37 (s, 3H), 3.28 - 3.18 (m, 3H), 3.25 - 3.22 (m, 2H), 3.08 - 3.01 (m, 1H), 2.03 - 1.94 (m, 1H), 1.91 - 1.80 (m, 1H), 1.24 (d, J= 7.0 Hz, 3H).

II. Biological tests

Functional activity

1. Human 5-HT 2 c Functional Assay

The functional activity of compounds of formula I was assayed by incubation with U20S_HTR 2C _ -Arrestin cells (DiscoverX, 93-0289C3) to induce beta-arrestin2 recruitment to the 5-HT 2 c receptor. The agonist-induced recruitment and proximity of the receptor and beta-arrestin2 leads to complementation and formation of active β- galactosidase. The enzyme complementation results in enzyme activity, which is measured following the termination of the agonist incubation using DiscoveRx's detection reagent, which contains a chemiluminescent substrate which produces a high intensity signal. Cells were plated and a medium-change to a 1% serum containing medium was performed 24h later. The next day, test compounds were added and incubated for 1.5 h before addition of detection reagent.

The response produced was measured and compared with the response produced by 10 [mu]M 5-HT or the maximal effect induced by 5-HT (defined as 100%) to which it was expressed as a percentage response (relative efficacy). Dose response curves were con- structed using Graphpad Prism (Graph Software Inc.) or using in house adapted software using a 4 parameter dose response model with variable slope (fit = (Bottom + (Top-Bottom)/(l+10 A ((LogEC50-x)*HillSlope))res = (y-fit)). Results are compiled in the table below. 2. Human 5-HT 2A Functional Assay

Functional activity on the 5-HT 2A receptor was determined by testing the effect of the compounds I on calcium mobilisation in CHO-K1 cells, stably transfected with human 5-HT 2A receptor. Cells were seeded into sterile black 384-well plates with clear bottom at 25,000 cells/well in a volume of 25 μΐ and grown for 5-6 hours at 37°C, in 5% C0 2 in tissue culture medium ("Ultra CHO" by LONZA), containing 1% dialysed FCS and 50 μg/ml gentamicin (Invitrogen). After this incubation, medium was replaced by a serum free version of the same tissue culture medium followed by incubation overnight at 37°C and in 5% C0 2 . Cells were then loaded with a fluorescent calcium-sensitive dye in the presence of 0.07% probenecid for an hour at 37°C, according to the manufacturer's protocol (Ca5-Assay Kit, Molecular Devices), followed by an additional 60 min incubation at room temperature. Serial compound dilutions (final concentrations of 10 "10 to 10 "5 M, prepared in HBSS + 50 mM HEPES) were first added to the cells alone ("first addition" to assess agonism on the 5 -HT 2A receptor) , then after 8 min, serotonin was added to the same wells at a final concentration of 3x10 "8 M ("second addition" to see potential antagonistic effect) and the maximal calcium response was determined using a FLIPR® Tetra instrument (Molecular Devices) in each of the two steps. The relative efficacy of the compounds was calculated as a percentage of the maximal effect induced by serotonin alone (defined as 100%). To determine EC 50 / IC 50 values, concentration-response curves were fitted using a four-parameter logistic equation (IDBS Bio- book™). Kb values were calculated from IC 50 values, according to Cheng & Prusoff 3. Human 5 -HT 2B Functional Assay

Functional activity on the 5-HT 2B receptor was determined by testing the effect of the compounds I on calcium mobilisation in CHO-Flpln cells, stably transfected with human 5-HT 2B receptor. Cells were seeded into sterile black 384-well plates with clear bottom at 30,000 cells/well in a volume of 25 μΐ and grown overnight at 37°C, in 5% C0 2 in tissue culture medium ("CHO-S-SFM Π" by Invitrogen), containing 1% dialysed FCS and 50 μg/ml gentamicin (Invitrogen). On the next morning, medium was replaced by a serum free version of the same tissue culture medium for a further incubation for 4 hours at 37°C and in 5% C0 2 . Cells were then loaded with a fluorescent calcium-sensitive dye in the presence of 0.07%> probenecid for an hour at 37°C, according to the manufacturer's protocol (Ca5-Assay Kit, Molecular Devices), followed by an additional 60 min incubation at room temperature. Serial compound dilutions (final concentrations of 10 "10 to 10 "5 M, prepared in HBSS + 50 mM HEPES) were first added to the cells alone ("first addition" to assess agonism on the 5 -HT 2 B receptor) , then after 8 min, serotonin was added to the same wells at a final concentration of 10 ~8 M ("second addition" to see potential antagonistic effect) and the maximal calcium response was determined using a FLIPR® Tetra instrument (Molecular Devices) in each of the two 5 steps. The relative efficacy of the compounds was calculated as a percentage of the maximal effect induced by serotonin alone (defined as 100%). To determine EC 50 / IC 50 values, concentration-response curves were fitted using a four-parameter logistic equation (IDBS Biobook™). K b values were calculated from IC 50 values, according to Cheng & Prusoff

0

Table

# EC50 % EC50 % Selectivity 4 EC50 % Selectivity 5

5-HT2C 1 effi5-HT 2A 2 effi(based on 5-HT 2B 3 effi(based on cacy cacy agonism) cacy agonism)

1 ++ 85 antagonist 6 antagonist l .A ++ 57 antagonist antagonist l .B ++ 88 antagonist + 34 3

2 + 72 full 7 antagonist

3 ++ 93 antagonist full

4 ++ 104 antagonist antagonist

4.A +++ 101 antagonist antagonist

4.B ++ 87 antagonist antagonist

4.C +++ 101 ++ 24 2 ++ 44 12

4.D + 86 antagonist + 30 2

5 ++ 95 antagonist antagonist

5. A +++ 109 +++ 21 0.5 + 36 20

5.B ++ 1 10 antagonist antagonist

5.C + 89 full antagonist

5.D + 84 full antagonist

7 ++ 107 antagonist antagonist

8 + 93 full full

9 + 82 full full Potency (EC50 5-HT 2 c) in functional assay

Potency (EC50 5-HT 2A ) in functional assay

Potency (EC50 5-HT 2B ) in functional assay

EC50 5-HT 2A / EC50 5-HT 2C

EC50 5-HT 2B / EC50 5-HT 2C

"antagonist" indicates functional antagonism with no measurable agonistic effect "full" indicates no measurable agonistic effect at ΙΟμΜ (highest concentration in assay)

Potency (EC50):

* > 10 μΜ

+ from 200 nM to < ΙμΜ

++ from 20 nM to < 200 nM

+++ < 20 nM