NZ623727B2 - Inhibitors of phosphodiesterase type 10a - Google Patents

Inhibitors of phosphodiesterase type 10a Download PDF

Info

Publication number
NZ623727B2
NZ623727B2 NZ623727A NZ62372712A NZ623727B2 NZ 623727 B2 NZ623727 B2 NZ 623727B2 NZ 623727 A NZ623727 A NZ 623727A NZ 62372712 A NZ62372712 A NZ 62372712A NZ 623727 B2 NZ623727 B2 NZ 623727B2
Authority
NZ
New Zealand
Prior art keywords
ethyl
quinolinyl
pyridinyl
thieno
phthalazin
Prior art date
Application number
NZ623727A
Other versions
NZ623727A (en
Inventor
Berthold Behl
Lawrence A Black
Jurgen Dinges
Karla Drescher
Herve Geneste
Clarissa Jakob
Katja Jantos
Loic Laplanche
Michael Ochse
Sean Turner
Original Assignee
Abbvie Deutschland Gmbh & Co Kg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbvie Deutschland Gmbh & Co Kg filed Critical Abbvie Deutschland Gmbh & Co Kg
Priority claimed from PCT/EP2012/072150 external-priority patent/WO2013068470A1/en
Publication of NZ623727A publication Critical patent/NZ623727A/en
Publication of NZ623727B2 publication Critical patent/NZ623727B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/08Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing alicyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The disclosure relates to novel compounds of the formula (I) which are inhibitors of phosphodiesterase type 10A and to their use for the manufacture of a medicament and which thus are suitable for treating or controlling of medical disorders selected from neurological disorders and psychiatric disorders, for ameliorating the symptoms associated with such disorders and for reducing the risk of such disorders. wherein Q is O or S, X1 is N or CH, X2 is N or C-R7; X3 is O, S, -X4=C(R8)-, where C(R8) is bound to the carbon atom which carries R2, or -X5=C(R9)-, where X5 is bound to the carbon atom which carries R2; X4 is N or C-R9; X5 is N; Het is selected from optionally substituted phenyl, monocyclic hetaryl and fused bicyclic hetaryl; R1 is selected inter alia from hydrogen, halogen, OH, C1-C4-alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4- alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, the moiety Y1-Cyc1; R2 is selected inter alia from hydrogen, halogen, OH, C1-C4-alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, etc; A represents one of the following groups A1, A2, A3, A4 or A5: where * indicates the points of attachment to Het and to the nitrogen atom, respectively; and where R3 to R9, R3e, R3f, A', Y1 and Cyc are defined in the claims. Exemplary compounds are: 3,7-di(pyridin-4-yl)-5-[2-(quinolin-2-yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 8-(furan-3-yl)-2-[2-(quinolin-2-yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[l,2-a]pyridin-2-yl)ethyl]-8-(morpholin-4-yl)phthalazin-l(2H)-one. ders, for ameliorating the symptoms associated with such disorders and for reducing the risk of such disorders. wherein Q is O or S, X1 is N or CH, X2 is N or C-R7; X3 is O, S, -X4=C(R8)-, where C(R8) is bound to the carbon atom which carries R2, or -X5=C(R9)-, where X5 is bound to the carbon atom which carries R2; X4 is N or C-R9; X5 is N; Het is selected from optionally substituted phenyl, monocyclic hetaryl and fused bicyclic hetaryl; R1 is selected inter alia from hydrogen, halogen, OH, C1-C4-alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4- alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, the moiety Y1-Cyc1; R2 is selected inter alia from hydrogen, halogen, OH, C1-C4-alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, etc; A represents one of the following groups A1, A2, A3, A4 or A5: where * indicates the points of attachment to Het and to the nitrogen atom, respectively; and where R3 to R9, R3e, R3f, A', Y1 and Cyc are defined in the claims. Exemplary compounds are: 3,7-di(pyridin-4-yl)-5-[2-(quinolin-2-yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 8-(furan-3-yl)-2-[2-(quinolin-2-yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[l,2-a]pyridin-2-yl)ethyl]-8-(morpholin-4-yl)phthalazin-l(2H)-one.

Description

INHIBITORS OF PHOSPHODIESTERASE TYPE 10A The present invention relates to novel compounds which are inhibitors of phosphodiesterase type 10A and to their use for the manufacture of a medicament and which thus are suitable for treating or controlling of medical disorders selected from neurological disorders and psychiatric disorders, for ameliorating the symptoms associated with such disorders and for reducing the risk of such disorders.
Background of the Invention 10 odiesterase type 10A (hereinafter ) is a dual-substrate phosphodiesterase that can convert both CAMP to AMP and cGMP to GMP. PDE10A is highly prominent in the mammalian brain. In the rat, as well as in other mammalian species, PDE10A and the mRNA of PDE10A are highly ed in the GABAergic 15 medium spiny tion neurons (MSNs) ofthe striatal complex (caudate nucleus, nucleus accumbens, and olfactory tubercle) where the output is ted by the effect ofPDE10A on cAMP and cGMP signalling cascades (see e.g. C. J. Schmidt et al, The Journal of Pharmacology and Experimental Therapeutics 325 (2008) 681-690, A. Nishi, The Journal ofNeuroscience 2008, 28, 10450-10471). 20 MSNs express two functional classes ofneurons: the D1 class expressing D1 dopamine receptors and the D2 class expressing D2 dopamine receptors. The D1 class of neurons is part of the 'direct' al output pathway, which broadly ons to facilitate behavioral responses. The D2 class of neurons is part of the 'indirect' striatal output pathway, which functions to suppress behavioral responses that e with 25 those being facilitated by the 'direct' pathway. PDElOA regulation of cAMP and/or cGMP signaling in the dendritic tment of these neurons may be involved in ng the cortico/thalamic input into the MSN. Furthermore, PDEI 0A may be involved in the regulation of GABA release in the substantia nigra and globus pallidus (Seeger, T.F. et al. Brain Research, 2003, 985, 1 13-126). Inhibition ofPDEIOA s 30 in striatal activation and behavioral suppression such as dampened locomotion, inhibition of conditioned avoidance se (CAR), and activity in the rat auditory 2 gating model, suggesting that inhibitors ofphosphodiesterase type 10A represent a novel class of antipsychotic agents.
The hypotheses around the physiological role ofPDElOA and the therapeutic utility ofPDElOA inhibitors derive in part from studies with rine (J. A. k et al. loc. cit.), the first extensively d pharmacological tool compound for this target. The PDElOA inhibitor papaverine was shown to be active in several ychotic models. Papaverine potentiated the cataleptic effect of the D2 receptor antagonist haloperidol in rats, but did not cause catalepsy on its own (WO 03/093499).
Papaverine reduced hyperactivity in rats induced by PCP, while reduction of 10 amphetamine—induced hyperactivity was insignificant (WO 03/093499). These models suggest that PDElOA inhibition has the classic antipsychotic potential that would be expected from theoretical considerations. Papaverine, however has significant limitations in this regard with vely poor potency and selectivity and a very short exposure half-life after systemic administration. It was found that tion of PDElOA 15 reverses subchronic PCP-induced deficits in attentional set-shifting in rats ting that PDElOA inhibitors might alleviate cognitive deficits associated with schizophrenia.
(Rodefer et al., Eur. J. Neurosci, 4 (2005) 1070—1076).
The discovery of a new class ofPDE10A inhibitors with improved potency, selectivity, and pharmacokinetic properties, ed an opportunity to r explore 20 the physiology ofPDE 1 0A and the potential therapeutic utility of inhibiting this . The new class of inhibitors are exemplified by MP-lO (PF-2545920: 2- {4-[1- methylpyridineyl— l -H-pyrazol—3—31y]phenoxymethyl} -quino line) and TP- 1 0, i.e. 2- {4-[pyridineyl-l-(2,2,2-trifluoroethyl)—1-H-pyrazol—3 -3ly]phenoxymethyl} - quino line. The compounds offer a therapeutic approach to the ent of 25 schizophrenia (see C. J. Schmidt et al., loc cit.; S.M. Grauer eta1., Journal of acology and Experimental Therapeutics, fast forward DOI 10.1124 JPET 109.155994). Positive signals in rodent models of schizophrenia include the: attenuation of conditioned avoidance response (CAR), inhibition of hyperactivity caused by amphetamine—induced dopamine release or phencyclidine (PCP) mediated NMDA 30 receptor blockade, attenuation ofpharmaco logically impaired social or object recognition, and antagonism of apomorphine—induced climbing. Taken together, these data suggest a broad suppression of all 3 symptoms clusters (positive symptoms, 3 negative symptoms & cognitive dysfunctions) linked to schizophrenia (see C. J.
Schmidt et a1., loc cit.; S.M. Grauer et al., loc. cit).
Beyond schizophrenia, selective PDE10 inhibitiors may have the ial for the treatment of Huntington's disease (S. H. Francis et al., l. Rev., 91 (2011) 651- 690) and they may be a therapeutic option for substance abuse disorders (F. Sotty et al., J. Neurochem., 109 (2009) 766—775). Furthermore, it has been suggested that PDE10A inhibitors may be useful for treatment of y and non—insulin dependent es (see e.g. W0 2005/120514, W0 2005/012485, Cantin et a1, Bioorganic & Medicinal Chemistry Letters 17 (2007) 2869-2873). 10 In summary, inhibitors of PDElOA offer a promising therapeutic approach to the treatment or prevention of ogical and psychiatric disorders, in particular schizophrenia and related disorders, including symptoms linked to schizophrenia such as cognitive dysfunction.
Several classes of compounds which are inhibitors of PDElOA have been 15 described in the art, the recent compound groups are: Pyrido[3,2-e]pyridazines - see W0 2007/137819, W0 2007/137820, W0 68246, W0 2009/068320, WO 2009/070583 and W0 2009/070584; 4-substiuted phtha1azines and quinazolines WO 2007/085954, WO 2007/022280, W0 2007/096743, W0 2007/103370, WO 2008/020302, W0 2008/006372 and W0 20 2009/036766; 4-substiuted cinnazolines - see WO 2006/028957, WO 2007/098169, W0 2007/098214, WO 2007/103554, WO 2009/025823 and WO 2009/025839; Isoquinolines and isoquinolinones - see W0 2007/100880 and WO 2009/029214 MP10 and MP10 like ds: US 2007/0155779, WO 2008/001182 and WO 25 2008/004117; and Benzodiazepines - see W0 2007/082546.
For a further review see also T. Chappie et a1. t Opinion in Drug ery & Development 12(4), (2009) 458-467) and the literature cited therein.
Although some of the compounds of prior art are known to inhibit PDE10A 30 effectively having IC50 values of less than 50 nM, there is still an ongoing need for compounds which inhibit . In particular, there is an ongoing need for compounds which have one ofthe ing characteristics: 4 Selective inhibition of PDElOA, in particular Vis-a-vis tion of the other ten phosphodiesterase families PDEl-9, 11 and their different gene variants; suitable selectivity with regard to molecular receptors, transporters channels, enzymes or other biomolecules whose interaction with the PDElOA ligand might cause undesired side effects; ii. metabolic stability, in particular omal ity, e.g. measured in vitro, in liver microsomes from various species (e.g. rat or human) in human cells, such as hepatocytes; iii. no or only low inhibition of rome P450 (CYP) enzymes: cytochrome 10 P450 (CYP) is the name for a superfamily ofheme 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 ofthe types and subtypes of CYP in the human body are: CYP 1A2, CYP 2C9, CYP 2D6 15 and CYP 3A4. If CYP 3A4 inhibitors (e.g. grapefruit juice, cimetidine, 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 , the degradation fmay be slowed down and thus effects and side effects of the stered medicinal substance may be 20 undesirably enhanced; iv. a suitable lity in water (in mg/ml); 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, 25 volume of distribution (in 1~kg‘l), plasma clearance (in l-h'l -kg'1), AUC (area under the curve, area under the concentration—time curve (in ng-h-l'l), oral bioavailability, (the dose-normalized ratio ofAUC after oral administration and AUC after intravenous administration), the so-called brain—plasma ratio (the ratio ofAUC in brain tissue and AUC in plasma); 30 Vi. no or only low blockade of the hERG channel: compounds which block the hERG l may cause a prolongation of the QT interval and thus lead to serious bances of cardiac rhythm (for example so-called "torsade de 5 pointes"). The potential of nds 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 logical 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, 10 50 , 9).
Vii. high free fraction in brain, i.e. the fraction of the compound bound to proteins should be low. viii. low ilicity. 15 Brief Description of the Invention The present invention is thus based on the object of providing compounds which inhibit PDElOA at low concentrations.
The compounds are further intended to display at least one of the properties i. to 20 viii. ned above, in particular high selectivity with regard to inhibition of PDElOA, high selectivity Vis-a-Vis other phosphodiesterases such as enhanced , metabolic ity, in particular microsomal and/or cytosolic stability, low affinity to the HERG receptor, low inhibition of cytochrome P450 (CYP) enzymes, suitable solubility in water and suitable pharmacokinetics. 25 This object and further objects are ed by the compounds of the general formula I described below, the N—oxides, the prodrugs, the hydrates and the tautomers thereof and the ceutically suitable salts thereof: Q R1 (I) N 2 X: |\ R k2 (.0 X wherein is O or S; >< is N or CH; >1, is N or C-R7; >3, is O, S, -X4=C(R8)-, where C(RS) is bound to the carbon atom which - 2 carries R or , -X5=C(R9)-, where X5 is bound to the carbon atom which carries R2; is N or C-R9; is N; 10 is selected from i. monocyclic hetaryl having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, which is unsubstituted or may carry 1, 2, 3 or 4 identical or 15 different substituents RX, ii. fused bicyclic hetaryl having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, benzothienyl or benzofuryl, where bicyclic hetaryl, hienyl and benzofuryl are, independently of each other, 20 unsubstituted or may carry 1, 2, 3 or 4 identical or ent substituents RX, and iii. phenyl, which carries a monocyclic hetaryl radical having 1 or 2 nitrogen atoms and optionally a fiarther heteroatom selected from O, S and N as ring members, which in on to 25 monocyclic hetaryl, may carry 1, 2 or 3 identical or different substituents RX, where RX is selected from the group consisting of H, halogen, alkyl, C1-C4-alkoxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C3-C5- 30 cycloalkyl, alkoxy-C1—C4-alkoxy, C1-C4-alkoxy-C1-C4- alkyl, OH, hydroxy—Cl—C4—alkyl, 6-cycloalkyl, benzyloxy, C(O)O—(C1—C4—alkyl), O-(Cl-C4-alkyl)-C02H, 7 N(RX1)(RX2), C(O)N(RX1)(RX2), C1-C4-alkyl-N(RX1)(RX2), -NRX3-C(O)—N(RX1)(RX2), NRX3-C(O)O-(C1-C4-alkyl), -N(RX3)-SOg—RX4, phenyl, CN, -SF5, -OSF5, —sosz4, -SR"4 and trimethylsilyl, where R“, RXZ, RX3 and RX4, independently of each other are selected from the group consisting of hydrogen, C1-C4-alkyl, C1—C4-fluoroalky1 and C3-C6-cycloalky1 or RX1 and RX2 form together with the N atom to which they are attached a 3- to 7-membered, nitrogen heterocycle which may have 1, 2 or 3 further different or identical heteroatoms or heteroatom 10 containing groups ed from the group of O, N, S, SO and SOZ as ring members and which may carry 1, 2, 3, 4, 5 or 6 substituents selected from C1-C4-alkyl, or two radicals Rx which are bound at adjacent carbon atoms may form a fused 5— or 6—membered saturated carbocyclic 15 radical or a fused 5— or 6—membered heterocyclic radical having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N; is selected from the group consisting of hydrogen, halogen, OH, C1- 20 C4-alkyl, trimethylsilyl, alkylsulfanyl, C1-C4-alkoxy-C1-C4- alkyl, C1-C4-alkoxy, C1—C4—alkoxy-C1-C4-alkoxy, C1-C4-alkylsulfanyl- C1-C4-alkoxy, alkenyloxy, fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRXIRXZ, NRXIRXZ-Cl-C4-alkoxy 25 and the moiety Yl-Cyc]; is selected from the group consisting of hydrogen, halogen, OH, C1- C4-alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1- C4-a1koxy-C1-C4-alkoxy, alkenyloxy, fluoroalkyl, C1-C4- 30 fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl, CN and NRXIRXZ; 8 A represents one of the following groups A1, A2, A3; A4 or A5: R3e R3f R4 R3 (A1) (A2) (A3) (A4) (A5) where * tes the points of attachment to Het and to the nitrogen atom, tively; R3, R4, R5, R6 independently of each other are selected from the group consisting of hydrogen, halogen, C1—C4—alkyl, trimethylsilyl, C1-C4-fluoroalkyl, 10 C1-C4-fluoroalkoxy, C3—C6—cycloalkyl, or the radicals together with the carbon atoms to which they are bound form a saturated 3- to 6- membered carbocycle or a saturated 3- to 6-membered heterocycle having 1 or 2 non-adjacent heteroatoms as ring members, where the ycle and the heterocycle are unsubstituted or may carry 1, 2, 3 15 or 4 substituents ed from fluorine and methyl or either the radicals R3, R4 or the radicals R5, R6 together with the carbon atom to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6-membered heterocycle having 1 or 2 non- nt heteroatoms as ring members, where the carbocycle and the 20 heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents selected from fluorine and methyl; AV is a O, NR3a, CR3bR3C or linear C2-C3-alkandiy1, where one of the ieties of C2-C3-alkandiyl may be replaced by oxygen or NR”, 25 and where l, 2, 3, or 4 of the hydrogen atoms of Cg-Cg-alkandiyl may be replaced by a radical R”, where R3a is hydrogen or C1—C4—alkyl, 9 R3b, R30 independently of each other are ed from the group consisting of hydrogen, halogen, C1-C4-alkyl or R3b and R30 together form C2-C3-alkandiyl; R3d is selected from the group consisting of halogen and C1-C4- alkyl; independently of each other are selected from the group consisting of hydrogen and C1-C4-alkyl; 10 is selected from the group consisting of hydrogen, halogen, OH, C1- C4—alkyl, trimethylsilyl, alkylsulfanyl, C1-C4-alkoxy-C1-C4- alkyl, C1—C4—alkoxy, C1—C4—alkoxy—C1-C4-alkoxy, C1-C4-alkylsulfanyl- alkoxy, C2-C4-alkenyloxy, C1—C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl 15 groups, fluorinated cyclopropyl, 1RX2, NRXlez-Cl-C4-alkoxy and the moiety Yz-Cycz; is selected from the group consisting of hydrogen, halogen, OH, C1- C4-alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, alkoxy, C1- 20 oxy-C1-C4—alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- lkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl, CN and NRXIRXZ; is selected from the group consisting of hydrogen, halogen, OH, C1- 25 C4-alkyl, trimethylsilyl, C1-C4—alkylsulfanyl, C1-C4-alkoxy-C1-C4- alkyl, C1-C4-alkoxy, C1-C4—alkoxy-C1—C4-alkoxy, C1-C4-alkylsulfanyl- C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRXIRXZ, NRXlez-Cl-C4-alkoxy 30 and the moiety Y3—Cyc3 ; 10 Y1, Y2, Y3 independently of each other are selected from a chemical bond, CH2, 0, O-CH2, NRy, NRy—CHg, NRy-S(O)2, S, S(O), S(O)2, 1,2-ethandiyl, 1,2-ethendiyl or 1,2-ethyndiyl, where Ry is selected from the group consisting of en, C1—C4—alkyl, C1-C4-alkylcarbonyl, C1-C4- alkylsulfonyl, C1-C4-fluoroalkylsulfonyl; Cycl, Cycz, Cyc3 ndently of each other are selected from the group consisting of phenyl, naphthyl, 4- to 8-membered saturated or partially unsaturated heteromonocyclic radicals, saturated or partially 10 unsaturated 7- to 10 membered bicyclic radicals, 5- or 6- membered monocyclic hetaryl, and 8— to 10 membered bicyclic hetaryl, where the saturated or partially rated heteromonocyclic and heterobicyclic radicals have 1, 2, 3 or 4 heteroatoms or heteroatom containing groups as ring s, which are selected 15 from O, S, SO, S02 and N, and where the 5- or 6-membered monocyclic hetaryl and the 8— to lO-membered ic l have 1, 2, 3 or 4 heteroatoms as ring members, which are selected from O, S and N, where phenyl, naphthyl, the saturated or partially unsaturated 20 monocyclic and heterobicyclic radicals and the mono and bicyclic heteroaromatic radicals are unsubstituted or carry 1, 2, 3, 4 or 5 radicals RC1 or one radical Y'—RC2 and 0, l, 2, 3 or 4 radicals RC1; where 25 RCl is selected from hydrogen, halogen, OH, CN, N02, C1-C4-alkyl, C1-C4-alkoxy, C1-C4-alkylsulfanyl, y—Cl-C4-alkyl, C1-C4- alkoxy-Cl-C4-alkyl, C1-C4-alkoxy-C1-C4-alkoxy, cyano-Cl-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C1-C4- alkylsulfonyl, C(O)Ra, Z-C(O)ORb, Z-C(O)NR°Rd, S(O)2NR°Rd 30 and Z—NReRf, where R81 is selected from the group consisting of C1-C4-alkyl and C1—C4—fluoroalkyl, 1 1 Rb is selected from the group consisting of hydrogen, C1-C4- alkyl, C2—C4-alkenyl and C1-C4-fluoroalkyl, RC, Rd is selected from the group consisting of hydrogen, C1-C4- alkyl, C1—C4—fluoroalkyl, C1-C4-alkoxy and C1-C4- fluoroalkoxy, Re, Rf is selected from the group consisting of hydrogen, C1-C4- alkyl, C1-C4—fluoroalkyl, C1-C4—alkoxy and C1-C4- fluoroalkoxy, Z is a nt bond or C1-C4-alkandiyl, 10 or two radicals RCl which are bound at adjacent carbon atoms may form a fused 5— or 6—membered carbocyclic radical or a fused 5— or 6—membered heterocyclic l having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N; 15 or two radicals RC] which are bound at the same carbon atom may form a spiro 5— or ered carbocyclic radical or a spiro 5- or 6-membered cyclic radical having 1 or 2 heteroatoms as ring members, which are ed from O, S and N, 20 or two radicals RC1 which are bound at the same carbon atom may form an oxygen atom, where the fused and the spiro radicals are unsubstituted or carry 1, 2, 3 or 4 radicals RC3; 25 Y! is a chemical bond, CH2, 0, O-CHz, S(O)2, NRy', NRy'-CH2 or NRy'-S(O)2, where R)" is selected from the group consisting of hydrogen, C1-C4-alkyl, C1-C4-alkylcarbonyl, C1-C4- alkylsulfonyl, C1-C4-fluoroalkylsulfonyl; 30 is a carbocyclic or heterocyclic l selected from the group consisting ofphenyl, 3- to 7-membered saturated or partially unsaturated monocarbocyclic radicals, 3- to 7-membered 12 saturated or partially unsaturated heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, and 5- or 6-membered heteroaromatic ls, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, where the carbocyclic and the heterocyclic radical is unsubstituted or carries 1, 2, 3, 4 or 5 radicals RC3; RC3 is selected from hydrogen, halogen, OH, CN, C1-C4-alky1, C1- 10 C4-alkoxy, hydroxy-Cl-C4-alkyl, C1-C4-alkoxy-C1-C4-alkyl, cyano—CI—C4—alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C2- C6—alkenyl, C(O)Ra, benzyl, Z-C(O)ORb, NR°Rd, S(O)2NR°Rd and Z-NReRf, where, z, Ra, Rb, R“, Rd, Re and Rf are as defined above or two radicals RC3 which are bound at the 15 same atom may form an oxygen atom; provided that for X3 being 0 or S, at least one of the ls R1 and R7 is a moiety Yl-Cyc1 or YZ-Cycz, respectively; 20 further provided that for X3 being X4=C(R8), one or two of the radicals R1, R7 and R9 are a moiety Yl-Cycl, YZ-Cyc2 or Y3-Cyc3, respectively; r provided that for X3 being 9), one or two of the radicals R1, R7 and R9 are a moiety Yl-Cycl, Yz-Cyc2 or Y3-Cyc3, respectively; 25 and the N—oxides, the prodrugs, the tautomers and the hydrates thereof, and the ceutically acceptable salts thereof.
The present invention therefore relates to the nds of the general formula 1, 30 their tautomers, the hydrates thereof, the pharmaceutically suitable salts of the compounds of formula I, the gs of the compounds of formula I and the 13 pharmaceutically suitable salts of said prodrugs, tautomers or es of the compounds of formula I.
The compounds of the formula I, their salts, their prodrugs, their hydrates and their ers effectively inhibit PDE10A even at low concentrations. They are additionally distinguished by a high selectivity in relation to the inhibition of the PDE10A vis-a-vis tion of other phosphodiesterease, such as PDE3 or PDE4. The compounds of the invention may additionally have one or more of the properties ii. to viii. ned above.
The compounds of the a I, their salts, their prodrugs, their hydrates and their tautomers are therefore particularly suitable for treating disorders and conditions in creatures, especially human creatures, which can be treated or controlled by inhibition of phosphodiesterase type 10A.
The invention therefore also relates to the use of carboxamide compounds of the formula I, their tautomers, their hydrates and their pharmaceutically suitable salts for the manufacture of a medicament, in particular of a ment which is le for the treatment of a disorder or a condition which can be treated by inhibition of phosphodiesterase type 10A.
The invention further relates to a medicament, in particular a medicament which is suitable for the treatment of a disorder or a condition which can be treated by inhibition of odiesterase type 10A. The medicament comprises at least one compound of the formula I, as described , or a tautomer, or a hydrate or a prodrug of said compound I, or a pharmaceutically suitable salt of the compound of the formula I or a pharmaceutically suitable salt of the tautomer, the hydrate or the prodrug of compound of the formula I.
The invention further relates to the compound of formula I Q R1 Het A (I) N R2 X1 X2 X3 n Q is O or S; X1 is N or CH; X2 is N or C-R7; (11352636_1):GGG 13a X3 is O, S, -X4=C(R8)-, where C(R8) is bound to the carbon atom which carries R2, or -X5=C(R9)-, where X5 is bound to the carbon atom which carries R2; X4 is N or C-R9; X5 is N; Het is ed from i. monocyclic C-bound 6-membered hetaryl having 1 or 2 nitrogen atoms as ring members, which is unsubstituted or may carry 1, 2, 3 or 4 cal or different substituents Rx, ii. fused bicyclic hetaryl having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, benzothienyl or benzofuryl, where bicyclic hetaryl, benzothienyl and benzofuryl are, ndently of each other, unsubstituted or may carry 1, 2, 3 or 4 identical or different substituents Rx, and iii. phenyl, which carries a monocyclic hetaryl radical having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, which in addition to monocyclic l, may carry 1, 2 or 3 cal or different tuents Rx, where Rx is selected from the group consisting of H, halogen, C1-C4-alkyl, C1-C4-alkoxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C3-C6- cycloalkyl, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkoxy-C1-C4-alkyl, OH, y-C1-C4-alkyl, O-C3-C6-cycloalkyl, benzyloxy, C(O)O- (C1-C4-alkyl), O-(C1-C4-alkyl)-CO2H, N(Rx1)(Rx2), Rx1)(Rx2), C1-C4-alkyl-N(Rx1)(Rx2), -NRx3-C(O)-N(Rx1)(Rx2), NRx3-C(O)O-(C1-C4-alkyl), -N(Rx3)-SO2-Rx4, phenyl, CN, -SF5, -OSF5, -SO2Rx4, -SRx4 and trimethylsilyl, where Rx1, Rx2, Rx3 and Rx4, independently of each other are selected from the group consisting of hydrogen, C1-C4- (11352636_1):GGG 13b alkyl, C1-C4-fluoroalkyl and C3-C6-cycloalkyl or Rx1 and Rx2 form together with the N atom to which they are attached a 3- to 7- membered, nitrogen heterocycle which may have 1, 2 or 3 further different or identical heteroatoms or heteroatom containing groups selected from the group of O, N, S, SO and SO2 as ring members and which may carry 1, 2, 3, 4, 5 or 6 substituents selected from alkyl; or two radicals Rx which are bound at nt carbon atoms may form a fused 5- or 6-membered saturated carbocyclic l or a fused 5- or 6-membered heterocyclic radical having 1, 2 or 3 heteroatoms as ring s, which are selected from O, S and N; R1 is a moiety Y1-Cyc1; R2 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl , fluorinated cyclopropyl, CN and NRx1Rx2; A represents one of the following groups A1, A2, A3, A4 or A5: R3e R3f R6 R5 R4 A' * * * * * * * * * * R4 R3 R5 R5 R4 R4 R5 (A1 ) (A2 ) (A3 ) (A4 ) (A5 ) where * indicates the points of attachment to Het and to the nitrogen atom, tively, R3, R4, R5, R6 independently of each other are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl, trimethylsilyl, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, C3-C6-cycloalkyl, or the radicals er with the carbon atoms to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6-membered heterocycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents (11352636_1):GGG 13c ed from ne and methyl or either the radicals R3, R4 or the radicals R5, R6 together with the carbon atom to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6- membered cycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents selected from fluorine and methyl; A' is a O, NR3a, CR3bR3c or linear C2-C3-alkandiyl, where one of the CH2- moieties of C2-C3-alkandiyl may be replaced by oxygen or NR3a, and where 1, 2, 3, or 4 of the hydrogen atoms of C2-C3-alkandiyl may be replaced by a radical R3d, where R3a is hydrogen or C1-C4-alkyl, R3b, R3c independently of each other are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl or R3b and R3c together form C2-C3-alkandiyl; R3d is selected from the group consisting of halogen and C1-C4- alkyl; R3e, R3f independently of each other are selected from the group consisting of hydrogen and C1-C4-alkyl; R7 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, alkoxy-C1-C4-alkyl, C1- oxy, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkylsulfanyl-C1-C4- alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRx1Rx2, NRx1Rx2-C1-C4-alkoxy and the moiety Y2-Cyc2; R8 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, alkoxy, C1-C4- -C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl , fluorinated cyclopropyl, CN and NRx1Rx2; (11352636_1):GGG 13d R9 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1- C4-alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, alkylsulfanyl-C1-C4- alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRx1Rx2, NRx1Rx2-C1-C4-alkoxy and the moiety Y3-Cyc3; Y1, Y2, Y3 ndently of each other are selected from a chemical bond, CH2, O, O-CH2, NRy, NRy-CH2, O)2, S, S(O), S(O)2, 1,2-ethandiyl, 1,2- ethendiyl or 1,2-ethyndiyl, where Ry is selected from the group consisting of hydrogen, C1-C4-alkyl, C1-C4-alkylcarbonyl, C1-C4-alkylsulfonyl, C1- C4-fluoroalkylsulfonyl; Cyc1, Cyc2, Cyc3 independently of each other are selected from the group consisting of phenyl, yl, 4- to ered saturated or partially unsaturated heteromonocyclic radicals, saturated or partially unsaturated 7- to 10 ed heterobicyclic radicals, 5- or 6-membered monocyclic hetaryl, and 8- to 10 membered bicyclic hetaryl, where the saturated or partially unsaturated heteromonocyclic and heterobicyclic radicals have 1, 2, 3 or 4 heteroatoms or heteroatom ning groups as ring s, which are selected from O, S, SO, SO2 and N, and where the 5- or 6-membered clic hetaryl and the 8- to 10-membered bicyclic hetaryl have 1, 2, 3 or 4 heteroatoms as ring members, which are selected from O, S and N, where phenyl, naphthyl, the saturated or partially unsaturated heteromonocyclic and heterobicyclic radicals and the mono- and bicyclic heteroaromatic radicals are unsubstituted or carry 1, 2, 3, 4 or 5 radicals RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4 radicals RC1; where RC1 is selected from hydrogen, halogen, OH, CN, NO2, C1-C4-alkyl, C1- C4-alkoxy, C1-C4-alkylsulfanyl, hydroxy-C1-C4-alkyl, C1-C4- alkoxy-C1-C4-alkyl, C1-C4-alkoxy-C1-C4-alkoxy, cyano-C1-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C1-C4-alkylsulfonyl, C(O)Ra, Z-C(O)ORb, NRcRd, S(O)2NRcRd and Z-NReRf, where (11352636_1):GGG 13e Ra is selected from the group consisting of C1-C4-alkyl and C1- oroalkyl, Rb is selected from the group consisting of hydrogen, C1-C4- alkyl, C2-C4-alkenyl and C1-C4-fluoroalkyl, Rc, Rd is selected from the group consisting of hydrogen, C1-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-alkoxy and C1-C4- fluoroalkoxy, Re, Rf is selected from the group consisting of hydrogen, C1-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-alkoxy and C1-C4- fluoroalkoxy, Z is a covalent bond or C1-C4-alkandiyl, or two radicals RC1 which are bound at adjacent carbon atoms may form a fused 5- or 6-membered carbocyclic radical or a fused 5- or 6-membered cyclic radical having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N; or two radicals RC1 which are bound at the same carbon atom may form a spiro 5- or 6-membered carbocyclic radical or a spiro 5- or 6-membered cyclic radical having 1 or 2 heteroatoms as ring members, which are selected from O, S and N, or two ls RC1 which are bound at the same carbon atom may form an oxygen atom, where the fused and the spiro radicals are unsubstituted or carry 1, 2, 3 or 4 radicals RC3; Y' is a chemical bond, CH2, O, O-CH2, S(O)2, NRy', NRy'-CH2 or NRy'-S(O)2, where Ry' is selected from the group ting of hydrogen, alkyl, alkylcarbonyl, C1-C4-alkylsulfonyl, C1-C4-fluoroalkylsulfonyl; RC2 is a carbocyclic or heterocyclic radical selected from the group consisting of phenyl, 3- to 7-membered saturated or partially unsaturated monocarbocyclic ls, 3- to 7-membered saturated or partially unsaturated heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, and 5- or 6-membered heteroaromatic radicals, having 1, 2 or 3 (11352636_1):GGG 13f heteroatoms as ring members, which are selected from O, S and N, where the carbocyclic and the heterocyclic radical is unsubstituted or carries 1, 2, 3, 4 or 5 radicals RC3; RC3 is selected from hydrogen, halogen, OH, CN, C1-C4-alkyl, C1-C4- alkoxy, hydroxy-C1-C4-alkyl, C1-C4-alkoxy-C1-C4-alkyl, cyano-C1- C4-alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C2-C6-alkenyl, C(O)Ra, , Z-C(O)ORb, Z-C(O)NRcRd, S(O)2NRcRd and ZNReRf , where, Z, Ra, Rb, Rc, Rd, Re and Rf are as defined above or two radicals RC3 which are bound at the same atom may form an oxygen atom; provided that for X3 being X4=C(R8), one or two of the radicals R1, R7 and R9 are a moiety Y1-Cyc1, Y2-Cyc2 or Y3-Cyc3, respectively; further provided that for X3 being X5=C(R9), one or two of the radicals R1, R7 and R9 are a moiety Y1-Cyc1, Y2-Cyc2 or 3, respectively; and the N-oxides, the prodrugs, the tautomers and the hydrates thereof, and the pharmaceutically acceptable salts thereof; wherein prodrugs are derivatives of the compounds of a I carrying an OH or NH2-group, where one of the hydrogen atoms of the OH or NH2-group is substituted by a alkylcarbonyl group, by benzoyl, or by an acyl group derived from an amino acid, which is linked to the oxygen or nitrogen of the OH or NH2-group via the carbonyl group of the amino acid; or wherein prodrugs are alkylcarbonyloxyalkyl carbonates or carbamates of compounds I carrying an OH- or NH2-group in which one of the hydrogen atoms of the OH- or oup has been replaced by a group of the formula -O-CHRp-O-C(=O)-Rq in which Rp and Rq are independently of one another C1-C4-alkyl; except for the following compounds: ylphenyl[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, yl[2-(4-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(4-Methylphenyl)[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, (11352636_1):GGG 13g 5-(2-Furanyl)[1-methyl(3-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(2-Thienyl)[1-methyl(3-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(3,4-Dimethylphenyl)[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(4-Methylphenyl)[2-(4-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-Phenyl[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, and 5-Phenyl[2-(4-oxophenylthieno[2,3-d]pyrimidin-(3H)yl)ethyl]-thieno[2,3- d]pyrimidin(3H)-one.
The invention further relates to the compound which is selected from the group consisting of idinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(pyrimidinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1,1-dioxidothiomorpholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyrimidinyl)phthalazin1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](morpholinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)- yl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyrimidinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](morpholinyl)phthalazin-1(2H)-one; 3-methyl(pyridinyl)(2-(quinolinyl)ethyl)thieno[3,2-c]pyridin-4(5H)-one; 5-(3-hydroxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 7-(pyridinyl)[2-(quinolinyl)ethyl]furo[3,2-c]pyridin-4(5H)-one; anti imidinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(pyrimidinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn idinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti 5-(morpholinyl)(2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(morpholinyl)(2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; (11352636_1):GGG 13h pholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(1H-benzimidazolyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 4-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1,4-dihydropyrimidinyl)[2-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin yl]phthalazin-1(2H)-one2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridin yl)phthalazin-1(2H)-one; 5-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; and the enantiomers thereof, the N-oxides thereof, the prodrugs thereof, the tautomers thereof and the hydrates thereof, and the ceutically acceptable salts thereof, wherein prodrugs are derivatives of the compounds of formula I ng an OH or NH2-group, where one of the hydrogen atoms of the OH or NH2-group is substituted by a C1-C4- alkylcarbonyl group, by benzoyl, or by an acyl group derived from an amino acid, which is linked to the oxygen or en of the OH or NH2-group via the carbonyl group of the amino acid; or wherein prodrugs are alkylcarbonyloxyalkyl carbonates or carbamates of compounds I carrying an OH- or NH2-group in which one of the hydrogen atoms of the OH- or oup has been replaced by a group of the formula -C(=O)-O-CHRp-OC (=O)-Rq in which Rp and Rq are ndently of one another C1-C4-alkyl.
The invention r relates to the use of at least one compound as claimed in the invention for the manufacture of a medicament for treating a medical disorder, selected from neurological and psychiatric disorders which can be treated by inhibition of phosphodiesterase type 10A.
Detailed Description of the Invention The terms "compound of the formula I" and "compounds I" are used as synonyms.
The term "prodrugs" means nds which are metabolized in vivo to the compounds I of the invention. Typical examples of prodrugs are described in C.G.
Wermuth (editor): The Practice of Medicinal Chemistry, Academic Press, San Diego, 1996, pages 671-715. These include for example phosphates, carbamates, amino acids, (11352636_1):GGG 14 esters, amides, peptides, ureas and the like. Suitable prodrugs in the present case may be for example tives of those compounds I ng an OH or oup, where the OH or NHz-group forms an ester/amide/peptide linkage, i.e. where one of the hydrogen atoms ofthe OH or NHz-group is substituted by a C1-C4-alkylcarbonyl group, e.g. by acetyl, propionyl, n-propylcarbonyl, isopropylcarbonyl, n—butylcarbonyl or tert- butylcarbonyl (pivaloyl), by benzoyl, or by an acyl group d from an amino acid, e.g. glycine, alanine, , phenylalanine and the like, which is linked to the oxygen or nitrogen of the OH or oup via the carbonyl group of the amino acid. Further suitable prodrugs are alkylcarbonyloxyalkyl carbonates or carbamates of compounds I 10 carrying an OH— or NHz-group in which one of the hydrogen atoms of the OH- or NH2- group has been ed by a group of the a —C(=O)-O-CHRp-O-C(=O)-R01 in which Rp and Rq are independently of one another C1—C4-alkyl. Such carbonates and carbamates are described for example in J. Alexander, R. Cargill, S. R. Michelson, H.
, J. Medicinal Chem. 1988, 31(2), 318—322. These groups can then be 15 eliminated under lic conditions and result in compounds 1. Therefore, said prodrugs and their pharmaceutically acceptable salts are also part of the invention.
The term "pharmaceutically acceptable salts" refers to cationic or anionic salts compounds, wherein the counter ion is derived from pharmaceutically acceptable non- toxic bases or acids including inorganic or c bases and inorganic or organic acids. 20 When the compound of formula I or its prodrug or N—oxide is acidic, salts may be ed from ceutically acceptable non-toxic bases, including inorganic and organic bases. Salts derived from inorganic bases include salts, wherein the counter ion is aluminium, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc ion and the like. Particularly preferred 25 are the ammonium, calcium, ium, potassium, and sodium ions. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, e, caffeine, e, dibenzylethylene-diamine, diethylamine, 2-diethylamino- 30 ethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N—ethyl-morpholine, N—ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, 15 procaine, purines, omine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of formula I or its prodrug or N—oxide is basic, salts may be prepared from pharmaceutically acceptable xic acids, including inorganic and organic acids. Such acids include acetic, roacetic acid, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, onic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, oric, succinic, sulfiiric, tartaric, p-toluenesulfonic acid, and the like. Particularly red are citric, hydrobromic, hydrochloric, maleic, 10 phosphoric, sulfuric, fumaric, and tartaric acids. It will be understood that, as used herein, references to the compounds of formula I are meant to also include the pharmaceutically acceptable salts.
The compounds of the invention may be in the form of a mixture of diastereomers, or of a mixture of diastereomers in which one of the two diastereomers is 15 enriched, or of essentially diastereomerically pure compounds (diastereomeric excess de > 90%). The compounds are ably in the form of essentially diastereomerically pure nds (diastereomeric excess de > 90%). The compounds I of the ion may furthermore be in the form of a mixture of enantiomers (for example as racemate), of a mixture of enantiomers in which one of the two enantiomers is enriched, or 20 essentially in enantiomerically pure compounds (enantiomeric excess ee > 90%). It is preferred to employ the nds enantiomerically pure or diastereomerically pure.
The present invention moreover relates to compounds as defined herein, wherein one or more ofthe atoms depicted in formula I have been replaced by its stable, preferably non-radioactive e (e.g., en by deuterium, 12C by 13C, 14N by 15N, 25 16O by 18O) and preferably wherein at least one hydrogen atom has been replaced by a deuterium atom. Of course, the compounds according to the invention contain more of the respective isotope than this lly occurs and thus is anyway present in the compounds I.
The compounds of the formula I and their salts in the solid form may exist in 30 more than one crystal structure (polymorphism), and may also be in the form of hydrates or other solvates. The present invention includes any polymorph of the compound I or its salt as well as any hydrate or other solvate. 16 In the context of the present description, unless stated otherwise, the terms ", "alkenyl", "alkoxy , alkenyloxy", "fluoroalkyl", alkoxy , cycloalkyl", "fluorinated cycloalkyl", ene", "alkandiyl", "hetaryl" and radicals derived therefrom, such as "alkylcarbonyl", "alkylsulfanyl", "alkylsulfonyl", "fluoroalkylsulfonyl", "hydroxylalkyl", "cyanoalkyl", "alkoxylalkyl", "alkoxyalkoxy", sulfanylalkyl", "alkylsulfanylalkoxy" and "hetarylmethyl" ent groups of individual radicals. The groups of noncyclic radicals "alkyl", "alkenyl", "alkoxy", "alkenyloxy", "fluoroalkyl", "fluoroalkoxy", "alkylene", "alkandiyl", and the groups of 10 ls derived therefrom always include both unbranched and branched "alkyl", "alkenyl", "alkoxy", "alkenyloxy", "fluoroalkyl", "fluoroalkoxy", "alkylene" and "alkandiyl", respectively.
The prefix Cn-Cm- indicates the respective number of carbons in the hydrocarbon unit. Unless indicated otherwise, fluorinated substituents preferably have one to five 15 identical or different fluorine atoms.
The term "halogen" ates in each case, e, bromine, chlorine or iodine, specifically fluorine, chlorine or bromine.
Examples of other meanings are: Alkyl, and the alkyl moieties for example in alkylcarbonyl, alkylsulfanyl, 20 alkylsulfonyl, alkylsulfanylalkyl and alkylsulfaylalkoxy: saturated, straight-chain or branched hydrocarbon radicals having one or more C atoms, e. g. l to 4 carbon atoms, e. g. C1-C4-a1kyl such as , ethyl, , l-methylethyl, n-butyl, l-methylpropyl, 2—methylpropyl and l l -dimethylethyl.
, Fluoroalkyl and the fluoroalkyl moieties for example in lkylsulfonyl: an 25 alkyl radical having ordinarily l to 4 C atoms, in particular 1 or 2 C-atoms (C1-C2- lkyl) as mentioned above, whose hydrogen atoms are partly or completely replaced by fluorine atoms such as fluoromethyl, difluoromethyl, trifluoromethyl, 2- fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 2-fluoro ethyl, 2,2—difluoro—1-methylethyl, 2,2-trifluoro-l-methylethyl, opropyl, 3- 30 fluoropropyl, 2,2—difluoropropyl, 2,3-difluoropropyl, 3,3,3-trifluoropropyl, 2,3,3,3- pentafluoropropyl, heptafluoropropyl, 1—(fluoromethyl)fluoroethyl, 4-fluorobutyl, and nonafluorobutyl. 17 Cycloalkyl, and the cycloalkyl moieties for example in cycloalkoxy or cycloalkyl- C1-C4-alkyl: clic, saturated hydrocarbon groups having three or more C atoms, e. g. 3, 4, 5, 6 or 7 carbon ring members, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. nated cycloalkyl: monocyclic, saturated hydrocarbon groups having three or more C atoms, e.g. 3, 4, 5, 6 or 7 carbon ring s, such as cyclopropyl, utyl, cyclopentyl, cyclohexyl and cycloheptyl, wherein at least one, e. g. l, 2, 3, 4, 5 or 6 of the hydrogen atoms are replaced by fluorine atoms, examples including 1- fluorocyclopropyl, rocyclopropyl, 2,2-difluorocyclopropyl, 1,2- 10 difluorocyclopropyl, 2,3—difluorocyclopropyl, etc..
Cycloalkoxy: a cycloalkyl radical as defined above which is linked Via an oxygen atom, e. g. cyclopropyloxy, cyclobutyloxy, cyclopentyloxy or cyclohexyloxy.
Cycloalkylalkyl: a cycloalkyl radical as defined above which is linked via an alkylene group, in particular via a methylene, 1,1—ethylene or 1,2-ethylene group, e. g. 15 cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl or cyclohexylmethyl.
Alkenyl, and alkenyl es for example in alkenyloxy: monounsaturated, straight-chain or branched hydrocarbon radicals having two or more C atoms, e.g. 2 to 4 carbon atoms and one C=C-double bond in any position, e. g. C2-C4-alkenyl such as ethenyl, l-propenyl, 2-propenyl, l-methylethenyl, l-butenyl, 2-butenyl, 3-butenyl, 1- 20 methyl- 1 nyl, 2-methyl- l -propenyl, l -methylpropenyl and 2-methyl propenyl.
Alkoxy or alkoxy es for example in alkoxyalkyl and alkoxyalkoxy: an alkyl radical as defined above having preferably 1 to 4 C atoms, which is connected to the remainder ofthe molecule via an O atom: e.g. methoxy, ethoxy, n- 25 propoxy, l-methylethoxy, butoxy, ylpropoxy, 2-methylpropoxy or 1,1- dimethylethoxy.
Fluoroalkoxy: alkoxy as described above, in which the hydrogen atoms of these groups are partly or completely replaced by e atoms, i.e. for example C1-C4- lkoxy, in particular C1—C2-fluoroalkoxy, such as fluoromethoxy, 30 difluoromethoxy, trifluoromethoxy, 2—fluoroethoxy, 2,2-difluoroethoxy, 2,2,2-trifluoroethoxy, pentafluoroethoxy, 2-fluoropropoxy, opropoxy, 2,2-difluoropropoxy, 2,3-difluoropropoxy, 3,3,3—trifluoropropoxy, 2,2,3,3,3- 18 pentafluoropropoxy, heptafluoropropoxy, l-(fluoromethyl)fluoroethoxy, specifically fluoromethoxy, difluoromethoxy, trifluoromethoxy, 2-fluoroethoxy, or 2,2,2- trifluoroethoxy.
Hydroxyalkyl: an alkyl radical ordinarily having 1 to 4 C atoms, in which one hydrogen atom is replaced by an OH radical. Examples thereof are CHz-OH, l- hydroxyethyl, 2-hydroxyethy1, 1 —hydroxypropyl, 2-hydroxypropyl, 1 -methyl hydroxyethyl, l-methyl-Z-hydroxyethyl, 3-hydroxypropy1, 2—hydroxybuty1, 3- hydroxybutyl, 4-hydroxybutyl, 1-methylhydroxypropyl, 1 ,1-dimethy1hydroxyetyl, 1-methy1hydroxypropyl etc. 10 lkyl: an alkyl radical ordinarily having 1 to 4 C atoms, in which one hydrogen atom is replaced by a CN radical. Examples thereof are CHg-CN, 1- cyanoethyl, 2-cyanoethyl, l—cyanopropyl, 2—cyanopropyl, l-methyl- l -cyanoethyl, l- methylcyanoethyl, 3-cyanopropyl, 2—cyanobutyl, 3-cyanobutyl, 4-cyanobutyl, l- methylcyanopropyl, l l -dimethyl—2—cyanoetyl, yl— l -cyanopropyl etc. , 15 Alkoxyalkyl: an alkyl l ordinarily having 1 to 4 C atoms, in which one hydrogen atom is replaced by an alkoxy radical rily having 1 to 4 C atoms.
Examples thereof are CHz-OCHg, sz, n—propoxymethyl, CH2-OCH(CH3)2, xymethyl, (l-methylpropoxy)methyl, (2—methylpropoxy)methyl, CH2-OC(CH3)3, 2-(methoxy)ethyl, 2-(ethoxy)ethyl, 2—(n—propoxy)ethyl, 2-(l-methylethoxy)ethyl, 20 2-(n-butoxy)ethyl, 2-(l-methylpropoxy)ethyl, 2-(2-methylpropoxy)ethyl, 2-(l,l-dimethylethoxy)ethyl, 2-(methoxy)propyl, 2-(ethoxy)propyl, 2-(n- propoxy)propyl, 2-(l-methylethoxy)propyl, 2-(n—butoxy)propyl, 2-(1- propoxy)propyl, 2-(2-methylpropoxy)propyl, 2-(1,l-dimethylethoxy)propyl, 3- (methoxy)propyl, 3-(ethoxy)propyl, ropoxy)propyl, 3-(l-methylethoxy)propyl, 3- 25 (n-butoxy)propy1, 3 -( l -methy1propoxy)propyl, 3-(2-methy1propoxy)propyl, 3 -(1 , 1 - dimethylethoxy)propyl, 2-(methoxy)butyl, oxy)butyl, 2-(n—propoxy)butyl, 2-(1 - methylethoxy)butyl, utoxy)butyl, 2-(1-methy1propoxy)butyl, 2-(2- methylpropoxy)butyl, 2-(1, 1-dimethylethoxy)butyl, 3-(methoxy)butyl, 3-(ethoxy)buty1, 3 -(n-propoxy)butyl, 3 —(1—methylethoxy)butyl, utoxy)butyl, 3 -(1- 30 methylpropoxy)butyl, 3—(2—methylpropoxy)butyl, 3—(1,l-dimethylethoxy)butyl, 4- (methoxy)butyl, 4-(ethoxy)butyl, 4—(n—propoxy)butyl, 4-(l-methylethoxy)butyl, 4-(n- 19 butoxy)butyl, 4-(l-methylpropoxy)butyl, 4-(2-methylpropoxy)butyl, 4-(l ,ldimethylethoxy )butyl, etc.
Alkoxyalkoxy: an alkyl radical as defined above ordinarily having 1 to 4 C atoms both in the alkoxy and the alkyl moiety which is connected to the remainder of the molecule via an O atom: Examples thereof are CHg, OCHz-OCsz, n- propoxymethoxy, OCHz-OCH(CH3)2, n—butoxymethoxy, (1—methylpropoxy)methoxy, (2-methylpropoxy)methoxy, OCH2-0C(CH3)3, 2-(methoxy)ethoxy, 2—(ethoxy)ethoxy, 2-(n-propoxy)ethoxy, 2-(1-methylethoxy)ethoxy, 2-(n—butoxy)ethoxy, 2-(1-methylpropoxy)ethoxy, ethylpropoxy)ethoxy, 2-(1 l -dimethyl- , 10 ethoxy)ethoxy, etc. arbonyl: alkyl as defined above preferably having 1 to 4 C atoms, which is connected via a carbonyl group to the remainder of the molecule, e.g. acetyl, propionyl, butyryl, isobutyryl, oyl, pivaloyl and the like.
Alkylsulfanyl and the alkylsulfanyl radicals in alkylsulfanylalkyl and 15 alkylsulfanylalkoxy: alkyl as defined above ably having 1 to 4 C atoms, which is connected via an S atom to the remainder of the molecule, e.g. methylsulfanyl, ethylsulfanyl, n-propylsulfanyl and the like.
Alkylsulfonyl: alkyl as defined above preferably having 1 to 4 C atoms, which is connected via an 802 group to the remainder of the molecule, e.g. methylsulfonyl, 20 ethylsulfonyl, n-propylsulfonyl and the like.
Fluoroalkylsulfanyl: lkyl as defined above preferably having 1 to 4 C atoms, which is connected via an S atom to the remainder of the molecule, e.g. fluoromethylsulfanyl, difluoromethylsulfanyl, trifluoromethylsulfanyl, 2- fluoroethylsulfanyl, 2,2-difluoroethylsulfanyl, 2,2,2-trifluoroethylsulfanyl, 25 pentafluoroethylsulfanyl, opropylsulfanyl, 3-fluoropropylsulfanyl, 2,2- difluoropropylsulfanyl, 2,3-difluoropropylsulfanyl, and heptafluoropropylsulfanyl.
Fluoroalkylsulfonyl: fluoroalkyl as defined above preferably having 1 to 4 C atoms, which is connected via an SO; group to the remainder of the molecule, e. g. fluoromethylsulfonyl, difluoromethylsulfonyl, trifluoromethylsulfonyl, 2- 30 fluoroethylsulfonyl, 2,2—difluoroethylsulfonyl, 2,2,2—trifluoroethylsulfonyl, uoroethylsulfonyl, 2—fluoropropylsulfonyl, 3—fluoropropylsulfonyl, 2,2- difluoropropylsulfonyl, 2,3-difluoropropylsulfonyl, and heptafluoropropylsulfonyl. 20 Alkylsulfanylalkyl: an alkyl radical ordinarily having 1 to 4 C atoms, in which one en atom is replaced by an alkylsulfanyl radical ordinarily having 1 to 4 C atoms. Examples thereof are CH2—SCH3, CH2-SC2H5, n-propylsulfanylmethyl, CH2-SCH(CH3)2, n-butylsulfanylmethyl, (1—methylpropsulfanyl)methyl, (2- methylpropsulfanyl)methyl, CH2—0C(CH3)3, 2—(methylsulfanyl)ethyl, 2- (ethylsulfanyl)ethyl, 2-(n-propylsulfanyl)ethyl, ethy1ethylsu1fanyl)ethyl, utylsulfanyl)ethyl, ethylpropylsulfanyl)ethyl, 2—(2- methylpropylsulfanyl)ethyl, 2-(1 , l -dimethylethylsulfanyl)ethyl, 2-(methylsulfanyl)propyl, 2-(ethylsulfanyl)propyl, 2-(n-propylsulfanyl)propyl, 10 2-(1-methylethylsulfanyl)propyl, 2-(n—butylsulfanyl)propyl, 2-(1- methylpropylsulfanyl)propyl, 2—(2—methylpropylsulfanyl)propyl, 2-(1,l-dimethylethylsulfanyl)propyl, 3—(methylsulfanyl)propyl, 3-(ethylsulfanyl)propyl, 3-(n-propylsulfanyl)propyl, 3—(1—methylethylsulfanyl)propyl, 3-(n-butylsulfanyl)propyl, 3-(1-methylpropylsulfanyl)propyl, 3—(2—methylpropylsulfanyl)propyl, 3-(1 , l - 15 dimethylethylsulfanyl)propyl, 2-(methylsulfanyl)butyl, 2-(ethylsulfanyl)butyl, 2-(npropylsulfanyl )butyl, 2—(1-methylethylsulfanyl)butyl, 2-(n-butylsulfanyl)butyl, 2-(1- methylpropylsulfanyl)butyl, 2-(2-methylpropylsulfanyl)butyl, 2-(1 ,l- dimethylethylsulfanyl)butyl, 3-(methylsulfanyl)butyl, 3-(ethylsulfanyl)butyl, 3-(npropylsulfanyl )butyl, ethylethylsulfanyl)butyl, utylsulfanyl)butyl, 3-(1- 20 methylpropylsulfanyl)butyl, 3-(2-methylpropylsulfanyl)butyl, 3-(l,l-dimethylethylsulfanyl )butyl, 4-(methylsulfanyl)butyl, 4-(ethylsulfanyl)butyl, 4-(n- propylsulfanyl)butyl, 4-(l-methylethylsulfanyl)butyl, 4-(n-butylsulfanyl)butyl, 4-(lmethylpropylsulfanyl )butyl, ethylpropylsulfanyl)butyl, 4-(1 ,ldimethylethylsulfanyl )butyl, etc. 25 "Alkylen" or "Alkandiyl": a ted hydrocarbon chain having rily from 1 to 4 carbon atoms, such as methylen (-CH2-), 1,2-ethylen (—CHzCH2—), 1,1-ethandiy1 (-CH(CH3)-), 1,2-propandiyl, 1,3-propandiyl, 1,4-butandiyl, 1,2-butandiyl, 1,3- butandiyl, l-methyl- 1 ,2-propandiyl, 2-methyl-1,3-propandiyl, l-methyl- l , 1 -ethandiyl, l-methyl— l ,2—propandiyl etc. 30 Saturated or partially unsaturated 4 to 7-membered monocarbocyclic radicals include cycloalkyl as defined above and lkenyl having ordinarily from 4 to 7 carbon atoms as ring members, e.g. l—cyclobuten—l—yl, 2-cyclobutenyl, l-cyclopentenyl, 21 2-cyclopentenyl, l-cyclohexenyl, 2—cyclohexenyl, 3-cyclohexenyl, l-cycloheptenyl, 2- cycloheptenyl, 3-cycloheptenyl.
Saturated or partially unsaturated 7 to 10-membered ocyclic radicals include bicyclic carbocyclic radicals which ordinarily have from 7 to 10 carbon atoms as ring s and which are saturated or which have one or more, e.g. one or two C=C double bonds, or which e a monounsaturated carbocycle where the double bond is part of a fused benzene ring, e.g. bicyclo[2,2,1]heptyl, bicyclo[2,2,1]—2- heptyl, bicyclo[2,2,1]heptyl, bicyclo[3,3,0]octyl, bicyclo[3,3,0]—2-octy1, bicyclo[3,3,0]octyl, bicyclo[2,2,2]octyl, bicyclo[2,2,2]octyl, bicyclo[3,2,1] 10 octyl, bicyclo[3 ,2, l ]—2—octyl, bicyclo[3,2,1]—6—octyl, bicyclo[3 ,2, l ]octyl, bicyclo[4,3,0]—l—nonyl, bicyclo[4,3,0]—2—nonyl, bicyclo[4,3,0]—3-nonyl, bicyclo[4,3,0]— 7-nonyl, bicyclo[4,3,0]—8—nonyl, bicyclo[4,4,0]—1—decyl, bicyclo[4,4,0]—2-decyl, bicyclo[4,4,0]—3-decyl, bicyclo[2,2,1]-hepten—l-yl, bicyclo[2,2, l ]-heptenyl, bicyclo[2,2, l ]-hepten—5-yl, bicyclo[2,2, l ]—hept—2—en—7-yl, bicyclo[2,2,2]—octen— l - 15 yl, bicyclo[2,2,2]-octenyl, o[2,2,2]—oct—2-en—5-yl, bicyclo[2,2,2]-octen yl, bicyclo[3,3,0]—2-octen—l-yl, bicyclo[3,3,0]—2—octen—2-yl, bicyclo[3,3,0]—2-octen—3-yl, o[3,3,0]—2-octen—4-yl, bicyclo[3,3,0]—2—octen—5-yl, bicyclo[3,3,0]—2-octen—6-yl, bicyclo[3,3,0]—2-octen—7-yl, bicyclo[3,3,0]—2-octenyl, inden— l -yl, 2-yl, inden- 4-yl, inden—6-yl, tetrahydro- l -naphthyl, tetrahydronaphthyl, tetrahydro-S-naphthyl, 20 tetrahydronaphthyl, etc..
Heterocyclyl: a heterocyclic radical which may be saturated or partly rated and which may be a monocyclic heterocyclic radical ordinarily having 3, 4, 5, 6, 7 or 8 ring atoms or a bicyclic radical rily having 7, 8, 9 or 10 ring atoms, where ordinarily l, 2, 3 or 4, in particular 1, 2 or 3, of the ring atoms are heteroatoms such as 25 N, S or O, or heteroatom groups such as S(=O) or S(=O)2 besides carbon atoms as ring members.
Examples of saturated heteromonocycles are in particular: - Saturated heteromonocyclic radical which rily has 3, 4, 5, 6 or 7 ring atoms, where ordinarily 1, 2 or 3 of the ring atoms are heteroatoms such as 30 N, S or O, besides carbon atoms as ring members. These include for example: C-bonded, 3- or 4—membered saturated rings such as 22 2-0Xiranyl, 2-oxetanyl, 3—0xetanyl, idinyl, 3-thiethanyl, idinyl, 2-azetidinyl.
C-bonded, 5-mernbered saturated rings such as tetrahydrofiJran-Z-yl, tetrahydrofiaran—3—y1, tetrahydrothien—Z-yl, tetrahydrothien—3-yl, tetrahydropyrrol—Z-yl, tetrahydropyrrol—S-yl, tetrahydropyrazo , tetrahydropyrazo 1y1, tetrahydroisoxazol—3-yl, tetrahydroisoxazol—4—yl, tetrahydroisoxazol-S-yl, 1,2—oxathi01anyl, 1,2- oxathiolanyl, athiolanyl, tetrahydroisothiazo1y1, tetrahydroisothiazolyl, tetrahydroisothiazo1y1, 1,2-dithiolan—3-yl, 1,2- 10 dithio 1any1, tetrahydroimidazol—2-yl, tetrahydroimidazol—4-y1, tetrahydrooxazo 1—2—yl, tetrahydrooxazo , tetrahydrooxazol—S-yl, tetrahydrothiazol—Z—yl, ydrothiazo , ydrothiazo 1y1, 1,3- dioxolan—Z-yl, 1,3—dioxolan—4—y1, 1,3—oxathi01anyl, 1,3-0xathi01an—4-yl, 1,3-oxathiolanyl, 1,3—dithiolan—2—yl, 1,3-dithi01an—4-yl, 1,3,2- 15 dioxathiolan—4-yl.
C-bonded, bered saturated rings such as: tetrahydropyran—2-yl, tetrahydropyran—3-yl, tetrahydropyran—4-yl, piperidin- 2-y1, piperidinyl, piperidin—4-yl, tetrahydrothiopyran—Z-yl, tetrahydrothiopyran—3-yl, tetrahydrothiopyran—4-yl, 1,3-di0xan—2-y1, 1,3- 20 dioxan—4-yl, 1,3-dioxan—5—yl, 1,4-di0xan—2-yl, thian—2-yl, 1,3-dithian— 4-yl, 1,3-dithian—5-yl, 1,4—dithian—2-yl, 1,3-oxathiany1, 1,3-oxathianyl, 1,3-oxathiany1, 1,3—oxathian-6—yl, 1,4-oxathiany1, 1,4-0xathian—3-yl, 1,2-dithian—3-yl, 1,2-dithian—4-yl, hexahydropyrimidin—Z-yl, hexahydropyrirnidin—4-yl, hexahydropyrimidin—S-yl, hexahydropyrazin—Z-yl, 25 hexahydropyridazin—3-yl, hexahydropyridazin—4-yl, tetrahydro-1,3-oxazin- 2-y1, tetrahydro- 1 ,3-0xazin—4-yl, tetrahydro-l ,3—0xazin—5-y1, tetrahydro- 1 ,3- oxazin—6-yl, tetrahydro-1,3-thiazinyl, tetrahydro-l ,3-thiaziny1, tetrahydro- 1 azinyl, tetrahydro- 1 ,3-thiaziny1, tetrahydro- 1 ,4- thiazin-Z-yl, tetrahydro- 1 ,4-thiazinyl, tetrahydro- 1 ,4-0xazin—2-yl, 30 tetrahydro— 1 ,4—oxazin—3—yl, tetrahydro— 1 ,2—0xazin—3 -y1, tetrahydro- 1 ,2- oxazin—4-yl, tetrahydro—l ,2—oxazin—5—yl, tetrahydro- 1 ,2-oxazinyl.
N—bonded, 5-membered saturated rings such as: 23 tetrahydropyrrol— l -yl, tetrahydropyrazo l- l -yl, tetrahydroisoxazo lyl, tetrahydroisothiazol-2—yl, tetrahydroimidazol— l -yl, tetrahydrooxazol—3-yl, tetrahydrothiazol—3-yl.
N—bonded, 6-membered saturated rings such as: piperidin— l -yl, hexahydropyrimidinyl, hexahydropyrazin- l -yl, hexahydro-pyridazin— 1 —yl, tetrahydro- l ,3-oxazin—3 -y1, tetrahydro- 1 ,3 - thiazinyl, tetrahydro— 1 ,4—thiazin—4—yl, tetrahydro— 1 ,4-oxazin—4-yl, tetrahydro- 1 ,2-oxazinyl.
Unsaturated heteromonocyclic radicals which ordinarily have 4, 5, 6 or 7 10 ring atoms, where ordinarily 1, 2 or 3 of the ring atoms are heteroatoms such as N, S or O, besides carbon atoms as ring s. These include for example: C-bonded, 5-membered, partially unsaturated rings such as: 2,3-dihydrofuran—2—yl, hydrofi1ran—3-yl, 2,5-dihydrofi1ran—2-yl, 15 2,5-dihydrofuran—3-yl, hydrofuran—2-yl, 4,5-dihydrofi1ran—3-yl, 2,3- dihydrothien—Z-yl, 2,3-dihydrothien—3—yl, hydrothien—2-yl, 2,5- dihydrothien—3-yl, 4,5-dihydrothien—2-yl, 4,5-dihydrothien—3-yl, 2,3- dihydro-lH-pyrrol—2-yl, 2,3-dihydro-lH-pyrrol—3-yl, 2,5-dihydro-1H- pyrrol—2-yl, 2,5-dihydro—lH-pyrrol—3-yl, 4,5-dihydro-lH-pyrrol—2-yl, 4,5- 20 dihydro-lH-pyrrol—3-yl, 3,4—dihydro-2H-pyrrol—2-yl, 3,4-dihydro-2H- —3-yl, 3,4-dihydro—5H—pyrrol—2-yl, 3,4-dihydro-5H-pyrrol—3-yl, 4,5- o- l H-pyrazo l—3—yl, 4,5-dihydro- l H-pyrazo lyl, 4,5 -dihydro- l H- pyrazol—S-yl, 2,5-dihydro-1H-pyrazol—3-yl, 2,5-dihydro-lH-pyrazol—4-yl, 2,5-dihydro-lH—pyrazol—S-yl, 4,5-dihydroisoxazol—3-yl, 4,5- 25 dihydroisoxazol—4-yl, 4,5-dihydroisoxazol—5—yl, 2,5-dihydroisoxazol—3-yl, 2,5-dihydroisoxazol—4-yl, 2,5—dihydroisoxazolyl, 2,3-dihydroisoxazol—3- yl, 2,3-dihydroisoxazolyl, 2,3-dihydroisoxazolyl, 4,5- dihydroisothiazol—3-yl, 4,5-dihydroisothiazolyl, 4,5-dihydroisothiazol—5- yl, 2,5-dihydroisothiazol—3-yl, 2,5-dihydroisothiazolyl, 2,5- 30 dihydroisothiazol—S—yl, 2,3—dihydroisothiazolyl, 2,3-dihydroisothiazol—4- yl, 2,3-dihydroisothiazol—5—yl, 4,5—dihydro-lH-imidazol—Z-yl, hydrolH-imidazol —4-yl, 4,5—dihydro—1H—imidazol—S-yl, 2,5-dihydro-lH-imidazol— 24 2-yl, 2,5-dihydro-1H-imidazol—4-yl, 2,5-dihydro-lH-imidazol-S-yl, 2,3- dihydro-lH-imidazol—Z—yl, 2,3—dihydro-1H-imidazol—4-y1, 4,5- dihydrooxazol—2-y1, 4,5-dihydr00xaz01—4-y1, 4,5-dihydrooxazol—5-yl, 2,5- dihydrooxazol—Z-yl, hydr00xazol—4-y1, 2,5-dihydrooxazol—5-yl, 2,3- dihydrooxazol—Z-yl, hydr00xazolyl, 2,3-dihydrooxazol—5-yl, 4,5- dihydrothiazol—2-yl, 4,5-dihydr0thiazol—4-yl, 4,5—dihydrothiazol—5-yl, 2,5- dihydrothiazol—Z-yl, 2,5—dihydr0thiazol—4-yl, 2,5—dihydr0thiazol—5-yl, 2,3- dihydrothiazol-Z-yl, 2,3-dihydrothiazolyl, 2,3-dihydrothiazol—5-yl, 1,3- dioxolyl, 1,3-dioxolyl, 1,3-dithioly1, 1,3-dithiolyl, 1,3-oxathi01- 10 2—y1, 1,3—oxathiol—4-yl, athiolyl.
C-bonded, ered, partially unsaturated rings such as: 2H-3,4-dihydropyran—6—yl, 2H—3,4—dihydropyran—5-yl, 2H-3,4- dihydropyran—4-y1, 2H—3,4—dihydr0pyran—3 -y1, 2H-3,4-dihydr0pyran—2-yl, 2H-3 ,4-dihydrothiopyran—6—yl, 2H—3,4—dihydr0thi0pyran—5-yl, 2H-3 ,4- 15 dihydrothiopyran—4-yl, 2H—3,4—dihydrothi0pyran—3-y1, 2H-3,4- dihydrothiopyran—2—yl, 1 ,2,3,4—tetrahydropyridinyl, 1 ,2,3 ,4- tetrahydropyridin-S-yl, 1 ,2,3,4—tetrahydropyridinyl, 1 ,2,3 ,4-tetra- hydropyridinyl, 1,2,3,4—tetrahydropyridiny1, 2H-5,6-dihydropyran—2- yl, 2H-5,6-dihydropyran—3—yl, 2H-5,6-dihydropyran—4-yl, - 20 dihydropyran—S-yl, —dihydropyran—6-yl, 2H-5,6-dihydr0thiopyran—2- yl, 2H-5,6-dihydrothiopyran—3—yl, 2H-5,6-dihydrothiopyran—4-yl, 2H-5,6- dihydrothiopyran—S-yl, 2H—5,6—dihydrothiopyran—6-yl, 1,2,5,6- tetrahydropyridinyl, 1,2,5,6-tetrahydropyridiny1, 1,2,5,6- ydropyridinyl, 1,2,5,6-tetrahydropyridiny1, 1,2,5,6-tetra- 25 hydropyridin-é-yl, 2,3,4,5—tetrahydr0pyridinyl, 2,3,4,5-tetrahydropyridin- 3-y1, 2,3,4,5-tetrahydr0pyridin—4-yl, 2,3,4,5—tetrahydr0pyridinyl, 2,3,4,5- tetrahydropyridinyl, anyl, 4H-pyrany1, 4H—pyrany1, 4H- thiopyran—2-yl, 4H-thiopyran—3-yl, 4H-thiopyrany1, 1,4-dihydropyridin—2- yl, 1,4—dihydropyridinyl, 1,4-dihydropyridiny1, 2H—pyran—2-yl, 2H- 30 pyran—3—y1, 2H—pyran—4—yl, 2H—pyran—5—yl, 2H-pyran—6-yl, 2H—thi0pyran—2- yl, opyran—3—yl, 2H—thiopyran—4—yl, 2H-thiopyran—5-yl, 2H-thi0pyran— 6-y1, 1,2-dihydr0pyridin—2—yl, 1,2—dihydropyridinyl, 1,2-dihydr0pyridin— 25 4-y1, 1,2-dihydropyridin—5—yl, hydropyridinyl, 3,4-dihydropyridin— 2-yl, 3,4-dihydropyridin—3—yl, 3,4—dihydropyridinyl, 3,4-dihydropyridin— 5-y1, 3,4-dihydropyridin—6-yl, 2,5—dihydropyridinyl, 2,5-dihydropyridin— 3-y1, hydropyridin—4-yl, 2,5—dihydropyridinyl, 2,5-dihydropyridin— 6-y1, 2,3-dihydr0pyridin—2-yl, 2,3—dihydropyridinyl, 2,3-dihydropyridin— 4-y1, 2,3-dihydr0pyridin—5—yl, 2,3-dihydropyridin—6-yl, 2H—5,6-dihydro-1,2- 0xazin—3-yl, 2H—5,6-dihydr0—1,2-0xazin—4-yl, 2H—5,6-dihydr0-1,2-oxazin yl, 2H-5,6-dihydro-1,2-oxazinyl, 2H-5,6-dihydro-1,2-thiaziny1, 2H-5,6-dihydro-1,2-thiazinyl, 2H-5,6-dihydro-1 ,2-thiaziny1, 2H—5 ,6- 10 dihydro— 1 ,2—thiazinyl, 4H-5,6-dihydro-1 ,2-oxazinyl, 4H-5 ,6-dihydro- 1,2—0xazin—4—yl, 4H—5,6—dihydro—1,2—0xazinyl, 4H-5 ,6-dihydr0-1,2- oxazin—6-y1, 4H—5,6—dihydro—1,2—thiazin—3—yl, 4H-5,6-dihydro-1,2-thiazin—4- yl, -dihydro—1,2—thiazin—5—yl, 4H—5,6-dihydr0-1,2-thiazinyl, 2H- 3,6-dihydro-1,2—oxazin—3—y1, 2H—3,6—dihydr0-1,2-oxazinyl, 2H-3,6- 15 dihydro-1,2-oxazinyl, 2H—3,6—dihydr0-1,2-0xazin—6-y1, 2H-3,6-dihydr0- 1,2-thiaziny1, 2H-3,6-dihydro—1,2—thiazin—4-y1, 2H-3,6-dihydr0-1,2- thiazin-S-yl, -dihydro-1,2—thiazin—6-y1, 2H-3,4-dihydr0-1,2-oxazin yl, 2H-3,4-dihydro-1,2-0xazin—4-yl, 2H-3,4-dihydr0-1,2-oxazinyl, 2H- 3,4-dihydr0-1,2-oxazin-6—yl, 2H-3,4-dihydro-1,2-thiaziny1, - 20 o- 1 ,2-thiazinyl, -dihydro-1,2-thiaziny1, 2H-3 ,4-dihydr0- 1,2-thiaziny1, 2,3,4,5—tetrahydropyridazin—3-y1, 5- tetrahydropyridazin-4—yl, 2,3,4,5—tetrahydropyridazin—5-y1, 2,3,4,5- tetrahydropyridazin—6-yl, 3,4,5,6—tetrahydropyridazin—3-y1, 3,4,5,6- tetrahydropyridazin—4-yl, 1 ,2,5,6-tetrahydr0pyridazin—3-y1, 1 ,2,5 ,6- 25 tetrahydropyridazinyl, 1 ,2,5,6-tetrahydr0pyridazin—5-y1, 1 ,2,5 ,6- tetrahydropyridazinyl, 1 -tetrahydr0pyridazin—3-y1, 1 ,2,3 ,6- tetrahydropyridazinyl, 4H-5,6-dihydro-1 ,3-oxazinyl, 4H—5 ,6-dihydro- 1,3-0xazin—4-yl, 4H-5,6-dihydro-1,3-oxaziny1, 4H-5,6-dihydro-1,3- oxazin—6-yl, 4H—5 ,6-dihydro- 1 ,3-thiaziny1, 4H-5,6-dihydr0-1,3-thiazin—4- 30 yl, 4H—5,6—dihydro—1,3—thiazin—5—yl, 4H—5,6-dihydr0-1,3-thiaziny1, 3,4,5- 6-tetrahydropyrimidin—Z—yl, 3,4,5,6—tetrahydr0pyrimidin—4-yl, 3,4,5,6-tetra— hydropyrimidin—S—yl, 3,4,5,6—tetrahydropyrimidin—6-y1, 1,2,3,4- 26 tetrahydropyrazinyl, l ,2,3,4-tetrahydropyrazinyl, l ,2,3 ,4- tetrahydropyrimidin—2—yl, l ,2,3,4-tetrahydropyrimidin—4-yl, l ,2,3 ,4- tetrahydropyrimidin—S—yl, l,2,3,4-tetrahydropyrimidin—6-yl, 2,3-dihydr0-l,4- thiazinyl, 2,3-dihydr0—l,4—thiazin—3-yl, 2,3-dihydro-l,4-thiazin—5-yl, 2,3- o- l ,4-thiazin-6—yl, 2H-l ,3—0xazinyl, 2H- 1 ,3-oxazinyl, 2H- 1 ,3- oxazin-S-yl, 2H— 1 ,3-0xazin—6-yl, 2H—1,3-thiazin—2-yl, 2H—1,3-thiazin—4-yl, 2H—1,3-thiazinyl, 2H—1,3-thiazinyl, 4H-l ,3-oxazinyl, 4H—1,3- oxazin—4-yl, 4H- 1 ,3-oxazinyl, 4H-1,3-oxazinyl, 4H—1,3-thiazin—2-yl, 4H-1,3-thiazinyl, 4H-1,3-thiazinyl, 4H-l ,3-thiazinyl, 6H—1,3- 10 oxazin—2-yl, 6H— 1 ,3-oxazinyl, 6H-1,3-oxazinyl, 6H-1,3-0xazin—6-yl, 6H— 1 ,3—thiazin—2—yl, 6H—1,3—oxazin—4—yl, 6H-l ,3-0xazinyl, 6H— 1 ,3- nyl, 2H— 1 ,4—oxazin—2—yl, 2H— 1 ,4—0xazin—3-yl, 2H- 1 ,4-0xazin—5-yl, 2H- 1 ,4-oxazinyl, 2H—1,4—thiazin—2—yl, 2H- 1 ,4-thiazinyl, 2H- 1 ,4- thiazin-S-yl, 2H- 1 ,4-thiazin—6—yl, 4H—1,4-0xazin—2-yl, 4H- 1 ,4-oxazinyl, 15 4H-l,4-thiazinyl, 4H-1,4—thiazin—3—yl, l,4-dihydr0pyridazin—3-yl, 1,4- dihydropyridazinyl, hydropyridazinyl, 1,4-dihydr0pyridazin yl, 1,4-dihydr0pyrazinyl, 1,2-dihydropyrazinyl, 1,2-dihydr0pyrazin yl, l,2-dihydr0pyrazin—5-yl, 1,2-dihydropyrazinyl, 1,4-dihydr0pyrirnidin- 2-yl, l,4-dihydropyrimidin—4-yl, 1,4-dihydropyrirnidin—5-yl, 1,4- 20 dihydropyrimidin—6-yl, 3,4—dihydropyrimidin—2-yl, 3,4-dihydropyrirnidin yl, 3,4-dihydropyrimidin—5-yl or 3,4-dihydropyrimidin—6-yl.
N—bonded, 5-membered, lly unsaturated rings such as: 2,3-dihydro- l H-pyrrol— l -yl, 2,5-dihydro- l H-pyrrol- l -yl, hydr0-1H- pyrazo l- l -yl, 2,5 -dihydr0— l H-pyrazo l— l -yl, 2,3 —dihydro- l H-pyrazo l- l -yl, 25 2,5-dihydroisoxazolyl, 2,3—dihydr0isoxazo1y1, 2,5-dihydroisothiazol—2- yl, 2,3-dihydr0isoxazol—2-yl, 4,5-dihydro- l azol— 1 -y1, 2,5-dihydro- lH-imidazol— l -yl, hydro- 1 H-imidazol- l -yl, hydrooxazol—3-yl, 2,3-dihydrothiazol—3-yl.
N—bonded, 6—membered, lly unsaturated rings such as: 30 l ,2,3 ,4—tetrahydropyridin—1—yl, 1 ,2,5 ,6—tetrahydr0pyridin— l -yl, l ,4- dihydropyridin— l —yl, l ,2—dihydropyridin— l —yl, 2H-5 ,6-dihydr0- l ,2-0xazin—2- yl, 2H-5,6-dihydro—l,2—thiazin—2—yl, 2H—3,6-dihydr0-l,2-0xazin—2-yl, 2H- 27 3,6-dihydro- l ,2-thiazin—2—yl, 2H-3,4-dihydro- l ,2-oxazinyl, - dihydro-l,2-thiazin-2—yl, 2,3,4,5-tetrahydropyridazinyl, l,2,5,6- ydropyridazin-l—yl, l,2,5,6-tetrahydropyridazin—2-yl, l,2,3,6- tetrahydropyridazin—l-yl, 3,4,5,6-tetrahydropyrimidin—3-yl, l,2,3,4- tetrahydropyrazin- l -yl, 1 ,2,3 ,4—tetrahydropyrimidin— l -yl, l ,2,3 ,4-tetrahydro- pyrimidinyl, 2,3—dihydro-1,4—thiazinyl, 2H—1,2-oxazin—2-yl, 2H—1,2- thiazinyl, 4H—l,4—oxazin—4-yl, 4H—1,4-thiazinyl, hydropyridazin- l-yl, 1,4-dihydropyrazinyl, 1,2-dihydropyrazin-l-yl, 1,4- dihydropyrimidin- l -yl or 3,4-dihydropyrimidinyl. 10 Examples of saturated or partially unsaturated bicycles are in particular radicals corresponding to saturated or partially unsaturated ocyclic radicals, wherein l, 2 or 3 CH or CH2 moieties have been replaced by N, NH, O, S, S(=O) or S(=O)2, such as 2-oxa—6-azaspiro-[3,4]octyl, 2-azabicyclo[2.2.l]heptyl, 5- yclo[2.2.l]heptyl, 2,5-diazabicyclo[2.2.l]heptyl, 3-azabicyclo[3.2.l]octyl, 8- 15 azabicyclo [3 .2. l]octyl, 3 , abicyclo[3 .2. l]octyl, dihydroindo lyl, dihydroindolizynyl, dihydroisoindolyl, oquinolinyl, dihydroisoquinolinyl, chromenyl and chromanyl.
Hetaryl: a 5- or 6-membered aromatic heteromonocyclic radical (also termed 5- or 6-membered monocyclic hetaryl) which ordinarily has 1, 2, 3 or 4 heteroatoms as ring 20 members, which are selected from O, S and N, and which has in particular 1, 2, 3 or 4 nitrogen atoms or a heteroatom selected from oxygen and sulfur and, if appropriate, 1 or 2 nitrogen atoms as ring members besides carbon atoms as ring members and a 8- to 10- ed aromatic heterobicyclic radical (also termed 8- to lO-membered bicyclic hetaryl) which ordinarily has 1, 2, 3 or 4 heteroatoms as ring members, which are 25 selected from O, S and N, and which has in particular 1, 2, 3 or 4 nitrogen atoms or a heteroatom selected from oxygen and sulfur and, if appropriate, 1 or 2 nitrogen atoms as ring members besides carbon atoms as ring members: for example C-bonded, 5-membered monocyclic hetaryl having 1, 2 or 3 or 4 nitrogen atoms or a heteroatom selected from oxygen and sulfur and, if appropriate, having 1, 2 30 or 3 nitrogen atoms as ring members, such as: l, 3-furyl, 2—thienyl, nyl, pyrrol—2—yl, pyrrol—3-yl, l—3-yl, pyrazol—4-yl, isoxazol—3—yl, isoxazol—4—yl, isoxazol-S-yl, isothiazol—3-yl, 28 isothiazol—4-yl, isothiazol—S-yl, imidazol—2-yl, imidazol—4-yl, oxazol—2-yl, oxazol— 4-yl, —S-yl, thiazol—2—yl, thiazol—4-yl, thiazol-S-yl, 1,2,3-oxadiazolyl, 1,2,3-oxadiazol—5-yl, 1,2,4—oxadiazol—3-yl, l,2,4,—oxadiazol—5-yl, 1,3,4-oxadiazol- 2-yl, 1,2,3-thiadiazol—4-yl, thiadiazol—5-yl, 1,2,4-thiadiazol—3-yl, 1,2,4- thiadiazol—S-yl, 1,3,4-thiadiazolylyl, 1,2,3-triazol—4-yl, 1,2,4-triazolyl, tetrazol—S-yl.
C-bonded, 6-membered monocyclic hetaryl having 1, 2 or 3 nitrogen atoms as ring members, such as: pyridinyl, pyridinyl, pyridinyl, pyridazinyl, pyridazinyl, pyrimidin- 10 2—yl, pyrimidin—4—yl, pyrimidin-S-yl, pyrazinyl, 1,3,5-triazinyl, 1,2,4-triazin- 3-yl, 1,2,4—triazin—5—yl, 1,2,4—triazin—6—yl, 1,2,4,5-tetrazin—3-yl.
N—bonded, 5-membered heteroaromatic radicals having 1, 2, 3 or 4 nitrogen atoms as ring members, such as: pyrrol— 1 -yl, pyrazol— 1 -yl, imidazol— 1 —y1, 1,2,3—triazol— l -yl, 1,2,4-triazolyl, 15 tetrazol— 1 -yl. bicyclic 8 to lO-membered hetaryl, hetaryl which has one of the aforementioned 5- or 6-membered aromatic rings and a r aromatic carbocycle or 5- or 6-membered heterocycle fused thereto, for example a fused benzene, thiophene, furane, pyrrole, pyrazole, imidazole, pyridine or pyrimidine ring. These bicyclic 20 hetaryl include for e inyl, isoquinolinyl, cinnolinyl, indolyl, indolizynyl, isoindolyl, indazolyl, benzofuryl, benzothienyl, b]thiazolyl, benzoxazolyl, benzthiazolyl, benzimidazolyl, imidazo[l,2-a]pyridineyl, thieno [3 yridine-5 -yl, imidazo—[2,1-b]-thiazolyl and 1 ,2,4-triazo lo [ 1 ,5 - dineyl. 25 Hetarylalkyl: a hetaryl radical as defined above which is linked via an alkylene group, in particular via a methylene, 1,1—ethylene or hy1ene group, to the remainder of the molecule.
The expression "optionally tuted" in the context of the present invention means that the tive moiety is unsubstituted or has 1, 2 or 3, in particular 1, 30 substituents Which are selected from halogen, C1-C4-alkyl, C1-C4-haloalkyl, OH, SH, CN, CF3, O-CFg, COOH, O—CHz—COOH, C1—C6—alkoxy, haloalkoxy, C1-C6- alkylthio, C3-C7-cycloalkyl, COO—Cl—C6—alkyl, CONHZ, CONH-Cl-Cg-alkyl, SOZNH- 29 C1-C6-alkyl, CON-(Cl-Cg-alkylh, SOzN—(Cl-C6-alkyl)2, NH-SOz-Cl-C6-alkyl, NH-COC1-C6-alkyl , -CG-alkyl, O-phenyl, O-CHg-phenyl, CONH-phenyl, SOZNH- phenyl, CONH-hetaryl, SOZNH-hetaryl, SOg-phenyl, NH-SOz-phenyl, NH-CO-phenyl, NH-SOz-hetaryl and NH-CO-hetaryl, where phenyl and hetaryl in the last 11 radicals mentioned are unsubstituted or may have 1, 2 or 3 substituents which are ed from halogen, C1-C4-alkyl, C1-C4-haloalkyl, C1-C4-alkoxy and C1-C4-haloalkoxy.
In relation to their use as inhibitors of PDEIOA, the variables Het, A, A', Q, X1, X2, X3, Y1, Y2, Y3, R1, R2, R3, R4, R5, R6, R7, R8, R9, Cycl, Cyc2 and Cyc3 preferably have the following meanings, where these represent, both considered on their own and 10 in combination with at least one other or all, special configurations of the compounds of the formula I: In a particular embodiment, Het is unsubstituted or may carry 1, 2, 3 or 4 identical or different tuents Rx. In this regard, Rx is selected from the group consisting of H, halogen, C1-C4-alkyl, C1-C4-alkoxy, C1—C4—fluoroalkyl, C1-C4-fluoroalkoxy, C3-C6- 15 lkyl, C1-C4-alkoxy-C1-C4-alkoxy, C1—C4—alkoxy-C1-C4-alkyl, OH, y—Cl- C4-alkyl, O-Cg-C6-cycloalkyl, benzyloxy, C(O)O-(C1-C4-alkyl), C4-alkyl)-C02H, N(RX1)(RX2), RX1)(R"2), C1-C4-alkyl-N(RX1)(RX2), -NRX3-C(O)-N(RX1)(RX2), NRX3-C(O)O-(C1-C4-alkyl), -N(RX3)-SOz-RX4, phenyl, CN, -SF5, -OSF5, -SOZRX4, -SR"4 and trimethylsilyl, where R“, sz, RX3 and RX4, independently of each other are selected 20 from the group consisting of hydrogen, alkyl, C1-C4-fluoroalkyl and C3-C6- cycloalkyl or RX1 and sz form together with the N atom to which they are attached a 3- to 7-membered, nitrogen heterocycle which may have 1, 2 or 3 further different or identical atoms or heteroatom containing groups selected from the group of O, N, S, SO and 802 as ring members and which may carry 1, 2, 3, 4, 5 or 6 substituents 25 selected from C1-C4-alkyl.
Het is preferably selected from the group consisting of C-bound 6-membered monocyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members, and C-bound, fused bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and ally a further heteroatom selected from O, S and N as ring member, where monocyclic 30 hetaryl and bicyclic hetaryl may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, l or 2 substituents Rx. In this regard, RX is preferably selected from n, alkyl, C1-C2-fluoroalkyl, C1—C4—alkoxy, C1-C2-fluoralkoxy, phenyl, C3- 30 C6-cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated C3-C6- cycloalkyl. In this regard, Rx is in particular selected from e, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, phenyl, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
In a particular embodiment of the invention, Het is selected from fiased bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring s and ally a further heteroatom selected from O, S and N as ring member and which may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in ular 0, 1 or 2 substituents RX. In this 10 regard, RX is ably selected from halogen, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4- , C1-C2-fluoralkoxy, C3—C6—cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated C3—C6—cycloalkyl. In this regard, RX is in particular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, romethoxy, ropyl, optionally substituted 15 by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
In another particular embodiment of the invention, Het is selected from 6- membered monocyclic hetaryl, which may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, l or 2 substituents RX. In this regard, RX is preferably selected from halogen, C1-C4-alkyl, C1—C2-fluoroalkyl, C1-C4-alkoxy, C1-C2- 20 fluoralkoxy, phenyl, C3-C6-cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and ated C3-C6-cycloalkyl. In this regard, RX is in particular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, omethoxy, romethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl or one Rx may also be phenyl, 25 ular preference is given to those Het radicals, which have at least one imino- nitrogen as ring member, which is located in the position adjacent to the carbon atom which is bound to A. Particular preference is given to those Het radicals, which have at least one imino-nitrogen as ring member, which is located in the position adjacent to the carbon atom which is bound to tA and which are ed from the group consisting of 30 d 6-membered monocyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members, uryl and C—bound, fused bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and optionally a further heteroatom selected from O, S and N as 31 ring , where monocyclic hetaryl, benzofuryl and bicyclic hetaryl may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, l or 2 tuents RX. In this , Rx is preferably ed from halogen, C1-C4-alkyl, C1-C2- fluoroalkyl, C1-C4-alkoxy, C1-C2—fluoralkoxy, , C3-C6-cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated C3-C6-cycloalkyl. In this regard, Rx is in particular selected from e, ne, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, phenyl, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, and fluorinated cyclopropyl. 10 Particular examples of Het are selected from the group consisting of 2-benzo furyl, 2-pyridyl, 2-pyrimidinyl, 4—pyrimidinyl, 2—pyrazinyl, 3-pyridazinyl, 2-quinolinyl, 3- nolinyl, 2-quinazolinyl, 2—quinoxalinyl, 1,5—naphthyridin—2-yl, 1,8-naphthyridin- 2-yl, benzothiazol— l -yl, benzoxazol— 1 —yl, benzimidazol—2-yl, l-methylbenzimidazol—2- yl, imidazo[l ,2-a]pyridineyl, thieno[3,2—b]pyridineyl, imidazo-[2, l -b]-thiazolyl 15 and l,2,4-triazolo[l,5-a]pyridineyl, where the entioned radicals are unsubstituted or may carry 1, 2 or 3 radicals RX as defined above, which are in particular selected from the group consisting of fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, romethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and 20 fluorinated cyclopropyl.
In a particular ment ofthe invention, Het has at least one imino-nitrogen as ring member, which is located in the position adjacent to the carbon atom which is bound to A and Het is selected from the group consisting of fiased bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and optionally a further heteroatom 25 selected from O, S and N as ring , where bicyclic hetaryl may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, 1 or 2 substituents RX. In this regard, RX is preferably selected from halogen, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4- alkoxy, C1-C2-fluoralkoxy, cycloalkyl, optionally substituted by 1, 2 or 3 methyl groups, and ated C3-C6-cycloalkyl. In this regard, Rx is in particular selected from 30 fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, ally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl. Particular examples of Het of 32 this embodiment are 2-quinolinyl, 3—isoquinolinyl, 2-quinazolinyl, 2-quinoxalinyl, 1,5- naphthyridin—2-yl, l ,8-naphthyridin—2—yl, benzothiazol— l -yl, benzoxazo l- l -yl, benzimidazol—2-yl, l-methylbenzimidazol—2—yl, imidazo[ l ,2-a]pyridineyl, thieno[3 ,2- b]pyridine-5 -yl, imidazo- [2, l -b]—thiazolyl and l,2,4-triazo lo [ l ,5 idineyl, where the aforementioned radicals are unsubstituted or may carry 1, 2 or 3 radicals RX as defined above, which are in particular selected from the group consisting of fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, and fluorinated cyclopropyl. 10 Particular preference is given to compounds, where Het is 2-quinolinyl, 1,8- naphthyridin—2-yl, benzothiazol—l—yl, benzoxazol— l —yl, benzimidazol—Z-yl, l- methylbenzimidazol—2-yl, imidazo[1,2—a]pyridine—2—yl, thieno[3,2-b]pyridineyl, and in particular 2-quinolinyl or imidazo[1,2—a]pyridine—2-yl, where these radicals are unsubstituted or may carry 1, 2 or 3 radicals Rx as defined above, which are in particular 15 selected from the group consisting of fluorine, chlorine, methyl, ethyl, difluoromethyl, trifluoromethyl, methoxy, ethoxy, difluoromethoxy, romethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and ated ropyl.
In particular ments of the invention, the variable X1 is CH. In these 20 particular embodiments, X2 is preferably C-R7, where R7 is as defined above and in particular H or YZ-Cycz. In these ular embodiments, X3 is preferably S or C(R9)=C(R8).
In further particular embodiments of the invention, the variable X1 is N. In these particular embodiments, X2 is preferably C-R7, where R7 is as defined above and in 25 particular H or z. In these particular embodiments, X3 is preferably S or C(R9)=C(R8).
In particular embodiments ofthe invention, the variable X2 is C-R7. In these particular embodiments R7 is as defined above and in particular H or Yz-Cycz. In these embodiments, X1 is CH or preferably N. In these particular embodiments, X3 is 30 ably 5 or C(R9)=C(R8). 33 In r particular embodiments ofthe invention, the variable X2 is N. In this embodiment, X1 is preferably CH. In these particular embodiments, X3 is preferably C(R9)=C(R8).
In particular embodiments ofthe invention X3 is O, S, -X4=C(R8)-, Where C(RS) is bound to the carbon atom which carries R2.
In particular ments of the invention X3 is S. In these embodiments, X1 is CH or preferably N and X2 is preferably C-R7, where R7 is as defined above and in particular H or Yz-Cycz.
In particular embodiments of the invention X3 is O. In these embodiments, X1 is N 10 or preferably CH, X2 is preferably C—R7, where R7 is as defined above and in particular H or 2 and R1 is as defined above and in particular H or Yl-Cycl.
In further particular ments of the invention, the variable X3 is C(R9)=C(R8). In these embodiments, X1 is CH or preferably N and X2 is preferably C- R7, where R7 is as defined above and in particular H or YZ-Cycz. 15 In further particular embodiments of the invention, the variable X3 is N=C(R9). In these embodiments, X1 is CH or ably N and X2 is preferably C-R7, where R7 is as defined above and in particular H and R1 is as defined above and in ular Yl-Cycl.
In the embodiments ofthe invention, where X3 is N=C(R9), R9 is preferably H.
In further particular embodiments of the invention, the variable X3 is N=C(R8). In 20 these embodiments, X1 is preferably CH and X2 is preferably C-R7, where R7 is as defined above and in particular H or YZ-Cycz.
In the embodiments ofthe invention, where X2 is C-R7, R7 is preferably H or Y2- Cyc2.
In the embodiments ofthe invention, where X3 is C(R9)=C(R8), R9 is preferably H 25 or Y3-Cyc3, while R8 is preferably en.
In the embodiments ofthe invention, where X3 is N=C(R8), R8 is preferably H.
In the embodiments ofthe invention, where X2 is C-R7, and where X3 is C(R8), R7 is preferably H or YZ-Cycz, R9 is preferably H or Y3-Cyc3, while R8 is ably hydrogen, where preferably either R7 is Yz-Cyc2 or R9 is Y3-Cyc3 While one 30 of R7 and R9 is different from Yz—Cyc2 or Y3—Cyc3, respectively, or both R7 and R9 are different from Yz-Cyc2 or Y3—Cyc3. 34 In this , those ls R], R7 and R9, which are different from Yl-Cycl, YZ-Cycz, Y3-Cyc3 , respectively, are in particular selected, independently of each other, from the group consisting of hydrogen, fluorine, C1-C4-alkyl, fluorinated C1-C2-alkyl, C1-C4-alkoxy, fluorinated C1-C2-alkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl. Those radicals R1, R7 and R9, which are different from Yl-Cycl, YZ-Cycz, Y3-Cyc3, respectively, are most preferably hydrogen.
In particular preferred embodiments of the invention, R1 is Yl-Cycl. In these embodiments X1 is CH or preferably N and X2 is preferably C-R7, where R7 is as defined above and in particular H or YZ-Cycz. In these embodiments, particular 10 preference is given to compounds, where X3 is S or X3 is C(R9)=C(R8), where R9 is ably H or Y3—Cyc3, while R8 is preferably en.
Y3-Cyc3, while R8 is preferably hydrogen.
In other particular preferred embodiments of the invention, R1 is different from Yl-Cyc1 and in particular hydrogen. In these embodiments X1 is CH or ably N. In 15 these embodiments X2 is preferably C—R7, where R7 is as defined above and in particular YZ-Cyc2 and X3 is S or X2 is preferably C-R7, where R7 is as defined above and in particular H or Yz-Cyc2 and X3 is X3 is C(R9)=C(R8), where R9 is preferably H or 3, while R8 is preferably hydrogen.
In the moiety Yl-Cycl, Y1 is ably selected from O, NH and a chemical 20 bond. In particular Y1 is a chemical bond.
In the moiety Yz-Cycz, Y2 is preferably selected from O, NH and a chemical bond. In particular Y2 is a chemical bond.
In the moiety Y3-Cyc3, Y3 is preferably selected from O, NH and a chemical bond. In particular Y3 is a al bond. 25 Preferably, Cyc1 is selected from the groups of (i) saturated 4-, 5-, 6-, 7- or 8—membered heteromonocycles or a saturated 7-, 8-, 9- or lO-membered heterobicycle, where the heteromonocycle and the heterobicycle have one nitrogen or oxygen atom as ring member and may have one fiirther heteroatom or atom group as ring member, which is selected from the group consisting of O, 30 S, S(=O), S(=O)2 and N, where the saturated monocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 ls RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in ular 0, l or 2 radicals RC1, where 35 RC1, RC2 and Y' are as defined herein and where Y', if t, is preferably a chemical bond or O; and (ii) phenyl or a 5- or 6 membered monocyclic hetaryl, which has one heteroatom, selected from O, S and N as ring member and optionally one or two further heteroatoms as ring members, and which is in particular selected from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, olyl, pyrazolyl, oxazolyl and thiazolyl, 9- or lO-membered bicyclic l which has one heteroatom, ed from O, S and N as ring member and optionally one or two further heteroatoms as ring members, and which is in particular selected from the group consisting of indolyl, 10 quinolinyl, isoquinolinyl, quinazolinyl, benzimidazolyl, benzotriazolyl, benzopyrazolyl and benzo furyl, where phenyl and hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one l Y'—RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O. 15 In this regard, RC1 is preferably selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2, or, if Cyc1 is , two radicals RC1 which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic heterocyclic radical, which is selected from 5- or 6-indolyl, 5- or 6-benzimidazolyl, 5- or 6-benzopyrazolyl, 5- or 6- 20 benzotriazolyl, 5- or 6-benzofuranyl, 2,3—dihydrobenzofuran—5-yl, 2,3- obenzofuran—6-yl, l,3-dihydroindol—2-on—5-yl, l,3-dihydroindol—2-on—6-yl, 5- or 6-quinolinyl, 5- or 6-isoquinolinyl, 5— or 6-quinazolinyl, 2-aminoquinazolinyl, and 2- aminoquinazolinyl.
In this regard, RCZ is ably selected from the group consisting of phenyl, C3- 25 Cs-cycloalkyl, optionally substituted by 1, 2, or 3 methyl groups, ated C3-C6- lkyl, and 5- or 6-membered ted heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, where phenyl the saturated heteromonocyclic radical is unsubstituted or carries 1, 2 or 3 radicals RC3, which are preferably selected from fluorine, chlorine, CN, methyl, difluoromethyl, 30 trifluoromethyl, methoxy and NH2.
In particular, Cyc1 is selected from the groups of 36 (i) saturated 4-, 5-, 6- or ered heteromonocycles, where the heteromonocycle has one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RCl is as defined herein; and (ii) phenyl or a 5- or 6 membered l, selected from pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and lyl, where phenyl and the 5- or 6 membered hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, 10 or 3 radicals RC1 or one radical Y‘-RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In particular embodiments of the invention, Cyc1 is selected from the group consisting of saturated 4-, 5-, 6- or 7—membered heteromonocycles or a ted 7-, 8-, 15 9- or 10-membered heterobicycle, where the heteromonocycle and the heterobicycle have one nitrogen or oxygen atom as ring member and may have one r heteroatom or heteroatom group as ring , which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the ted heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals 20 RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In special embodiments ofthe invention, Cycl is selected from the group consisting of saturated 4-, 5-, 6- or ered heteromonocycles, where the 25 heteromonocycle has one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O); and N, where the saturated heteromonocycle and the saturated heterobicycle are tituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein. 30 In this particular and special embodiment, Y1 is preferably selected from O, NH and a chemical bond, with particular preference given to Y1 being a chemical bond. 37 In this particular and special embodiment Yl-Cyc1 is e.g. selected from the group consisting of l-piperidinyl, 4,4-difluoropiperidinyl, 4-piperidinyl, l-methyl piperidinyl, l-piperazinyl, 4-methylpiperazinyl, morpholin—4-yl, 2-oxaazaspiro- ctyl, 2,5-diazabicyclo[2.2.I]heptan—2—yl, 3,8-diazabicyclo[3.2.l]octan—8-yl, thiomorpholinyl, l-oxothiomorpholin-4—yl, N-(oxetan—3-yl)amino, l,l- dioxothiomorpholinyl and oxetan—3—ylamino and especially from the group consisting of l-piperidinyl, 4,4-difluoropiperidinyl, 4-piperidinyl, 1—methy1—4- piperidinyl, 1-piperazinyl, 4-methylpiperazinyl, morpholinyl, thiomorpholinyl, l-oxothiomorpholinyl, N-(oxetanyl)amino, 1, 1 -dioxothiomorpholinyl and 10 oxetan—3-ylamino.
In other ular embodiments of the invention, Cyc1 is phenyl or a 5- or 6 membered heteroaromatic radical, which has one heteroatom, selected from O, S and N as ring member and optionally one or two further heteroatoms as ring s, and which is in particular ed from the group consisting of l, pyrimidinyl, fiiryl, 15 thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered heteroaromatic radical are tituted or either carry, independently of each other, carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'- RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O. 20 In special embodiments ofthe ion, Cycl is selected from the group consisting of phenyl or a 5- or 6 membered l, selected from pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, yl and thiazolyl, where phenyl and the 5- or 6 ed hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RCl or one radical Y'—RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals 25 RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a al bond or O.
In particular Cyc1 is selected from the group consisting of phenyl and 5- or 6- membered hetaryl selected from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and lyl, where phenyl and 30 hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2, or, if Cyc1 is Pheny1, two radicals RC1 which are bound to adjacent 38 carbon atoms, together with the phenyl ring to which they are bound, form a ic heterocyclic radical, which is selected from 5- or 6-indolyl, 5- or 6-benzimidazolyl, 5- or 6-benzopyrazolyl, 5- or otriazolyl, 5- or 6-benzofuranyl, 2,3- dihydrobenzofiaran—S-yl, 2,3-dihydrobenzofi1ran—6-yl, l,3-dihydroindolonyl, 1,3- dihydroindol—2-on—6-yl, 5- or 6-quinolinyl, 5— or 6-isoquinolinyl, 5- or 6-quinazolinyl, 2-aminoquinazolinyl, and 2-aminoquinazolinyl. Amongst these, particular preference is given to compounds, where Y1 is a chemical bond. Amongst these, particular preference is given to compounds, where Cyc1 is selected from the group consisting of phenyl and 5- or ered hetaryl selected from the group consisting of 10 pyridyl, pyrimidinyl, fiiryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are selected from the group ting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2.
Preferably, Cyc2 and Cyc3 are, independently from each other, selected from the 15 groups of (i) ted 4-, 5-, 6-, 7- or 8-membered heteromonocycles or a saturated 7-, 8-, 9- or lO-membered heterobicycle, where the heteromonocycle and the heterobicycle have one en or oxygen atom as ring member and may have one fiarther heteroatom or heteroatom group as ring member, which is ed from the group consisting of O, 20 S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in ular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O; and 25 (ii) phenyl or a 5- or 6 membered monocyclic hetaryl, which has one atom, selected from O, S and N as ring member and optionally one or two r heteroatoms as ring members, and which is in particular ed from the group consisting of pyridyl, pyrimidinyl, fiiryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, 9- or lO-membered bicyclic hetaryl which has one heteroatom, selected from 30 O, S and N as ring member and optionally one or two further heteroatoms as ring members, and which is in particular ed from the group consisting of indolyl, quinolinyl, isoquinolinyl, quinazolinyl, benzimidazolyl, benzotriazolyl, benzopyrazolyl 39 and benzo fiaryl, where phenyl and hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in ular 1, 2, or 3 radicals RC1 or one radical Y'—RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is ably a chemical bond or O.
In this regard, RCl is preferably selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, y and NH2, or, if Cyc2 or Cyc3 are phenyl, two radicals RC1 which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic heterocyclic radical, which is selected from 5- or 6-indolyl, 5- or 6-benzimidazolyl, 5- or 6-benzopyrazoly1, 10 5- or 6—benzotriazolyl, 5- or 6-benzofuranyl, 2,3-dihydrobenzofuran-S-yl, 2,3- dihydrobenzofuran—6—yl, 1,3—dihydroindol—2—on—5—yl, 1,3-dihydroindol—2-on—6-yl, 5- or olinyl, 5- or 6-isoquinolinyl, 5— or 6—quinazolinyl, 2-aminoquinazolinyl, and 2- aminoquinazolinyl.
In this regard, RC2 is preferably ed from the group consisting of , C3- 15 C6-cycloalkyl, optionally substituted by 1, 2, or 3 methyl groups, fluorinated C3-C6- lkyl, and 5- or 6-membered saturated heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, where phenyl the saturated heteromonocyclic radical is unsubstituted or carries 1, 2 or 3 radicals RC3, which are ably selected from e, chlorine, CN, methyl, difluoromethyl, 20 trifluoromethyl, methoxy and NH2.
In particular, Cyc2 and Cyc3 are, independently from each other, selected from the groups of (i) saturated 4-, 5-, 6- or 7-membered heteromonocycles, where the heteromonocycle has one nitrogen or oxygen atom as ring member and may have one 25 further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O); and N, where the saturated heteromonocycle and the saturated bicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein; and (ii) phenyl or a 5— or 6 membered hetaryl, selected from pyridyl, pyrimidinyl, 30 furyl, l, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y‘—RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals 40 RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In ular embodiments ofthe invention, Cyc2 and Cyc3 are, independently from each other, selected from the group consisting of ted 4-, 5-, 6- or 7- membered heteromonocycles or a saturated 7-, 8-, 9- or lO-membered heterobicycle, where the heteromonocycle and the heterobicycle have one en or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted 10 or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 ls RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y’ are as defined herein and where Y', if present, is preferably a chemical bond or O.
In special embodiments ofthe invention, Cyc2 and Cyc3 are, independently from each other, selected from the group consisting of saturated 4-, 5-, 6- or 7-membered 15 heteromonocycles, where the heteromonocycle has one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated bicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein. 20 In this particular and special ments Y2 and Y3 are, independently from each other preferably selected from O, NH and a chemical bond, with particular preference given to Y2 and Y3 being a chemical bond.
In this particular and special embodiments Cyc2 and Cyc3 are, independently from each other, e. g. selected from the group consisting of l-piperidinyl, 4,4-difluoro-l- 25 dinyl, 4-piperidinyl, 1 -methyl—4—piperidinyl, 1 -piperazinyl, 4-methy1— 1 - piperazinyl, linyl, 2-oxaazaspiro-[3,4]octy1, 2,5— diazabicyclo[2.2.1]heptanyl, 3,8-diazabicyclo[3.2.1]octanyl, thiomorpholinyl, 1-oxothiomorpholinyl, N-(oxetanyl)amino, 1,1-dioxothiomorpholinyl and —3—ylamino and especially from the group consisting of 1-piperidinyl, 4,4- 30 difluoro- l -piperidinyl, 4—piperidinyl, 1 l—4—piperidinyl, l -piperazinyl, 4-methyl- l - piperazinyl, morpholin-4—yl, thiomorpholin—4—yl, 1—oxothiomorpholin—4-yl, N-(oxetan— 3-yl)amino, l,l-dioxothiomorpholin—4—yl and oxetan—3-ylamino. 41 In other particular ments of the invention, YZ-Cyc2 and Y3-Cyc3 are, independently of each other, phenyl or a 5— or 6 membered heteroaromatic l, which has one heteroatom, selected from O, S and N as ring member and optionally one or two fiirther heteroatoms as ring members, and which is in particular selected from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5— or 6 ed heteroaromatic radical are unsubstituted or either carry, independently of each other, carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 ls RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, 10 is ably a chemical bond or O.
In special embodiments of the invention, YZ—Cyc2 and Y3-Cyc3 are, independently from each other, selected from the group consisting of phenyl or a 5- or 6 membered l, selected from pyridyl, pyrimidinyl, fiiryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered hetaryl are 15 unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In particular YZ-Cyc2 and 3 are, independently from each other, selected from the group consisting of phenyl and 5- or 6-membered hetaryl selected from the 20 group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, romethyl, methoxy and NH2, or, if one or both of Y2- Cyc2 and 3 are phenyl, two radicals RCl which are bound to adjacent carbon 25 atoms, er with the phenyl ring to which they are bound, form a bicyclic heterocyclic radical, which is selected from 5- or lyl, 5- or 6—benzimidazoly1, 5- or 6-benzopyrazolyl, 5- or 6-benzotriazolyl, 5- or 6-benzofurany1, 2,3- dihydrobenzofuran—S-yl, 2,3-dihydrobenzofuranyl, 1,3-dihydroindolony1, 1,3- dihydroindol—2—on—6-yl, 5— or 6-quinolinyl, 5- or 6-isoquinoliny1, 5- or 6-quinazolinyl, 30 2-aminoquinazolinyl, and 2—amino—6—quinazolinyl. Amongst these, particular preference is given to compounds, where Yz—Cyc2 and Y3-Cyc3 are, ndently from each other, selected from the group consisting of phenyl and 5- or 6-membered hetaryl 42 selected from the group consisting of pyridyl, dinyl, furyl, thienyl, pyrrolyl, olyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC] which are selected from the group consisting of fluorine, ne, CN, methyl, omethyl, trifluoromethyl, methoxy and NH2.
With regard to Cycl, Cyc2 and Cyc3, and in particular with regard to the aforementioned ular or special embodiments of Cycl, Cyc2 and Cyc3 RC1 is , preferably selected from the group consisting of fluorine, chlorine, CN, , difluoromethyl, trifluoromethyl, methoxy and NH2. 10 With regard to Cycl, Cyc2 and Cyc3, and in particular with regard to the aforementioned particular or special embodiments of Cycl, Cyc2 and Cyc3, RC2 is preferably selected from the group consisting ofphenyl, C3-C6-cycloalkyl, optionally substituted by l, 2, or 3 methyl groups, fluorinated C3-C6-cycloalkyl, and 5- or 6- ed saturated heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring 15 members, which are selected from O, S and N, where phenyl the saturated heteromonocyclic radical is unsubstituted or carries 1, 2 or 3 radicals RC3 , which are preferably selected from fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2.
Irrespectively from the entioned embodiments, R2 is preferably selected 20 from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4- alkoxy, fluoroalkoxy, cyclopropyl, ally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
Irrespectively from the aforementioned embodiments, R2 is in particular hydrogen. 25 Irrespectively from the aforementioned embodiments, Q is in particular oxygen.
Particular embodiments of the invention relate to the compounds of the following formulae 1-1 and 1-2: 0 1 R Het—A\ I}! l \ R2 (M) N\ 8 R7 43 O R1 Het—A\N l \ R2 (I-2) \ 8 R7 where Het, A, R1, R2 and R7 are as defined here and in the claims.
Other particular embodiments of the invention relate to the compounds of the following ae 1-3 and 1-4: 0 R Het—A\ R2 I? 0-3) N \ R8 R7 R9 o R1 Het—A\ R2 N (I-4) \ R8 R7 R9 where Het, A, R1, R2, R7, R8 and R9 are as defined here and in the claims.
Further particular embodiments of the invention relate to the compounds of the following formulae 1-5 and 1-6: 0 R Het—A\ R2 “I1 \ | 0-5) N \ / N R8 44 (l-6) where Het, A, R1, R2, R7 and R8 are as defined here and in the claims. ular embodiments of the invention relate to the compounds of the following formulae 1-7 and 1-8: 0 R1 Het—A\ I}! l \ R2 (I-7) 7 R 0 R1 N l \ R2 (I-8) 0 R7 where Het, A, R1, R2 and R7 are as defined here and in the claims.
Further particular embodiments of the invention relate to the compounds of the 10 following formulae 1-9 and 1-10: 0 R Het—A\ R2 “I1 \ | 0-9) N \ / N 45 o R Het—A\ R2 N \ l (I-10) \ / N R7 R9 where Het, A, R1, R2, R7 and R9 are as defined here and in the claims.
In a particular embodiment of the invention, A in formula I is Al, A2, A3 or A4.
In a particular embodiment of the invention, A in formula I, and likewise in formulae 1—1, 1—2, 1—3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10 is a bivalent radical A1. ence is given to compounds of the formula I, and likewise to compounds of formulae 1-1, 1—2, 1—3, 1—4, 1—5, 1-6, 1—7, 1-8, 1—9 and 1—10, where R3, R4 are selected from hydrogen and fluorine and in particular to those compounds, where both R3 and R4 are 10 hydrogen. Preference is given to compounds of the a I, where R5 and R6 are, independently of each other, selected from the group consisting of hydrogen, fluorine and methyl, and in particular to those compounds, where both R5 and R6 are en.
In another particular embodiment of the invention, A in a I, and likewise in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10, is a bivalent radical A2. 15 In another particular embodiment of the ion, A in formula I, and likewise in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1—7, 1—8, 1-9 and 1-10, is a bivalent radical A3.
Preference is given to compounds ofthe formula I, and likewise to compounds of formulae 1-1, 1—2, 1—3, 1—4, 1—5, 1-6, 1—7, 1-8, 1—9 and 1—10, where R4, R5 are selected from hydrogen and fluorine. In particular, both R4 and R5 are hydrogen. 20 In another particular embodiment of the invention, A in formula I, and likewise in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1—8, 1-9 and 1-10, is a bivalent radical A4.
Preference is given to nds of the formula I, and se to compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10, where R4, R5 are selected from hydrogen and e and in particular to those compounds, where both R4 and R5 are 25 hydrogen. In the bivalent radical A4 the moiety A' is ably CR3bR3c, where R3b and R30 are independently of each other selected from the group consisting of hydrogen, fluorine and methyl or together form CHZCHZ and where R3b and R3b are independently of each other in ular selected from the group consisting of hydrogen and fluorine. 46 Particular preference is given to compounds of the formula I, and likewise to nds of formulae 1-1, 1-2, 1—3, 1—4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10, where A is a bivalent radical A4, where the moiety A' is CR3bR3C, where R3b and R30 are as defined herein and where R3b and R3C are in particular, independently of each other, selected from the group ting of hydrogen, fluorine and methyl or together form CH2CH2 and ally where both R3b and R30 are hydrogen or fluorine.
In another particular ment of the ion, A in formula I and likewise in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10 is a bivalent radical A4, where A' is O. 10 In another particular embodiment of the invention, A in formula I, and likewise in formulae 1-1, 1—2, 1—3, 1—4, 1—5, 1—6, 1—7, 1—8, 1—9 and 1—10, is a bivalent radical A4, where A' is NR3a with R3a being as defined above in particular hydrogen or C1-C4-alkyl.
In r particular embodiment, A in formula I, and likewise in formulae 1-1, I- 2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1—10, is a bivalent radical A4, where A' is linear C2- 15 Cg-alkanediyl, where one of the CHz—moieties of C2-C3-alkandiyl may be replaced by oxygen or NR3a, and where l, 2, 3, or 4 of the hydrogen atoms of C2-C3-alkandiyl may be replaced by a l R3d. R3d is eably selected from halogen and C1-C4-alkyl.
In another particular embodiment of the invention, A in formula I, and likewise in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9 and 1-10 is a bivalent radical A5. 20 Preference is given to compounds ofthe formula I, and likewise to compounds of ae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1—7, 1-8, 1-9 and 1-10, where R4, R5 are selected from hydrogen and methyl and in particular to those compounds, where both R4 and R5 are hydrogen. Preference is given to compounds of the formula I, where R36 and R3f are, independently of each other, selected from the group consisting of hydrogen and 25 methyl, and in particular to those compounds, where both R36 and R3f are en.
A particular preferred embodiment of the invention relates to the compounds of formula I-1.A, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 Hetml N l \ R2 (I1.A)_ R7 47 where Het, R1, R2, R5, R6 and R7 are as defined here and in the claims.
Another particular red embodiment of the ion relates to the compounds of formula I-2.A, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 HetDC N I \ R2 (I 2.A)_ R R \ 8 R7 where Het, R1, R2, R5, R6 and R7 are as defined here and in the claims.
Another particular preferred embodiment of the invention relates to the compounds of formula 1—1 .B, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 10 R30 R3b 0 R1 Het N R5 I l \ R2 _ N\ 8 R7 where Het, R1, R2, R3b, R“, R5 and R7 are as defined here and in the claims and where R5 is in particular hydrogen.
Another particular preferred embodiment of the invention relates to the 15 compounds of formula I-2.B, described below, to the es, the prodrugs, the es and the tautomers thereof and to the pharmaceutically suitable salts thereof: R30 3b R O 1 R Het N R5 | \ R2 (I-2.B) \ 8 R7 where Het, R1, R2, R3b, R3c, R5 and R7 are as defined here and in the claims and where R5 is in ular hydrogen. In formulae I—l.B and I-2.B, the variables R3b and 48 R30 are independently of each other in particular selected from the group ting of hydrogen, fluorine and methyl or together form CHZCHZ. Particular preference is given to compounds of the ae 1-1 .B and I-2.B, where both R3b and R30 are hydrogen or fluorine. 5 Another particular preferred embodiment of the invention relates to the nds of formula I-l .C, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: RE>1 0 R1 _ Het N _ I l \ R2 (I1.C) N\ 8 R7 where Het, R1, R2, R5 and R7 are as defined here and in the claims and where R5 is 10 in particular en.
Another particular preferred embodiment of the invention relates to the nds of formula I-2.C, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: Fabio 1 R Het—N I \ R2 (I-2.C) \ 8 R7 15 where Het, R1, R2, R5 and R7 are as defined here and in the claims and where R5 is in particular hydrogen.
A particular preferred embodiment of the invention relates to the compounds of formula I-l .D, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 ! l \ R2 (I-1.D) N \ R3 R3f 8 R7 20 49 where Het, R1, R2, R36, R3f and R7 are as defined here and in the claims and where R3‘3 and R3f are in particular both hydrogen.
Another particular preferred embodiment of the invention s to the compounds of formula I-2.D, described below, to the N-oxides, the prodrugs, the 5 hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 Het N | |\ R2 (I-2.D) \ R3 R3f S R7 where Het, R1, R2, R36, R3f and R7 are as defined here and in the claims and where R36: and R3f are in particular both hydrogen.
Particular embodiments of the invention relate to compounds of the formulae 1-1, 10 1-2, I-l.A, I-2.A, 1-1 .B, I-2.B, I-l.C, I-2.C, I-1.D, and I-2.D described above, to the N- oxides, the prodrugs, the hydrates and the tautomers thereof and to the ceutically suitable salts thereof, where R1 is a l Yl—Cyc1 and R7 is as defined above and in particular selected from the group ting of hydrogen, fluorine, C1-C4-alkyl, C1-C2- fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, ropyl, optionally substituted by l, 15 2 or 3 methyl groups, fluorinated cyclopropyl and z, especially from the group consisting of hydrogen and Yz-Cycz.
Further particular embodiments of the invention relate to nds of the formulae 1- 1, 1-2, 1-1 .A, I-2.A, 1-1 .B, I-2.B, I-1.C, I-2.C, I-l .D, and I-2.D, described above, to the N—oxides, the gs, the hydrates and the tautomers thereof and to the 20 pharmaceutically suitable salts thereof, where R1 is selected from the group consisting of hydrogen, fluorine, C1-C4-a1kyl, C1-C2-fluoroalkyl, C1—C4—alkoxy, C1-C2- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups and fluorinated cyclopropyl and R7 is a moiety YZ-Cycz. In these embodiments R1 is in particular hydrogen. 25 A further particular preferred embodiment of the ion relates to the compounds of formula I—3.A, described below, to the N-oxides, the prodrugs, the es and the tautomers thereof and to the pharmaceutically suitable salts thereof: 50 o R Het R2 mlN (I 3.A)_ R R N\ R8 R7 R9 where Het, R1, R2, R5, R6, R7, R8 and R9 are as defined here and in the claims.
Another particular preferred embodiment of the invention relates to the compounds of formula I-4.A, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts f: 1 o R Het R2 xN (I 4.A)_ R R \ R8 R7 R9 where Het, R1, R2, R5, R6, R7, R8 and R9 are as defined here and in the claims.
Another particular preferred embodiment of the invention s to the compounds of formula I-3.B, described below, to the N—oxides, the prodrugs, the 10 hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 3C R 3b R 1 O R R2 Het N R5 (I _3. B) | N \ R8 R7 R9 where Het, R1, R2, R3b, R3°, R7, R8 and R9 are as defined here and in the claims and where R5 is in particular hydrogen. 15 Another particular preferred embodiment of the invention relates to the nds of formula I—4.B, described below, to the es, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 51 R R3b 1 o R R2 Het N R5 (I-4.B) \ R8 R7 R9 where Het, R1, R2, R3b, R3°, R7, R8 and R9 are as defined here and in the claims and where R5 is in particular hydrogen.
In formulae I-3.B and 1-43, the variables R3b and R“ are independently of each other in particular selected from the group consisting of hydrogen, fluorine and methyl or together form CHZCHZ. Particular preference is given to compounds ofthe formulae I-3.B and I-4.B, where both R3b and R30 are hydrogen or fluorine.
Another particular preferred embodiment ofthe invention relates to the compounds of formula I-3.C, described below, to the N-oxides, the prodrugs, the 10 hydrates and the tautomers thereof and to the ceutically suitable salts thereof: 5 O R N ) where Het, R1, R2, R5, R7, R8 and R9 are as defined here and in the claims and where R5 is in particular hydrogen. r particular preferred embodiment of the invention relates to the 15 nds of formula I-4.C, described below, to the es, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: R5 O R :3: R2 Het N (I-4.C) \ R8 R7 R9 where Het, R1, R2, R5, R7, R8 and R9 are as defined here and in the claims and where R5 is in particular hydrogen. 52 A further particular preferred embodiment of the invention relates to the nds of formula I-3.D, described below, to the N—oxides, the prodrugs, the es and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 Het R2 f:11N (I-3.D) \ 8 R3 R3f R R7 R9 5 where Het, R1, R2, R33, R”, R7, R8 and R9 are as defined here and in the claims and where R36 and R3f are in particular both hydrogen.
Another particular preferred embodiment of the invention relates to the compounds of formula I—4.D, described below, to the N—oxides, the prodrugs, the es and the tautomers thereof and to the ceutically suitable salts thereof: 0 R1 Het R2 R393 \Nl (I-4.D) R8 R7 R9 10 where Het, R1, R2, R36, R“, R7, R8 and R9 are as d here and in the claims and where R3‘3 and R3f are in particular both hydrogen.
Particular embodiments ofthe invention relate to compounds of the formulae 1-3, 1-4, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-3.D, I-4.C, and I-4.D, described above, to the N- 15 oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof, where R1 is a radical Yl—Cycl and R7, R8 and R9 are as defined above and where R7 is in particular selected from the group consisting of hydrogen, fluorine, C1-C4-a1kyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2—fluoroalkoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl 20 and Y2-Cyc2, especially from the group consisting of en and Yz-Cyc2 and where R9 is in particular selected from the group consisting of hydrogen, e, C1-C4-alkyl, C1-C2-fluoroalkyl, C1—C4—alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and Y3-Cyc3, especially from the group consisting of hydrogen and Y3—Cyc3, provided that none or only one of 53 R7 and R9 is a moiety Y3-Cyc3, or Y3—Cyc3, tively. R7 and R9 are in ular hydrogen.
Further particular embodiments of the invention relate to compounds of the formulae 1-3, 1-4, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D and I-4.D, bed above, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the ceutically suitable salts thereof, where R1 is selected from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1—C2-fluoroalkyl, alkoxy, C1-C2- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups and fluorinated cyclopropyl and where R7 and R9 are selected, independently of each other, 10 from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4- alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, ally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and Yz—Cyc2 or Y3—Cyc3 , respectively, provided that either R7 is Yz-Cyc2 or R9 is Y3—Cyc3. In these embodiments R1 is in particular hydrogen. In these embodiments the radical R7 or R9, which is different from Yz-Cyc2 15 or Y3-Cyc3 is in particular hydrogen. , respectively, A further particular preferred embodiment of the invention relates to the compounds of formula 1-5.A, described below, to the N—oxides, the prodrugs, the hydrates and the ers thereof and to the pharmaceutically suitable salts thereof: 0 R Het R2 9QN \ | " 5""_ \ / N R8 R7 20 where Het, R1, R2, R5, R6, R7 and R8 are as defined here and in the claims. r particular preferred embodiment of the invention relates to the compounds of a I-6.A, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the ceutically suitable salts thereof: 54 o R Het R2 ECN \ l (I 6.A)_ R R \ N/ R8 R7 where Het, R1, R2, R5, R6, R7 and R8 are as defined here and in the .
A further particular preferred ment of the invention relates to the compounds of formula I-5.B, described below, to the N-oxides, the prodrugs, the 5 hydrates and the tautomers thereof and to the pharmaceutically le salts thereof: 3b 30 R R 1 o R R2 Het \ R5 I}! (I-5.B) N \ / N R8 R7 where Het, R1, R2, R3b, R30, R5, R7 and R8 are as defined here and in the claims and where R5 is in ular hydrogen.
A further particular preferred embodiment of the invention relates to the 10 compounds of formula I-6.B, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: (I—6.B) where Het, R1, R2, R33, R3b, R5, R7 and R8 are as defined here and in the claims 15 and where R5 is in particular hydrogen. In formulae I-S.B and I-6.B, the variables R3b and R3C are independently of each other in particular selected from the group consisting of en, fluorine and methyl or together form CHZCHZ. Particular preference is given to compounds ofthe formulae I-5.B and I-6.B, where both R3b and R30 are hydrogen or fluorine. 55 A further particular preferred embodiment of the invention relates to the compounds of formula 1-5.C, described below, to the es, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: R5 O R :5: R2 Het I?! \ l (I-5.C) N \ N/ R8 R7 5 where Het, R1, R2, R5, R7 and R8 are as defined here and in the claims and where R5 is in particular hydrogen.
A further particular preferred embodiment of the invention relates to the nds of formula I-6.C, described below, to the N—oxides, the prodrugs, the 10 hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: (I-6.C) where Het, R1, R2, R5, R7 and R8 are as defined here and in the claims and where R5 is in particular hydrogen. 15 A further particular preferred ment of the invention relates to the compounds of formula I-5.D, described below, to the N—oxides, the prodrugs, the es and the tautomers thereof and to the pharmaceutically suitable salts f: 1 o R Het R2 fl |\ ("5-0) N / R3e \ Raf N R8 R7 where Het, R1, R2, R36, R“, R7, R8 and R9 are as d here and in the claims 20 and in particular, R36: and R3f are both hydrogen. 56 r particular red embodiment ofthe invention relates to the compounds of formula I-6.D, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers f and to the pharmaceutically suitable salts f: (I-6.D) where Het, R1, R2, R33, R”, R7, R8 and R9 are as defined here and in the claims and in particular, R36 and R3f are both hydrogen.
Particular embodiments of the invention relate to compounds of the formulae 1-5, 1-6, I-5.A, I-6.A, I-5.B, I—6.B, 1—5.C, I—6.C, I—5.D and I-6.D, described above, to the N- oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically 10 suitable salts thereof, where R1 is a radical Yl—Cyc1 and R7 is as defined above and in particular selected from the group consisting of hydrogen, e, C1-C4-alkyl, C1-C2- fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and YZ-Cycz, especially from the group consisting of hydrogen and Yz-Cycz. 15 Further particular embodiments of the invention relate to compounds of the formulae 1-5, 1-6, 1-5.A, I-6.A, 1-5.B, I—6.B, I-5.C, I-6.C, I-5.D, and I-6.D, bed above, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically le salts thereof, where R1 is ed from the group consisting of hydrogen, fluorine, alkyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2- 20 fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups and fluorinated cyclopropyl and R7 is a moiety Yz-Cycz. In these embodiments R1 is in particular hydrogen.
A particular red embodiment of the invention relates to the compounds of formula I-7.A, described below, to the N-oxides, the prodrugs, the hydrates and the 25 tautomers thereof and to the pharmaceutically suitable salts thereof: 57 o R1 HetXI N I \ R2 (I 7.A)_ R7 where Het, R1, R2, R5, R6 and R7 are as defined here and in the claims.
Another particular red embodiment of the invention relates to the compounds of formula I-8.A, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R1 Hetx N I \ R2 (I 8.A)_ R R \ 0 R7 where Het, R1, R2, R5, R6 and R7 are as defined here and in the claims.
Another particular preferred embodiment ofthe invention relates to the 10 compounds of a I-7.B, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically le salts f: R30 R3b 0 R1 Het N R5 (I 7.3)_ I I \ R2 N\ 0 R7 where Het, R1, R2, R3b, R3°, R5 and R7 are as defined here and in the claims and where R5 is in particular hydrogen. 15 Another particular preferred embodiment of the invention relates to the nds of formula I-8.B, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 58 R30 R3b O R1 Het N R5 I \ R2 (I 8.8)_ \ o R7 where Het, R1, R2, R3b, R3°, R5 and R7 are as defined here and in the claims and where R5 is in particular hydrogen.
In formulae I-7.B and I-8.B, the variables R3b and R“ are independently of each other in particular selected from the group consisting of hydrogen, fluorine and methyl or er form CHZCHZ. Particular preference is given to compounds ofthe formulae I-7.B and I-8.B, where both R3b and R30 are hydrogen or fluorine.
Another particular preferred embodiment of the invention relates to the compounds of formula I-7.C, described below, to the es, the prodrugs, the 10 es and the tautomers thereof and to the pharmaceutically suitable salts thereof: REgj O R1 _ Het I}! l \ R2 (I-7.C) N\ 0 R7 where Het, R1, R2, R5 and R7 are as defined here and in the claims and where R5 is in particular hydrogen. 15 Another particular preferred embodiment of the ion relates to the compounds of a I-8.C, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: R5 O R1 \ 0 R7 where Het, R1, R2, R5 and R7 are as defined here and in the claims and where R5 is 20 in particular hydrogen. 59 A particular red embodiment of the invention relates to the compounds of formula I-7.D, described below, to the N—oxides, the prodrugs, the hydrates and the ers thereof and to the pharmaceutically suitable salts f: 0 R1 Hetfill N l \ R2 (I-7.D) 3e \ R O R3f 7 R 5 where Het, R1, R2, R31: R3e and R7 are as defined here and in the claims where R3e and R3f are both in particular hydrogen.
Another particular preferred embodiment of the invention relates to the compounds of a I—8.D, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 10 0 R1 Het Rae R3f \ O R7 where Het, R1, R2, R3f, R36 and R7 are as defined here and in the claims where R3‘3 and R3f are both in ular hydrogen.
Particular embodiments ofthe invention relate to compounds of the formulae 1-7, 15 1-8, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, and I-8.D described above, to the N- oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof, where R1 is a radical Yl—Cycl and R7 is as defined above and in particular selected from the group ting of hydrogen, fluorine, C1—C4-alkyl, C1-C2- fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, ropyl, optionally substituted by 1, 20 2 or 3 methyl groups, fluorinated cyclopropyl and YZ-Cycz, ally from the group consisting of hydrogen and YZ-Cycz.
Further particular embodiments of the invention relate to nds of the formulae 1-7, 1-8, I-7.A, I—8.A, I—7.B, I—8.B, I—7.C, I—8.C, I-7.D and I-8.D, described above, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the 25 pharmaceutically suitable salts thereof, where R1 is selected from the group consisting 60 of hydrogen, fluorine, C1-C4-alkyl, C1—C2-fluoroalkyl, C1-C4-alkoxy, C1-C2- fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups and fluorinated cyclopropyl and R7 is a moiety YZ—Cycz. In these embodiments R1 is in particular hydrogen. 5 A further particular preferred embodiment ofthe invention relates to the compounds of formula I-9.A, described below, to the N-oxides, the gs, the es and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R Hetml R N \ | (I 9.A)_ R R N\ /N 7 9 R R where Het, R1, R2, R5, R6, R7 and R9 are as defined here and in the claims. 10 r particular preferred ment of the invention relates to the compounds of formula I-lO.A, described below, to the es, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 1 o R Het R2 ECN \ l (I 10.A)_ R7 R9 where Het, R1, R2, R5, R6, R7, and R9 are as defined here and in the claims. 15 Another particular preferred embodiment of the invention relates to the compounds of formula I-9.B, described below, to the es, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts f: 30 R 3b R 1 o R 2 R Het N \ 5 R | | (I-9.B) N \ / N R7 R9 61 where Het, R1, R2, R3b, R“, R7, and R9 are as defined here and in the claims and where R5 is in particular hydrogen. r particular preferred embodiment of the invention relates to the 5 compounds of formula I-lO.B, described below, to the N—oxides, the prodrugs, the hydrates and the ers thereof and to the pharmaceutically le salts thereof: (I-10.B) where Het, R1, R2, R3b, R3c, R7, and R9 are as defined here and in the claims and where R5 is in particular hydrogen. 10 In formulae I-9.B and I-lOB, the variables R3b and R30 are independently of each other in particular selected from the group consisting of hydrogen, fluorine and methyl or together form CHZCHZ. Particular preference is given to compounds ofthe formulae I-3.B and I-4.B, where both R3b and R30 are hydrogen or fluorine.
Another particular preferred embodiment of the invention relates to the 15 compounds of formula I-9.C, described below, to the N—oxides, the prodrugs, the es and the tautomers f and to the pharmaceutically suitable salts thereof: R5 O R :3: R2 Het [TI \ | ) N \ / N R7 R9 where Het, R1, R2, R5, R7, and R9 are as defined here and in the claims and where R5 is in particular hydrogen. 20 Another particular preferred ment of the invention relates to the compounds of formula I—lO.C, described below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 62 (I-10.C) where Het, R1, R2, R5, R7, and R9 are as defined here and in the claims and where R5 is in particular hydrogen.
A r particular preferred embodiment of the invention relates to the 5 compounds of formula I-9.D, bed below, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof: 0 R 2 Het R \ I [Tl I (I-9.D) N\ /N R3 R3f R7 R9 where Het, R1, R2, Rh, R3f, R7, and R9 are as defined here and in the claims and where R3‘3 and R3f are in particular both hydrogen. 10 Another particular preferred embodiment of the invention relates to the compounds of formula I-lO.D, described below, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically le salts thereof: (I-10.D) where Het, R1, R2, R36, R3f, R7, and R9 are as defined here and in the claims and 15 where R36 and R3f are in particular both hydrogen.
Particular embodiments of the invention relate to compounds of the formulae 1-9, 1-10, I-9.A, I—lO.A, I—9.B, , I—9.C, 1—10.C, I—9.D and I-lOD, described above, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof, where R1 is a radical Yl-Cyc1 and R7, and R9 are 20 as defined above and where R7 is in particular selected from the group consisting of 63 en, fluorine, C1-C4-alkyl, C1—C2—fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and YZ-Cycz, ally from the group consisting of hydrogen and Yz-Cyc2 and Where R9 is in particular selected from the group consisting of hydrogen, e, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, ropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and Y3-Cyc3, especially from the group consisting of hydrogen and Y3-Cyc3, provided that none or only one of R7 and R9 is a moiety Y3-Cyc3, or Y3-Cyc3, respectively. R7 and R9 are in ular hydrogen. 10 Further particular embodiments of the invention relate to compounds of the formulae 1-9, 1-10, I-9.A, 1—10.A, I-9.B, 1-10.B, I-9.C, 1-10.C, I-9.D and I-lO.D, described above, to the N—oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically le salts thereof, Where R1 is selected from the group consisting of hydrogen, fluorine, C1—C4—alkyl, C1—C2-fluoroalkyl, C1-C4-alkoxy, C1-C2- 15 lkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups and ated cyclopropyl and where R7 and R9 are ed, independently of each other, from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4- alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, fluorinated cyclopropyl and YZ-Cyc2 or Y3-Cyc3 , respectively, provided that 20 either R7 is YZ-Cyc2 or R9 is Y3-Cyc3. In these embodiments R1 is in particular hydrogen. In these embodiments the radical R7 or R9, which is different from 2 or Y3-Cyc3 is in particular en. , respectively, In formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I-l.B, I-2.A, I-2.B, I-l.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I- 25 6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I—7.A, I-8.A, I—7.B, I—8.B, I-7.C, I-8.C, I- 7.D, I-8.D, I-9.A, , I-9.B, I-lO.B, I-9.C, I-10.C, I-9.D and I—10.D, R2 is preferably selected from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2- fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, and fluorinated cyclopropyl. R2 is in particular hydrogen. 30 Preference is given to compounds of the formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, I- 8, 1-9, 1-10, I-1.A, I-l.B, I-2.A, I-2.B, I-1.C, I-2.C, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I- 4.C, I-5.A, I-6.A, I-5.B, I-6.B, I—S.C, I—6.C, I—7.A, I—8.A, I-7.B, I-8.B, I-7.C, I-8.C, I- 64 9.A, I-lO.A, I-9.B, I-lO.B, I-9.C and I—10.C, where R3b, R“, R5 and R6, ifpresent, are, independently of each other, selected from the group consisting of hydrogen, fluorine and methyl, and in particular to those compounds, where both R5 and R6 are hydrogen.
In ae 1-3, 1-4, 1-5, 1-6, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I—6.C, I—5.D, I-6.D, R8 is in ular hydrogen.
With regard to formulae I—l 1-1 .A, 1-1 .B, I- , 1-2, 1-3, 1—5, 1—4, 1-6, 1—7, 1-8, 1—9, 1-10; 2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I-3.A, I-4.A, I—3.B, I-4.B, I—3.C, I—4.C, I-3.D, I- 4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I- 7.C, I-8.C, I-7.D, I-8.D, I-9.A, 1-10.A, I-9.B, I-lO.B, I-9.C, I-lO.C, I-9.D and I-lO.D, the 10 variable Het is as defined above and ably selected from the group consisting of C- bound 6-membered monocyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members, and C-bound, fused bicyclic l, which has 1 or 2 nitrogen atoms as ring members and optionally a further heteroatom selected from O, S and N as ring member, where monocyclic hetaryl and bicyclic hetaryl may be unsubstituted or may carry 1, 2, 3 15 or 4 substituents RX, in ular 0, l or 2 substituents RX. In this regard, RX is preferably selected from halogen, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2- fluoralkoxy, phenyl, C3-C6-cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated C3-C6-cycloalkyl. In this regard, RX is in ular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, 20 ethoxy, difluoromethoxy, trifluoromethoxy, phenyl, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
In formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I- 6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I- 25 7.D, I-8.D, I-9.A, I-lO.B, I-9.A, ,I-9.C,I-10.C,I-9.D and I-lO.D, Het is in particular selected from the group consisting of fused bicyclic hetaryl, which has 1 or 2 en atoms as ring s and optionally a fiarther heteroatom selected from O, S and N as ring member and which may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, 1 or 2 substituents Rx. In this , Rx is preferably 30 selected from halogen, C1—C4—alkyl, C1-C2-fluoroalkyl, alkoxy, C1-C2- fluoralkoxy, C3-C6-cycloalkyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated C3-C6-cycloalkyl. In this regard, Rx is in particular selected from fluorine, 65 chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, optionally tuted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
In another particular embodiments of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, I- 9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I- 3.C, I-4.C, I-3.D, I-4.D, I-5.A, I—6.A, I-5.B, I-6.B, I-5.C, I—6.C, I—5.D, I-6.D, I-7.A, 1- SA, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I—9.A, 1-10.A, I—9.B, I—lO.B, I—9.C, I-10.C, I- 9.D and 1-10.D Het is selected from 6-membered monocyclic hetaryl, which may be unsubstituted or may carry 1, 2, 3 or 4 substituents Rx, in particular 0, 1 or 2 substituents 10 RX. In this regard, Rx is preferably selected from n, C1-C4-alkyl, C1-C2- fluoroalkyl, C1—C4—alkoxy, C1—C2—fluoralkoxy, phenyl, cycloalkyl, ally substituted by l, 2 or 3 methyl groups, and fluorinated C3-C6-cycloalkyl. In this regard, Rx is in particular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, romethyl, methoxy, fluoromethoxy, difluoromethoxy, 15 trifluoromethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl or one Rx may also be phenyl.
Particular preference is given to those compounds of 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I-1.C, I-2.C, I-l .D, I-2.D, I-3.A, I-4.A, I-3.B, I- 4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-S.A, I—6.A, I-5.B, I-6.B, I-S.C, I-6.C, I-5.D, I-6.D, I- 20 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I—7.D, I-8.D, I-9.A, I-lO.A, I-9.B, I-lO.B, I-9.C, I- lO.C, I-9.D and I-lO.D, to the N—oxides, the gs, the hydrates and the tautomers thereof and to the ceutically suitable salts thereof, where the Het radical has at least one imino-nitrogen as ring member, which is located in the position adjacent to the carbon atom which is bound to the group CR5. Amongst these, particular preference is 25 given to those, where the Het l has at least one imino-nitrogen as ring member, which is located in the position adjacent to carbon atom bound to the group CR5 and which is ed from the group consisting of C-bound 6-membered monocyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members, and C-bound, fused bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and ally a fiarther 30 heteroatom selected from O, S and N as ring member, where monocyclic l and ic hetaryl may be unsubstituted or may carry 1, 2, 3 or 4 substituents RX, in particular 0, l or 2 substituents Rx. In this regard, RX is preferably selected from 66 n, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4-a1koxy, C1-C2-fluoralkoxy, phenyl, C3- C6-cycloalkyl, ally substituted by 1, 2 or 3 methyl groups, and ated C3-C6- cycloalkyl. In this regard, Rx is in particular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, phenyl, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
Particular examples of Het in ae 1-], 1-2, 1—3, 1—4, 1-5, 1-6, 1—7, 1-8, 1-9, 1-10, I-1.A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I- 4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I- 10 73, I—8.B, I—7.C, I—8.C, I—7.D, I—8.D, I—9.A, 1—10.A, I—9.B, 1-10.B, I-9.C, I-lO.C, I-9.D and I-lO.D are ed from the group consisting of 2-benzofuryl, 2-pyridyl, 2- pyrimidinyl, 4-pyrimidinyl, 2—pyrazinyl, 3—pyridazinyl, 2-quinolinyl, 3-isoquinolinyl, 2- quinazolinyl, 2-quinoxalinyl, 1,5—naphthyridin—2—yl, 1,8-naphthyridinyl, benzothiazol—l-yl, azol—l-yl, benzimidazol—2—yl, l-methylbenzimidazolyl, 15 imidazo[l ,2-a]pyridineyl, thieno[3,2—b]pyridine—5-yl, imidazo-[2, l -b]-thiazolyl and l,2,4-triazolo[l,5-a]pyridineyl, where the aforementioned radicals are unsubstituted or may carry 1, 2 or 3 ls Rx as defined above, which are in particular ed from the group consisting of fluorine, chlorine, methyl, fluoromethyl, omethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, 20 trifluoromethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
In a particular embodiment ofthe compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, I- 6, 1-7,I-8,I-9,I-10,I-l.A,I-l.B,I-2.A,I-2.B,I-1.C,I—2.C,I-l.D,I-2.D,I-3.A,I-4.A,I- 3.B, I-4.B, I-3.C, I-4.C, I-3.D, I—4.D, I—5.A, I—6.A, I—5.B, I-6.B, I-5.C, I-6.C, I-5.D, I- 25 6.D, I-7.A, I-8.A, I-7.B, I-8.B, I—7.C, I—8.C, I-7.D, I-8.D, I-9.A, I—lO.A, I-9.B, I-lO.B, I- 9.C, I-lO.C, I-9.D and I-lO.D Het has at least one imino—nitrogen as ring member, which is located in the position adjacent to carbon atom bound to the group CR5 and Het is selected from the group consisting of fused bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and optionally a further heteroatom selected from O, S and N as 30 ring member, where bicyclic hetaryl may be unsubstituted or may carry 1, 2, 3 or 4 tuents RX, in ular 0, l or 2 tuents Rx. In this regard, RX is preferably selected from halogen, C1-C4—alkyl, C1—C2—fluoroalkyl, C1-C4-alkoxy, C1-C2- 67 fluoralkoxy, C3-C6-cycloalkyl, optionally tuted by l, 2 or 3 methyl groups, and fluorinated C3-C6-cycloalkyl. In this regard, Rx is in particular selected from fluorine, chlorine, methyl, fluoromethyl, difluoromethyl, romethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, ally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl. ular examples of Het of this embodiment are olinyl, 3—isoquinolinyl, 2-quinazolinyl, 2-quinoxalinyl, 1,5- naphthyridin—2-yl, 1,8-naphthyridinyl, benzothiazol— l -y1, benzoxazo 1y1, benzimidazolyl, 1-methylbenzimidazol—2-yl, imidazo[l ,2-a]pyridineyl, thieno[3 ,2- b]pyridineyl, o-[2, l -b]-thiazolyl and 1,2,4-triazolo [l ,5-a]pyridineyl, 10 Where the aforementioned radicals are unsubstituted or may carry 1, 2 or 3 radicals Rx as defined above, Which are in particular selected from the group consisting of fluorine, chlorine, methyl, fluoromethyl, omethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl. 15 ular preference is given to compounds, Where Het in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I-l.B, I—2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I- 3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I—4.D, I-5.A, I-6.A, I-SB, I-6.B, 1-5.C, I- 6.C, I-SD, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I- 9.B, I-lO.B, I-9.C, , I-9.D and I—lO.D is selected from the group consisting of 2- 20 quinolinyl, 1,8-naphthyridinyl, benzothiazol—l-yl, benzoxazol—l-yl, benzimidazol—2- yl, l-methylbenzimidazolyl, imidazo[l,2—a]pyridineyl, thieno[3,2-b]pyridineyl, and in particular 2-quinolinyl or o[l ,2-a]pyridineyl, where these radicals are unsubstituted or may carry 1, 2 or 3 radicals Rx as defined above, which are in particular selected from the group ting of fluorine, chlorine, methyl, fluoromethyl, 25 difluoromethyl, trifluoromethyl, methoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, optionally substituted by l, 2 or 3 methyl groups, and fluorinated cyclopropyl.
With regard to formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 1-1 .A, I-1.B, I- 2.A, I—2.B, I—l.C, I—2.C, I—1.D, I—2.D, I—3.A, I—4.A, I—3.B, I-4.B, I-3.C, I-4.C, I-3.D, I- 30 4.D, I-5.A, I-6.A, 1—5.B, I—6.B, I—S.C, I—6.C, I—5.D, I—6.D, I-7.A, I-8.A, I-7.B, I-8.B, I- 7.C, I-8.C, I-7.D, I-8.D, I—9.A, I—lO.A, I—9.B, 1—10.B, I-9.C, 1-10.C, I-9.D and 1-10.D, the 68 variables Y1, Y2, Cyc1 and Cyc2 are as defined above and in ular have the red meanings.
In particular, Cycl, ent in formulae I-l, I-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, I- 10, 1-1 .A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I—5.B, I—6.B, I-5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I- 7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I—10.A, I—9.B, I-10.B, I-9.C, I-10.C, I-9.D and I-10 D is selected from the groups of (i) saturated 4-, 5-, 6- or 7-membered heteromonocycles, where the heteromonocycle has one en or oxygen atom as ring member and may have one 10 further heteroatom or heteroatom group as ring , which is selected from the group consisting of O, S, S(=O), S(=O); and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein; and (ii) phenyl or a 5- or 6 membered hetaryl, selected from pyridyl, pyrimidinyl, 15 furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'—RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if t, is preferably a chemical bond or O. 20 In particular embodiments ofthe compounds of formulae 1-1, I-2, L3, L4, 1-5, 1-6, 1-7, 1-8, 1-9, I-10, I-1.A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I- 3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-SD, I- 6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I—8.C, I-7.D, I-8.D, I-9.A, , I-9.B, I-10.B, I- 9.C, I-10.C, I-9.D and I-10.D, Cyc], ifpresent, is selected from the group consisting of 25 saturated 4-, 5-, 6- or 7-membered heteromonocycles or a saturated 7-, 8-, 9- or 10- membered heterobicycle, where the heteromonocycle and the heterobicycle have one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the ted heteromonocycle and the saturated 30 heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where 69 RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In special embodiments ofthe compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, I- 7, 1-8,I-9,I-10,I-1.A,I-1.B,I-2.A,I-2.B,I-1.C,I-2.C,I-1.D,I-2.D,I-3.A,I-4.A,I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I- 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I—8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I—9.B, I-10.B, I-9.C, I- lO.C, I-9.D and I-10.D, Cycl, ifpresent, is selected from the group consisting of saturated 4-, 5-, 6- or 7-membered heteromonocycles, where the heteromonocycle has one nitrogen or oxygen atom as ring member and may have one further heteroatom or 10 heteroatom group as ring , which is ed from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are tituted or carry 1, 2, or 3 ls RC1, where RC1 is as defined herein.
In these particular and special embodiments of the compounds of formulae 1-1, I- 15 2, 1—3, 1—4, 1—5, 1-6, 1—7, 1-8, 1—9, 1—10, I-1.A, 1—1.3, I—2.A, 1—2.3, I-1.C, I-2.C, I-1.D, 1— 2D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I—3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I- 5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I-7.B, I—8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I- 10.A, I-9.B, I-lOB, I-9.C, , I-9.D and I—10.D, Y1, ifpresent, is preferably selected from O, NH and a chemical bond, with particular preference given to Y1 being a 20 chemical bond.
In these particular and special embodiments of the compounds of formulae 1-1, I- 2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, I—1.A, I—l.B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I- 2D, I-3.A, I-4.A, 1—3.3, 1—4.3, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, 1—5.3, I-6.B, 1— 5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I—7.B, I—8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I- 25 10.A, I-9.B, I-lO.B, I-9.C, I-lO.C, I—9.D and I—lO.D, Yl-Cycl. ifpresent, is e.g. selected from the group consisting of l-piperidinyl, 4,4-difluoropiperidiny1, 4-piperidinyl, 1- methylpiperidinyl, 1-piperazinyl, 4-methylpiperaziny1, morpholiny1, 6- azaspiro-[3,4]octyl, 2,5-diazabicyclo[2.2.1]heptanyl, 3,8-diazabicyclo[3.2. l]octan yl, thiomorpholinyl, 1—oxothiomorpholinyl, tanyl)amino, 1,1- 30 dioxothiomorpholin—4—yl and oxetan—3—ylamino and especially from the group consisting of l-piperidinyl, 4,4—difluoro—1—piperidinyl, 4-piperidinyl, l-methyl piperidinyl, l-piperazinyl, 4-methyl—1—piperazinyl, lin—4-yl, thiomorpholinyl, 70 l-oxothiomorpholin—4-yl, N—(oxetan—3—y1)amino, 1, l -dioxothiomorpholinyl and oxetan—3-ylamino.
In other particular embodiments of the compounds of formulae 1-1, 1-2, 1-3, 1-4, I- 5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I-l.B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I-3.A, I- 4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I—5.A, I-6.A, I—5.B, I-6.B, I-5.C, I-6.C, I- 5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I—8.D, I-9.A, 1-10.A, I-9.B, I- 10.B, I-9.C, I-lO.C, I-9.D and 1—10.D, Cycl, ifpresent, is phenyl or a 5- or 6 ed heteroaromatic radical, which has one heteroatom, selected from O, S and N as ring member and optionally one or two further heteroatoms as ring members, and which is in 10 ular selected from the group consisting of pyridyl, pyrimidinyl, fiaryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered heteroaromatic radical are unsubstituted or either carry, independently of each other, carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as 15 defined herein and where Y', if t, is preferably a chemical bond or O.
In other l embodiments ofthe compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I-l.B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, 1-5.C, I-6.C, I-SD, I- 6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I—8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I-9.B, I-lO.B, I- 20 9.C, I-lO.C, I-9.D and , Cyc], ifpresent, is selected from the group consisting of phenyl or a 5- or 6 membered hetaryl, selected from pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, olyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 ed hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RCl or one radical Y'-RC2 and 0, 1, 2, 3 or 4, in particular 0, l or 2 radicals RC1, 25 where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or 0.
More ularly, Cycl, if present in formulae 1-], 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, I- 9, 1-10, I-l.A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I- 3.C, I—4.C, I—3.D, I—4.D, I—5.A, I—6.A, I—5.B, I—6.B, I—5.C, I-6.C, I-5.D, I-6.D, I-7.A, I- 30 8.A, I-7.B, I-8.B, I—7.C, I—8.C, I—7.D, I—8.D, I—9.A, I—lO.A, I-9.B, 1-10.B, I-9.C, 1-10.C, I- 9.D and I-lO.D, is selected from the group consisting of phenyl and 5- or 6-membered hetaryl ed from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, 71 pyrrolyl, olyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC] which are selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, romethyl, methoxy and NH2, or, if Cyc1 is phenyl, two radicals RCl which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic cyclic radical, which is selected from 5- or 6-indolyl, 5- or 6-benzimidazolyl, 5- or 6-benzopyrazolyl, 5- or 6-benzotriazolyl, 5- or 6-benzofuranyl, 2,3— dihydrobenzofuran—S-yl, 2,3-dihydrobenzofuranyl, l,3-dihydroindolonyl, 1,3- oindolonyl, 5- or 6-quinolinyl, 5- or 6-isoquinolinyl, 5- or 6-quinazolinyl, 10 2-amino—5—quinazolinyl, and 2-aminoquinazolinyl. Amongst these, particular preference is given to compounds, where Y1 is a chemical bond. Amongst these, particular preference is given to compounds, where Cyc1 in formulae 1-1, 1-2, 1-3, 1-4, I- 5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I—l.B, I—2.A, I—2.B, I—l.C, I-2.C, I-l.D, I-2.D, I-3.A, I- 4.A, I-3.B, I-4.B, I-3.C, I-4.C, l-3.D, I-4.D, I—5.A, l—6.A, I-5.B, I-6.B, I-5.C, I-6.C, I- 15 5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I—7.C, I—8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I-9.B, I- lO.B, I-9.C, I-lO.C, I-9.D and , is selected from the group consisting of phenyl and 5- or 6-membered hetaryl selected from the group ting of pyridyl, pyrimidinyl, furyl, thienyl, yl, imidazolyl, pyrazolyl, yl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are 20 ed from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2.
In particular, Cyc2 and Cyc3, if present in formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, I- 8, 1-9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I-1.C, I-2.C, I-l .D, I-2.D, I-3.A, I-4.A, I-3.B, l- 4.B, l-3.C, l-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, 1-5.C, I-6.C, 1-5.D, I-6.D, I- 25 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I—7.D, I-8.D, l-9.A, I—lO.A, I-9.B, I-lO.B, I-9.C, I- lO.C, I-9.D and I-lO.D, are, independently from each other, selected from the groups of (i) saturated 4-, 5-, 6- or 7-membered heteromonocycles, where the heteromonocycle has one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the 30 group consisting of O, S, S(=O), S(=O); and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein; and 72 (ii) phenyl or a 5- or 6 membered hetaryl, selected from pyridyl, pyrimidinyl, fiiryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5- or 6 membered hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RCl or one radical Y'—RCZ and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RCZ and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In particular embodiments of the compounds of formulae I-l, I—2, I-3, I-4, I-5, I-7, 1-8, I-9,I1.A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I- 43, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I- 10 7.A, I—8.A, I—7.B, I—8.B, I—7.C, I—8.C, I—7.D, I—8.D, I—9.A, I-10.A, I-9.B, I-10.B, I-9.C, I- NC, I-9.D and I—10.D, Y2—Cy02 and Y3—Cyc3, if present, are, ndently from each other, selected from the group consisting of saturated 4-, 5-, 6- or ered heteromonocycles or a saturated 7—, 8—, 9— or 10—membered heterobicycle, where the heteromonocycle and the heterobicycle have one nitrogen or oxygen atom as ring 15 member and may have one further atom or heteroatom group as ring member, which is ed from the group ting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated bicycle are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 radicals RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and 20 where Y', if present, is preferably a chemical bond or O.
In special embodiments ofthe compounds of formulae I-l, I-2, L3, L4, 1-5, 1-6, I- 7, 1-8, 1-9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I-1.C, I-2.C, I-l .D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, 1-5.C, I-6.C, 1-5.D, I-6.D, I- 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I-9.B, , I-9.C, I- 25 lO.C, I-9.D and I-lO.D, YZ-Cyc2 and Y3-Cyc3, if present, are, independently from each other, selected from the group consisting of saturated 4-, 5-, 6- or 7-membered heteromonocycles, where the heteromonocycle has one en or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is ed from the group consisting of O, S, S(=O), S(=O)2 and N, where the 30 saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, or 3 radicals RC1, where RC1 is as defined herein. 73 In this particular and special embodiments of the compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, I—1.A, I-1.B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I- 3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-S.C, I- 6.C, I-5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I- 9.B, I-lO.B, I-9.C, I-lO.C, I-9.D and I-lO.D, Yz-Cyc2 and Y3-Cyc3, ifpresent, are, independently from each other preferably selected from O, NH and a chemical bond, with ular preference given to Y2 and Y3 being a chemical bond.
In this particular and special embodiments of the compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10,I-1.A,I-1.B,I-2.A,I-2.B,I-1.C,I-2.C,I-1.D,I-2.D,I- 10 3.A, I-4.A, I-3.B, I-4.B, I-3.C, I—4.C, I—3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I- 6.C, I-5.D, I-6.D, I—7.A, I—8.A, I—7.B, I—8.B, I—7.C, I—8.C, I-7.D, I-8.D, I-9.A, 1-10.A, I- 9.B, I-lO.B, I-9.C, I-lO.C, I—9.D and I—10.D, YZ—Cyc2 and Y3-Cyc3, ifpresent, are, independently from each other, e.g. selected from the group consisting of l-piperidinyl, 4,4-difluoro-l-piperidinyl, 4-piperidinyl, 1—methyl—4-piperidinyl, l-piperazinyl, 4- 15 methyl-l-piperazinyl, morpholinyl, 2—oxa—6—azaspiro-[3,4]octyl, 2,5- diazabicyclo[2.2. l]heptan—2-yl, 3,8-diazabicyclo[3 .2. l]octan—8-yl, thiomorpholinyl, l-oxothiomorpholin—4-yl, tan—3-yl)amino, l, l thiomorpholinyl and oxetan—3-ylamino and especially from the group consisting of l-piperidinyl, 4,4- difluoro- l -piperidinyl, 4-piperidinyl, l —methylpiperidinyl, l -piperazinyl, 4-methyl- l - 20 piperazinyl, morpholinyl, thiomorpholin—4-yl, 1-oxothiomorpholin—4-yl, N—(oxetan— mino, oxothiomorpholin—4-yl and oxetan—3-ylamino.
In other particular embodiments of the compounds of formulae 1-1, 1-2, 1-3, 1-4, I- 5, 1-6, 1-7, 1-8, 1-9, 1-10, I-l.A, I-1.B, I-2.A, I-2.B, I-l.C, I-2.C, I-l.D, I-2.D, I-3.A, I- 4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I—5.A, I-6.A, I—5.B, I-6.B, I-5.C, I-6.C, I- 25 5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I—8.D, I-9.A, I-lO.A, I-9.B, I- lO.B, I-9.C, I-lO.C, I-9.D and , Yz-Cyc2 and Y3-Cyc3, ifpresent, are, independently of each other, phenyl or a 5- or 6 membered aromatic radical, which has one heteroatom, selected from O, S and N as ring member and optionally one or two further atoms as ring members, and which is in particular selected from the 30 group consisting of pyridyl, pyrimidinyl, furyl, l, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5— or 6 ed heteroaromatic radical are unsubstituted or either carry, independently of each other, carry 1, 2, 3, 4 or 5, in 74 particular 1, 2, or 3 radicals RCl or one l Y'—RC2 and 0, l, 2, 3 or 4, in particular 0, l or 2 radicals RC1, where RC1, RC2 and Y' are as defined herein and where Y', if present, is preferably a chemical bond or O.
In special ments ofthe compounds of formulae 1-1, 1-2, 1-3, 1-4, 1-5, 1-6, I- 7, 1-8, 1-9, 1-10, I-l.A, I-l.B, I-2.A, I-2.B, I-1.C, I-2.C, I-l.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I—6.C, I-5.D, I-6.D, I- 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I—8.C, I-7.D, I-8.D, I-9.A, 1—10.A, I—9.B, , I-9.C, I- 10.C, I-9.D and I-10.D, 2 and Y3-Cyc3, if present, are, independently from each other, selected from the group consisting ofphenyl or a 5- or 6 membered hetaryl, 10 selected from pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and the 5— or 6 membered hetaryl are unsubstituted or carry 1, 2, 3, 4 or 5, in particular 1, 2, or 3 ls RC1 or one radical Y'-RC2 and 0, l, 2, 3 or 4, in particular 0, 1 or 2 radicals RC1, where RC1, RC2 and Y’ are as defined herein and where Y', if present, is preferably a chemical bond or O. 15 In ular, Yz-Cyc2 and Y3-Cyc3, ifpresent in formulae 1-1, 1-2, 1-3, 1-4, 1-5, I- 6, 1-7, 1-8, 1-9, 1-10, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I—1.C, I-2.C, I-l .D, I-2.D, I-3.A, I-4.A, I- 3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I—6.A, I-5.B, I-6.B, 1-5.C, I-6.C, I-SD, I- 6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I-7.D, I-8.D, I-9.A, I-lO.A, I-9.B, I-lO.B, I- 9.C, I-lO.C, I-9.D and I-lO.D, YZ-Cyc2 and Y3-Cyc3, are, ndently from each 20 other, selected from the group consisting ofphenyl and 5- or 6-membered hetaryl selected from the group consisting of pyridyl, pyrimidinyl, fiaryl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC] which are selected from the group ting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy 25 and NH2, or, if one or both of Yz—Cyc2 and Y3-Cyc3 are phenyl, two radicals RCl which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic heterocyclic l, which is selected from 5- or 6-indoly1, 5- or 6-benzimidazolyl, 5- or 6-benzopyrazolyl, 5- or 6-benzotriazolyl, 5- or 6-benzofi1rany1, 2,3-dihydrobenzofuran—5—yl, hydrobenzofi1ran—6-yl, 1,3-dihydroindol—2-on—5-yl, 30 l,3-dihydroindol—2—on—6—yl, 5— or 6—quinolinyl, 5— or 6-isoquinolinyl, 5- or 6- quinazolinyl, 2-amino-5—quinazolinyl, and 2—amino—6-quinazolinyl. Amongst these, particular preference is given to nds, where YZ-Cyc2 and Y3-Cyc3 are, 75 independently from each other, selected from the group consisting of phenyl and 5- or 6-membered hetaryl selected from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, lyl, oxazolyl and thiazolyl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RCl which are selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2.
Particular embodiment of the invention relates to the compounds of formula I, to the N-oxides, the prodrugs, the hydrates and the tautomers thereof and to the pharmaceutically suitable salts thereof, where the compounds of the formula I are 10 selected from the group ting of: 3,7-di(pyridin—4—yl)—5—[2—(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 7-(pyridin—4-yl)-5—[2—(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one; 3-(pyridinyl)[2-(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one; 3-(pyridinyl)[2-(pyridin—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 15 (pyridinyl)[2-(pyridin—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; quinolinyl)ethyl][4-(trifluoromethyl)phenyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(4-methylphenyl)[2-(quino1inyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-[4-(propan—2-yl)phenyl][2—(quinolinyl)ethyl]thieno[2,3-d]pyridazin— 20 4(5H)-one; 3-(4-ethylphenyl)[2-(quino1iny1)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 4- {4-oxo[2-(quinolin-2—yl)ethy1]—4,5—dihydrothieno[2,3-d]pyridazin—3- yl}benzonitrile; 3-(4-methoxyphenyl)[2—(quino1inyl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one; 25 3-(4-fluorophenyl)[2-(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(4-ethoxyphenyl)[2-(quinolin—2-yl)ethy1]thieno[2,3—d]pyridazin-4(5H)-one; 3-[4-(dimethylamino)phenyl][2-(quinolin—2-y1)ethy1]thieno[2,3-d]pyridazin- one; (4— {4—oxo—5- ino lin—2-yl)ethyl]-4,5-dihydrothieno [2,3 -d]pyridazin-3 - 30 yl}phenyl)acetonitrile; 3-(4-hydroxyphenyl)—5—[2—(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one; 3-(2-chlorophenyl)[2—(quino1in—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 76 3 -(2-rnethylphenyl)-5 -[2-(quin01in—2-y1)ethy1]thieno[2,3 -d]pyridazin-4(5H)-one; 3 -(2-ethy1phenyl)-5 -[2-(quin01iny1)ethy1]thieno[2,3 -d]pyridazin-4(5H)-one; 3 -(2-flu0ropheny1)-5 - [2-(quin01in—2-y1)ethyl]thieno [2,3 -d]pyridazin-4(5H)-one; 3-(2-methoxypheny1)-5 - [2—(quin01in—2-yl)ethy1]thieno [2,3 -d]pyridazin—4(5H)-one; 3 hoxypheny1)-5 - [2-(quin01in—2-yl)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 3 -(2-hydroxyphenyl)-5 -[2-(quin0lin—2-yl)ethy1]thien0[2,3 —d]pyridazin—4(5H)-one; 5 - [2-(quin0 liny1)ethyl]—3—[2-(triflu0r0methy1)phenyl]thieno ]pyridazin- 4(5H)-one; 3 - [3 oxymethyl)phenyl][2-(quino lin—2-y1)ethy1]thieno ]pyridazin- 10 4(5H)—one; 3 -(3 -methoxyphenyl)—5 — [2—(quin0 lin—2—yl)ethy1]thien0 [2,3 -d]pyridazin—4(5H)-one; 3 -(3 -ethoxypheny1)—5 — [2—(quino1in—2—yl)ethyl]thien0 [2,3 -d]pyridazin-4(5H)-one; 3 - [3 -(dimethylamino)pheny1]—5—[2—(quin0lin—2—y1)ethy1]thien0[2,3-d]pyridazin- 4(5H)-one; 15 3 - [4-OX0(2-quino 1iny1—ethy1)—4,5—dihydr0-thien0 [2,3 idazin—3 -y1} - benzonitrile; 3 -(3 -fluor0phenyl)-5 - [2-(quino1inyl)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 3 -(3 -hydroxyphenyl)-5 - [2-(quino1iny1)ethy1]thieno [2,3 -d]pyridazin—4(5H)-one; N,N—dimethy1—3 - {4-0X0 [2-(quino1iny1)ethy1] -4,5 -dihydr0thieno [2,3 - 20 d]pyridazin—3-y1}benzamide; 3 -(3 -rnethylphenyl)-5 - [2-(quin01in—2-yl)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 5 - ino 1iny1)ethy1] -3—(thi0phen—2-yl)thieno [2,3 -d]pyridazin—4(5H)-one; 3 -(1-methy1—1H-indol—5 -y1)(2-quin01iny1-ethy1)-5H-thieno [2,3 -d]pyridazin- 4-one; 25 3 -(1H-ind01—6-y1)-5 - [2-(quin01in—2-yl)ethyl]thieno [2,3 —d]pyridazin-4(5H)-one; 3 -(pyrimidin—5 -y1)-5 - [2-(quin0 lin—2—yl)ethyl]thien0 [2,3 -d]pyridazin-4(5H)-one; 3 -(2-methoxypyridin-3 -yl)[2-(quino liny1)ethy1]thieno [2,3-d]pyridazin— 4(5H)-one; 3 —(pyridin—3 —y1)—5 — [2—(quino yl)ethyl]thieno [2 ,3 idazin—4(5H)-0ne; 30 3 -(4-methoxypyridin—3 —y1)—5—[2—(quinolin—2—yl)ethy1]thien0[2,3-d]pyridazin— 4(5H)-one; 3 -(furan—3 -y1)-5 - [2-(quino1in—2—y1)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 77 3 -(quin01in-3 -y1)-5 - [2-(quino1in—2—y1)ethy1]thieno [2,3 -d]pyridazin—4(5H)-one; 3 -(isoquinolinyl)-5 - [2-(quin01inyl)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 3 -(isoquinolin-5 -y1)-5 - [2-(quin01iny1)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 3 -(1H-ind01y1)-5 -(2-quin01iny1—ethyl)—5H-thieno [2,3 idazin—4-one; 3 -(2,3-dihydrobenzo furan—S-yl)—5—(2-quin0 liny1—ethy1)-5H-thieno [2,3- d]pyridazin—4-one; 3 -(quin0 1111-5 -y1)-5 - [2-(quin0 lin—2—yl)ethyl]thien0 [2,3 -d]pyridazin—4(5H)-one; 3 -(3,5-dimethyl-1,2-oxazolyl)[2-(quino1iny1)ethy1]thieno[2,3 - d]pyridazin—4(5H)-one; 10 3 thoxypyridin-3 -yl)—5-[2-(quino liny1)ethy1]thien0 [2,3-d]pyridazin— 4(5H)-one; 3 -(2,3-dihydr0-1 ,4—benzodioxin—6—yl)—5—[2—(quin01iny1)ethyl]thieno[2,3 - d]pyridazin—4(5H)-one; 3 -(2-methy1pyridinyl)-5—[2—(quino1in—2—y1)ethyl]thieno [2,3-d]pyridazin-4(5H)- 15 one; 3 -(5 -meth0xypyridin-3 -y1)[2-(quino1in—2-y1)ethy1]thieno [2,3-d]pyridazin— 4(5H)-one; 3 - rph01inyl)pyridiny1][2—(quino y1)ethy1]thieno [2,3 - d]pyridazin—4(5H)-0ne; 20 3 -(1 ,3-benz0di0X01-5 -y1)[2—(quin01in—2-y1)ethyl]thieno [2,3-d]pyridazin-4(5H)- one; 3 -(quino 1iny1)-5 - [2-(quin01in—2-yl)ethy1]thieno [2,3 -d]pyridazin—4(5H)-one; 3 -(1-methy1—1H-pyrazo1-4—yl)—5-[2—(quinolin—2-y1)ethy1]thieno[2,3-d]pyridazin- 4(5H)-one; 25 3 -[1-(2-methy1propy1)-1H-pyrazol—4-yl]—5-[2-(quin01in—2—y1)ethy1]thieno[2,3 - d]pyridazin—4(5H)-one; tert-butyl 2- {4-oxo-5 - [2-(quino lin—2-yl)ethyl]-4,5 rothieno [2,3-d]pyridazin— 3 -y1}-1H-pyrrole- l-carboxylate; 3 —(2—methoxypyrimidin—5-yl)—5-[2-(quin0 lin—2-y1)ethy1]thien0 [2,3 -d]pyridazin— 30 4(5H)-one; 5 - [2-(quino liny1)ethy1] 3 ,4—triflu0r0phenyl)thien0 [2,3-d]pyridazin-4(5H)- one; 78 3-(4-fluoron1ethylphenyl)-5—[2—(quino1iny1)ethyl]thieno[2,3-d]pyridazin— 4(5H)-one; 3-(4-fluoromethylphenyl)-5—[2-(quino1iny1)ethy1]thieno[2,3-d]pyridazin— 4(5H)-one; hlorofluoropheny1)[2-(quinolin—2-y1)ethy1]thieno [2,3-d]pyridazin— 4(5H)-one; hlorofluoropheny1)[2—(quinolin—2-yl)ethyl]thieno[2,3-d]pyridazin— 4(5H)-one; 3-(3,4-dimethylphenyl)[2-(quinolin—2-yl)ethy1]thieno[2,3-d]pyridazin-4(5H)- 10 one; 3-(2,4-dimethylpheny1)—5—[2—(quinolin—2—yl)ethy1]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,4-difluoropheny1)-5—[2—(quino1in—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 15 3-(2,4-din1ethoxyphenyl)[2-(quino1in—2—y1)ethyl]thieno [2,3-d]pyridazin-4(5H)- one; 3-(2,5-din1ethoxyphenyl)[2-(quino1in—2-y1)ethy1]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,3-difluorophenyl)[2-(quino1iny1)ethy1]thieno[2,3-d]pyridazin-4(5H)- 20 one; 3-(3,4-din1ethoxyphenyl)[2—(quino1iny1)ethy1]thieno[2,3-d]pyridazin-4(5H)- one; 3-(3,4-difluorophenyl)[2-(quino1in—2-yl)ethy1]thieno[2,3-d]pyridazin-4(5H)- one; 25 3-(5-fluoromethoxyphenyl)—5-[2-(quinoliny1)ethy1]thieno [2,3-d]pyridazin- 4(5H)-one; 3-(4-fluoromethoxyphenyl)[2-(quinoliny1)ethy1]thieno[2,3-d]pyridazin- 4(5H)-one; 3—(3,5—din1ethoxyphenyl)[2-(quinoliny1)ethy1]thieno[2,3-d]pyridazin-4(5H)- 30 one; 3-(2,5-difluoropheny1)—5—[2—(quinolin—2—yl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 79 3 -(2,3-din1ethoxyphenyl)[2—(quino1iny1)ethy1]thieno [2,3-d]pyridazin-4(5H)- one; 3 -(3 -fluorornethoxyphenyl)—5-[2—(quino liny1)ethy1]thieno [2,3-d]pyridazin- 4(5H)-one; 3 -(2-fluorornethoxyphenyl)—5-[2—(quino y1)ethy1]thieno [2,3-d]pyridazin- 4(5H)-one; 3 -(3 ,5-difluorophenyl)-5 -[2—(quino linyl)ethyl]thieno [2,3 idazin-4(5H)— one; 3 -(3 -fluoromethoxyphenyl)[2-(quino liny1)ethy1]thieno [2,3-d]pyridazin- 10 4(5H)—one; 3-(2-methoxy—5—methy1phenyl)—5—[2—(quino lin—2-y1)ethy1]thieno [2,3 -d]pyridazin— 4(5H)-one; 3 -(2,5-dichloropheny1)—5 —[2—(quino1in—2—yl)ethyl]thieno [2,3 -d]pyridazin-4(5H)- one; 15 3 -(naphtha1en—2-y1)-5 - [2-(quinolin—2—y1)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 3 -pheny1-5 -[2-(quino1iny1)ethy1]thieno[2,3-d]pyridazin—4(5H)-one; 3 nzo furan—2-y1)-5 - [2-(quino1in—2—y1)ethy1]thieno [2,3 idazin—4(5H)- one; 3 -(1H-indazol—5 -y1)-5 - [2-(quino1iny1)ethy1]thieno [2,3 -d]pyridazin-4(5H)-one; 20 3 -(1 -methyl- 1 H-pyrazo1yl)—5—[2—(quino1iny1)ethy1]thieno[2,3-d]pyridazin- 4(5H)-one; 3 -(4,5-difluorornethoxypheny1)—5—[2-(quinoliny1)ethy1]thieno[2,3 - d]pyridazin—4(5H)-one; 3 -(2-fluorornethy1pheny1)—5-[2-(quino liny1)ethy1]thieno [2,3-d]pyridazin— 25 4(5H)-one; 3-(2-fluoromethoxyphenyl)—5-[2—(quinoliny1)ethy1]thieno [2,3—d]pyridazin- 4(5H)-one; 3 1 {4-oxo-5 -[2-(quinolinyl)ethyl]-4,5 -dihydrothieno [2,3-d]pyridazin— 3 —y1}benzonitrile; 30 5 - [2-(6-fluoroquino lin—2—y1)ethyl]—3—(pyridin—4—yl)thieno [2,3 -d]pyridazin-4(5H)- one; 80 5 - [2,2-difluor0(quino yl)ethy1](pyridinyl)thieno [2,3 idazin— 4(5H)-one; 3 -(pyridin—4-y1)-5 - [2-(thien0[3 ,2-b]pyridin—5-y1)ethy1]thieno[2,3-d]pyridazin— 4(5H)-one; 5 - [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—3-(pyridin—4—y1)thieno [2,3 -d]pyridazin— 4(5H)-one; 5 - [2-(7-flu0roimidazo [1 ,2-a]pyridin—2-yl)ethyl]-3 -(pyridinyl)thieno [2,3 - dazin—4(5H)-one; 8-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 10 2—[2-(quinoliny1)ethyl][4-(trifluoromethy1)phenyl]phtha1azin-1(2H)-0ne; 8-(4-methylpheny1)—2—[2—(quino lin—2—yl)ethy1]phthalazin- 1 (2H)-0ne; 8-[4-(propan—2-y1)pheny1]—2—[2—(quinolin—2—yl)ethyl]phthalazin- 1 (2H)-0ne; 8-(4-ethy1phenyl)[2—(quino1in—2—y1)ethyl]phtha1azin—1(2H)-0ne; 4- {4-oxo-3 -[2-(quino linyl)ethy1]—3 ,4—dihydrophthalazin-5 -y1}benzonitrile; 15 ethoxyphenyl)[2-(quino1in—2—y1)ethy1]phthalazin- 1 (2H)-one; 8-(4-flu0rophenyl)[2-(quinolin-2—y1)ethy1]phthalazin- 1 (2H)-one; (4- {4-oxo [2-(quino linyl)ethyl]-3 ,4—dihydrophthalazin-5 - y1}phenyl)acetonitrile; 8-(4-hydroxyphenyl)[2-(quin0liny1)ethyl]phthalazin- 1 (2H)-one; 20 8-(2-chlorophenyl) [2-(quino1in—2-y1)ethy1]phthalazin- 1 (2H)-one; 8-(2-rnethy1pheny1)[2-(quin01in—2-y1)ethy1]phthalazin- 1 (2H)-one; 8-(2-ethy1pheny1)[2-(quin01iny1)ethyl]phthalazin- 1 (2H)-one; 8-(2-fluoropheny1)[2-(quin01in—2-y1)ethyl]phthalazin- 1 (2H)-one; 8-(2-methoxyphenyl)[2-(quin01in—2—yl)ethy1]phthalazin- 1 (2H)-one; 25 8-(3 -methoxyphenyl)[2-(quin0lin—2-yl)ethyl]phthalazin-1(2H)-one; 3 - {4-ox0-3 -[2-(quin01in—2—yl)ethyl]—3 ,4-dihydrophthalazin—5 —y1}benzonitrile; 8-(3-fluorophenyl)[2-(quino linyl)ethyl]phtha1azin- 1 (2H)-one; 8-(3 -hydroxyphenyl)[2-(quinolinyl)ethy1]phtha1azin-1 (2H)-one; N,N—dimethy1—3 — {4-oxo[2-(quino yl)ethy1] -3 ,4-dihydr0phthalazin-5 - 30 y1}benzamide; 8-(3 -methy1pheny1)—2—[2—(quino1in—2—yl)ethy1]phthalazin- 1 (2H)-0ne; 2-[2-(quino1iny1)ethy1]—8—(thi0phen—2—yl)phtha1azin—1(2H)-0ne; 8 1 8-(1-methy1—1H-indol—5-y1)-2—[2—(quino1iny1)ethy1]phthalazin—1(2H)-0ne; 8-(3 ,S-dimethyl-1H-pyrazol—4—y1)—2-[2-(quino1iny1)ethy1]phthalazin-1(2H)-0ne; 8-(1H-ind01y1)[2-(quin0lin—2—y1)ethyl]phthalazin—1(2H)-one; 8-(1H-indo1—6-y1)[2-(quin01in—2-y1)ethyl]phthalazin—1(2H)-one; 8-(pyrimidin—5-y1)[2-(quin01in-2—yl)ethyl]phthalazin-1(2H)-one; 8-(2-methoxypyridiny1)[2-(quin0[iny1)ethy1]phthalazin- 1 (2H)-one; 8—(pyridin—3 -y1)[2-(quin0[in—2—yl)ethyl]phtha1azin-1 (2H)—0ne; 8-(furan-3 -y1)[2-(quinolin—2-yl)ethyl]phthalazin-1(2H)-one; 8-(quino 1in-3 -[2-(quinolin—2-yl)ethyl]phtha1azin- 1 (2H)-one; 10 8—(1H—indo1—4—y1)—2—[2—(quino[inyl)ethyl]phthalazin- 1 (2H)-0ne; 8-(2,3-dihydr0— 1 —benzofi1ran—5—yl)—2—[2—(quino liny1)ethyl]phthalazin- 1 (2H)- one; 8-(3 ,4-dihydr0-2H-1 ,5-benzodi0xepin—7—yl)—2—[2-(quin01iny1)ethy1]phthalazin- 1 (2H)-one; 15 8-(1-benzofuran—S-y1)[2-(quino1in—2—y1)ethy1]phthalazin-1(2H)-0ne; nethoxypyridin-3 -y1)[2-(quinolin—2—y1)ethyl]phthalazin— 1 (2H)-one; 8-(2,3-dihydr0-1 ,4-benzodioxiny1)—2—[2—(quino1iny1)ethy1]phthalazin— 1 (2H)- one; 8-(2-methy1pyridinyl)[2—(quino1iny1)ethy1]phthalazin- 1 (2H)-one; 20 8-(5-meth0xypyridiny1)-2—[2—(quino1iny1)ethyl]phthalazin— 1 (2H)-one; 8-(5 -flu0r0pyridin—3 -y1)[2—(quin01in—2-y1)ethy1]phthalazin- 1 (2H)-one; 8-(1 ,3-benzodioxo 1—5 -y1)-2—[2—(quin01in-2—y1)ethyl]phthalazin-1(2H)-0ne; 8-(1-methy1—1H-pyrazo1—4—y1)—2-[2—(quin0lin—2-yl)ethy1]phthalazin-1(2H)-0ne; 8-[ 1 -(2-methy1propyl)- 1H-pyrazol—4-yl]—2-[2-(quin01iny1)ethy1]phthalazin— 25 1 (2H)-one; utyl 2- {4-0x0-3 - [2-(quin0 lin—2-yl)ethyl]-3 ,4-dihydr0phthalazin—5 -y1}-1H— pyrrolecarboxylate; 8-(3 ofluorophenyl)[2-(quinoliny1)ethy1]phtha1azin-1(2H)-one; 8-(2—chloro—4—fluorophenyl)[2-(quinolin—2-y1)ethy1]phthalazin- 1 (2H)-0ne; 30 8-(3 ,4-dimethylpheny1)—2—[2—(quino lin—2—yl)ethy1]phthalazin- 1 (2H)-0ne; 8-(2,4-dimethoxypheny1)—2—[2—(quino yl)ethyl]phthalazin- 1 (2H)-0ne; 8-(2,5-dimethoxypheny1)—2—[2—(quino1in—2—y1)ethyl]phthalazin- 1 (2H)-0ne; 82 8-(2,3-difluor0pheny1)[2-(quin01iny1)ethy1]phthalazin-1(2H)-0ne; 8-(3 ,4-dimethoxypheny1)-2—[2—(quin01iny1)ethy1]phthalazin- 1 (2H)-one; 8-(3 ,4-difluoropheny1) [2—(quin01iny1)ethy1]phtha1azin- 1 (2H)-one; 8-(5-flu0r0methoxypheny1)—2-[2—(quin0 lin—Z-yl)ethy1]phtha1azin- 1 ne; 8-(4-fluorometh0xyphenyl)—2-[2-(quin0 liny1)ethy1]phtha1azin- 1 (2H)-one; 8-(3,5-dimeth0xyphenyl)—2—[2-(quin0[in—2—y1)ethy1]phthalazin-1(2H)-one; 8-(2,5-diflu0r0pheny1)[2—(quin0[inyl)ethy1]phthalazin-1(2H)-0ne; 8-(3 -fluoromethoxyphenyl)[2-(quinoliny1)ethy1]phtha1azin-1(2H)-one; 8-(2-fluoromethoxyphenyl)[2-(quinoliny1)ethy1]phtha1azin-1(2H)-one; 10 8—(3,5—difluoropheny1)[2-(quino[inyl)ethy1]phthalazin-1(2H)-0ne; 8-(3-flu0r0—5—methoxyphenyl)—2—[2—(quin0lin—2—y1)ethy1]phtha1azin- 1 (2H)-0ne; 8-(naphthaleny1)—2—[2—(quinolin—2—yl)ethy1]phtha1azin-1(2H)-0ne; y1—2-[2-(quino1111—2—y1)ethy1]phthalazin— 1 (2H)-0ne; 8-(1-benzofuran—Z-y1)[2-(quino1in—2—y1)ethy1]phtha1azin-1(2H)-one; 15 8-(1-rnethy1—1H-pyrazo1yl)-2—[2—(quino1in—2-y1)ethy1]phtha1azin-1(2H)-0ne; 8-(4,5-diflu0romethoxyphenyl)-2—[2—(quin0liny1)ethy1]phtha1azin- 1 (2H)- one; 8-(2-fluor0rnethy1pheny1)—2-[2-(quinoliny1)ethy1]phtha1azin— 1 (2H)-one; 8-(2-flu0r0meth0xyphenyl)—2—[2-(quino1iny1)ethy1]phtha1azin- 1 (2H)-one; 20 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl](pyridin—3 -y1)phtha1azin—1(2H)-one; 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl](pyridin—4-y1)phtha1azin—1(2H)-0ne; 2- [2-(imidazo [1 yridin—2-yl)ethyl]-8—(3-methoxypyridin—4-y1)phtha1azin— 1 ne; imidazo[1 ,2-a]pyridin—2-yl)ethyl]-8—(pyrimidin—5 -y1)phtha1azin-1(2H)-one; 25 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl](1-methy1—1H—pyrazol—3 -y1)phtha1azin- 1 (2H)-one; 8-(furan-3 -y1) [2-(imidazo[1 ,2-a]pyridiny1)ethy1]phtha1azin—1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-oxo-2,3-dihydro-1H-ind01 y1)phtha1azin-1(2H)—one; 30 8-(3 ,4-dihydro—2H—chromen—6—yl)—2—[2—(imidazo [1 ,2-a]pyridin—2- y1)ethy1]phtha1azin— 1 (2H)—one; 83 8-(1 , 1 -dioxidothiomorpho1in—4—y1)—2-[2-(quino1iny1)ethy1]phthalazin- 1 (2H)- one; 2- [2-(imidazo [1 yridin—2-yl)ethyl]-8—(morpho1iny1)phtha1azin-1(2H)-0ne; 8-(1 , 1-dioxidothiomorpholin—4-y1)—2—[2-(imidazo [1 ,2-a]pyridin—2- y1]phtha1azin— 1 (2H)—0ne; 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—8—(tetrahydr0— 1 H—furo [3 ,4-c]pyrr01- 5(3H)-y1)phtha1azin—1(2H)—0ne; 8-(5 ,5-difluorohexahydrocyclopenta[c]pyrrol—2( 1 H)-y1)[2-(imidazo [1 ,2- a]pyridinyl)ethyl]phthalazin-1(2H)-one; 10 2— [2—(imidazo [1 ,2—a]pyridin—2-yl)ethyl](piperaziny1)phtha1azin-1(2H)-0ne; 8-(4,4-difluoropiperidin—1—y1)—2—[2—(imidazo[ 1 ,2-a]pyridin—Z-y1)ethy1]phtha1azin- 1 (2H)-one; 8- [4-(ch10romethy1)(hydroxymethyl)piperidin— 1 -y1] [2-(irnidaz0 [1 ,2- a]pyridiny1)ethy1]phtha1azin— 1 ne; 15 2- [2-(irnidazo [1 ,2-a]pyridinyl)ethy1]—8—(piperidin—1-y1)phtha1azin-1(2H)-0ne; 8-(2,3-dihydro-4H- 1 zoxazin—4—y1)—2—[2-(imidazo [1 ,2—a]pyridin—2- y1)ethy1]phtha1azin— 1 (2H)-one; 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethy1][4-(triflu0r0rnethy1)piperidin— 1 - y1]phtha1azin—1(2H)-one; 20 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl](4-methy1piperaziny1)phtha1azin- 1 (2H)-one; 8-(1 ,3-dihydro-2H-isoind01—2-y1)-2—[2-(imidazo [1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)—0ne; 8-(7-benzy1—2,7-diazaspir0[4.4]n0nyl)-2—[2-(imidazo [1 ,2-a]pyridin—2- 25 yl)ethy1]phtha1azin— 1 (2H)-0ne; 8-( { [(3 aR,4S,6aS)benzyloctahydrocyclopenta[c]pyrr01y1]methyl} amino) idazo[1 ,2-a]pyridin-Z-yl)ethyl]phthalazin-1(2H)-one; tert-butyl (3 R)-3 -( {3 -[2-(imidazo[1 ,2-a]pyridiny1)ethy1] oxo-3 ,4- dihydrophthalazin—S —y1} amino)pyrrolidinecarb0xy1ate; 30 8-(2,6-dimethy1morpho1in—4—yl)—2—[2—(imidazo[ 1 ,2-a]pyridin y1]phtha1azin— 1 (2H)—one; 84 2- [2-(in1idazo [1 ,2-a]pyridin—2—yl)ethy1](1,4-oxazepan—4-y1)phtha1azin—1(2H)- one; tert-butyl 4- {3 - [2-(irnidazo[1 yridiny1)ethy1] oxo-3 ,4- dihydrophthalazin—S -y1} -3 ,6—dihydropyridine- 1 (2H)-carboxy1ate; 5 -(pyridin—4-y1) [2-(quino1in—2—y1)ethyl]phtha1azin— 1 (2H)-one; 5 -(pyrimidin—5 -y1)[2-(quino1inyl)ethyl]phthalazin-1(2H)-one; 5 -(1-methy1—1H—pyrazo1yl)[2-(quinolin—2-y1)ethy1]phthalazin—1(2H)-one; 5 -(1,1-dioxidothiomorpholinyl)[2-(quino1iny1)ethy1]phthalazin-1(2H)- one; 10 2— [2—(in1idazo [1 ,2-a]pyridin—2-yl)ethyl](pyridin-3 -y1)phtha1azin—1(2H)-one; 2- [2-(imidazo [1 ,2—a]pyridin—2—yl)ethyl]—5—(pyrimidin-5 -y1)phtha1azin1 (2H)-one; 2- [2-(imidazo [1 ,2—a]pyridin—2—y1)ethyl]—5—(morpho1iny1)phtha1azin-1(2H)-one; 2- [2-(imidazo [1 yridin—2—y1)ethy1]—5—(tetrahydro- 1 H-furo [3 ,4-c]pyrro 1- 5 (3H)-y1)phtha1azin— 1 (2H)-one; 15 2- [2-(in1idazo [1 ,2-a]pyridinyl)ethy1]—4—(pyrin1idin-5 -y1)phtha1azin-1(2H)-one; 2- [2-(in1idazo [1 ,2-a]pyridin—2-yl)ethyl]—4—(morpho1iny1)phtha1azin-1(2H)-one; 3 -(3 -n1ethoxypyridiny1)[2-(quino1in—2-y1)ethy1]thieno [3 ,2-c]pyridin—4(5H)- one; 3 -(3 -hydroxypyridiny1)[2—(quino1iny1)ethy1]thieno [3 ,2-c]pyridin—4(5H)- 20 one; 3 -(1-n1ethy1—1H-pyrazo1y1)—5—[2-(quino1iny1)ethy1]thieno[3 ,2-c]pyridin— 4(5)H-one; 3 -(pyridin—4-y1)-5 - [2-(quino1in—2-y1)ethyl]thieno [3 ,2-c]pyridin—4(5H)-one; 3 -(pyrirnidin—5 -y1)-5 ino1in—2-y1)ethyl)thieno [3 yridin-4(5H)-one; 25 3 -(2-oxoindo liny1)-5 - [2-(quinolinyl)ethy1]thieno [3 yridin-4(5H)-one; 3 -(3 -hydroxypheny1)-5 - [2-(quino linyl)ethy1]thieno [3 ,2-c]pyridin-4(5H)-one; 5 - [2-(5-ethy1pyridinyl)ethyl](pyridiny1)thieno [3 ,2-c]pyridin-4(5H)-one; 5 - idazo[1 ,2-a]pyridinyl)ethyl](pyridiny1)thieno [3 ,2-c]pyridin- 4(5H)—one; 30 3 -(morpholin—4—y1)—5 — [2—(quinolin—2—yl)ethy1]thieno [3 ,2-c]pyridin-4(5H)-one; tert-buty1(4-oxo—5 —(2—quino1in—2—yl)ethyl)—4,5 -dihydrothieno [3 ,2-c]pyridin—3 - y1)-5 ,6-dihydropyridine— 1 (2H) carboxylate; 85 5 -(2-(quino1iny1)ethy1)(1 ,2,3 rahydropyridin—4-y1)thieno [3 ,2-c]pyridin— 4(5H)-one; tert-butyl(4-oxo-5 -(2-quinoliny1)ethyl)—4,5 -dihydrothieno [3 ,2-c]pyridin—3 - y1)-piperidine-1 carboxylate; 5 3 -(piperidin—4-y1)-5 - ino1iny1)ethyl]thieno [3 ,2-c]pyridin-4(5H)-one; 3 -methy1—7-(pyridiny1)(2-(quino lin—2—yl)ethyl)thieno [3 ,2-c]pyridin—4(5H)- one; 3 diny1)-5 - [2-(5 ,6,7,8-tetrahydroquino lin-2 -y1)ethy1]thieno [2,3 - d]pyridazin-4(5H)-one; 10 (R)—tert—buty1 3 —(3 —(2—(imidazo[1 yridin—2-y1)ethy1)oxo-3 ,4- dihydrophthalazin—S—y1amino)pyrrolidine— 1 —Carboxylate; 2- idazo [1 ,2—a]pyridin—2—yl)ethyl]—8—[(3 rolidin-3 -y1arnino]phthalazin- 1 (2H)-one; 5 -(3 -hydroxypheny1)-2— [2-(quino1in—2—y1)ethyl]phthalazin- 1 (2H)-one; 1 5 (E)—8-(pyridinyl)(2-(quino1in—2—y1)viny1)isoquinolin- 1 (2H)-one; anti (rac) 8-(pyridiny1)(2-(quino1in—2—y1)cyclopropy1)isoquinolin- 1 (2H)-one; anti (+)(pyridiny1)(2-(quino1in—2—y1)cyclopropyl)isoquino lin- 1 (2H)-one; anti (-)(pyridin—4-y1)(2-(quino1in—2—y1)cyclopropy1)isoquinolin-1(2H)-one; 8-(pyridin—4-y1)(2-quino1in—2—y1—ethy1)isoquino lin- 1 (2H)-one; 20 anti (rac) 3 -(pyridiny1)(2—(quino1iny1)cyclopropy1)thieno [3 ,2-c]pyridin- 4(5H)-one; anti (+) 3-(pyridiny1)-5—(2—(quino1in-2—yl)cyclopropy1)thieno[3 ,2-c]pyridin- 4(5H)-one; anti (-) 3-(pyridin—4-y1)-5—(2-(quinolin—2-yl)cyclopropy1)thieno[3 ,2-c]pyridin- 25 4(5H)—one; (E)—8-pyridin—4-y1—2-(2-quino linyl—vinyl)-2H-phthalazin- 1 —one; anti (rac) 8-pyridinyl(2-quinolinyl-cyclopropy1)-2H-phtha1azinone; anti (+) 8-pyridinyl(2-quino linyl-cyclopropy1)-2H-phtha1azinone; anti (—) 8-pyridiny1—2—(2-quino lin—2-yl-cyclopropy1)-2H-phthalazinone; 3 0 and the N—oxides, the prodrugs, the tautomers and the hydrates thereof, and the pharmaceutically acceptable salts thereof. 86 Further particular embodiments ofthe invention relate to the compounds of formula I, where the nds ofthe formula I are selected from the group consisting of: 7-(pyridin—4-yl)-5 - [2-(quinolinyl)ethyl]furo[3 ,2-c]pyridin-4(5H)-one; 3 -(pyridinyl)-5 -(2-(quino1in—2—y1)ethyl)fi1ro[3 ,2—c]pyridin-4(5H)-one; 5 -(2-( l H—benzo dazo1yl)ethyl)(pyridiny1)furo [3 ,2-c]pyridin—4(5H)- one; 5 -(2-(imidazo [1 ,2-a]pyridinyl)ethyl)(pyridiny1)furo [3 ,2-c]pyridin-4(5H)- one; 10 3 —(pyrimidin—5 —yl)—5 uino linyl)ethyl)furo [3 ,2-c]pyridin—4(5H)-one; 4-(pyridin—4—yl)—6—(2—(quinolin—2—yl)allyl)pyrido [2 ,3 -d]pyridazin-5 (6H)-one; syn 8-(pyridin—4-yl)—2—(2—(quinolin—2—yl)cyclopropyl)isoquino lin- l (2H)-one; syn 3 -(pyridin—4-yl)-5 -(2—(quinolin—2—yl)cyclopropyl)thieno [3 ,2-c]pyridin—4(5H)- one; 15 anti 3 dazin—4-yl)-5 -(2-(quinolin—2—yl)cyclopropyl)furo [3 ,2-c]pyridin-4(5H)- one; syn 3 -(pyridazinyl)-5 -(2-(quinolin-2—yl)cyclopropyl)furo [3 ,2-c]pyridin-4(5H)- one; anti 3 -(pyridinyl)-5 -(2-(quino1inyl)cyclopropyl)furo [3 ,2-c]pyridin-4(5H)- 20 one; syn 3 -(pyridinyl)-5 -(2-(quinolin-2—yl)cyclopropyl)furo [3 ,2-c]pyridin-4(5H)- one; anti 8-(oxetan—3 -ylamino)(2—(quino1inyl)cyclopropyl)isoquino lin- l (2H)-one; syn 8-(oxetan—3-ylamino)(2-(quinoliny1)cyclopropyl)isoquinolin- l (2H)-one; 25 anti 8-(pyridazin—4-y1)(2—(quinolinyl)cyclopropy1)isoquinolin- 1 (2H)-one; syn 8-(pyridazinyl)(2-(quinolin—2-yl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(6-fluoropyridinyl)(2-(quinoliny1)cyclopropyl)isoquinolin-1(2H)- one; syn 8—(6—fluoropyridin—3-yl)—2-(2-(quinoliny1)cyclopropyl)isoquinolin-1(2H)- 30 one; anti uoropyridin—4—yl)—2—(2—(quino lin—2—yl)cyclopropyl)isoquino lin- 1 (2H)- one; 87 syn 8-(2-fluoropyridin—4-y1)—2—(2—(quino1iny1)cyclopropy1)isoquino 1in- 1 (2H)- one; anti 8-(pyridin-3 -y1)(2-(quino1iny1)cyclopropy1)isoquino1in- 1 (2H)-one; syn idin—3 -y1)(2—(quinolinyl)cyclopropy1)isoquino1in- 1 (2H)-one; anti 8-(pyrirnidin-5 -(2—(quino1inyl)cyclopropy1)isoquino 1in- 1 (2H)-one; syn 8-(pyrimidin—5 -y1)(2—(quino linyl)cyclopropy1)isoquino lin- 1 (2H)-one; anti 8-(1-methy1—1H—pyrazol—4—yl)(2-(quinolin-2—y1)cyclopropyl)isoquino1in- 1 (2H)-one; syn 8-(1-methyl- 1H-pyrazolyl)(2-(quinolin-2 -y1)cyclopropy1)isoquino1in- 10 1 (2H)-one; anti 8-(3 -fluoropyridin—4—y1)—2—(2—(quinolin—2—y1)cyclopropyl)isoquinolin- 1 (2H)- one; syn 8-(3 -fluoropyridin—4—y1)—2—(2—(quinolin—2—yl)cyclopropyl)isoquinolin- 1 (2H)- one; 15 anti uoropyridiny1)(2—(quino1in—2-y1)cyclopropy1)isoquino 1in- 1 (2H)- one; syn 8-(2-fluoropyridin—4-y1)(2-(quino1in—2-y1)cyclopropy1)isoquino 1in- 1 (2H)- one; anti 8-((3 S)hydroxypiperidin—1-y1)(2-(quinolin 20 y1)cyclopropy1)isoquino1in- 1 (2H)—one; syn 8-((3 S)-3 -hydroxypiperidin—1—y1)—2—(2-(quinolin—2- y1)cyclopropyl)isoquino1in- 1 (2H)—one; anti 8-(3 -rnethoxypiperidin-1 —y1)(2-(quino 1iny1)cyclopropy1)isoquino 1in- 1 (2H)-one; 25 syn 8-(3-methoxypiperidiny1)(2-(quinoliny1)cyclopropyl)isoquino1in- 1 ne; anti 8-morpholino(2-(quinolinyl)cyclopropyl)isoquino1in-1(2H)-one; syn 8-morpho lino(2-(quinolin—2-yl)cyclopropyl)isoquino1in- 1 (2H)-one; anti 1-(1—oxo—2—(2—(quinolin-Z-yl)cyclopropy1)-1 ,2-dihydroisoquino 1in 30 y1)piperidineoarbonitrile; syn 1-( 1 -oxo(2—(quino1in—2—y1)cyclopropyl)— 1 ,2-dihydroisoquino 1in y1)piperidinecarbonitrile; 88 anti 8-((3R,4R)fluorohydr0xypiperidinyl)(2-(quin01in yl)cyc10pr0pyl)isoquino lin- 1 (2H)-0ne; syn 8-((3R,4R)fluorohydr0xypiperidin—1-y1)(2-(quin01in 10propyl)isoquino lin- 1 (2H)—0ne; anti 8-((3 S)hydroxypyrr01idin— 1 -y1)(2-(quin01in—2- y1)cyc10propyl)isoquinolin- 1 (2H)—0ne; syn 8-((3 S)-3 -hydr0xypyrrolidinyl)—2-(2—(quin01in—2— y1)cyclopropyl)isoquinolin- 1 (2H)-one; anti 8-((3 R)-3 xypyrrolidinyl)(2-(quinolin 10 y1)cyclopropy1)isoquino lin- 1 (2H)—one; syn )—3 —hydroxypyrrolidin—1—yl)—2—(2—(quin01in—2- yl)cyc10propyl)isoquino lin— 1 (2H)—0ne; anti 8-(methy1(oxetan—3—y1)amino)—2—(2—(quino1iny1)cyclopr0pyl)isoquinolin- 1 (2H)-one; 15 syn 8-(niethy1(oxetany1)amino)—2—(2—(quin01iny1)cyclopropyl)isoquino lin- 1 (2H)-one; anti 8-(4-niethoxypiperidin-1 -(2—(quino 1iny1)cyc10pr0py1)is0quino lin- 1 (2H)-one; syn 8-(4-niethoxypiperidiny1)—2-(2-(quino1iny1)cyc10pr0py1)is0quino lin- 20 1 (2H)-one; anti 8-(4-hydroxypiperidin— 1 —y1)—2—(2-(quinoliny1)cyc10pr0py1)is0quino lin- 1 (2H)-one; syn 8-(4-hydroxypiperidin-1 -y1)—2-(2—(quino1in—2-y1)cyc10pr0py1)isoquinolin- 1 (2H)-one; 25 anti 8-(1 -acetylpiperidin-4—ylamin0)—2—(2-quinolin—2-cyclopropy1)isoquino lin- 1 (2H)-one; syn 8-(1-acetylpiperidin—4-ylamino)(2-quino1incyclopropy1)isoquinolin- 1 (2H)-one; anti 8-(piperidiny1amino)(2-(quin0liny1)cyclopr0pyl)isoquin0lin-1(2H)- 30 one; syn 8-(piperidin—4—y1amino)—2—(2—(quin0lin—2—yl)cyclopr0pyl)isoquino lin- 1 (2H)- one; 89 anti 2-(2-(quino1iny1)cyc10pr0pyl)—8-(tetrahydro-2H-pyran—4-y1)isoquinolin- 1 (2H)-one; syn 2-(2-(quino1iny1)cyclopr0pyl)(tetrahydro-2H-pyran—4-y1)is0quino1in- 1 ne; anti 2-(2-(quino1iny1)cyclopr0pyl)—8-(2—0xa-6—azaspiro[3 .4]octan y1)isoquin01in-1(2H)-0ne; syn 2-(2-(quin01iny1)cyc10pr0pyl)—8-(2-0xa—6—azaspir0[3 .4]0ctan—6- y1)isoquino lin- 1 (2H)-one; anti 8-(dihydro- 1 H-furo [3 ,4-c]pyrrol-5(3H,6H,6aH)-y1)(2-(quino 1in 10 y1)cyclopropy1)isoquino 1in- 1 (2H)—one; syn 8-(dihydro— 1 H—furo [3 ,4-c]pyrro1-5(3H,6H,6aH)-y1)(2-(quino 1in yl)cyclopropy1)isoquino1in— 1 (2H)—0ne; anti 8-(4,4-difluoropiperidin— 1 —y1)—2—(2—(quino 1iny1)cyc10pr0pyl)isoquino 1in- 1 (2H)-one; 15 syn -difluoropiperidiny1)—2—(2—(quino 1iny1)cyc10propyl)isoquino 1in- 1 (2H)-one; anti 8-n10rpholino(2-(quinoliny1)cyclopropy1)isoquino1in- 1 (2H)-0ne; syn 8-n10rpho1in0(2-(quinolin—2-y1)cyclopropyl)is0quino 1in- 1 (2H)-0ne; anti 8-(3 0r0methyl)pyrr0lidin— 1 -y1)(2-(quin01in—2- 20 y1)cyclopropyl)isoquino1in- 1 (2H)—0ne; syn 8-(3 -(difluorornethyl)pyrr01idin— 1 —y1)(2-(quino 1in lopropyl)isoquino1in- 1 (2H)—0ne; anti 8-((1R,5 S)—3 -oxa—8-azabicyclo [3 .2. 1 ]octany1)(2-(quino y1)cyc10- propyl)isoquin01in- 1 (2H)-0ne; 25 syn 8-((1R,5 S)oxaazabicyclo [3 .2. ny1)—2—(2—(quino 1iny1)cyclo- propy1)is0quin0 lin- 1 (2H)-0ne; anti 8-(4-methylpiperazinyl)(2-(quinoliny1)cyc10propy1)isoquino1in- 1 (2H)-one; syn 8—(4—methy1piperazin— 1 -yl)—2-(2-(quino lin—2-y1)cyc10pr0py1)is0quin0 1in- 30 1 (2H)-one; anti 8-(3 -(fluor0methy1)pyrrolidin— 1 —yl)—2—(2—(quin01in—2- yl)cyclopropyl)isoquino1in- 1 (2H)—0ne; 90 syn 8-(3 -(fluoron1ethy1)pyrrolidin— 1 -y1)(2-(quino1in lopropy1)isoquino lin- 1 (2H)—one; anti 8-((1R,5 S)—8-oxa—3 -azabicyclo[3.2.1]octan—3 -(2-(quino 1in y1)cyclopropy1)isoquino lin- 1 (2H)—one; syn 8-((1R,5 S)oxa—3 -azabicyclo[3.2.1]octan-3 -y1)(2-(quino 1in y1)cyclopropy1)isoquino lin- 1 (2H)—one; anti 8-(4-fluoropheny1)-2—(2-(quinolin—2—yl)cyclopropy1)isoquinolin-1(2H)-one; syn 8-(4-fluorophenyl)(2-(quinolin—2-yl)cyclopropy1)isoquinolin-1(2H)-one; anti 8-(furanyl)(2-(quinolinyl)cyclopropy1)isoquinolin-1 (2H)-one; 10 syn 8—(furan—3 -y1)(2—(quino linyl)cyc lopropy1)isoquino lin- 1 (2H)-one; anti 8-(4,5 —dihydrofuran—3—yl)—2—(2—(quino lin—2—y1)cyclopropy1)isoquino lin- 1 (2H)- one; syn 8-(4,5 -dihydro furan—3—yl)—2—(2—(quino lin—2—y1)cyclopropy1)isoquino lin- 1 (2H)- one; 15 anti 8-(4-n1ethoxypheny1)—2-(2-(quino1in—2—y1)cyclopropy1)isoquino lin- 1 (2H)-one; syn 8-(4-n1ethoxypheny1)(2-(quino1in—2—y1)cyclopropy1)isoquino lin- 1 (2H)-one; anti 6-fluoroquinoliny1)cyclopropy1)morpholinoisoquinolin— 1 (2H)- one; syn 2-(2-(6-fluoroquinoliny1)cyclopropy1)niorpholinoisoquino lin- 1 (2H)- 20 one; anti 2-(2-(6-fluoroquino1in-2—y1)cyclopropy1)—8-(pyridin—3 -y1)isoquino lin- 1 (2H)- one; syn 2-(2-(6-fluoroquino1in—2-y1)cyclopropyl)(pyridin—3 -y1)isoquino lin- 1 (2H)- one; 25 anti 2-(2-(6-fluoroquino1iny1)cyclopropyl)—8-(pyrimidiny1)isoquinolin- 1 (2H)-one; syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyrimidiny1)isoquinolin- 1 (2H)-one; anti 2-(2—(6—fluoroquino [inyl)cyclopropyl)(pyridiny1)isoquino lin- 1 (2H)- 30 one; syn 2-(2-(6-fluoroquinolin—2—y1)cyclopropyl)—8—(pyridin—4-y1)isoquinolin- 1 (2H)- one; 91 anti 4-fluoro(pyridin—4-y1)—2—(2—(quino1iny1)cyclopropy1)isoquino lin- 1 (2H)- one; syn 4-fluoro(pyridiny1)—2—(2-(quino1iny1)cyclopropy1)isoquino lin- 1 (2H)- one; anti 4-chloro(pyrimidin-5—y1)(2—(quino1iny1)cyclopropy1)isoquinolin- 1 (2H)-one; syn 4-chloro(pyrimidin—5-yl)—2-(2—(quinoliny1)cyclopropyl)isoquinolin- 1 ne; anti 4-(pyridinyl)(2-(quinolinyl)cyclopropyl)pyrido ]pyridazin- 10 5(6H)—one; syn4-(pyridin—4—y1)—6—(2—(quinolin—2—yl)cyclopropyl)pyrido[2,3-d]pyridazin- 5(6H)-one; anti 2-(2-(6-fluoroquino 1in—2—y1)cyclopropyl)—8-(pyridin—4-y1)phthalazin— 1 (2H)- one; 15 syn 2-(2-(6-fluoroquino1inyl)cyclopropyl)—8-(pyridin—4-y1)phtha1azin— 1 (2H)- one; anti 7-fluoro(pyridin—4-y1)—5-(2-(quino1iny1)cyclopropy1)thieno[3 ,2- c]pyridin-4(5H)-one; syn 7-fluoro(pyridiny1)-5—(2-(quino1iny1)cyclopropyl)thieno[3 ,2- 20 c]pyridin-4(5H)-one; anti 3-(2-fluoropyridiny1)—5—(2-(6—fluoroquino1iny1)cyclopropy1)thieno[3 ,2- c]pyridin-4(5H)-one; syn 3-(2-fluoropyridin—4-y1)—5-(2-(6—fluoroquino1iny1)cyclopropy1)thieno[3 ,2- c]pyridin-4(5H)-one; 25 anti 5-(2-(6-fluoroquino1inyl)cyclopropyl)(pyridin—4—y1)thieno[3 ,2- c]pyridin-4(5H)-one; syn 5-(2-(6-fluoroquino linyl)cyclopropyl)(pyridinyl)thieno [3 ,2-c]pyridin- 4(5H)-one; anti 3—(pyrimidin—5—y1)—5-(2-(quino[inyl)cyclopropyl)thieno[3 ,2—c]pyridin— 30 one; syn 3-(pyrimidin—5—yl)—5—(2—(quinolin—2—yl)cyclopropyl)thieno[3 ,2-c]pyridin— 4(5H)-one; 92 anti 3-(pyrin1idiny1)(2-(quin01iny1)cyc10pr0py1)thieno[3 ,2-c]pyridin— 4(5H)-one; syn 3-(pyrirnidin—5-y1)(2—(quin01iny1)cyc10propy1)thieno[3 ,2-c]pyridin— 4(5H)-one; anti 3-(pyrirnidiny1)-5—(2-(quin01in—2—yl)cyclopr0py1)thieno[3 ,2-c]pyridin— 4(5H)-one; syn 3-(pyrimidin—5-y1)(2-(quin0[inyl)cyc10pr0pyl)thien0[3 ,2-c]pyridin— one; anti 3-(6-fluoropyridinyl)(2-(quinoliny1)cyc10propy1)thieno[3,2- 10 c]pyridin-4(5H)—one; syn 3-(6-flu0ropyridin—3—y1)—5—(2—(quin0lin—2—yl)cyclopropyl)thien0[3 ,2-c]pyridin- 4(5H)-one; anti 3-(2-methy1pyrimidin—5—y1)—5—(2—(quin0lin—2-y1)cyc10pr0pyl)thien0[3 ,2- c]pyridin-4(5H)-one; 15 syn 3-(2-methylpyrimidiny1)—5—(2—(quinoliny1)cyc10pr0pyl)thieno[3 ,2- c]pyridin-4(5H)-0ne; anti 3 -(pyridazin—4-yl)-5 -(2-(quinolin—2—yl)cyclopr0pyl)thieno [3 yridin— 4(5H)-one; syn 3 -(pyridaziny1)-5 -(2-(quino1iny1)cyclopropyl)thieno [3 ,2-c]pyridin— 20 4(5H)-one; anti 3-(2-fluoropyridiny1)—5—(2—(quin01iny1)cyclopropyl]thieno[3 ,2- c]pyridin-4(5H)-one; syn 3-(2-fluoropyridin—4-y1)—5-(2—(quin0liny1)cyclopropy1]thieno[3 ,2-c]pyridin- 4(5H)-one; 25 anti 3 -(rnorph01iny1)-5 [2—(quin0 linyl)cyclopr0pyl]thieno [3 ,2-c]pyridin— 4(5H)-one; syn 3-(morpholin—4-yl)-5[2-(quinolinyl)cyclopropy1]thieno[3,2-c]pyridin- one; anti 3—(pyridin—3—y1)—5—[2-(thieno[3,2-b]pyridiny1)cyc10pr0py1]thien0[3 ,2- 30 din-4(5H)—one; syn 3-(pyridin—3-y1)—5—[2—(thieno[3,2—b]pyridin—5-y1)cyclopr0pyl]thieno[3 ,2- c]pyridin-4(5H)-one; 93 anti 3-(pyridiny1)[2-(quino1in—2-y1)cyclopropyl]thieno[3 ,2-c]pyridin—4(5H)- one; syn idin—3-y1)[2-(quinolin-2—y1)cyclopropy1]thieno[3 ,2-c]pyridin—4(5H)- one; anti 3-(pyrirnidiny1)-5—[2-(quino1inyl)cyclopropy1]thieno [2,3-d]pyridazin— 4(5H)-one; syn 3-(pyrin1idin—5-y1)[2—(quino[inyl)cyclopropyl]thieno[2,3—d]pyridazin— 4(5H)-one; anti 3-(pyridinyl)[2-(thieno[3,2-b]pyridiny1)cyclopropy1]thieno[2,3- 10 d]pyridazin—4(5H)—one; syn -(pyridin—4—yl)—5—[2—(thieno[3,2—b]pyridin—5—y1)cyclopropy1]thieno[2,3- d]pyridazin—4(5H)-one; anti 3-(pyridiny1)[2—(thieno[3,2—b]pyridiny1)cyclopropyl]thieno[3 ,2- din-4(5H)-one; 15 syn 3-(pyridin—4-yl)[2-(thieno[3,2—b]pyridin—5-y1)cyclopropyl]thieno[3 ,2- c]pyridin-4(5H)-one; anti 3-(pyridiny1)[2-(quino1iny1)cyclopropy1]thieno[3 ,2-c]pyridin—4(5H)- one; syn 3-(pyridin—4-y1)[2-(quinoliny1)cyclopropyl]thieno[3 ,2-c]pyridin—4(5H)- 20 one; anti 3-(pyridiny1)[2-(quinoliny1)cyclopropy1]thieno[2,3-d]pyridazin— 4(5H)-one; syn 3-(pyridin—4-y1)[2—(quinolinyl)cyclopropyl]thieno[2,3-d]pyridazin— 4(5H)-one; 25 anti 5-(pyrimidiny1)[2-(quino lin—2-yl)cyclopropy1]isoquino lin- 1 (2H)-one; syn 5 -(pyrin1idin—5 -y1)[2-(quinolinyl)cyclopropy1]isoquinolin-1(2H)-one; anti 5 -(pyridiny1)[2-(quinolinyl)cyclopropy1]isoquinolin-1(2H)-one; syn 5 -(pyridinyl)[2-(quinolinyl)cyclopropy1]isoquinolin-1(2H)-one; anti idiny1)[2-(quinolinyl)cyclopropy1]isoquinolin-1(2H)-one; 30 syn 5-(pyridin—3—y1)—2—[2—(quinolin—2—yl)cyclopropy1]isoquinolin-1(2H)-one; anti 5 -(morpholin—4—y1)—2—(2—(quinolin—2—yl)cyclopropyl]isoquinolin- 1 (2H)-one; syn 5 -(morpho1iny1)—2—(2—(quino1in—2—yl)cyclopropyl]isoquino lin- 1 (2H)-one; 94 anti o(pyrirnidin—5—y1)—2—(2-(quino1iny1)cyclopropyl)isoquino lin- 1 (2H)-one; syn 4-fluoro(pyrirnidin—5—y1)(2-(quino liny1)cyclopropy1)isoquino lin- 1 (2H)-one; idin—4-y1) [2-(quino1iny1)ethyl]pyrido [3 ,4-d]pyridazin—1(2H)-one; 5 - idazo [1 ,2-a]pyridin—2-yl)ethyl](1-methy1—1H—imidazol—4-y1)-3 - (pyridiny1)thieno [2,3 -d]pyridazin-4(5H)—one; 5 - [2-(imidazo [1 ,2-a]pyridinyl)ethyl](1-methy1-1H-pyrazoly1)-3 -(pyridin- 4-y1)thieno [2,3 -d]pyridazin-4(5H)-one; 10 5 — [2—(imidazo [1 ,2-a]pyridin—2-yl)ethyl](pyridin-3 -y1)-3 -(pyridin—4- y1)thieno [2,3-d]pyridazin—4(5H)—one; 5 - [2-(6-chloroquino lin—2—y1)ethy1]—3—(pyridin—4—y1)thieno [3 ,2-c]pyridin-4(5H)- one; 5 - [2-(3-n1ethquuino 1iny1)ethy1]—3—(pyridin—4-yl)thieno [3 ,2-c]pyridin—4(5H)- 15 one; 5 - [2-(8-fluoroquino 1iny1)ethyl]-3—(pyridinyl)thieno [3 ,2-c]pyridin-4(5H)- one; 5 - [2-(6-fluoroquino 1iny1)ethyl]—3-(pyridin-3 -y1)thieno [3 yridin-4(5H)- one; 20 5 - [2-(6-fluoroquino liny1)ethy1]—3-(pyridinyl)thieno [3 ,2-c]pyridin-4(5H)- one; 5 - [2-(in1idazo [1 ,2-a]pyridin—2-yl)ethyl](pyridin—3 -y1)thieno [3 ,2-c]pyridin- 4(5H)-one; 3 din—4-y1)-5 - [2-(quinoxalin—2-yl)ethyl]thieno [2,3 -d]pyridazin-4(5H)-one; 25 5 - [2-(1 ,5 -naphthyridiny1)ethyl](pyridinyl)thieno [2,3 -d]pyridazin-4(5H)- one; 5 -[2-(1H-indazoly1)ethyl](pyridinyl)thieno[2,3 -d]pyridazin-4(5H)-one; 3 -(1-rnethy1-1 H-pyrazo lyl)[2-(quino liny1)ethy1]thieno [2,3-d]pyridazin- 4(5H)-one; 30 3 yrazol—3 —y1)—5 —[2—(quinolin—2—yl)ethy1]thieno[2,3 -d]pyridazin-4(5H)-one; 5 -[2-(1H-benzimidazol—1—y1)ethy1]—3—(pyridin—4—yl)thieno[2,3-d]pyridazin-4(5H)- one; 95 5 -[2-( 1 H-benzimidazo1y1)ethy1]—3-(pyridin—4-y1)thieno[2,3-d]pyridazin-4(5H)- one; 5 -[2-(6-chloroquino1iny1)ethyl](pyridiny1)thieno[2,3 -d]pyridazin-4(5H)- one; 3 -(pyridin—3 -y1ethyny1)-5 -[2-(quino1inyl)ethy1]thieno [2,3 -d]pyridazin-4(5H)- one; 3 -(pyridin—4-y1ethynyl)[2-(quino lin—2—yl)ethyl]thieno [2,3 -d]pyridazin-4(5H)- one; 5 - [2-(3 ,5 -dimethylpyridinyl)ethyl](pyridinyl)thieno [2 ,3 -d]pyridazin— 10 4(5H)—one; 5 - [2-(7-fluoroquino lin—2—y1)ethyl]—3—(pyridin—4—yl)thieno [2,3 -d]pyridazin-4(5H)- one; 5 - [2-(pyrazin—2-y1)ethyl] —3—(pyridin—4—yl)thieno [2,3 -d]pyridazin-4(5H)-one; 2- [2-(1 ,6-naphthyridiny1)ethy1]—8—(pyridin—4-y1)isoquinolin-1(2H)-one; 15 2-[2-(8-fluoroquino1iny1)ethy1]—8—(pyridin—4-y1)isoquinolin- 1 (2H)-one; 8-(pyridin—4-y1)[ 1 -(quino1iny1)propan—2-y1]isoquinolin— 1 (2H)-one; 2-[2-(3-n1ethy1quinoliny1)ethy1](pyridin—4-y1)isoquino1in- 1 (2H)-one; 2- [2-(in1idazo [1 ,2-a]pyridin—2-yl)ethy1]—8-(1 H-pyrazol—3 tha1azin-1(2H)-one; 8-(morpholin—4-y1) [2-(quinoliny1)ethy1]phtha1azin- 1 (2H)-one; 20 2-[2-(in1idazo[1,2-a]pyridin—2—yl)ethyl](1-oxa-4,9-diazaspiro[5.6]dodec y1)phtha1azin— 1 (2H)-one; 2- [2-(in1idazo [1 ,2-a]pyridin—2-yl)ethyl]-8—(2-oxaazaspiro [3 .5 ]non y1)phtha1azin— 1 (2H)-one; 8- [(3 R)hydroxypiperidiny1][2-(imidazo [1 ,2—a]pyridin 25 y1]phtha1azin— 1 ne; 8- [(3 S)-3 -hydroxypiperidin—1—yl][2—(imidazo[1 ,2-a]pyridin y1)ethy1]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-oxaazaspiro[3.3]hept y1)phtha1azin— 1 ne; 30 2- [2-(imidazo [1 yridin—2—yl)ethyl]—8—(1 ,2—oxazo1idiny1)phtha1azin- 1 (2H)- one; 96 8-(hexahydrocyclopenta[b] [1 zin—4(4aH)-y1) [2-(irnidazo [1 ,2-a]pyridin—2- y1)ethy1]phtha1azin— 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl](tetrahydrofuran—3 -y1)phtha1azin- 1 (2H)- one; 2- [2-(imidazo [1 yridin—2-yl)ethyl]-8—(3-oxaazabicyclo [3 .2. 1 ]oct y1)phtha1azin— 1 (2H)-0ne; 2-[2-(imidazo[1,2-a]pyridin—2-yl)ethyl](2-oxaazaspir0[3.4]0ct y1)phthalazin-1(2H)-one; imidazo[1 ,2-a]pyridin—2-yl)ethyl](2,2,6,6-tetrafluoromorpholin 10 ha1azin— 1 (2H)-one; 8-(4-hydroxypiperidin—1—y1)—2—[2—(imidaz0[1 ,2—a]pyridin-Z-y1)ethy1]phthalazin- 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—8—(2—methylpyrirnidin-5 -y1)phtha1azin- 1 (2H)-one; 15 8-(2-cyclopropy1pyrimidiny1)—2—[2—(imidazo [1 ,2-a]pyridin-2— y1)ethy1]phtha1azin— 1 (2H)-one; 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethy1]—8—(pyridaziny1)phtha1azin-1(2H)-0ne; 8-(5 -flu0r0pyridin—3 -y1) idazo [1 ,2-a]pyridiny1)ethy1]phtha1azin— 1 (2H)- one; 20 8- [2-(3-fluoropheny1)morpholin—4—y1][2-(imidazo [1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl](2-methoxypyrimidin-5 -y1)phtha1azin- 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—8-[2-(trifluoromethy1)pyridin 25 y1]phtha1azin—1(2H)-one; 2-[2-(imidazo[1,2-a]pyridin—2-yl)ethyl](8-oxaazabicyc10[3.2.1]oct y1)phthalazin-1(2H)-one; 2-[2-(imidazo[1 ,2-a]pyridin—2-yl)ethyl][2-(trifluoromethy1)morpho1in y1]phtha1azin—1(2H)-one; 30 8-(2,2—dimethy1morpho1in—4—yl)—2—[2—(imidazo [1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)—one; 97 8- [2-(4-ch10ropheny1)morpho1in—4—y1][2-(imidazo [1 yridin y1)ethy1]phtha1azin— 1 ne; 8- [2-(3 ,4-difluoropheny1)morph01iny1][2-(imidazo [1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)-one; 2-[2-(imidazo[1 yridin—2—yl)ethyl]—8-(piperidin—4-y1)phtha1azin-1(2H)-one; 2- [2-(imidazo [1 yridin—2—yl)ethyl]—8-(tetrahydr0—2H-pyran—4-y1)phtha1azin- 1 (2H)-one; 8-(2,6-diazabicyclo[3 .2. 1]octyl)[2-(imidazo[1 ,2-a]pyridin y1)ethy1]phtha1azin- 1(2H)-one; 10 8— [(1 8,5 S)—3 ,6—diazabicyclo[3 .2.0]heptyl][2-(imidazo [1 ,2-a]pyridin—2- y1)ethy1]phtha1azin— 1 (2H)—one; 8-(furan—2-y1) [2—(imidazo[1 ,2—a]pyridin—2—yl)ethy1]phtha1azin-1(2H)-0ne; 2- [2-(imidazo [1 ,2-a]pyridin—2—y1)ethyl]—8—(1—methy1—1H-pyrazo1y1)phtha1azin- 1 (2H)-one; 15 8-(hexahydropyrro10[3 ,4-c]pyrr01—2(1H)—yl)—2-[2-(irnidaz0[1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)-one; 8-(2,7-diazaspiro[4.4]non—2—yl)[2-(imidazo[1,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)-0ne; 8-[(1S,4S)—2,5-diazabicyc10[2.2.1]hepty1]—2-[2-(irnidazo[1,2-a]pyridin—2- 20 y1)ethy1]phtha1azin— 1 (2H)-one; 8-(2,7-diazaspiro [3 .5 ]n0n—7—y1)—2—[2-(imidazo [1 ,2-a]pyridin y1)ethy1]phtha1azin— 1 (2H)-one; 8-(2,6-diazaspiro [3 .5 ]n0n—6-y1)—2-[2—(imidazo[1 ,2-a]pyridin y1]phtha1azin— 1 (2H)-0ne; 25 8-(piperidin—4-y1)[2-(5,6,7,8-tetrahydroimidazo[1 ,2-a]pyridin—2- yl)ethy1]phtha1azin— 1 (2H)-0ne; 8- [2-(aminomethyl)chloropyrrolidinyl][2-(imidazo [1 ,2-a]pyridin—2- y1)ethy1]phtha1azin- 1(2H)-one; 2— [2—(imidazo [1 ,2—a]pyridin—2-yl)ethyl]-4,8-di(pyridiny1)phtha1azin-1(2H)-one; 30 2- idazo [1 ,2—a]pyridin—2—yl)ethyl]—8—[(3aR,4S,7R,7aS)-0ctahydro-1H—4,7- epiminoisoindo1y1]phtha1azin— 1 (2H)—one; 98 8- [5 -(4-ch10r0pheny1)-2,5-diazabicyclo[2.2. 1]hepty1] [2-(irnidazo [1 ,2- din-Z-y1)ethy1]phtha1azin- 1 (2H)—0ne; 4-br0m0[2-(imidazo [1 yridin—2-yl)ethy1] -8 -(pyridin—4-y1)phtha1azin— 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl][(3aS,8aS)-octahydropyrr010[3 ,4- c]azepin—2(1H)-y1]phtha1azin—1(2H)—0ne; 2- idazo [1 ,2-a]pyridin—2-yl)ethyl][(3aS,8aR)—0ctahydr0pyrro10[3 ,4- c]azepin-2(1H)-y1]phthalazin-1(2H)-one; tert-butyl (3 aR,4S,7R,7aS){3-[2-(imidazo[1,2-a]pyridiny1)ethy1]oxo-3,4- 10 dihydrophthalazin—S-y1}octahydro-ZH-4,7—epiminoisoindo16carboxy1ate; 8-(hexahydro—5H—furo[2,3—c]pyrrol—S—yl)—2—[2—(imidazo [1 ,2-a]pyridin—2- y1)ethy1]phtha1azin— 1 (2H)—one; 2- [2-(imidazo [1 ,2-a]pyridin—2—y1)ethyl]—8—( 1 ,2,3 ,6-tetrahydr0pyridin y1)phtha1azin— 1 (2H)-one; 15 2- [2-(irnidazo [1 ,2-a]pyridinyl)ethy1]—8—[(3 S)-tetrahydrofuran—3 - y1arnino]phtha1azin—1(2H)-one; 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethy1]—8—[(3 R)-tetrahydr0furan—3 - y1amino]phtha1azin— 1 (2H)-one; 8- {[5 -(hydr0xyrnethy1)- 1 ,4-dioxan—2-yl]methoxy} [2-(irnidazo [1 ,2-a]pyridin 20 y1]phtha1azin— 1 (2H)-one; 2- [2-(irnidazo [1 ,2-a]pyridin—2—y1)ethyl]—8-(oxetan—3 -y10xy)phtha1azin—1(2H)-0ne; 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl]—8-(pyridin—4-y1methoxy)phtha1azin— 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl]—8—(morph01iny1methy1)phtha1azin— 25 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl](pyridin-3 -y10xy)phtha1azin—1(2H)-one; 2- idazo [1 ,2-a]pyridinyl)ethyl][(oxetan-3 -y1methy1)amino]phtha1azin— 1 (2H)-one; 2— [2—(imidazo [1 ,2-a]pyridin—2-yl)ethyl](tetrahydr0-2H-pyran—4- 30 y1amino)phtha1azin— 1 (2H)—one; 2- [2-(imidazo [1 ,2—a]pyridin—2—y1)ethyl]—8—[( 1 —methy1azetidin-3 - y1)amino]phtha1azin— 1 (2H)-one; 99 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1](1 ,3 -oxazol—2-y1arnino)phtha1azin- 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl][methy1(oxetan—3 -y1)amino]phtha1azin- 1 (2H)-one; 2- idazo [1 ,2-a]pyridin—2-yl)ethyl]—8-(piperidin—4-y1amino)phtha1azin-1(2H)- one; 8-[(1-acety1piperidin—3 -y1)amino]—2-[2-(imidazo[1 ,2-a]pyridin y1)ethy1]phthalazin-1(2H)-one; 8-[(1-acety1piperidin—4-yl)amino][2-(imidazo[1 yridin 10 y1)ethy1]phtha1azin—1(2H)—one; 2- [2-(imidazo [1 ,2—a]pyridin—2—yl)ethyl]—8—(tetrahydrofuran—3 -y1amino)phtha1azin- 1 (2H)-one; 2- [2-(imidazo [1 ,2-a]pyridin—2—y1)ethy1]—8—(tetrahydro-2H-pyran—3 - y1arnino)phtha1azin— 1 (2H)-one; 15 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethy1]—8—(piperidin—3 -y1arnino)phtha1azin-1(2H)- one; 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethyl]—8— {methy1[(3 -methyloxetan-3 - y1)rnethy1]arnino } phthalazin- 1 (2H)—one; 2- [2-(irnidazo [1 yridin—2—yl)ethy1](oxetan—3 -y1arnino)phtha1azin—1(2H)- 20 one; 8- { [(3 aS ,4S ,6aS)-octahydrocyclopenta[c]pyrro1y1methy1]amino} [2-(5 ,6,7,8- tetrahydroimidazo[ 1 ,2-a]pyridin—2—y1)ethy1]phthalazin— 1 (2H)-one; 5 holin—4-y1) [2-(quinoliny1)ethyl]phtha1azin- 1 (2H)-one; 2-[2-(1H-benzimidazo1y1)ethy1](pyridiny1)phthalazin-1(2H)-one; 25 4-(pyridin—3 -y1) [2-(quino1in—2—yl)ethyl]phtha1azin- 1 (2H)-one; 5 -(1 ,4-dihydropyrimidinyl)—2-[2—(5 ,6,7,8-tetrahydroimidazo[1 ,2-a]pyridin—2- y1)ethy1]phthalazin-1(2H)-one2-[2-(imidazo[1 yridiny1)ethy1] -5 -(pyridin y1)phtha1azin-1(2H)-one; 5 —(pyridin—3 — [2—(quino [inyl)ethyl]phtha1azin- 1 (2H)-one; 30 2- [2-(imidazo [1 ,2—a]pyridin—2—yl)ethyl]—8— { [(3—methyloxetan-3 -y1)methy1]amino} - phthalazin-1(2H)-one; 4-(pyridin—4-y1) [2—(quino1in—2—y1)ethyl]pyrido [2,3 -d]pyridazin—5 (6H)-one; 100 4-(morpholin—4-yl)[2-(quinolin—2-yl)ethyl]pyrido[2,3-d]pyridazin-5(6H)-one; 4-(oxetan—3-ylamino)[2-(quinolinyl)ethyl]pyrido[2,3-d]pyridazin—5(6H)- one; 2- [2-(6-methoxypyridin—2-yl)ethyl](pyridin—4-yl)isoquino lin- l ne; 2- [2-(1 ,3 -benzothiazo l-2—yl)ethyl](pyridinyl)isoquino lin- l (2H)-one; 2-[2-(5-methylpyridin—2-y1)ethy1](pyridinyl)isoquinolin-1(2H)-one; 5-[(E)(6-methoxyquinolin—2—yl)ethenyl](pyridinyl)thieno[3,2-c]pyridin— one; 8-(pyridinyl)[(E)(quinazolinyl)ethenyl]isoquinolin-1 ne; 10 5—[(E)—2-(6—chloroquinolinyl)ethenyl](pyridinyl)thieno[3,2-c]pyridin- 4(5H)-one; 5-[(E)(3 -methquuinolin—2—yl)ethenyl]—3—(pyridin—4-yl)thieno[3,2-c]pyridin- 4(5H)-one; 8-(pyridin—4-yl) [(E)-2—(quinolin—2—yl)ethenyl]isoquino lin- l (2H)-one; 15 5 - [(E)(l zothiazolyl)ethenyl]—3—(pyridin—4-yl)thieno [3 ,2-c]pyridin- 4(5H)-one; 3-(pyridinyl)[(E)(quinolinyl)ethenyl]thieno[3,2-c]pyridin-4(5H)-one; and the omers, the N-oxides, the prodrugs, the tautomers and the hydrates thereof, and the pharmaceutically acceptable salts thereof. 20 The compounds of the invention of the general formula I and the starting materials used to prepare them can be prepared in analogy to known processes of c chemistry as are described in standard works of organic chemistry, e. g. — Weyl, "Methoden der Organischen Chemie", Thieme-Verlag, Stuttgart, Jerry March "Advanced Organic Chemistry", 5th edition, Wiley & Sons and the literature cited 25 therein, and R. Larock, "Comprehensive Organic Transformations", 2Ild edition, Weinheim, 1999 and the literature cited therein. The compounds of the invention of the general formula I are advantageously prepared by the methods described below and/or in the experimental section. nds of the formula I, wherein Q is oxygen, can be prepared e. g. by 30 reacting a compound of the formula II O R1a Het—A\ (n) T | \ R2 X§X23 X3":l wherein X2a is N or C-R7a; X3a is s, o, N=C(R8), C(R93)=C(R8) or ); Het, A, X1, R2, R6 and R8 are as defined for formulae I, 1-1 .A, I-1.B, I-2.A, I-2.B, I-1.C, I-2.C, I-1.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I-6.A, I-5.B, I-6.B, I-5.C, I-6.C, I-5.D, I-6.D, I-7.A, I-8.A, I—7.B, I—8.B, I—7.C, I—8.C, I—7.D, I—8.D, I-9.A, I-lO.A,I-9.B, 1-10.B, I- 9.C, I-lO.C, I—9.D or 1—10.D; 10 R”, R”, R921 independently of each other, are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl, trimethylsilyl, C1-C4- alkylsulfanyl, alkoxy—C1—C4-alkyl, C1-C4-alkoxy, C1-C4-alkoxy- C1-C4-alkoxy, C1-C4-alkylsulfanyl-C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, CN,NRX1RX2, NRXlez-Cl-C4- 15 alkoxy; ed that l or 2 of the ls Rla, R7a and Rga, in particular exactly one of these radicals is bromine or iodine, while the others are ent from bromine or iodine; with a nd of formula III, 20 M-Y-Cyc (111) where Y has one of the meanings given for Y1, Y2, and Y3 and Cyc has one of the meanings given herein for Cyc], Cycz, and Cyc3 and wherein 25 M is a Li, B(ORBI)(ORBZ) radical or an Sn(Rsn)3 radical, where RBl and R132 are, independently of each other, hydrogen or C1-C4-alkyl or RBl and R132 together form a C2—C6—alkandiyl moietyl, e.g. ethan-l,2-diyl, propan—l,3-diyl or l,l ,2,2-tetramethylethan—1,2—diyl, and wherein R811 is C1-C6-alkyl or C3- C6-cycloalkyl or phenyl. 102 Amongst the compounds of formula III, where Y is a chemical bond, particular ence is given to the compounds of formula 111a and, if RBl and R132 are hydrogen, the trimers thereof. 31 R —O\ /B—Y—Cyc (”13) RBZ—O The reaction ofthe nd II with the compound 111 can be performed by analogy to known coupling reactions in the presence of suitable transition metal catalysts, in particular palladium catalysts. Typical reactions conditions are those of Stille coupling (see e.g. Stille et al. Angew. Chem. Int. Ed. Engl. 1986, 25,508; J. Eluguero et al.; Synthesis 1997, 5, 563—566) or Suzuki coupling (see e.g. A. Suzuki et 10 a1, Chem. Rev. 1995, 95, 2457—2483, N. Zhe et al.; J. Med. Chem. 2005, 48 (5), 609; Young et al.; J. Med. Chem. 2004, 47 (6), 1547-1552; C. Slee et al.; Bioorg.
Med. Chem. Lett. 2001, 9, 253).
In a similar manner, compounds of the formula I, where Y1, Y2 or Y3 is NH or were Cycl-Yl, Cycz-Y2 or Cyc3-Y3 (Y1, Y2 or Y3 are single bonds) an N—bound 15 heterocycle (Y1, Y2 or Y3 are single bonds) can be prepared by reacting a compound of the formula II, as defined above, with a compound of the formula III' H-Y-Cyc (111') where Y and Cyc are as defined for formula III. The reaction of II with 111' is ably carried out in an aprotic solvent, such as dimethylsulfoxide, acetonitrile, N- 20 methylpyrrolidone, dimethylformamide, dimethylacetamide, ethyl urea, or mixtures thereof or mixtures f with halogenated hydrocarbons such as dichloromethane. The reaction is preferably carried out in the presence of a suitable base, e.g. an alkalimetal carbonate such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate or an metal alkoxide. 25 Compounds of the formula I, where Q is O, can also be prepared e. g. by reacting a compound of the formula 11a 0 1b R Het \A\ ( Ila ) N 1 | \ R2 X§ 2b X3b 103 wherein X2b is N or C-R7b; X3b is s, o, N=C(R8), C(Rgb)=C(R8) or N=C(R9); Het, A, X1, R2 and R8 are as defined for formulae I, 1-1 .A, 1-1 .B, I-2.A, I-2.B, I- l.C, I-2.C, I-l.D, I-2.D, I-3.A, I-4.A, I-3.B, I-4.B, I-3.C, I-4.C, I-3.D, I-4.D, I-5.A, I—6.A, I-5.B, I-6.B, I-5.C, I—6.C, I—5.D, I-6.D, I-7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, I—7.D, I-8.D, I—9.A, I—lO.A,I—9.B, I-lO.B, I- 9.C,I-10.C,I-9.D or 1-10.D; Rlb, R713, R9b independently of each other, are selected from the group 10 consisting of hydrogen, alkyl, trimethylsilyl, C1-C4- ulfanyl, C1-C4-alkoxy-C1-C4-alkyl, alkoxy, C1-C4-alkoxy- C1-C4—alkoxy, alkylsulfanyl—C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1—C4—fluoroalkoxy, CN,NRX1RX2, NRXlez-Cl-C4- alkoxy or a moiety M; 15 provided that l or 2 of the radicals Rlb, R7b and Rgb, in particular exactly one of these radicals is moiety M, while the others are different from M, where M is as defined for formula III and in particular a B(ORB1)(ORB2) radical; with a compound of formula IIIb, 20 Hal-Y-Cyc (IIIb) where Y and Cyc are as defined herein and wherein Hal is bromine or .
The reaction of the compound IIa with the compound IIIb can be performed by analogy tot the reaction of compound II with compound III. 25 The compounds 11, Ha, III, III', 111a and IIIb are known or can be prepared by standard methods of organic chemistry.
Compounds of the formula I, where Yl-Cycl, YZ-Cyc2 or 3 is a N-bound radical can be obtained by a coupling reaction between the compound II and the corresponding amine in the presence of a palladium catalyst in terms of a Buchwald- 30 Hartwig reaction. Suitable ium catalyst are for example tris-(dibenzylidene- acetone)dipalladium(0) (Pd2(dba)3), [1,1—bis(diphenylphosphino)ferrocene]dichloropalladium (II) (PdC12(dppf)) or palladium acetate (Pd(OAc)2). The reaction is usually 104 carried out in the presence of a bstituted)phosphine, e.g. a triarylphosphine such as triphenylphosphine, tritolylphosphine or 2,2'-bis(diphenylphosphino)-1,1'-binaphthalene (BINAP), tri(cyclo)alkylphosphine such as tris—n-butylphosphine, ertbutyl hine or tris(cyclohexylphosphine), or ohexyl-(Z',4',6'-tri-iso-propyl— biphenylyl)-phosphane (X-Phos). Usually, the reaction is performed in the presence of a base such as an alkaline alkoxide, earth alkine alkoxide, alkaline carbonate or earth ne carbonate such as or sodium tert—butoxide or cesium carbonate.
Compounds of the formula I (or likewise the compounds 11), where Q is O and A is a radical A1 can be prepared according to the following reaction schemes 1 and 2 by 10 successively reacting compounds of the formulae V or Va, respectively with a le hydroxy compound IV, in terms of a Mitsunobu reaction.
Schemel: 6 R R5 0 R1 R4 R3 O R1 Hetitem ”TiaHet HN MT |\ R2 R R3 X§X2 X3 R R X§X2 X3 (IV) (V) (I) 15 Scheme2: (IV) (Va) (II) In schemes 1 and 2, X1, X2, X3, X23, X”, R], R”, R2, R3, R4, R5, R6 and Het are as 20 defined above. Compounds of the formulae V and Va, respectively, can be prepared in analogy to known methods, e. g. as described in Natl. Symposium Vacuum Technol, Chicago, (Trans), 161-3; 1956, Journal of Organic Chemistry, , 3849-3 855; 2009, or Journal of Heterocyclic Chemistry, 8(1), 57—60; 1971. 105 Alternatively, compounds ofthe formula I (or likewise the compounds II), where Q is O and A is a radical A1 can be prepared by hydrogenation of nds of the formula I (or likewise the nds II), where A is A3.
Compounds of the formula I (or se the compounds II), where Q is O and A is a radical A3 can be prepared according to the following reaction schemes 3 and 4 by sively reacting compounds of the formulae V or Va, respectively with a suitable halocompound VI.
Scheme 3: R5 0 R1 R4 0 R1 HetM‘g‘Hal Het + HN | \ R2 —> \ 2 4 X1\ ’LYA'T‘ 3 1 l R R \XZ X R5 X§X2 X3 10 (VI) (V) (I) Scheme 4: R5 0 R13 R4 O R1a HetNyHal + HTJfiRZ —> HetQ/5 'l‘ l \ R2 R X§X23 X3a R X1§X28 X3a (VI) (Va) (II) In schemes 3 and 4, X1, X2, X3, X23, X33, R1, R13, R2, R4, R5 and Het are as defined 15 above. Hal is halogen, preferably bromine or iodine. The reaction is usually performed in the presence of a base. Suitable bases are alkali metal carbonates and hydrogen carbonates or earth metal carbonates and hydrogencarbonates such as cesium carbonate.
Compounds of the formula I (or likewise the compounds II), where Q is O and A is a radical A4 can be ed according to the following reaction schemes 5 and 6 by 20 successively reacting compounds of the formulae V or Va, respectively with a suitable halocompound VII.
Scheme 5: O 1 R Het A. 4 R5 AI R WHEN + HN \ 1, I R2 _> O 5 Het R4 X§X2 X3 R N x1\< R (VII) X2 / (V) \ (I) X3 R2 Scherne6: O R1a Het A' R4 5 R:{§.HalAl + '3‘ |\ Rz_,R5 o \ 3a R4 \X2a X Xl/N R1 a Het \\2a / (VII) (Va) (n) x 3\ X3 R2 In schemes 5 and 6, X1, X2, X3, X23, X33, R1, R”, R2, R4, R5, A’ and Het are as defined above. Hal is halogen, preferably bromine or iodine. The reaction is usually performed in the presence of a base. Suitable bases are alkali metal carbonates and hydrogen carbonates or earth metal carbonates and hydrogencarbonates such as cesium carbonate. 10 Alternatively, compounds ofthe formula I (or likewise the nds II), where Q is O and A is a radical A4 with A' being a CR3bR3c (R3b and R30 are as defined above) can be ed by cyclopropanation of compounds of the formula I (or likewise the compounds II), where A is A3, in terms of a Simmon—Smith reaction. Compounds of the formula I (or likewise the compounds 11), where Q is O and A is a radical A4 with A' 15 being a O can be prepared by epoxidation of compounds ofthe formula I (or likewise the compounds 11), where A is A3, using hydrogen peroxide.
Compounds of the formula I (or likewise the compounds 1]), where A is a l A4 can be ed in the form of enantiomers, for example as racemate, in the form of a mixture of enanantiomers, as pure enantiomers or in the form of diastereomers. 20 nds of the formula I (or likewise the compounds 11), where Q is O and A is a radical A5 can be prepared according to the following on schemes 7 and 8 by reacting compounds of the formulae V or Va, respectively with a suitable mpound VIII under basic conditions for a longer period. 107 Scheme7 3e 5 R36 R3f O R1 R R3f R Het R4 4 o R5 Het R _.
Hal + HI}! I \ R2 _> o / R36 N 1 X1 R5 3fX1/ R 3 Ht R4 \X2 x N R1 R X1] \\)(2 / \\ \ (VIII) (V) x2 /3\ x3 R2 (I) X R2 (I) A : A4 A=A5 Scheme8 5 0 RS‘a Ref Het R5 3e R16‘ flR4 R 3f R Het 4 R 0 5 R Hal + W l \ R2 _> O _> R38 R3fN 1, X1\ 5 3a R N XI’ R Het R4 \Xza X x1< R13 \\X2a / \ l (VII) (Va) (II) X2a / \ X3:11 2 x33 R2 (II) A=A4 A=A5 In schemes 7 and 8, X1, X2, X3, Xza, X3a, R1, R”, R2, R36, R“, R4, R5, and Het are as defined above. Hal is halogen, preferably bromine or iodine. The reaction is usually 10 med in the presence of a base. Suitable bases are alkali metal carbonates and hydrogen ates or earth metal carbonates and encarbonates such as cesium carbonateApart from that, compounds of the formula I and likewise compounds ofthe formula II, where Q is S can be prepared by successively reacting compounds of the formulae I and II, where Q is O with a suitable sulfurizing agent, such as Lawenson's 15 reagent or P285.
The N—oxides of compound I may be prepared from the compounds of formula 1 according to conventional oxidation methods, for e by treating said compounds with an organic d; such as metachloroperbenzoic acid or 3-chloroperbenzoic acid [Journal of Medicinal Chemistry 38(11), 1892-1903 (1995), WO 03/64572]; or with 20 inorganic oxidizing agents; such as hydrogen peroxide [cf. l of Heterocyclic Chemistry 18 (7), 1305—1308 (1981)] or oxone [cf. Journal ofthe American al Society 123(25), 5962—5973 (2001)]. The oxidation may lead to pure mono-N—oxides or to a mixture of different N—oxides, which can be separated by conventional methods; such as chromatography. 108 nds of the formula Ila can be prepared from compounds of the formula II by suitable metal-halogen exchange reactions.
The nds of the formulae III, IIIa, IV, V and Va are well known in the art or can be prepared by anology to well established reactions of organic synthetic chemistry or by analogy to the methods as bed in standard works of organic chemistry, e.g. —Weyl, "Methoden der Organischen Chemie", -Verlag, Stuttgart, Jerry March "Advanced Organic Chemistry", 5th edition, Wiley & Sons and the literature cited therein, and R. Larock, "Comprehensive Organic ormations", 2nd edition, im, 1999 and the literature cited therein. Compounds of the formula 10 VI can be prepared in analogy to known methods, e.g in terms of a Wittig reaction.
Compounds of the formula VII can be prepared in analogy to known methods. E. g., compounds of the formula VII, where A' is CR3bR3C can be prepared by cyclopropanation of compounds VI in terms of a Simmon—Smith reaction.
The reactions are usually performed in an organic t, including aprotic 15 organic solvent, e.g. substituted amides, lactames and ureas; such as dimethylformamide, dimethylacetamide, N—methylpyrrolidone, tetramethyl urea, cyclic ethers; such as dioxane, tetrahydrofurane, halogenated hydrocarbons; such as romethane, and mixtures thereof as well as mixtures thereof with C1-C6-alkanols and/or water. 20 The ons described above will be usually med at temperatures ranging from -10°C to 100°C, depending on the reactivity of the used compounds.
The reaction mixtures are worked up in a conventional way, e.g. by mixing with water, ting the phases and, where appropriate, ing the crude products by chromatography. The intermediates and final products in some cases result in the form 25 of colorless or pale brownish, viscous oils which are freed of volatiles or purified under reduced pressure and at moderately elevated temperature. If the intermediates and final products are obtained as solids, the purification can also take place by recrystallization or digestion.
Due to their capability of inhibiting PDE10A at low concentrations, the 30 compounds of the formula 1, their N—oxides, their hydrates, their tautomers and their prodrugs and the pharmaceutically acceptable salts thereof, are particularly suitable for treating disorders or ions, which can be treated by inhibition ofphosphodiesterase 109 type 10A. The terms "treating" and ment" in terms of the present invention have to be understood to e both curative treatment of the cause of a disease or disorder, the treatment of the symptoms associated with a disease or disorder, i.e. controlling the e or disorder or ameliorating the conditions or symptoms associated with a disease or disorder, and prophylactic treatment, i.e. a treatment for reducing the risk of a disease or disorder.
Neurological and psychiatric disorders or conditions which can be treated by inhibition of PDElOA, including curative treatment, control or amelioration and prophylaxis, include CNS disorders, in particular schizophrenia, depression, bipolar 10 disorders, cognitive dysfunctions associated with phrenia, cognitive ctions associated with Alzheimer's disease, Huntington's disease (Huntington chorea), anxiety and substance-related disorders, especially substance use disorder, substance tolerance conditions associated with substance withdrawal. Disorders or conditions which can be treated by inhibition of PDElOA, including curative treatment, control or amelioration 15 and laxis, also include treatment of diet induced obesity.
Thus, the invention relates to the use of compounds of formula 1, their N—oxides, their hydrates, their tautomers and their prodrugs and the pharmaceutically acceptable salts thereof, for ent of disorders or conditions, which can be treated by inhibition phodiesterase type 10A, i.e. the invention relates to the use of such compounds 20 for curative treatment of such a disease or disorder, controlling such a disease or disorder, ameliorating the ms associated with such a disease or disorder and reducing the risk for such a disease or disorder.
The present invention also relates to a method for the ent of a l disorder, selected from neurological and psychiatric disorders which can be d by 25 tion ofphosphodiesterase type 10A, said method comprising administering an effective amount of at least one compound, selected from the group of compounds of formula 1, their N-oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically acceptable salts thereof, to a mammal in need thereof.
The present ion in ular relates to: 30 o a method for treating, controlling, ameliorating or reducing the risk of schizophrenia in a mammalian; 110 o a method for treating, controlling, ameliorating or reducing the risk of cognitive disturbances associated with schizophrenia in a mammalian; o a method for treating, controlling, rating or ng the risk of sion in a mammalian; o a method for treating, controlling, ameliorating or reducing the risk of bipolar disorders in a mammalian; o a method for treating or ameliorating the symptoms associated with substance use ers in a mammalian; o a method for treating or ameliorating the symptoms associated with diet- 10 induced y in a mammalian; o a method for treating, controlling, ameliorating or reducing the risk of cognitive disturbances associated with Alzheimer's e in a mammalian; o a method for treating, lling, ameliorating or reducing the risk of behavioral symptoms in Alzheimer's disease; 15 0 a method for ng, controlling, ameliorating or reducing the risk of anxiety in a mammalian; o a method for treating, controlling, ameliorating or reducing the risk of Huntington's disease in a mammalian; which methods comprising administering an effective amount of at least one 20 compound, selected from the group of compounds of a 1, their N—oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically acceptable salts thereof, to a mammal in need thereof.
The t treated in the present methods is generally a mammal, preferably a human being, male or female, in whom tion of PDElOA is desired. The terms 25 "effective amoun " and "therapeutically effective amount" mean the amount ofthe subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. It is recognized that one skilled in the art may affect the neurological and psychiatric disorders by treating a patient presently ed with the disorders or 30 by prophylactically ng a patient afflicted with the disorders with an effective amount of the compound of the present invention. As used herein, the terms "treatment" and "treating" refer to all processes, wherein there may be a slowing, interrupting, 1 1 1 arresting, controlling, or stopping of the progression of the disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the ned ions, ularly in a patient who is predisposed to such disease or disorder. The term "composition" as used herein is intended to encompass a product comprising the specified ingredients in the specified s, as well as any product which results, directly or indirectly, from combination ofthe specified ingredients in the specified amounts. Such term in relation to pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product 10 which results, directly or indirectly, from ation, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types ofreactions or ctions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the t invention encompass any composition made by ng a compound of the present invention 15 and a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" it is meant the carrier, t or excipient must be compatible with the other ingredients of the formulation and not rious to the recipient thereof.
The terms "administration of and or "administering a" compound should be understood to mean providing a compound of the invention or a prodrug of a compound 20 ofthe invention to the individual in need of treatment.
A preferred ment ofthe present invention provides a method for treating schizophrenia, comprising: administering to a patient in need thereof an ive amount of at least one compound, selected from the group of compounds of formula 1, their N—oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically 25 acceptable salts thereof.
In another preferred ment, the present invention provides a method for treating cognitive disturbances associated with schizophrenia, comprising: administering to a t in need thereof an effective amount of at least one compound, selected from the group of compounds of formula 1, their N-oxides, their hydrates, their 30 tautomers, their prodrugs and the pharmaceutically able salts thereof At present, the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) (1994, American Psychiatric Association, Washington, DC), 1 12 provides a stic tool including schizophrenia and other tic disorders. These include: disorders having psychotic symptoms as the g feature. The term psychotic refers to delusions, prominent hallucinations, disorganized , disorganized or catatonic behavior. The disorder includes: paranoid, anized, catatonic, undifferentiated, and residual schizophrenia, schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic er, psychotic disorder due to a general l ion, substance-induced psychotic er, and psychotic disorder not otherwise specified. The skilled artisan will recognize that there are alternative nomenclatures, nosologies, and classification 10 systems for neurological and psychiatric disorders, and particular schizophrenia, and that these systems evolve with medical scientific progress. Thus, the term "schizophrenia" is intended to include like disorders that are described in other diagnostic sources.
In another preferred embodiment, the present invention provides a method for 15 treating substance-related disorders, comprising: administering to a patient in need thereof an effective amount of at least one compound, selected from the group of compounds of formula 1, their es, their es, their tautomers, their prodrugs and the pharmaceutically able salts thereof.
In another preferred embodiment, the present invention provides a method for 20 treating anxiety, comprising: administering to a patient in need thereof an effective amount of at least one compound, selected from the group of compounds of formula 1, their N—oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically acceptable salts thereof. At present, the fourth n of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) (1994, American Psychiatric Association, 25 Washington, DC), provides a diagnostic tool ing y and related disorders.
These include: panic disorder with or without agoraphobia, agoraphobia without history ofpanic disorder, specific phobia, social phobia, obsessive-compulsive er, post- tic stress disorder, acute stress disorder, generalized anxiety disorder, anxiety disorder due to a general medical condition, substance-induced anxiety disorder and 30 anxiety disorder not otherwise specified. As used herein the term "anxiety" es treatment of those anxiety disorders and related disorder as described in the DSM-IV.
The skilled artisan will recognize that there are alternative nomenclatures, nosologies, 1 13 and classification systems for neurological and psychiatric ers, and particular anxiety, and that these systems evolve with medical scientific progress. Thus, the term "anxiety" is intended to include like ers that are described in other diagnostic sources.
In another preferred embodiment, the present invention provides a method for treating depression, comprising: administering to a patient in need thereof an effective amount of at least one compound, selected from the group of compounds of formula 1, their N-oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically acceptable salts thereof. At present, the fourth edition of the Diagnostic and Statistical 10 Manual of Mental Disorders (DSM-IV) (1994, American atric Association, gton, DC), provides a diagnostic tool ing depression and d disorders. Depressive ers include, for example, single episodic or ent major sive disorders, and dysthymic disorders, depressive neurosis, and neurotic depression; holic depression including anorexia, weight loss, insomnia and early 15 morning waking, and psychomotor retardation; atypical depression (or reactive depression) including increased appetite, hypersomnia, psychomotor ion or irritability, anxiety and phobias; seasonal affective disorder; or bipolar disorders or manic depression, for example, bipolar] disorder, bipolar II disorder and cyclothymic er. As used herein the term "depression" includes treatment of those depression 20 disorders and related er as described in the DSM-lV.
In another preferred embodiment, the present invention provides a method for treating nce-related disorders, especially substance dependence, substance abuse, substance tolerance, and substance withdrawal, comprising: administering to a patient in need thereof an effective amount at least one compound, selected from the group of 25 compounds of formula 1, their N—oxides, their hydrates, their tautomers, their prodrugs and the pharmaceutically acceptable salts thereof. At present, the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) (1994, American Psychiatric Association, Washington, DC), provides a stic tool including disorders related to taking a drug of abuse ding alcohol), to the side effects of a 30 medication, and to toxin exposure. Substances e alcohol, amphetamine and similarly acting sympathomimetics, caffeine, cannabis, cocaine, hallucinogens, inhalants, nicotine, opioids, phencyclidine (PCP) or similarly acting 1 14 arylcyclohexylamines, and sedatives, hypnotics, or anxiolytics. Also, polysubstance dependence and other n substance-related disorders are included. The skilled artisan will recognize that there are alternative nomenclatures, nosologies, and classification systems for neurological and psychiatric disorders, and particular substance-related disorders, and that these systems evolve with medical scientific ss. Thus, the term "substance-related disorder" is intended to include like ers that are described in other diagnostic s.
In the treatment, prevention, control, amelioration, or reduction of risk of conditions which require inhibition of PDE10A an appropriate dosage level will 10 generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses. Preferably, the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day. A le dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 15 0.5, 0.5 to 5 or 5 to 50 mg/kg per day. For oral administration, the compositions are preferably provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be 20 treated. The compounds may be administered on a regimen of l to 4 times per day, ably once or twice per day. When ng, preventing, controlling, rating, or reducing the risk of neurological and psychiatric disorders or other diseases for which compounds of the present invention are indicated, generally satisfactory results are obtained when the compounds ofthe present invention are administered at a daily 25 dosage of from about 0.1 milligram to about 100 milligram per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. For most large s, the total daily dosage is from about 1.0 milligrams to about 1000 milligrams, preferably from about 1 milligram to about 50 milligrams, in the case of a 70 kg adult human, the total daily dose will 30 generally be from about 7 rams to about 350 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient 1 15 may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the ular condition, and the host undergoing therapy.
The compounds of the present invention may be administered by conventional routes of administration, including parenteral (e.g., intramuscular, intrapentoneal, intravenous, ICV, intracisternal injection or infiision, subcutaneous ion, or implant), oral, by inhalation spray, nasal, vaginal, rectal, sublingual, or l routes of 10 administration.
The compounds according to the present invention are further useful in a method for the prevention, treatment, control, ration, or reduction of risk of the aforementioned diseases, disorders and conditions in combination with other agents.
The nds of the present invention may be used in combination with one or 15 more other drugs in the treatment, prevention, control, amelioration, or ion of risk of diseases or conditions for which compounds of Formula I or the other drugs may have utility, where the combination ofthe drugs together are safer or more effective than either drug alone. Such other drug(s) may be administered, by a route and in an amount commonly used therefore, poraneously or tially with a compound 20 of Formula I. When a compound of formula I is used contemporaneously with one or more other drugs, a ceutical composition in unit dosage form containing such other drugs and the compound of formula I is preferred. However, the combination therapy may also include therapies in which the nd of formula I and one or more other drugs are administered on different overlapping schedules. It is also contemplated 25 that when used in combination with one or more other active ingredients, the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly. Accordingly, the ceutical compositions of the present invention include those that contain one or more other active ingredients, in on to a compound of a I. The above combinations 30 include combinations of a compound ofthe present invention not only with one other active compound, but also with two or more other active compounds. 1 16 Likewise, compounds of the present invention may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction ofrisk of the es or conditions for which compounds of the present invention are useful. Such other drugs may be stered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the present invention. When a compound ofthe t invention is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of the present ion is preferred. Accordingly, the pharmaceutical compositions ofthe present invention include those that also n 10 one or more other active ingredients, in addition to a compound of the present invention.
The weight ratio of the compound of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a 15 compound of the present invention is combined with another agent, the weight ratio of the compound of the present invention to the other agent will generally range from about 1000: l to about , preferably about 200:1 to about 1:200. Combinations of a nd of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an ive dose of each active 20 ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in ction. In addition, the administration of one element may be prior to, rent to, or subsequent to the stration of other agent(s).
The present invention also relates to pharmaceutical compositions (i.e. 25 medicaments) which comprise at least one compound of the present invention and, where appropriate, one or more suitable excipients.
These excipients/drug carriers are chosen according to the pharmaceutical form and the desired mode of administration.
The compounds of the present invention can be used to manufacture 30 pharmaceutical compositions for parenteral (e.g., intramuscular, intrapentoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), oral, sublingual, intratracheal, intranasal, l, transdermal, vaginal or 1 17 rectal administration, and be administered to animals or humans in unit dose forms, mixed with conventional pharmaceutical carriers, for the prophylaxis or treatment of the above impairments or diseases.
In the pharmaceutical compositions, the at least one compound of the present invention may be formulated alone or together with fiarther active compounds, in suitable dosage unit formulations containing conventional ents, which generally are non-toxic and/or pharmaceutically acceptable. Carriers or excipients can be solid, semisolid or liquid materials which serve as vehicles, carriers or medium for the active compound. le excipients are listed in the specialist medicinal monographs. In 10 addition, the formulations can se pharmaceutically acceptable carriers or ary auxiliary substances, such as glidants; wetting ; emulsifying and suspending agents; preservatives; antioxidants; antiirritants; chelating agents; coating auxiliaries; emulsion stabilizers; film formers; gel s; odor g agents; taste corrigents; resin; hydrocolloids; solvents; solubilizers; neutralizing ; diffusion 15 accelerators; pigments; quaternary ammonium compounds; refatting and overfatting agents; raw materials for ointments, creams or oils; silicone derivatives; spreading auxiliaries; stabilizers; ants; suppository bases; tablet auxiliaries, such as binders, fillers, glidants, disintegrants or coatings; propellants; drying agents; opacifiers; thickeners; waxes; plasticizers and white mineral oils. A formulation in this regard is 20 based on specialist knowledge as described, for example, in r, H. P., Lexikon der Hilfsstoffe fur Pharmazie, Kosmetik und angrenzende Gebiete lopedia of auxiliary substances for pharmacy, cosmetics and related fields], 4th edition, Aulendorf: ECV-Editio-Kantor-Verlag, 1996.
Suitable unit dose forms include forms for oral administration, such as tablets, 25 gelatin capsules, powders, granules and solutions or suspensions for oral , forms for sublingual, buccal, intratracheal or intranasal administration, aerosols, implants, forms of subcutaneous, intramuscular or intravenous administration and forms of rectal administration.
The compounds of the invention can be used in creams, nts or s for 30 topical administration. 1 18 If a solid composition is ed in the form of tablets, the main ingredient is mixed with a pharmaceutical carrier such as gelatin, starch, lactose, magnesium te, talc, silicon dioxide or the like.
The s may be coated with sucrose, a cellulose derivative or another suitable substance or be treated otherwise in order to display a prolonged or delayed ty and in order to release a predetermined amount of the active basic ingredient continuously.
A preparation in the form of gelatin capsules is obtained by mixing the active ingredient with an extender and taking up the resulting mixture in soft or hard gelatin capsules. 10 A preparation in the form of a syrup or elixir or for administration in the form of drops may comprise active ingredients together with a sweetener, which is preferably calorie-free, methylparaben or propylparaben as antiseptics, a flavoring and a suitable coloring.
The water-dispersible powders or granules may comprise the active ingredients 15 mixed with dispersants, wetting agents or suspending agents such as polyvinylpyrrolidones, and ners or taste improvers.
Rectal administration is achieved by the use of suppositories which are prepared with binders which melt at the rectal temperature, for example cocobutter or polyethylene s. Parenteral administration is effected by using aqueous 20 sions, isotonic salt solutions or sterile and injectable solutions which comprise pharmacologically suitable dispersants and/or wetting , for example propylene glycol or polyethylene glycol.
The active basic ingredient may also be formulated as microcapsules or mes/centrosomes, if suitable with one or more carriers or additives. 25 In addition to the compounds of the general formula I, their prodrugs, their N- , their tautorners, their hydrates or their pharmaceutically suitable salts, the compositions ofthe invention may comprise further active basic ingredients which may be beneficial for the ent of the impairments or diseases indicated above.
The present invention thus further relates to pharmaceutical compositions in 30 which a plurality of active basic ingredients are present together, where at least one thereof is a compound of the invention. 1 19 When producing the pharmaceutical compositions, the compounds according to the invention are optionally mixed or diluted with one or more rs.
The compounds of the invention also include those compounds in which one or more atoms have been replaced by their , non-radioactive es, for example, a hydrogen atom by deuterium.
Stable isotopes (e.g., deuterium, 13’C, 15 N, 18O) are nonradioactive isotopes which contain one additional neutron than the ly abundant isotope of the respective atom. Deuterated compounds have been used in ceutical research to investigate the in vivo metabolic fate of the compounds by evaluation of the mechanism of action 10 and metabolic pathway of the non deuterated parent compound (Blake et al. J. Pharm.
Sci. 64, 3, 367-391 (1975)). Such metabolic studies are important in the design of safe, effective therapeutic drugs, either because the in vivo active compound administered to the patient or because the metabolites ed from the parent compound prove to be toxic or carcinogenic (Foster et al., Advances in Drug ch Vol. 14, pp. 2-36, 15 Academic press, London, 1985; Kato et al., J. Labelled Comp. Radiopharmaceut., :927-932 (1995); Kushner et al., Can. J. Physiol. Pharmacol., 77, 79-88 (1999).
Incorporation of a heavy atom particularly substitution of deuterium for en, can give rise to an isotope effect that could alter the pharmacokinetics of the drug. This effect is usually insignificant if the label is placed at a metabolically inert position of the 20 molecule.
Stable isotope labeling of a drug can alter its physico-chemical properties such as pKa and lipid solubility. These changes may influence the fate of the drug at different steps along its passage through the body. Absorption, distribution, metabolism or excretion can be changed. Absorption and distribution are processes that depend 25 primarily on the molecular size and the lipophilicity ofthe substance. These effects and alterations can affect the pharmacodynamic response of the drug molecule if the isotopic substitution affects a region involved in a -receptor interaction.
Drug metabolism can give rise to large isotopic effect if the ng of a chemical bond to a deuterium atom is the rate limiting step in the process. While some 30 ofthe physical properties of a stable isotope—labeled le are different from those ofthe unlabeled one, the chemical and biological properties are the same, with one important exception: because of the increased mass of the heavy isotope, any bond 120 ing the heavy isotope and another atom will be stronger than the same bond between the light isotope and that atom. In any reaction in which the breaking of this bond is the rate limiting step, the on will proceed slower for the molecule with the heavy isotope due to "kinetic isotope effect". A reaction involving breaking a C--D bond can be up to 700 percent slower than a similar reaction involving breaking a C--H bond. If the C--D bond is not involved in any of the steps leading to the metabolite, there may not be any effect to alter the behavior of the drug. If a deuterium is placed at a site involved in the metabolism of a drug, an isotope effect will be observed only if ng of the C--D bond is the rate limiting step. There is evidence to t that 10 whenever cleavage of an aliphatic C--H bond occurs, usually by oxidation catalyzed by a mixed-function oxidase, replacement of the en by deuterium will lead to able e effect. It is also important to understand that the incorporation of deuterium at the site of metabolism slows its rate to the point where another metabolite produced by attack at a carbon atom not substituted by ium becomes the major 15 pathway a process called "metabolic switching".
Deuterium tracers, such as deuterium—labeled drugs and doses, in some cases edly, of thousands of milligrams of deuterated water, are also used in healthy humans of all ages, including neonates and pregnant women, without reported incident (e.g. Pons G and Rey E, rics 1999 104: 633; Coward W A et al., Lancet 1979 7: 20 13; Schwarcz H P, Control. Clin. Trials 1984 5(4 Suppl): 573; Rodewald L E et al., J.
Pediatr. 1989 114: 885; Butte N F et al. Br. J. Nutr. 1991 65: 3; MacLennan A H et al.
Am. J. Obstet l. 1981 139: 948). Thus, it is clear that any deuterium released, for instance, during the metabolism of compounds of this invention poses no health risk.
The weight percentage of hydrogen in a mammal (approximately 9%) and natural 25 abundance of deuterium (approximately 0.015%) indicates that a 70 kg human normally contains nearly a gram of deuterium. Furthermore, replacement of up to about 15% of normal hydrogen with ium has been effected and maintained for a period of days to weeks in mammals, including rodents and dogs, with minimal observed adverse effects (Czajka D M and Finkel A J, Ann. NY. Acad. Sci. 1960 84: 770; Thomson J F, 30 Ann. New York Acad. Sci 1960 84: 736; Czakja D M eta1., Am. J. l. 1961 201: 357). Higher deuterium concentrations, usually in excess of 20%, can be toxic in animals. However, acute replacement of as high as 15%-23% of the hydrogen in 121 humans' fluids with deuterium was found not to cause toxicity j evic N et al. in "Dosimetry & Treatment Planning for Neutron Capture Therapy", Zamenhof R, Solares G and Harling O Eds. 1994. Advanced Medical Publishing, Madison Wis. pp. 125-134; es Metab. 23: 251 (1997)).
Increasing the amount of deuterium present in a compound above its natural abundance is called ment or deuterium-enrichment. Examples of the amount of enrichment include from about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 16, 21, 25, 29, 33, 37, 42, 46, 50, 54, 58, 63, 67, 71, 75, 79, 84, 88, 92, 96, to about 100 mol %.
The hydrogens present on a particular c nd have different ties 10 for ge with ium. Certain hydrogen atoms are easily exchangeable under physiological conditions and, if replaced by deuterium atoms, it is expected that they will readily exchange for protons after administration to a patient. Certain hydrogen atoms may be exchanged for deuterium atoms by the action of a deuteric acid such as D2S04/D20. Alternatively, deuterium atoms may be incorporated in various 15 combinations during the synthesis of compounds ofthe invention. Certain hydrogen atoms are not easily exchangeable for deuterium atoms. However, deuterium atoms at the remaining positions may be incorporated by the use of deuterated ng materials or intermediates during the construction of compounds of the invention.
Deuterated and deuterium-enriched compounds of the invention can be prepared 20 by using known methods described in the literature. Such methods can be carried out utilizing corresponding deuterated and optionally, other isotope-containing reagents and/or intermediates to synthesize the compounds delineated herein, or invoking standard synthetic protocols known in the art for introducing ic atoms to a chemical structure. Relevant procedures and intermediates are disclosed, for instance in 25 Lizondo, J et al., Drugs Fur, , 1116 (1996); Brickner, S J et al., JMed Chem, 39(3), 673 (1996); Mallesham, B et al., Org Lett, 5(7), 963 (2003); PCT publications W01997010223, WO2005099353, W01995007271, W02006008754; US Patent Nos. 7538189; 7534814; 7531685; 7528131; 1; 7514068; 3; and US Patent Application Publication Nos. 20090137457; 20090131485; 20090131363; 30 20090118238; 20090111840; 20090105338; 05307; 20090105147; 20090093422; 88416; 20090082471, the methods are hereby incorporated by reference. 122 The following examples are intended for further illustration of the present invention.
Abbreviations which have been used in the descriptions of the s and the Examples that follow are: BINAP for 2,2'—bis (diphenylphosphino)-l,l'—binaphthyl; DCM for dichloromethane; DEAD for diethyl azodicarboxylate; DIAD for diisopropyl azodicarboxylate; DMF for dimethylformamide; EA for ethyl acetate; EDCI l-ethyl (3-dimethylaminopropyl)carbodiimide; Et for ethyl; EX. for EXAMPLE; HMPA for hexamethylphosphoramide; HOBT for hydroxybenzotriazole; i-Pr for isopropyl; LDA for lithium diisopropylamide; MeOH for methanol; PE for petroleum ether; Pd2(dba)3 10 for tris (dibenzylideneacetone)dipalladium(0); PdClz(dppf) for 1,1'-bis (diphenylphosphino)ferrocene—palladium (ID—dichloride; Rt for retension time; TEA for triethylamine; THF for ydrofuran; TMEDA for N,N,N’,N’-tetramethyl 1,2- ethanediamine; TMSCl for hylsilyl chloride. 15 LC-MS measurements were run on Agilent 1200 HPLC/6100 SQ .
The compounds I of the invention were d in some cases by preparative HPLC. The compounds I then result as the salts.
Preparation Examples 20 1. Preparation of intermediates The starting materials used in the examples are either cially available or can be synthesized by the average skilled person trained in organic chemistry following e laboratory practice as outlined, for example in the examples below. 25 a) Preparation of compounds of the general formula Het—Al—OH a1) 2-Quinolinyl-ethanol \ / N OH 30 al . l) Quinolin—2-yl-acetic acid ethyl ester 123 To a suspension ofvacuum dried Zn dust (6.0 g, 93.8 mmol) in dry THF (100 mL) was added TMSCl (0.5 mL) dropwise over 5 min under N2 atmosphere and under stirring. The e was d for 30 min and warmed to 45°C. Ethyl bromoacetate (5.2 mL, 46.9 mmol) was added dropwise Via a e. After addition, the mixture was stirred at the same temperature for 1 h. After sedation at room temperature for 2 h, a clear orange solution was formed. The orange solution (50 mL) was carefully sucked into a syringe through a long needle and added to a mixture of 2—bromoquinoline (2.0 g, 9.6 mmol) and PdC12(dppf) (200 mg, 0.27 mmol) in a three-neck flask. The mixture was refluxed under N2 for 3 h. The reaction was red with LC-MS. Ethyl acetate (200 10 mL) was added to dilute the mixture and water (50 mL) was added to quench the reaction. The mixture was filtered through a celite pad. The ion was partitioned n brine and ethyl acetate. The organic layer was ted, washed with brine (100 mL), dried over sodium sulfate and concentrated. The residue was purified with silica column (PE/EA=3: l) to give the title compound as orange oil (1.0 g, 48%). LC- 15 MS (ESI+): m/e 216 (M+H)+, R: 0.62 min. al .2) 2-Quinolinyl-ethanol To a cold (0 0C) solution of the compound from Example a1 . la (10 g, 45 mmol) in THE (200 mL) was added LiAlH4 (2.65 mg, 70 mmol) in small portions over a period 20 of 5 min. The resulting mixture was stirred for 1h. Water was added dropwise very slowly. Then more water and EA were added. The organic phase was ted, dried and concentrated. The residue was purified by silica gel chromatography (PE/EA=2: l) to give the title compound as a yellow solid (2.5 g, 30%). LC-MS (ESI+): m/e 174 (M+H)+, R: 0.75 min. 25 a2) 2-(6-Fluoroquinolinyl)-ethanol Fm/N OH 6-Fluoro—2-methquuinoline (1.00 g, 6.20 mmol) and sodium hydroxide were each added sequentially to the mixture ofHCHO in water. Then 2 mL ofEtOH were added 30 to the mixture. The resulting solution was stirred at about 85 °C overnight. The organic layer was extracted with EA (3 x 10 mL), collected and dried with anhydrous Na2804, 124 d and concentrated to afford a pink oil. The crude material was purified by chromatography on silica-gel (eluent: PE/EA=6/1) and then r purified by combi- fiash chromatography (NH4HC03/Hgo, MeOH/HZO=40%~60%) to give 360 mg of the title t (yield: 30.3%).
LC-MS: m/e (M+H): 192.7, R: 1.63 min. a3) 2-Thieno[3,2-b]pyridin-5—yl—ethanol NH2 NH2 ””2 N 000“ / / N COOCH | 213.1} (TL 213.2) I L3)- qj/ 3 a3.4) qj/ S S 8 S / o 0 / OH s H 1 o 4 5 ‘CH3 2 3 10 OH CN Cl N N 5 fl / \ OH I a3.6) / \ a3.8) N\ l a3.7) N\ / I / / I 3 / O s / / s s 6 7 8 9 N\ 9 fl / O‘CHS N\ OH I a3.10) —> / O I / 3 / s 10 11 a3.l) Compound (2) 15 To a suspension of LiAlH4 (1.39 g, 36.58 mmol) in anhydrous THF (30 mL) was added a solution of methyl 3-aminothiophene 2-carboxylate (compound 1, 5.00 g, 31.81 mmol) dropwise at 0 0C. The reaction mixture was stirred at room temperature overnight. Water (4 mL) was added dropwise to quench the reaction. The mixture was stirred for 30 min. and then more water was added (10 mL). The solid was filtered off 20 and then washed with NaOH solution (50 mL, 5 N). The filtrate was concentrated in vacuo and the residue was dissolved in EtOAc (200 mL). The solution was dried over NaZSO4, d and concentrated in vacuo. The crude solid was used in the next step without fiirther purification (2.71 g, yield 66%). LC—MS (ESI+): m/e 130 (M+H)+, R: 1.57 min. 25 125 a3.2) Compound (3) A mixture of compound 2 (8.14 g, 63.00 mmol) and MnOz (32.8 g, 0.378 mol) in EtOAc (100 mL) was stirred at 30°C for 48 h. The mixture was filtered and the filtrate was concentrated in vacuo. The residue was used in the next step without further purification (6.82 g, yield 85%). LC—MS (ESI+): m/e 128 (M+H)+, R: 1.55 min. a1.3) Compound (4) To a solution of compound 3 (6.82 g, 53.62 mmol) in EtOH (70 mL) was added a e of pyruvic acid (9.44 g, 0.107 mol) and NaOH (10.7 g, 0.268 mol) in H20 (70 10 mL) in one portion. The mixture was heated at 60 °C for 2 h, then cooled and extracted with EtzO/EtOAc (l: l, 30 mL). The aqueous layer was acidified with HCl (2 N) to pH = 3 at 0°C and the water was removed under d pressure. The residue was co- evaporated with toluene (50 mL><3) and then used in the next step without further purification. LC-MS (ESI+): m/e 180 (M+H)+, Rt: 1.50 min. 15 a3.4) Compound (5) To a mixture of crude nd 4 (7 g, 39 mmol) in methanol (60 mL) was added thionyl chloride (10 mL) se at 0°C. The reaction mixture was then heated at 65 °C for 3 h. The excess of solvent was removed under reduced pressure. The 20 residue was diluted with EtOAc (100 mL) and washed with ted NaHC03 aqueous solution (30 mL><4) and brine (30 mL). The organic layer was dried over Na2S04, filtered and concentrated in vacuo. The residue was purified on a silica column (PE/EtOAc = 5: 1, V/V) to afford the title product as an off-white solid (715 mg, total yield 9.5%). LC-MS (ESI+): m/e 194 (M+H)+, R,: 1.78 min. 25 a3.5) Compound (6) To a on of compound 5 (100 mg, 0.52 mmol) in THF (2 mL) was added LiBH4 (11 mg) in one portion. The mixture was allowed to stir at room temperature overnight. The reaction was quenched with saturated NH4C1 solution, and then extracted 30 with EtOAc (20 mL). The organic layer was washed with brine (10 mL), dried over Na2S04, d and concentrated in vacuo. The yellow residue was used in the next step without fiirther purification. LC—MS : m/e 166 (M+H)+, R: 1.44 min. 126 a3.6) Compound (7) A e of compound 6 (100 mg, crude) and thionyl chloride (1 mL) in DCM (3 mL) was stirred at room temperature for 3 h. The mixture was trated in vacuo.
The residue was diluted with EtOAc (20 mL) and washed with saturated NaHC03 solution (6 mLX4) and brine (6 mL). The organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The red residue was used in the next step without further ation. LC-MS (ESI+): m/e 184 (M+H)+, Rt: 1.88 min. 10 a3.7) Compound (8) A mixture of compound 7 (580 mg, 3.157 mmol) and NaCN (170 mg, 3.473 mmol) in EtOH (12 mL) and H20 (4 mL) was stirred at 50 0C for 60 h. The mixture was diluted with EtOAc (50 mL) and washed with brine (15 mL><4). The organic layer was dried over , filtered and concentrated in vacuo. The e was purified on a 15 silica column (PE/EtOAc = 10: 1, v/v) to afford the title product as an off-white solid (280 mg, yield 51%). LC-MS (ESI+): m/e 175 (M+H)+, Rt: 1.89 min. a3.8) Compound (9) A mixture of compound 8 (200 mg, 1.148 mmol), sodium hydroxide (459 mg, 20 11.48 mmol) in EtOH (4 mL) and water (0.5 mL) was stirred at 700C overnight. The solvent was concentrated, the residue was adjusted to pH=3. The solution was concentrated. The t was used in the next step without r purification LC-MS (ESI+): m/e 194 (M+H)+, Rt: 1.16 min 25 a3.9) Compound (10) To a mixture of 2-(thieno[3,2-b]pyridinyl)acetic acid from Example a3.8 (77 mg, 0.399 mmol) in MeOH (5 mL) was added S002 (0.5 mL, 6.85 mmol) dropwise at 0 0C. The reaction mixture was d at room temperature for 5h. The excess of solvent was removed under reduced pressure. The residue was diluted with EtOAc (100 mL) 30 and washed with saturated NaHC03 aqueous solution (5 mL><2) and brine (30 mL). The organic layer was dried over NaZSO4, filtered and concentrated in vacuo. The residue 127 was purified by Pre-TLC (PE/EtOAc = 2:1, v/v) to afford the title product as an oil (55 mg, yield 65%).
LC-MS (ESI+): m/e 208 (M+H)+, Rt: 1.69 min. a3. 10) 2-Thieno [3 ,2-b]pyridin—5-y1—ethanol To a on of methyl 2—(thieno[3,2-b]pyridinyl)acetate (70 mg, 0.338 mmol) from Example a3.9) in THF (4 mL) was added LiBH4 (8.09 mg, 0.372 mmol) in one portion. The mixture was allowed to stir at room temperature overnight. The reaction was quenched with saturated NH4Cl solution, and then extracted with EtOAc (3>< 10 10 mL). The organic layer was washed with brine (10 mL), dried over NaZSO4, filtered and concentrated in vacuo. The yellow residue was used in the next step without further purification.
LC-MS (ESI+): rn/e 180 (M+H)+, Rt: 157 min 15 a4) 2-(Imidazo[ 1 ,2-a]pyridinyl)ethanol a4. 1) Ethyl 2-(imidazo[1,2-a]pyridin—2-yl)acetate A mixture of ethyl 4-chloro-3—oxobutanoate (15 g, 91 mmol) and pyridinamine (8.58 g, 91 mmol) in THF (80 mL) was refluxed overnight. Then, the reaction mixture 20 was concentrated and the residue purified by silica gel chromatography =1 :1) to afford ethyl 2-(imidazo[1,2-a] pyridine—2-y1)acetate (5 g, yield 26.9 %).
LC-MS (ESI+): m/e 205 (M+H)+, Rt: 0.54 min. a4.2) 2-(Imidazo[1 ,2-a]pyridinyl)ethanol 25 To a solution of ethyl 2-(imidazo[1,2-a]pyridinyl)acetate (3.1 g, 15.18 mmol) in THF (30 mL), LiBH4 (0.661 g, 30.4 mmol) was added and the mixture was stirred at room ature overnight. Saturated NH4C1 was added dropwise to quench the reaction. The solution was extracted with EA (3X 100 mL). The organic phase was collected, dried over , filtered and trated. The title compound was used 30 t fiirther purification in the next step.
LC-MS (ESI+): rn/e 163 (M+H)+, Rt: 135 min 128 a5) 2-(7-Fluoroimidazo[1 ,2-a]pyridin-2—y1)ethanol a5. 1) 4-Fluoropyridin—2-amine A mixture ofN—(diphenylmethylene)—4-fluoropyridin—Z—amine (2.0 g, 7.24 mmol) in THF (50 mL) and 1N HCl (aq) (50 mL) was stirred at about room ature overnight. The aqueous layer was adjusted to pH>10 with 5N NaOH. The aqueous layer was extracted with ethyl acetate (3x50 mL), concentrated. The mixture was purified by column chromatography (PE/EA=4/1) to give the title compound (300 mg, yield: 37%). 10 LC-MS: m/e 113 (M+H); 6 min. lH NMR(CDC13) : 4.62 (s, 2H), 6.16 (dd, J=10.8 Hz, 2.4, 1H) 6.38-6.42 (m, 1H), 8.00 (dd, J=9.2 Hz, 6 Hz, 1H). a5 .2) Ethyl 2-(7-fluoroimidazo[1,2-a]pyridin—2—yl)acetate A mixture of 4-fluoropyridinamine (1.0 g, 8.92 mmol) and ethyl 4-chloro 15 oxobutanoate (1.46 mL, 10.7 mmol) in THF (20 mL) was stirred at about reflux overnight. The residue was concentrated and was chromatographed on the C18 ISCO Combiflash system using the following gradient: A: Water (0.1% NH4HC03); B: Methanol; 10%B to 60% B over 20 min to give 350 mg product as brown oil (350 mg, yield: 17.6%). 20 LC-MS: m/e 223 (M+H); Rt:1.62 min; 1H NMR (CDC13): 1.29 (t, 3H), 3.84 (s, 2H), 4.21 (q, 2H), 7.66-7.69 (m, 1H), 7.19 (dd, J=8.2, 2.4 Hz, 1H), 7.56 (s, 1H), 8.01- 8.04 (m, 1H). a5 .3) 2-(7-Fluoroimidazo[1,2-a]pyridinyl)ethanol 25 LiAlH4 (0.171 g, 4.50 mmol) was added to the solution of ethyl 2-(7- fluoroimidazo[1,2-a]pyridinyl)acetate (0.5 g, 2.25 mmol) in THF (15 mL) and the reaction was stirred for about 2 h. The reaction was quenched with H20 and concentrated. The residue was d with methanol (3 mL) and filtered. The e was purified by C18 ISCO Combiflash system using the following gradient: A: Water 30 (0.1% NH4HC03); B: ol; 10%B to 50%B over 20 min (100 mg, yield:24.6%). 129 a6) 2-(5,6,7,8-Tetrahydroquinolin—2—yl)ethanol \ l / N OH a6. 1) Ethyl 2-(5 ,6,7,8-tetrahydroquinolinyl)acetate A solution of 7.56 g (74.7 mmol) of diisopropylamine in 100 mL of water-free THF was cooled to -30°C and n—BuLi (31.9 g, 15% solution in n-hexane, 74.7 mrnol mmol) was added via a syringe. The mixture was allowed to stir for 30 min. After cooling to -70°C, 8.68 g (74.7 mmol) tetramethylethylenediamine in 20mL ofTHF were added and the mixture was allowed at —70°C for 1 h. Then, 5 g (34.00 mmol) of 2- -5,6,7,8-tetrahydroquinoline and 3.87 g (35,7 mmol) of ethyl carbonochloridate 10 were added. The mixture was d to warm to room temperature within 2 h while being stirring. The reaction mixture was poured onto conc. cold aqueous um chloride solution and extracted with EA (3 times). The combined organic phase was once extracted with a saturated um de solution, washed with a saturated sodium bicarbonate solution and dried (magnesium sulfate). After removal of the 15 solvent, the crude product was purified by flash column chromatography (eluent: heptane/EA 1/2) to provide 2.16 g (29%) of the title compound). a6.2) 2-(5,6,7,8-Tetrahydroquinolin—2—yl)ethanol Under N2, 4.56 mL (4.56 mmol) of lithium aluminium hydride (1M in THF) was 20 cooled to 0°C and ethyl 2-(5,6,7,8-tetrahydroquinolin—2-yl)acetate (1 g, 4.56 mmol) dissolved in a small quantity ofTHF was slowly added. The e was allowed to stir for 1.5 h. To the reaction mixture 173 mg ofH20 in THF,173 mg of 10%ige NaOH and 3x 173 mg ofH20 were added. The mixture was then stirred for 30 min. ium sulfate was added. After removal of the solvent, the crude product (0.78 g, 96%) was 25 purified by chromatography (eluent: EA/heptane 3/ 1) to give the title compound. a7) 2-( l H—benzo [d]imidazo l—2—yl)ethanol H I 61W“ 130 Commercially available from Sigma-Aldrich. a8) 1-(quinolinyl)propan—2-ol \ CH3 / N OH 5 GAS-No.2 1565388, commercially available from e (Ukraine, Kiew, order number EN300-95420. a9) 2—(8—fluoroquinolin—2—yl)ethanol \ / N OH F 10 To a solution of formaldehyde and paraformaldehyde (0.187 g, 6.20 mmol) in 2 mL ofwater was added -fluoromethquuinoline (1 g, 6.20 mmol) and then sodium ide (0.37 g, 9.31 mmol) dissolved in 2 mL of water. Then 2-3 mL of l were added. The mixture ws stirred at 85°C for 48 hours. After cooling to room temperature, the reaction mixture was poured onto ice-water. The on mixture was 15 mixed with DCM and water. The organic phase was washed with water, dried and concentrated. The residue was purified by chromatography (eluent: DCM/methanol) to give 0.316 g (26.6%) of the title compound as bright yellow oily compound. a10) 2-(7-fluoroquinolinyl)ethanol m/ F N OH 20 a10. 1) tert—butyl 2—(7—fluoroquinolin—2—yl) acetate To a solution of diisopropylamine (0.753 g, 7.45 mmol) in THF was added at -78°C l lithium (0.437 g, 6.82 mmol, 2.5 molar) over a period of 15 min. Then 25 the mixture was stirred for further 30 min at -78°C. A solution of 7-fluoro 131 methquuinoline (1 g, 6.20 mmol) in 2 mL ofTHF was added at -78°C. After stirring for a filrther hour solution of di—tert-butyl dicarbonate (1.49 g, 6.82 mmol) in 1 mL of THF was added. The reaction mixture was allowed to warm up to room temperature over a period of 2 h. The reaction mixture was mixed with water and EA. The aqueous phase was extracted with EA. The combined organic phases were dried and concentrated to give the title compound (790 mg, 48.7%) as red oil. a10.2) 2-(7-fluoroquino linyl)ethanol To 3.03 mL of lithiumaluminiumhydride in THF (1 molar, 115 mg, 3.03 mmol) 10 was added the compound of example a10.1 at 0°C. Then the mixture was stirred for 2 h at 0°C and then stirred at room temperature for 20 h. The reaction e was mixed with water and 10% by weight aq. NaOH solution. The mixture was stirred for 30 min, dried and concentrated to give 250 mg of the title compound (43.2%). 15 a1 1) 2-(1 ,6-naphthyridinyl)ethanol N OH The title compound was ed in analogy to the method described in example a9). 20 a12) 2-(1 ,5-naphthyridinyl)ethanol /N \ \ / N OH The title compound was prepared in analogy to the method described in example a9). 25 a1 3) 2-( l o[d]imidazo le—2—yl)ethanol 132 (INM/OHN | H cially available from Matrix Scientific, catalogue number 27653 al4) 2-(1H-benzo[d]imidazol—1 hanol Commercially available from Matrix ific, catalogue number 054768. b) Preparation of compounds ofthe general formula Het-Al-OS(O)2CF3 10 bl) Trifluoromethanesulfonic acid 2,2-difluoro—2-quinolin—2-yl ethyl ester \ O / II N F F O’beF3 bl.l) Difluoroquinolin—2-yl acetic acid ethyl ester 2-Bromoquinoline (5.0 g, 24.0 mmol), ethyl 2-bromodifluoroacetate (5.8 g, 28.8 mmol) and copper powder (3.5 g, 55.2 mmol) in DMSO (20 mL) were stirred at 55°C 15 for 5 hours. The solid was filtered off, water (100 mL) and EA (150 mL) were added.
The organic layer was separated, dried over sodium sulfate and concentrated to give the title compound as a yellow oil (4.2 g, 70%), which was used in the next step without further purification. LC-MS (ESI+): m/e 252 (M+H)+, R: 0.93 min. 20 b1.2) 2,2-Difluoroquinolinylethanol To a solution of difluoroquinolin—Z—yl acetic acid ethyl ester (2 g, 7.9 mmol) in ethanol (20 mL) was added NaBH4 (317 mg, 1.0 mmol) at 0°C under N2. The mixture was stirred for 1 hour and then at room ature for 1.5 hours. The solution was quenched with dilute HCl (0.1 N, 20 mL). The mixture was neutralized with saturated 133 NaHC03 solution and extracted with EtOAc (3* 100 mL). The combined organic layer was dried over sodium sulfate. The solvent was evaporated and the residue was purified by column chromatography on silica gel (PE: EA=10: l) to give the title compound as a yellow solid (0.7 g, 44%). LC-MS (1331+); m/e 210 (M+H)+, Rt: 0.75 min. bl .3) Trifluoromethanesulfonic acid 2,2-difluoro—2-quinolin—2—yl ethyl ester To a on of 2,2-difluoroquinolin—2-ylethanol (300 mg, 1.4 mmol) and triethylamine (217 mg, 2.1 mmol) in ous DCM (5 mL) was added dropwise trifluoromethanesulphonic ide (606 mg, 2.1 mol) at -70 0C. The reaction 10 mixture was stirred for 1 hour. The resulting solution was warmed slowly to room ature and stirred for 1 hour. The solid was removed by filtration. Water (5 mL) and DCM (30 mL) were added, the organic layer was separated, dried over sodium sulfate and evaporated to give the crude title compound as an orange oil (450 mg, 92%), which was used in the next step without further purification. LC-MS (ESI+): m/e 342 15 (M+H)+, R: 1.01 min. 0) Preparation of compounds ofthe l a Het-A4-Br cl) syn(2-Bromocyclopropyl)quinoline 20 cl . l) (Z)(2-Bromovinyl)quinoline A sion of 24.97 g (57.3 mmol) of (bromomethyl)triphenylphosphonium bromide in 160 mL ofTHF was chilled to -70°C under argon. Then, 6.43 g (57.3 mmol) ofpotassium tert-butoxide were added portionwise and the suspension was allowed to 25 stir at this temperature for 1 h. A solution of 7.5 g of (47.7 mmol) quinoline carbaldehyde in 40 mL ofTHF was slowly added dropwise. The mixture was allowed to stir at -75°C for further 5 hours and then warmed to room temperature overnight. 160 mL ofPE were added. The precipitate formed was sucked off. The mother liquid was evaporated and the residue was stirred with diisopropyl ether. The residue formed was 134 sucked off. Purification by chromatography (heptane/EA 3/1) d 6.6 g (59.1%) of the title compound. cl .2) syn romocyclopropyl)quinoline A solution of lzinc in hexane (5.54 g, 44.9 mL, 44.9 mmol) was chilled to 0°C under argon. Trifluoroacetic acid (5.11 g, 3.46 mL, 44.9 mmol) was added dropwise over 20 minutes. The mixture was stirred for further 20 minutes at 0° C. Then, a solution of diiodomethane (12.01 g, 3.62 mL, 44.9 mmol) in dichloromethane was added at 0°C over a period of 20 minutes. A solution of (Z)(2-bromovinyl)quino line 10 (2.1 g, 8.97 mmol) in DCM was added dropwise and the e was d over night at room temperature. The total amount ofDCM was 350 mL. The reaction mixture was quenched by addition of saturated aqueous ammonium chloride and then extracted with DCM. The organic phase was washed with water. The combined organic phases were ated. The residue was stirred with EA. Active charcoal was added and the 15 mixture was stirred for some further s. The solids were filtered off and the filtrate was concentrated. Purification by chromatography (CombiFlash Rf, normal phase chromatography, gradient elution using cyclohexane in EA up to a concentration of 15%) d 0.78 g (34.8%) of the title compound. 20 c2) syn 2-(2-bromocyclopropyl)—6—fluoroquinoline F c2. 1) (Z)(2-bromovinyl)fluoroquino line To as suspension of (bromomethyl)triphenylphosphonium bromide (14.94 g, 34.3 mmol) in 100 mL of THF was added potassium tert-butoxide (3.84 g, 34.3 mmol) 25 portionwise under Ar. Then the reaction mixture was stirred for 1 h at this temperature. 6-Fluoroquinolinecarbaldehyde (5 g, 28.5 mmol) in 40 mL ofTHFwas added. The resulting reaction mixture was stirred for 5 h at -75°C and allowed to warm up overnight. The reaction mixture was diluted with petrol ether and the solid was sucked off. The mother liquid was concentrated, diisopropyl ether was added and the formed 30 itate was sucked off. The mother liquidwas purified by column chromatography 135 hexane/EA) to give 4.7 g (65.3%) of the title compound as brown solid. LC-MS: m/e 254.0. c2.2) syn romocyclopropyl)fluoroquinoline The title compound was prepared in analogy to the method described for example c1.2. LC-MS: 268.0 03) syn romocyclopropyl)thieno[3,2-b]pyridine S \ Br \l/ N H H 10 The title compound was prepared in analogy to the method described in example c1.2 starting from (Z)(2-bromovinyl)thieno[3,2—b]pyridine. LC-MSM m/e 255.9 (M+H)+ d) Preparation of nds ofthe general formula Het-A3-Br 15 d1) (Z)(2-bromovinyl)methy1quinoline To a suspension of (bromomethyl)triphenylphosphonium bromide (3.06 g, 7.01 mmol) in 30 mL of THF, potassium tert-butoxide (0.78 g, 7.01 mmol) was added 20 portionwise at -75°C under argon. After completion ofthe addition, the mixture was d for 1 h a -75°C. Then, a solution of 3—methquuinolinecarbaldehyde (1 g, 5.84 mmol) in 20 mL of THF was added. The reaction mixture was stirred for 5 h at -75°C and then allowed to warm up to room temperature overnight. For work-up, the reaction mixture was diluted with diisopropyl ether (1 :1) and the precipitate formed was sucked 25 off. The filtrate was concentrated and triturated in diisopropyl ether. The precipitate was sucked off. The filtrate was purified by column chromatography (normal phase, eluent: 136 cyclohexane/EA) to give 492.5 mg (34%) of the title compound as yellow solid. LC- MS: m/e 248.0 ; Rt 1.561 min. d2) (Z)(2-bromovinyl)methoxyquinoline o H3C’ \ Br / N H H The title compound was prepared in analogy to the method described in example d1 but using 6-methoxyquinolinecarbaldehyde instead of 3-methquuinoline carbaldehyde. LC—MS: m/e 265.1 ; R 1.78 min 10 d3) (Z)(2-bromovinyl)—6—chloroquinoline Cl \ Br / N H H The title compound was prepared in analogy to the method described in example dl but using 6-chloroquinolinecarbaldehyde instead of 3-methquuinoline carbaldehyde.LC-MS: m/e 268.1 Rt 4.609 min. 15 d4) (Z)(2-bromovinyl)thieno[3,2—b]pyridine The title compound was prepared in analogy to the method described in example d1 but using thieno[3,2-b]pyridinecarbaldehyde. LC-MS: m/e 239.9. 20 11. Preparation of compounds of the formula I 11.1 Preparation of nds of the formula I in which A is Al, X1 is N, and X3 is S 137 EXAMPLE 1 : 3 ,7-Di(pyridin-4—yl)—5—[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one 1.1 4-Bromothiophenecarboxylic acid To a mixture ofMg (1.4 g, 60 mmol) and 12 (0.1 g) in ous THF (2 mL) was added dropwise a solution of 2—bromo—propane (7.4 g, 60 mmol) in anhydrous THF (60 mL) at room temperature under nitrogen during a period of 30 min. After the addition, the mixture was d until the most of magnesium was consumed. The resulting Grignard reagent was added dropwise to a solution of 3,4-dibromo-thiophene (12.1 g, 10 50 mmol) in anhydrous THF (60 mL) at 0 0C under nitrogen within about 30 min. The mixture was allowed to stir at 0 °C for 1.5 h. Excessive CO2 was purged into the mixture at -30 0C and the reaction mixture was stirred until the temperature rose to room temperature. Then the reaction was quenched with water (30 mL) and basified with 8% aq. NaOH solution to pH 11 and was washed with ethyl acetate (3 ><60 mL). The 15 aqueous layer was acidified with 5% aq. HCl to pH l~2, the precipitate was filtered and was dried to give the title compound as off-white solid (5.8 g, yield 56%).
LC-MS (ESI+): m/e 209 (M+H)+, Rt: 0.69 min. 1.2 4-Bromoisonicotinoylthiophenecarboxylic acid 20 To a solution of diisoproplyamine (5.3 g, 53 mmol) in anhydrous THF (40 mL) at —30°C was added n—BuLi (23.2 mL, 58 mmol, 2.5M in hexanes) dropwise. The e was stirred at the same temperature for 0.5 h, then cooled to -78 0C and HMPA (0.86 g, 4.8 mmol) was added slowly. Then the solution of 4-bromothiophenecarboxylic acid (5.0 g, 24 mmol) in anhydrous THF (50 mL) was added slowly. The mixture was stirred 25 at the same temperature for l h, N—methoxy—N—methyl—4-pyridinecarboxamide (8.0 g, 48 mmol) was added dropwise into the stirring mixture at —78°C. The reaction mixture was stirred for another 1 h at room ature and was then quenched with H20 (10 mL).
The aqueous layer was acidified with 5% aq. HCl to pH l~2, the precipitate was 138 collected by filtration. The filter cake was washed with DCM (10*50 mL). The title compound was ved in DCM; the solid insoluble in DCM was the side product.
The filtrate was extracted with DCM (3 X200 mL). The organic layers were dried over Na2SO4 and concentrated under d pressure. The crude product was washed with DCM to give the title compound (0.8 g, yield 10.7 %) as a yellow solid.
LC-MS (ESI+): m/e 312 (M+H)+, Rt: 1.45 min. 1.3 Ethyl 4-bromoisonicotinoylthiophenecarboxy1ate To a solution of the compound from example 1.2 (2.5 g, 8 mmol) and CszC03 (5.2 g, 16 10 mmol) in CH3CN (500 mL) was added CH3CHZI (3.0 g, 19.2 mmol) dropwise . The mixture stirred at 30 0C for 48 h. The mixture was filtered and trated to give the title compound as yellow oil (2 g, 73.5% yield).
LC-MS (ESI+): m/e 340 (M+H)+, Rt: 0.81 min 15 1.4 Ethyl 2-isonicotinoyl(pyridin—4—yl)thiophenecarboxylate A mixture of the compound from example 1.3 (500 mg, 1.47 mmol), 4-pyridineboronic acid (271 mg, 2.21 mmol), Na2C03 (390 mg, 3.68 mmol) and Pd(dppf)C12 (122 mg, 0.15 mmol) in dioxane/HZO (3:1) (12 mL) was stirred at 100°C under argon for 2 h. The mixture was concentrated and the residue was purified by PLC to give the title 20 product as white solid (318 mg, 64 % .
LC-MS (ESI+): m/e 339 (M+H)+, 11,; 0.61 min 1 .5 3 ,7-Di(pyridinyl)thieno[2,3-d]pyridazin—4(5H)-one A mixture of the compound from example 1.4 (200 mg, 0.59 mmol) in 25 NHzNHz-HzO (2 mL) and EtOH (10 mL) was stirred at room temperature for 30 min.
The mixture was filtered and the solid was dried to obtain the title compound as a white solid (150 mg, 83.1%). LC-MS (ESI+): m/e 307 (M+H)+, R: 1.56 min. 1 .6 3 ,7—Di(pyridin—4—yl)—5—[2—(quinolinyl)ethy1]thieno[2,3-d]pyridazin-4(5H)—one 30 To a solution of 3,7—di(pyridin—4—yl)thieno[2,3—d]pyridazin—4(5H)-one from example 1.5 (100 mg, 0.32 mmol), 2—quinolin—2—yl—ethanol from example a1 (58 mg, 0.33 mmol) and PPh3 (256 mg, 0.98 mmol) in DCM (10 mL), DIAD (198 mg, 0.98 139 mmol) was added dropwise. The mixture was stirred at room temperature for 3h, concentrated and the residue was purified by Prep-HPLC to give the title compound as white solid (26 mg, 17.6% yield).
LC-MS (ESI+): m/e 462 (M+H)+, R: 2.00 min; 1H-NMR(DMSO-d, 400MHz): 5 3.46 (t, J: 7.2 Hz, 2H), 4.71 (t, J= 7.2 Hz, 2H), 7.49-7.51 (m, 3H), 7.54-7.57 (m, 1 H), 7.62 (dd, J: 4.4, 1.6 Hz, 2H), .72 (m, 1 H), 7.84 (d, J= 8.4 Hz, 1H), 7.94 (d, J: 7.2 Hz, 1H), 8.28-8.30 (m, 2H), 8.60 (dd, J= 4.6, 1.6 Hz, 2H), 8.70 (dd, J: 4.6, 1.6 Hz, 2H). 10 EXAMPLE 2: 7-(Pyridin—4-yl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin— 4(5H)—one \ o / N N \ s / | \ N 2.1 2-Isonicotinoylthiophenecarboxylic acid To a solution of diisopropylamine (5.2 g, 51.5 mmol) in anhydrous THF (40 mL) 15 at —30°C was added n-BuLi (23.2 mL, 56.2 mmol, 2.5M in THF) dropwise. The mixture was stirred at the same temperature for 0.5 h, then cooled to -78°C and HMPA (0.8 g, 4.7 mmol) was added slowly. Then a solution of thiophenecarboxylic acid (3.0 g, 23.4 mmol) in anhydrous THF (50 mL) was added slowly. The mixture was d at the same temperature for l h, N—methoxy—N-methyl—4-pyridinecarboxamide (5.0 g, 46.9 20 mmol) was added drop wise into the stirring e at —78°C. The reaction mixture was stirred for another 1 h at room temperature and was then quenched with H20 (10 mL). The aqueous layer was acidified with 5% aq. HCl to pH 1~2, the precipitate was collected by filtration. The filter cake wash with DCM and the filtrate was extracted with DCM (3 X200 mL). The organic layers were dried over NaZSO4 and trated 25 under reduced pressure. The crude product was washed with DCM to give the title product (2.2 g, yield 40 %) as a white solid.
LC-MS (ESI+): m/e 234 (M+H)+, Rt: 0.57 min. 140 2.2 Ethyl 2-isonicotinoylthiophene—3—carboxy1ate To a solution of icotinoylthiophenecarboxylic acid (2.2 g, 9.4 mmol) from e 2.1 and CS2CO3 (6.2 g, 18.9 mmol) in CH3CN (500 mL) was added CH3CH21 (2.9 g, 18.9 mmol) dropwise. The mixture was d at 30 0C for 48 h. The mixture was filtered and concentrated to give the title compound as yellow oil (2.1 g, 85.5% yield).
LC-MS (ESI+): m/e 340 (M+H)+, Rt: 0.81 min 2.3 7-(Pyridinyl)thieno[3,2-d]pyridazin-4(5H)-one 10 The mixture of ethyl 2-isonicotinoylthiophenecarboxylate (200 mg, 0.77 mmol) from example 2.2 in NHzNH2~HzO (2 mL) and EtOH (10 mL) was d at room temperature for 30 min. The mixture was filtered and the solid dried to obtain the title compound (150 mg, 85.1% yield).
LC-MS (ESI+): m/e 230 , Rt: 1.52 min. 15 2.4 7-(Pyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one To a solution of 7-(pyridinyl)thieno[3,2-d]pyridazin—4(5H)-one (100 mg, 0.44 mmol) from example 2.3, 2-quinoliny1—ethanol from example a1 (83 mg, 0.48 mmol) and PPh3 (343 mg, 1.31 mmol) in DCM (10 mL), DEAD (228 mg, 1.31 mmol) was 20 added dropwise. The mixture stirred at room temperature for 3 h, concentrated and the purified by Prep-HPLC to give the title compound as a white solid (20 mg, 11.8% yield).
LC-MS (ESI+): m/e 385 (M+H)+, Rt: 1.87 min; lH-NMR(DMSO-d6, 400 MHz): 8 3.49 (t, J: 7.2 Hz, 2H), 4.76 (t, J: 7.2 Hz, 2H), 7.49—7.57 (m, 2H), 7.58 (dd, J: 4.4, 25 1.6 Hz, 2H), 7.67-7.72 (m, 1 H), 7.78 (d, J: 5.2 Hz, 1H), 7.85 (d, J: 8.0 Hz, 1H), 7.93 (d, J: 7.6 Hz, 1H), 8.21 (d, J= 5.2 Hz, 1H), 7.93 (d, J: 8.8 Hz, 1H), 8.66 (dd, J: 4.2, 1.6 Hz, 2H).
EXAMPLE 3: 3—(Pyridin—4—yl)[2-(quinolinyl)ethy1]thieno[2,3-d]pyridazin—4(5H)— 30 0116 141 N / \ \ O / / N 3.1 4-Bromoformyl-thiophene—3—carboxylic acid To a solution of (i—Pr)2NH (1.09 g, 10.8 mmol) in anhydrous THF (15 mL) was added dropwise n-BuLi (5.0 mL, 12.5 mmol, 2.5M in hexane) at -30°C. The mixture was d at the same temperature for 0.5 h. Then, the mixture was cooled to -78°C and the solution of 4-bromothiophenecarboxylic acid from example 1.1 (1.0 g, 4.85 mmol) and HMPA (0.17 g, 0.95 mmol) in anhydrous THF (20 mL) was added slowly.
The mixture was stirred at the same ature for l h, anhydrous DMF (0.6 g, 8.22 mmol) was added dropwise into the stirring mixture at —78°C. The reaction mixture was 10 stirred for another 45 min at room ature and then quenched with water. The aqueous layer was acidified with 5% aq. HCl to pH 1~2, the precipitate was collected by filtration, the filtrate was extracted with DCM (3 X50 mL). The organic layers were dried over Na2S04 and concentrated under reduced pressure. The crude product was washed with CHzClz to give the title t (0.68 g, 60.2 %) as a solid. 15 LC-MS: m/e : 235.7; R: 0.64 min. 3.2 4-Bromoformyl-thiophene—3—carboxylic acid ethyl ester 4-Bromoformyl-thiophene—3—carboxylic acid (0.16 g, 0.68 mmol), K2C03 (0.188 g, 1.36 mmol), and 4 mL of ous DMF were stirred at room temperature 20 for 10 min, then iodoethane was added (0.128 g, 0.8 mmol) dropwise. The reaction solution was stirred at 50 0C for 3h. The reaction mixture was cooled, extracted with EA (3 x 50 mL), concentrated and the residue was purified by TLC (PE/EA =8/ 1) to give 0.4 g of the title compound (yield: 63.5%).
LC-MS: m/e (M+H)+: 263.7, R: 0.88 min. 25 3.3 3-Bromo—5H—thieno[2,3-d]pyridazinone 4-Bromo-2—formy1—thiophenecarboxylic acid ethyl ester (1.0 g, 3.8 mmol), NHZNHZ'HZO (0.27 g, 4.58 mmol) and 20 mL of EtOH were refluxed under nitrogen 142 for 5h. The reaction mixture was cooled and precipitate formed, then filtered to give the title compound (0.2 g, yield: 23%).
LC-MS: m/e (M+H)+: 231.7, R: 1.29 min 3.4 3-Bromo[2-(quinolin—2-y1)ethyl]thieno[2,3-d]pyridazin—4(5H)-one To a stirred solution of PPh3 (977 mg, 3.73 mmol) and DEAD (1.13 mL) in 15 mL ofanhydrous THF was added a solution of 3-bromo-5H-thieno[2,3-d]pyridazin one (429 mg, 1.86 mmol) and 2-quinolinyl-ethanol from example al (355 mg, 2.05 mmol) in 15 mL of ous THF while being cooled with an th. Then the 10 resulting mixture was stirred in nitrogen atmosphere at 45°C overnight. The reaction mixture was concentrated and the product was recrystalized from EA to give the title nd (370 mg, 53.6%).
LC-MS: m/e (M+H)+: 386.7, R: 2.02 min 15 3.5 idinyl)[2-(quinolin-2—yl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one 3-Bromo[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one (130 mg, 0.33 mmol), pyridineylboronic acid (45.7 mg, 0.37 mmol), Pd(dppt)C12 (15 mg) and K2C03 (93 mg, 0.67 mmol) were dissolved in dioxane/HZO (3/1, 2.8 mL). The mixture was stirred in nitrogen atmosphere at 120°C for 1h in a microwave tube. The solution 20 was concentrated and purified by TLC eOH =10/ 1) and recrystalized from MeOH to give the title product (90 mg, 69.4%).
LC-MS: m/e (M+H)+: 385.7, R: 1.90 min. 1H NMR (DMSO, 400MHz) 5: 8.48- 8.45 (m, 3H), 8.25 (d, J: 8.4Hz, 1H), 8.02 (s, 1H), 7.93-7.88 (m, 2H), 7.72 (t, J: 8.4Hz, 1H), 7.58 (t, J=7.2Hz, 1H), 7.48-7.42 (m, 3H), 4.72 (t, J: 7.0 Hz, 2H), 3.49 (t, J: 7.0 25 Hz, 2H) EXAMPLE 4: 3-(Pyridinyl)[2-(pyridinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- 0116 N / \ \ O / \ / N 143 4.1 3-Bromo(2-pyridinyl-ethyl)—5H-thieno[2,3-d]pyridazin—4-one The title compound was prepared in analogy to the process described in Example 3.4 starting from 3-bromo-5H-thieno[2,3-d]-pyridazin—4-one and dinyl-ethanol.
Yield: 76.3%.
LC-MS: m/e (M+H)+: 336.7, Rt: 1.51 min 4.2 3-(Pyridin—4-yl)[2-(pyridinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one The title compound was prepared in analogy to the process bed in Example 3.5 starting from o(2-pyridinyl-ethyl)-5H-thieno[2,3-d]pyridazinone 10 and pyridineylboronic acid. Yield: 33.1%.
LC-MS: m/e (M+H): 335.7, R: 1.69 min. 1H NMR (DMSO, 400MHz) 5: 8.60 (d, J: 5.2Hz, 2H), 8.47 (d, J: 4.4Hz, 1H), 8.16 (s, 1H), 7.53-7.48 (m, 2H), 7.43 (d, J=5.2Hz, 2H), 7.11-7.04 (m, 2H), 4.57 (t, J: 7.6 Hz, 2H), 3.24 (t, J: 7.4 Hz, 2H). 15 EXAMPLE 5: 3,7-Di(pyridinyl)—5—[2—(pyridin—2-yl)ethyl]thieno[2,3-d]pyridazin- 4(5H)—one 5. l 3-Bromopyridinyl-5H-thieno[2,3—d]pyridazin—4-one The title compound was prepared in analogy to the process described in Example 20 3.3 starting from ethyl 4-bromo-2—isonicotinoyl—thiophene—3-carboxylic acid from Example 1.3. Yield: 56.8%.
LC-MS: m/e (M+H) +3087, Rt: 1.59 min. 5.2 3-Bromopyridinyl[2-(pyridinyl)ethy1]thieno[2,3-d]pyridazin- 25 4(5H)—one The title compound was prepared in y to the process described in Example 3.4 starting from 3-bromo—7—pyridin—4—yl—5H—thieno[2,3-d]pyridazinone and 2- pyridin—2-yl—ethanol. Yield: 70%. 144 LC-MS: m/e (M+H)+: 413.7; R: 1.84 min. 5 .3 3 ,7-Di(pyridinyl)-5 - [2-(pyridinyl)ethyl]thieno [2,3 -d]pyridazin-4(5H)-one The title compound was prepared in analogy to the process described in Example 3.5 starting from 3-bromopyridinyl-5—[2-(pyridin—2-yl)ethyl]thieno[2,3- dazin—4(5H)-one and pyridineylboronic acid. Yield: 75.2%.
LC-MS: m/e (M+H)+: 412.7, Rt: 1.71 min; 1H NMR (DMSO, 400MHz) 8: 8.78 (d, J= 5.2Hz, 2H), 8.71 (d, J= 4.4Hz, 2H), 8.56 (d, J= 4.8Hz, 1H), 7.70-7.67 (m, 3H), 7.60 (t, J=7.6Hz, 1H), 7.51 (d, J= 5.2 Hz, 2H), 7.21 (d, J= 7.6 Hz, 1H), .14 (m, 10 1H), 4.75 (t, J= 7.6 Hz, 2H), 3.37 (t, J= 7.4 Hz, 2H).
EXAMPLE 6: 5-[2-(Quinolin—2—yl)ethyl]—3—[4—(trifluoromethyl)phenyl]thieno[2,3- dazin—4(5H)-one FFF \ o / N N I |\ N\ s 15 3-Bromo[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin—4(5H)-one Example 3.4 (100 mg, 0.26 mmol), 4-(trifluoromethyl)phenylboronic acid (0.26 mmol), Pd(dppf)Clz (12.7 mg) and K2C03 (72 mg, 0.527 mmol) were dissolved in dioxane/HZO (3/1, 2.8 mL). The mixture was d at 110°C for 0.5 h in a microwave tube. The solution was concentrated and purified by prep—TLC to give the title product. 20 LC-MS: m/e (M+H)+: 452, R: 2.36,1H NMR (CDC13, 400 MHZ) 8: 3.50 (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=7.6 Hz), 7.33 (d, 1H, J=4.4 Hz), 7.48-7.53 (m, 2H), 7.59-7.69 (m, 5H), 7.78 (d,1H, J=7.6 Hz), 8.00 (d, 1H, J=8.0), 8.06 (d, 1H, J=8.0 Hz), 8.23 (s, 1H) Examples 7 to 92 were prepared analogously to the method described for Example 25 145 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 3-(4- 398 /2.32 2.40 (s, 3H), 3.50 (t, 2H, J=7.6 Hz), 4.73 (t, methylpheny1) 2H, J=7.6 Hz), 7.22 (d, 2H, J=8.0 Hz), 7.32 [2-(quinolin—2- (d, 1H, J=7.6 Hz),7.40 (d, 2H,J=7.6 Hz), 7 y1)ethy1]thieno[2,3- 7.45 (s,1H), 7.49 (t, 1H,J=7.2 Hz),7.67 (t, d]pyridazin-4(5H)- 1H, J=7.2 Hz), 7.77 (d, 1H, J=7.6 Hz),8.01- one 8.06 (m, 2H), 8.19 (s,1H) 3-[4-(propan—2- 426 / 2.46 1.29 (d, 6H, J=6.8 Hz), 2.95 (m, 1H), 3.49 y1)pheny1]-5—[2— (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=8 Hz), (quinolin 7.27~7.31 (m, 3H), 7.44~7.49 (m, 4H), 7.66 8 y1)ethy1]thieno[2,3- (t, 1H, J=8.4 Hz), 7.75 (d, 1H, J=8 Hz), d]pyridazin—4(5H)- 8.01~8.04 (m, 2H), 8.18 (s, 1H) one 412 / 2.34 1.28 (t, 3H, J=7.6 Hz), 2.70 (q, 2H, J=7.6 3-(4-ethy1pheny1)- Hz), 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, 5-[2-(quinolin J=7.6 Hz), 7.25 (d, 2H, J=8.0 Hz), 7.31 _ (d, 9 y1)ethy1]th1eno[2,3- 1H, J=8.4 Hz), 7.44-7.50 (m, 4H), 7.65-7.69 d]pyr1daz1n—4(5H)-_ _ (m, 1H), 7.77 (d, 1H, J=8.4 Hz), 8.00-8.05 one (m, 2H), 8.19 (s, 1H) 4- [2- 409 /2.11 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 (quinolin Hz), 7.33 (d, 1H, J=8.0 Hz), 7.50-7.58 (m, y1]-4,5- 4H), 7.66-7.68 (m, 3H), 7.79 (d, 1H, J=7.6 10 dihydrothieno[2,3- Hz), 7.98 (d, 1H, J=8.0 Hz), 8.07 (d, 1H, d]pyridazin—3- J=8.0 Hz), 8.25 (s, 1H) y1}benzonitrile 146 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 3-(4- 414 / 2.18 3.50 (t, 2H, J=7.6 Hz), 3.85 (s, 3H), 4.74 (t, methoxypheny1) 2H, J=7.6 Hz), 6.94 (d, 2H, J=8.4 Hz), 7.31 [2-(quinolin—2- (d, 1H, J=8.4 Hz), 7.42—7.51 (m, 4H), 7.65- 11 y1)ethy1]thieno[2,3- 7.69 (m, 1H), 7.77 (d, 1H, J=8.4 Hz), 8.00- d]pyridazin-4(5H)- 8.06 (m, 2H), 8.19 (s, 1H) one 3-(4—fluoropheny1)— 402 / 2.21 3.50 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 5-[2-(quinolin—2— Hz), 7.06—7.11 (m, 2H), 7.32 (d, 1H, J=8.4 12 y1)ethy1]thieno[2,3— Hz), 7.45—7.52 (m, 4H), 7.65-7.69 (m, 1H), d]pyridazin—4(5H)— 7.78 (d, 1H, J=8.4 Hz), 8.01 (d, 1H, J=8.4 one Hz), 8.06 (d, 1H, J=8.4 Hz), 8.21 (s, 1H) 3-(4- 428 /2.26 1.44 (t, 3H, J=6.8 Hz), 3.49 (t, 2H, J=7.6 ethoxypheny1) Hz), .11 (m, 2H), 4.74 (t, 2H, J=7.6 [2-(quinolin—2- Hz), 6.93 (d, 2H, J=8.4 Hz), 7.32 (d, 1H, 13 y1)ethy1]thieno[2,3- J=8.4 Hz), 7.42~7.51 (m, 4H), 7.67 (t, 1H, dazin—4(5H)- J=7.6 Hz), 7.77 (d, 1H, J=8 Hz), 8.00~8.06 one (m, 2H), 8.19 (s, 1H) 3-[4- 427 /2.26 2.99 (s, 6H), 3.50 (t, 2H, J=7.6 Hz), 4.75 (t, (dimethylamino)ph 2H, J=7.6 Hz), 6.77 (d, 2H, J=8.8 Hz), 7.31 eny1][2- (d, 1H, J=8.8 Hz), 7.38 (s, 1H), 7.43-7.51 14 (quinolin (m, 3H), 7.65—7.69 (m, 1H), 7.77 (d, 1H, y1)ethy1]thieno[2,3- J=8.0 Hz), 8.01-8.05 (m, 2H), 8.16 (s, 1H). d]pyridazin—4(5H)- 0116 147 LC-MS: 1H NMR ) 5: EX. Name m/ e (M+H)+/ Rt [min] (4- {4-oxo[2- 423 /2.10 3.49 (t, 2H, J=7.6 Hz), 3.79 (s, 3H), 4.73 (t, (quinolin 2H, J=7.6 Hz), 7.31-7.37 (m, 3H), 7.48-7.51 y1]-4,5- (m, 4H), 7.65—7.69 (m, 1H), 7.99 (d, 1H, 15 dihydrothien0[2,3- J=8.4 Hz), 8.05 (d, 1H, J=8.4 Hz), 8.22 (s, d]pyridazin—3 - 1 H) y1}pheny1)acetonitr ile 3-(4-hydr0xy— 400 / 2.00 2.17 (s, 1H), 3.53 (t, 2H, J=7.6 Hz), 4.77 (t, phenyl)[2- 2H, J=7.6 Hz), 7.80 (d, 2H, J=8.8 Hz), (quinolin 7.33~7.42 (m, 4H), 7.51 (t, 1H, J=7.2 Hz), 16 y1)ethy1]thieno[2,3- 7.69 (t, 1H, J=7.2 Hz), 7.79 (d, 1H, J=7.6 d]pyridazin—4(5H)- Hz), 8.08 (d, 2H, J=8.4 Hz), 8.22 (s, 1H) one 3-(2-ch10r0pheny1)- 418 /2.22 3.47 (t, 2H, J=7.6 Hz), 4.71 (t, 2H, J=7.6 5-[2-(quinolin Hz), .36 (m, 4H), .50 (m, 3H), 17 y1)ethy1]thieno[2,3- 7.64-7.68 (m, 1H), 7.76 (d, 1H, J=8.0 Hz), d]pyridazin—4(5H)- 8.00-8.04 (m, 2H), 8.21 (s, 1H). one 3-(2- 398 /2.29 2.09 (s, 3H),3.48 (t, 2H, J=7.6 Hz), 4.69 (t, methylphenyl) 2H, J=7.6 Hz), 7.17-7.33 (m, 5H), 7.37 (s, [2-(quin01in 1H), 7.49 (t, 1H, J=7.2 Hz), 7.67 (t, 1H, 18 y1)ethy1]thien0[2,3- J=7.2 Hz), 7.76 (d, 1H, J=8.0 Hz), 7.99- d]pyridazin—4(5H)- 8.05 (m, 2H), 8.22 (s, 1H) 0116 3-(2-ethy1phenyl)- 412/2.36 1.02 (t, 3H, J=7.6 Hz), 2.36~2.49 (m, 2H), 5-[2—(quin0 1111—2— 3.47 (t, 2H, J=7.6 Hz), 4.65~4.71 (m, 2H), 19 y1)ethy1]thieno[2,3— 7.15 (d, 1H, J=7.2 Hz), 7.02~7.38 (m, 6H), d]pyridazin—4(5H)— 7.49 (m, 1H, J=7.6 Hz), 7.77 (d, 1H, J=8 0116 Hz), 7.99~8.04 (m, 2H), 8.22 (s, 1H) 148 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 402 /2.16 3.49 (t, 2H, J=7.6 Hz), 4.73 (t, 2H, J=7.6 3-(2-fluoropheny1)- Hz), 7.14~7.21 (m, 2H), .31 (In, 5-[2-(quinolin 7.38 (t, 2H, J=7.6 Hz), 7.48 (t, 1H, _ 1H), 20 y1)ethy1]th1eno[2,3- J=7.6 Hz), 7.54 (s, 1H), 7.67 (t, 1H, J=7.6 d]pyr1da21n-4(5H)-. .
Hz), 7.76 (d, 1H, J=8 Hz), 8.02 (t, 2H, one J=7.6 Hz), 8.20 (s, 1H) 3-(2- 414 / 2.22 3.47 (t, 2H, J=7.2Hz), 3.74(s, 3H), 4.69 (t, ypheny1)—5— 2H, J=7.6Hz), 6.97~7.02 (m, 2H), 7.28 (t, [2-(quinolin—2- 2H, J=8.4Hz), 7.38 (t, 1H, J=8Hz), 7.48 (t, 21 y1)ethy1]thieno[2,3- 2H, J=7.6Hz), 7.67 (t, 1H, J=7.6Hz), 7.76 dazin—4(5H)- (d, 1H, J=8Hz), 8.03 (d, 2H, J=8.4Hz), 8.17 one (s, 1H) 3-(2- 428 /2.31 1.18 (t, 3H, J=6.8 Hz), 3.47 (t, 2H, J=7.6 ethoxypheny1) Hz), 4.01~4.06 (m, 2H), 4.69 (t, 2H, J=7.6 [2-(quinolin—2- Hz), 6.96~7.01 (m, 2H), 7.27~7.36 (n1, 22 y1)ethy1]thieno[2,3- 3H), 7.48 (t, 2H, J=8.8 Hz), 7.67 (t, 1H, d]pyridazin—4(5H)- J=7.2 Hz), 7.76 (d, 1H, J=8.4 Hz), 8.03 ((1, one 2H, J=8 Hz), 818 (s, 1H) 23 3-(2- 400 / 2.04 3.52 (t, 2H, J=7.6 Hz), 4.83 (t, 2H, J=7.6 hydroxypheny1)-5 - Hz), 7.05 (t, 1H, J=8 Hz), 7.17 (d, 1H, [2-(quino lin J=7.6 Hz), 7.27~7.38 (m, 3H), 7.48~7.55 y1)ethy1]thieno[2,3- (m, 2H), 7.68 (t, 1H, J=7.6 Hz), 7.78 (d, d]pyridazin—4(5H)- 1H, J=7.2 Hz), 8.01~8.08 (m, 2H), 8.32 (s, 0116 1H), 8.71 (s, 1H) 149 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 24 5-[2-(quinolin 452 /2.30 3.47 (t, 2H, J=7.6 Hz), 4.67 (t, .6 y1)ethy1][2- Hz), 7.25-7.29 (m, 2H), 7.47-7.57 (m, 4H), (trifluoromethyl)ph 7.66—7.58 (m, 3H), 8.02:8.04 (m, 2H), 8.22 hieno[2,3- (s, 1H) d]pyridazin-4(5H)- one 25 3-[3— 428 / 2.16 3.42 (s,1H), 3.49 J=7.6Hz),4.52 xymethy1)ph (s,2H),3.79 (s,3H),4.74(t,2H,J=7.6Hz),7.31 eny1][2- (d,1H,J=8.4Hz), 7.35-7.51 (m,6H),7.65- (quinolin 7.69 (m,1H), 7.77 (d,1H,J=8.0Hz),7.99-8.05 yl)ethy1]thieno[2,3- (m,2H), 8.20 (s,1H) d]pyridazin—4(5H)- one 26 3-(3- 414 / 2.19 3.50 (t, 2H, J=7.6 Hz), 3.83 (s, 3H), 4.74 (t, methoxypheny1) 2H, J=7.6 Hz), 6.92-6.95 (m, 1H), 7.07-7.12 [2-(quinolin—2- (m, 2H), 7.31-7.35 (m, 2H), 7.47-7.51 (m, yl)ethy1]thieno[2,3- 2H), 7.66-7.70 (m, 1H), 7.77 (d, 1H, J=8.4 d]pyridazin—4(5H)- Hz), 8.01-8.06 (m, 2H), 8.20 (s, 1H) one 27 3-(3- 428 /2.27 1.43 (t, 3H, J=6.8 Hz), 3.50 (t, 2H, J=7.2 ethoxypheny1) Hz), 4.06 (q, 2H, J=6.8 Hz), 4.74 (t, 2H, [2-(quinolin—2- J=7.2 Hz), 6.91—6.94 (m, 2H), 7.07-7.09 (m, y1)ethy1]thieno[2,3- 2H), 7.29-7.33 (m, 2H), 7.47—7.51 (m, 2H), d]pyridazin-4(5H)- 7.65-7.78 (m, 2H), 8.01-8.06 (m, 2H), 8.20 one (s, 1H) 150 LC-MS: 1H NMR ) 5: EX. Name m/ e (M+H)+/ Rt [min] 28 3-[3- 427 / 2.26 2.97 (s, 6H), 3.49 (t, 2H, J=7.2 Hz), 4.73 (t, (dimethylamino)ph 2H, J=7.6 Hz), 6.77 (dd, 1H, J=2.4 Hz, eny1][2- J=8.4 Hz), 6.85—6.88 (m, 2H), 7.26-7.32 (m, (quinolin 2H), 7.47-7.51 (m, 2H), 7.65-7.77 (m, 2H), y1)ethyl]thieno[2,3- 8.03 (t, 2H, J=7.6 Hz), 8.19 (s, 1H) d]pyridazin—4(5H)- one 29 3-[4-Ox0-5—(2— 409 / 2.11 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 quinolin—2-y1— Hz), 7.35 (d, 1H, J=8.4 Hz), 7.47~7.52 (m, ethy1)-4,5-dihydro- 3H), 7.65~7.73 (m, 4H), 7.79 (d, 1H, J=8 thieno[2,3- Hz), 7.97 (d, 1H, J=8.4 Hz), 8.08 (d, 1H, d]pyridazin—3-y1}- J=8.4 Hz), 8.24 (s, 1H) benzonitrile 30 3-(3-fluoropheny1)- 402 / 2.21 3.49 (t, 2H, J=7.6 Hz), 4.47 (t, 2H, J=7.6 5-[2-(quin01in Hz), 7.06-7.11 (m, 1H), 7.20-7.24 (m, 1H), y1)ethy1]thieno[2,3- 7.27-7.39 (m, 3H), 7.47-7.51 (m, 2H), d]pyridazin—4(5H)- .69 (m, 1H), 7.77 (d, 1H, J=8.0 Hz), one 8.00 (d, 1H, J=8.4 Hz), 8.06 (d, 1H, J=8.4 Hz), 8.21 (s, 1H) 31 3-(3- 400/200 3.51 (t, 2H, J=7.6 Hz), 3.76 (s, 1H), 4.72 (t, hydroxypheny1)-5 - 2H, J=7.6 Hz), 6.85 (d,1H,J=1.6 Hz, J=8.0 [2-(quino 1111 Hz), 6.99-7.02 (m, 2H), 7.23 (t, 1H, J=8.0 y1]thien0[2,3- Hz), 7.34 (d, 1H, J=8.8 Hz), 7.48-7.53 (m, d]pyridazin—4(5H)- 2H), 7.66-7.70 (m, 1H), 7.78 (d, 1H, J=8.0 0116 Hz), 8.03-8.08 (m, 2H), 8.21 (s, 1H) 151 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 32 N,N—dimethy1—3-{4- 455 / 1.97 3.09 (d, 6H, J=17.6 Hz), 3.50 (t, 2H, J=7.6 [2-(quinolin- Hz), 4.74 (t, 2H, J=7.6 Hz), 7.31 (d, 1H, 2-y1)ethy1]—4,5- J=8.4 Hz), 7.43~7.51 (m, 6H), .69 dihydrothien0[2,3- (m, 1H), 7.78 (d, 1H, J=8.4 Hz), 8.00 (d, d]pyridazin—3- 1H, J=8.4 Hz), 8.06 (d, 1H, J=8.4 Hz), 8.21 y1}benzamide (s, 1H) 33 3-(3— 398 / 2.08 2.40 (s,3H), 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, methylphenyl)—5— 2H, J=7.6 Hz), 7.19-7.21 (m, 1H), 7.30-7.33 [2-(quinolin—2- (m, 4H), 7.45 (s, 1H), 7.49 (t, 1H, J=7.6 yl)ethyl]thieno[2,3- Hz), 7.65—7.69 (m, 1H), 7.77 (d, 1H, J=7.6 d]pyridazin—4(5H)- Hz), 8.01 (d, 1H, J=8.4 Hz), 8.04 (d, 1H, one J=8.4 Hz), 8.19 (s, 1H) 34 5-[2-(quin01in 390/ 1.53 3.92 (t, 2H, J=6.4 Hz), 4.81 (t, 2H, J=6.4 yl)ethy1]—3- Hz), 7.02-7.04 (m, 1H), 7.32 (d, 1H, J=3.6 (thiophen—2- Hz), 7.38 (d,1H, J=3.6 Hz), 7.62 (s, 1H), yl)thieno[2,3- 7.71 (d, 1H, J=8.4 Hz), 7.82 (t, 1H, J=7.6 d]pyridazin—4(5H)- Hz), 8.01-8.05 (m, 2H), 8.17 (s, 1H), 8.52 one (d, 1H, J=8.4 Hz), 8.64 (d, 1H, J=8.4 Hz) 35 3-(1-methy1—1H- 437 / 2.23 3.49 (t, 2H, J=7.8 Hz), 3.82 (s, 3H), 4.73 (t, ind01y1)(2- 2H, J=8 Hz), 6.51 (d, 1H, J=2.8 Hz), 7.07 quinolin—2-y1— (d, 1H, J=2.8 Hz), 7.26~7.48 (m, 5H), ethy1)-5H- 7.65~7.77 (m, 3H), 8.01~8.04 (m, 2H), 8.20 thien0[2,3- (s, 1H) d]pyridazin—4-one 36 indolyl)- 423 /2.14 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 quin0 1111—2— Hz), 6.57 (s, 1H), 7.25—7.32 (m, 2H), 7.48- y1)ethy1]thieno[2,3— 7.52 (m, 2H), 7.59 (s, 1H), 7.65-7.70 (m, d]pyridazin—4(5H)— 2H), 7.78 (d, 1H, J=8.0 Hz), 8.04 (t, 2H, 0116 J=8.4 Hz), 8.22 (s, 1H), 8.28 (s, 1H) 152 LC-MS: 1H NMR (CDC13) 5: EX. Name 111/ e (M+H)+/ Rt [min] 37 3-(pyrimidin—5-y1)— 386/ 1.73, 3.50 (t, 2H, J=7.6 Hz), 4.76 (t, 2H, J=7.6 5-[2-(quinolin Hz),7.34 (d, 1H, J=8.4 Hz), 7.49 (t, 1H, y1)ethy1]thien0[2,3- J=8.0 Hz), 7.61 (s, 1H), 7.66 (t, 1H, J=8.0 d]pyridazin—4(5H)- Hz), 7.78 (d, 2H, J=8.4 Hz), 7.96 (d, 1H, 0116 J=8.4 Hz), 8.07 (d, 1H, J=8.4 Hz), 8.26 (s, 1H), 8.76 (s, 2H), 9.22 (s, 1H) 38 3 -(2— 415/2.06 3.48 (t, 2H, J=7.6Hz), 3.90 (s, 3H), 4.71 (t, methoxypyridin—3 — 2H, J=7.6 Hz), 6.93-6.96 (m, 1H), 7.30 (d, y1)[2-(quinolin- 1H, J=8.4 Hz), 7.49 (t, 1H,J=7.6 Hz), 7.56- 2- 7.59 (m, 2H), 7.67 (t, 2H, J=7.6 Hz), 7.77 yl)ethyl]thieno[2,3- (d, 1H, J=8.0 Hz), 8.00-8.05 (m, 2H), 8.19 d]pyridazin—4(5H)- (s, 1H), 8.21 (dd, 1H, J=1.2, J=5.2 Hz) one 39 3-(pyridin—3-y1) 385 / 1.91 3.49 (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=7.6 [2-(quinolin—2- Hz), 7.32-7.36 (m, 2H), 7.49 (t, 1H, J=80 yl)ethy1]thieno[2,3- Hz), 7.55 (s, 1H), 7.64-7.69 (m, 1H), 7.77 d]pyridazin—4(5H)- (d, 1H, J=8.0 Hz), 7.88 (d, 1H, J=8.0 Hz), one 8.06 (d, 1H, J=8.8 Hz), 8.23 (s, 1H), 8.62 (br, 1H), 8.73 (br, 1H). 40 3-(4- 415 / 1.90 3.47 (t, 2H, J=7.6 Hz), 3.79 (s, 3H), 4.70 (t, ypyridin-3 - 2H, J=7.6 Hz), 6.91 (d, 1H, J=5.6 Hz), 7.30 y1)[2-(quinolin- (d, 1H, J=8 Hz), 7.47~7.51 (m, 2H), 7.68 (t, 2- 1H, J=7.2Hz), 7.77 (d, 1H, J=7.6 Hz), yl]thieno[2,3- 8.00~8.06 (m, 2H), 8.20 (s, 1H), 8.37 (s, d]pyridazin—4(5H)- 1H), 8.54 (d, 1H, J=5.6 Hz) 0116 153 LC-MS: 1H NMR (CDC13) 5: EX. Name 111/ e (M+H)+/ Rt [min] 41 3-(fi11rany1)[2- 374/214 3.53 (t, 2H, J=7.6 Hz), 4.77 (t, 2H, J=7.6 lin Hz), 6.76 (d, 1H, J=1.2 Hz), 7.35 (d, 1H, y1)ethy1]thien0[2,3- J=8.8 Hz), 7.46~7.52 (m, 3H), 7.68 (t, 1H, d]pyridazin—4(5H)- J=7.6 Hz), 7.78 (d, 1H, J=8 Hz), 8.02~8.08 0116 (m, 2H), 8.15 (s, 1H), 8.23 (s, 1H) 42 3-(quinolinyl) 435 / 1.93 3.50 (t, 2H, J=7.6 Hz), 4.76 (t, 2H, J=7.6 [2-(qu1nolin—2- Hz), 7.32 (d, 1H, J=8.4 Hz), 7.49 (t, 1H, y1)ethy1]thieno[2,3— J=7.6 Hz), 7.57 (t, 1H, J=7.6 Hz), 7.65-7.69 d]pyridazin—4(5H)— (m, 4H), 7.85 (d, 1H, J=8.4 Hz), 7.99 ((1, one 1H, J=8.4 Hz), 8.05 (d, 1H, J=8.4 Hz), 8.14 (d, 1H, J=8.4 Hz), 8.31 (s,1H), 9.06 (s, 1H) 43 3-(isoquinolin 435 / 1.89 3.42 (t, 2H,J=7.6 Hz), 4.62-4.68 (m, 2H), yl)[2-(quinolin— 7.25 (d, 1H, J=8.0 Hz),7.47-7.68 (m, 7H), thy1]— 7.76 (d, 1H, J=8.4 Hz), 7.95-8.04 (m, 3H), thieno[2,3-d]pyri- 8.29 (s, 1H), 8.45 (s, 1H), 8.30 (s, 1H) dazin—4(5H)-one 44 3-(isoquinolin 435 / 1.87 3.43 (t, 2H, J=7.6 Hz), 4.65 (t, 2H, J=7.6 yl)[2-(quinolin— Hz), 7.25-7.30 (m, 2H), 7.47-7.51 (m, 1H), 2- 7.56 (s, 1H), 7.63-7.68 (m, 3H), 7.76 (d, y1]thieno[2,3- 1H, J=8.0 Hz), 7.95-8.05 (m, 2H), 8.30 (s, d]pyridazin—4(5H)- 1H), 8.36 (d, 1H, J=7.0 Hz), 8.30 (s,1H) one 45 3-(1H-ind01—4-y1)- 423 /2.10 3.47 (t, 2H, J=7.6 Hz), 4.71 (t, 2H, J=7.6 5-(2-quinolin—2-yl— Hz), 6.26 (s, 1H), 7.05 (s, 1H, J=2.4 Hz), ethy1)-5H- 7.15~7.33 (m, 4H), 7.46 (t, 1H, J=7.2 thieno[2,3- Hz),7.61~7.67 (m, 2H), 7.74 (d, 1H, J=8.4 d]pyridazin—4—one Hz), 8.00 (d, 2H, J=8.4 Hz), 8.22 (s, 1H), 8.49 (s, 1H) 154 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 46 3-(2,3- 426 / 2.19 3.26 (t, 2H, J=8.4 Hz), 3.50 (t, 2H, J=7.6 dihydrobenzofuran— Hz), 4.61 (t, 2H, J=8.8 Hz), 4.74 (t, 2H, 5-y1)(2-quinolin- J=7.6 Hz), 6.81 (d, 1H, J=8.4 Hz), 2-y1-ethy1)-5H- 7.23~7.40 (m, 4H), 7.49 (t, 1H, J=7.6 Hz), thieno[2,3- 7.67 (t, 1H, J=7.6 Hz), 7.77 (d, 1H, J=8 d]pyridazinone Hz), 8.00~8.06 (m, 2H), 8.19 (s, 1H) 47 3-(quinolin—5—y1)—5— 435/ 1.88 3.43 (t, 2H, J=7.6 Hz), 3.98 (s, 3H), 4.74 (t, [2-(quin0 1111—2— 2H, J=7.6 Hz), .27 (m, 2H), 7.47-7.51 yl)ethy1]thieno[2,3— (m, 2H), 7.55 (s, 1H), 7.64-7.77 (m, 3H), d]pyridazin—4(5H)- 7.83 (d, 1H, J=8.4 Hz), 7.97 (d, 1H, J=8.4 0116 Hz), 8.01 (d, 1H, J=8.4 Hz), 8.18 (d, 1H, J=8.8 Hz), 8.29 (S, 1H), 8.90 (d, 1H, J=2.8 Hz) 48 3-(3,5-dirnethy1— 403 /2.01 2.07 (s, 3H), 2.25 (s, 3H), 3.49 (t, 2H, J=7.6 1,2-0xaz01—4-y1) Hz), .75 (m, 2H), 7.35 (t, 2H, J=8.4 [2-(quin01in—2- Hz), 7.49 (t, 1H, J=8 Hz), 7.66 (t, 1H, y1]thieno[2,3- J=8.4 Hz), 7.77 (d, 1H, J=8.4 Hz), 7.98 (d, d]pyridazin—4(5H)- 1H, J=8 Hz), 8.06 (d, 1H, J=8.4 Hz), 8.22 one (s, 1H) 49 3-(6- 415 /2.09 3.49 (t, 2H, J=7.6 Hz), 4.65 (t, 2H, J=7.6 methoxypyridin-3 - Hz), 6.79 (d, 1H, J=8.4 Hz), 7.33 (d, 1H, y1)[2-(quinolin- J=8.8 Hz), 7.47—7.52 (m, 2H), 7.65-7.69 (m, 2- 2H), 7.75-7.79 (m, 2H), 8.00 (d, 1H, J=8.4 yl)ethyl]thieno[2,3- Hz), 8.06 (d, 1H, J=8.4 Hz), 8.22 (s, 1H), d]pyridazin—4(5H)- 8.28 (d, 1H, J=2.0 Hz) 0116 155 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e / Rt [min] 50 3-(2,3-dihydro-1,4- 442 /2.17 3.50 (t, 2H, J=7.6 Hz), 4.30 (s, 4H), 4.74 (t, benzodioxiny1)- 2H, J=7.6 Hz), 6.90 (d, 1H, J=8.8 Hz), 5-[2-(quinolin 7.00~7.02 (m, 1H), 7.08 (d, 1H, J=2 Hz), y1)ethy1]thieno[2,3- 7.32 (d, 1H, J=8 Hz), 7.42 (s, 1H), 7.49 (t, dazin-4(5H)- 1H, J=8Hz), 7.67 (t, 1H, J=6.8 Hz), 7.77 ((1, one 1H, J=8 Hz), 8.01~8.06 (m, 2H), 8.18 (s, 1H) 51 3-(2-rnethylpyridin— 399/ 1.97 2.61 (s, 3H), 3.50 (t, 2H, J=7.6 Hz), 4.75 (t, 4-y1)[2- 2H, J=7.6 Hz), 7.22-7.28 (m, 2H), 7.33 (d, lin 1H, J=8.0 Hz), 7.47-7.51 (m, 1H), 7.56 (s, yl)ethy1]thieno[2,3- 1H), 7.65-7.69 (m, 1H), 7.78 (d, 1H, J=7.6 d]pyridazin—4(5H)- Hz), 7.98 (d, 1H, J=8.4 Hz), 8.06 (d, 1H, one J=8.4 Hz), 8.23 (s, 1H), 8.52 (d, 1H, J=5.2 Hz). 52 3-(5- 415 / 1.96 3.50 (t, 2H, J=7.6 Hz), 3.87 (s,3H), 4.75 (t, methoxypyridin 2H, J=7.6 Hz), 7.45-7.51 (m, 2H), 7.57 (s, y1)[2-(quinolin- 1H), 7.67 (t, 1H, J=7.6 Hz), 7.77 (d, 1H, 2- J=8.0 Hz), 7.99 (d, 1H, J=8.4 Hz), 8.05 (d, yl)ethy1]thieno[2,3- 1H, J=8.4 Hz), 8.23 (s,1H), 8.33 (m, 2H) d]pyridazin—4(5H)- one 53 3-[6-(morpholin 470 / 2.05 3.50 (t, 2H, J=7.6Hz), 3.58 (t, 4H, J=4.8 yl)pyridiny1]—5- Hz), 3.84 (t, 4H, J=4.8 Hz), 4.75 (t, 2H, [2-(quino 1in J=7.6 Hz), 6.69 (d, 1H, J=8.8 Hz), 7.32 (d, y1)ethy1]thieno[2,3- 1H, J=8.4 Hz), 7.44 (s, 1H), 7.49 (t, 1H, d]pyridazin—4(5H)— J=7.2 Hz), 7.68 (m, 1H, J=8.4 Hz), 0116 7.76~7.79 (m, 2H), 8.00~8.07 (m, 2H), 8.20 (s, 1H), 8.33 (d, 1H, J=2 Hz) 156 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 54 3-(1,3-benzodioxol— 428 /2.16 3.50 (t, 2H, J=7.6Hz), 4.74(t, 2H, z), 5-y1)[2- 6.00 (s, 2H), 6.84 (d, 1H, J=8Hz), (quinolin 6.95~7.01 (m, 2H), 7.33 (d, 1H, J=8.4Hz), y1]thieno[2,3- 7.42 (s, 1H), 7.49 (t, 1H, J=7.6Hz), 7.67 (t, d]pyridazin-4(5H)- 1H, J=7.2Hz), 7.77 (d, 1H, J=8.4Hz), one 8.00~8.06 (m, 2H), 8.19 (s, 1H) 55 3-(quinolin—6—y1)—5— 435 /2.03 3.50 (t, 2H, J=7.6 Hz), 3.95 (d, 2H, J=7.2 [2-(quinolin—2— Hz), 4.75 (t, 2H, J=7.6 Hz), 7.21 (d, 1H, yl)ethy1]thieno[2,3— J=8.4 Hz), 7.40-7.50 (m, 2H), 7.60 (s, 1H), d]pyridazin—4(5H)- 7.65—7.69 (m, 1H), 7.77-7.84 (m, 2H), 7.94 one ((1, 1H, J=2.4 Hz),7.99 (d, 1H, J=8.4 Hz), 8.05 (d, 1H, J=8.4 Hz), 8.12 (d, 1H, J=8.4 Hz), 8.17 (d, 1H, J=8.0 Hz), 8.25 (s, 1H), 8.93—8.94 (m, 1H) 56 3-(1-rnethy1—1H- 388 / 1.93 3.52 (t, 2H, J=7.6 Hz), 3.95 (s, 3H), 4.77 (t, l—4-yl)[2- 2H, J=7.6 Hz), 7.34 (d, 1H, J=8.4 Hz), 7.48- (quinolin 7.55 (m, 2H), 7.66-7.71 (m, 1H),7.77-7.80 yl)ethy1]thieno[2,3- (m, 2H), 8.02-8.08 (m, 2H), 8.16 (s, 1H), d]pyridazin—4(5H)- 8.23 (s, 1H) one 57 3-[1-(2- 430 / 2.17 0.95 (d, 6H, J=6.8 Hz), 2.26 (m, 1H), 3.54 methylpropyl)- 1 H- (t, 2H, J=7.6 Hz), 3.95 (d, 2H, J=7.2 Hz), pyrazo 1—4-y1][2- 4.78 (t, 2H, J=7.6 Hz), 7.34 (d, 1H, J=8.4 (quinolin Hz), 7.50-7.54 (m, 2H), 7.69 (t, 1H, J=7.6 y1)ethy1]thieno[2,3- Hz), 7.78 (d, 1H, J=8.4 Hz), 7.82 (s,1H), d]pyridazin—4(5H)— 8.02-8.08 (m, 2H), 8.14 (s, 1H), 8.28 (s, 0116 1H) 157 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 58 tert-buty12-{4-0X0- 473 /2.11 1.35 (s, 9H), 3.46 (t, 2H, J=7.6 Hz), 4.68 (t, 5-[2-(quinolin 2H, J=7.6 Hz), 6.23~6.28 (m, 2H), 7.29 (d, y1)ethy1]-4,5- 1H, J=8.4 Hz), .51 (m, 3H), 7.69 (t, dihydrothien0[2,3- 1H, J=8 Hz), 7.77 (d, 1H, J=7.6 Hz), 8.05 d]pyridazin—3-yl}- (d, 2H, J=8 Hz), 8.15 (s, 1H) lH-pyrrole-l- carboxylate 59 3-(2- 416/ 1.95 3.50 (t, 2H, J=7.6 Hz), 4.07 (s, 3H), 4.75 (t, methoxypyrimidin— 2H, J=7.6 Hz), 7.35 (d, 1H, J=8.4 Hz), 7.54 5-y1)[2- (s, 1H), 7.67 (t, 2H, J=9.2 Hz), 7.79 (d, 1H, (quinolin J=7.6 HZ), 7.98 (d, 1H, J=8.4 Hz), 8.07 (d, yl)ethy1]thieno[2,3- 1H, J=8.4 Hz), 8.24 (s, 1H), 8.67 (s, 2H) d]pyridazin—4(5H)- one 60 5-[2-(quinolin 438 / 1.60 3.88 (t, 2H, J=6.0 Hz), 4.76 (t, 2H, J=6.0 y1]—3-(2,3,4- Hz), 6.88-7.02 (m, 2H), 7.55 (s, 1H), 7.68 trifluorophenyl)thie (d, 1H, J=7.6 Hz), 7.82 (t, 1H, J=7.6 Hz), no[2,3-d]pyridazin- 7.98-8.06 (m, 2H), 8.24 (s, 1H), 8.48 (d, 4(5H)-one 1H, J=8.4 Hz), 8.63 (d,1H,J=8.4 Hz) 61 u0r0 416 / 2.28 2.24 (d, 3H, J=1.6 Hz), 3.42 (t, 2H, J=7.6 methylpheny1)-5 - Hz), 4.66 (t, 2H, J=7.6 Hz), 6.95 (t, 1H, [2-(quino 1111 J=8.8 Hz), 7.19~7.24 (m, 3H), 7.35 (s, 1H), y1)ethy1]thien0[2,3- 7.44 (t, 1H, J=6.8 Hz), 7.58~7.62 (m, 1H), d]pyridazin—4(5H)- 7.70 (d, 1H, J=8.4 Hz), 7.93~7.99 (m, 2H), 0116 8.13 (s, 1H) 158 LC-MS: 1H NMR (CDC13) 5: EX. Name 111/ e (M+H)+/ Rt [min] 62 3-(4-fluoro 416/ 1.61 3.87 (t, 2H, J=6.0 Hz), 4.73 (t, 2H, J=6.0 methylpheny1) Hz), 6.78-7.03 (m, 3H), 7.38 (s, 1H), 7.68 [2-(quinolin—2- (d, 1H, J=8.4 Hz), 7.82 (t, 1H, J=7.6 Hz), y1)ethy1]thieno[2,3- 7.98-8.04 (m, 2H), 8.25 (s, 1H), 8.48 (d, dazin-4(5H)- 1H, J=8.4 Hz), 8.60 (d, 1H, J=8.4 Hz) one 63 hloro—4— 436 / 2.09 3.50 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 fluoropheny1)—5—[2— Hz), 7.15 (t, 1H, J=8.8 Hz), 7.33-7.38 (m, (quinolin 2H), 7.47—7.53 (m, 3H), 7.68 (t, 1H, J=7.6 y1)ethy1]thieno[2,3- Hz), 7.79 (d, 1H, J=7.6 Hz), 7.99-8.08 (m, d]pyridazin—4(5H)- 2H), 8.22 (s, 1H) one 64 3-(2-chloro 436 / 2.24 3.47 (t, 2H,J=7.6 Hz), 4.71 (t, 2H, J=7.6 fluoropheny1)[2- Hz), 6.99-7.04 (m, 1H), 7.31-7.44 (m, 2H), (quinolin 7.47-7.51 (m, 2H), .69 (m, 1H), 7.76 y1)ethy1]thieno[2,3- (d, 1H, J=8.0 Hz), 7.98 (d, 1H, J=8.8 Hz), d]pyridazin—4(5H)- 8.03 (d, 1H, J=8.8 Hz), 8.22 (s, 1H) one 65 3-(3,4- 412 /2.13, 2.30 (s, 6H), 3.49 (t, 2H, J=7.6 Hz), 4.73 (t, dimethylpheny1)-5 - 2H, J=7.6 Hz), 7.18 (d, 1H, J=7.6 Hz), 7.24— [2-(quino 11n 7.27 (m, 2H), 7.31 (d, 1H, J=8.4 Hz), 7.43 y1)ethy1]thieno[2,3- (s, 1H), 7.46-7.50 (m, 1H), 7.65-7.69 (m, d]pyridazin—4(5H)- 1H), 7.77 (d, 1H, J=8.4 Hz), 8.03 (t, 2H, 0116 J=8.8 Hz), 8.18 (s, 1H). 159 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 66 3-(2,4- 412 /2.12 2.05 (s, 3H), 2.36 (s, 3H), 3.47 (t, 2H, J=7.6 dimethylpheny1) Hz), 4.69 (t, 2H, J=7.6 Hz), 7.02~7.08 (In, [2-(quinolin—2- 4H), 7.29 (d, 1H, J=8.8 Hz), 7.35 (s, 1H), y1]thieno[2,3- 7.48 (t, 1H, J=7.2 Hz), 7.66 (t, 1H, J=7.2 d]pyridazin-4(5H)- Hz), 7.76 (d, 1H, J=8 Hz), 8.01 (t, 1H, one J=8.8 Hz), 8.21 (s, 1H) 67 3-(2,4— 420 / 2.19 3.48 (t, 2H, J=7.6 Hz),4.73 (t, 2H, J=7.6 difluoropheny1)—5— Hz), 6.89—6.94 (m, 2H), 7.29-7.36 (m, 2H), [2-(quinolin—2- 7.47—7.53 (m, 2H), 7.67-7.69 (m, 1H) 7.76 y1]thieno[2,3- (d, 1H, J=8.0 Hz), 8.00 (d, 1H, J=8.4 Hz), d]pyridazin—4(5H)- 8.04 (d, 1H, J=8.4 Hz), 8.21 (s, 1H) one 68 3-(2,4- 444 / 2.17 3.47 (t, 2H, J=7.6 Hz), 3.72 (s,3H), 3.84 (s, dimethoxypheny1)- 3H), 4.69 (t, 2H, J=7.6 Hz), 6.52-6.55 (m, 5-[2-(quinolin 2H), 7.19 (s, 1H, J=8.0 Hz), 7.30 (d, 1H, y1)ethy1]thieno[2,3- J=8.0 Hz), 7.44 (s, 1H) 7.46-7.50 (m, 1H), d]pyridazin—4(5H)- 7.65-7.77 (m, 2H), 8.03 (d, 2H, J=8.4 Hz), one 8.16 (s, 1H) 69 - 444 / 2.16 3.46~3.50 (m, 2H), 3.69 (s, 3H), 3.79 (s, dimethoxypheny1)- 3H), 4.69 (t, 2H, J=7.6 Hz), 6.87 (d, 1H, 5-[2-(quinolin J=2 Hz), 6.91 (s, 2H), 7.30 (d, 1H, J=8.4 y1)ethy1]thieno[2,3- Hz), 7.49 (t, 2H, J=6.8 Hz), 7.68 (t, 1H, d]pyridazin—4(5H)- J=8.4 Hz), 7.76 (d, 1H, J=8.4 Hz), 0116 8.02~8.05 (m, 2H), 8.17 (s, 1H) 160 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 70 3-(2,3- 420 /2.19 3.50 (t, 2H, J=7.6 Hz), 4.73 (t, 2H,J=7.6 difluoropheny1) Hz), 7.10-7.13 (m, 2H), 7.18-7.23 (m, 1H), [2-(quinolin—2- 7.31 (d, 1H, J=8.4 Hz), 7.49 (t, 1H, J=7.6 y1)ethy1]thieno[2,3- Hz), 7.57 (s,1H), 7.68 (t, 1H, J=7.6 Hz), d]pyridazin-4(5H)- 7.77 (d, 1H, J=7.6 Hz), 8.01-8.06 (m, 2H), one 8.22 (s, 1H) 71 3-(3,4— 444/ 1.95, 3.50 (t, 2H, J=7.6 Hz), 3.88 (s, 3H), 3.92 (s, dimethoxyphenyl)— 3H), 4.75 (t, 2H, J=7.6 Hz), 6.92 (d, 1H, 5-[2-(quinolin J=8.4 Hz), .11 (m, 2H), 7.32 (d, 1H, y1)ethy1]thieno[2,3- J=8.4Hz), .51 (m, 2H), 7.68 (t, 1H, d]pyridazin—4(5H)- J=7.6Hz), 7.77 (d, 1H, J=7.6 Hz), 8.04 (t, one 2H, J=8.8 Hz), 8.20 (s, 1H) 72 3-(3,4- 420 / 2.22 3.49 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 difluoropheny1) Hz), 7.15-7.21 (m, 2H), 7.28-7.35 (m, 2H), [2-(quinolin—2- 7.47 (s, 1H), 7.50 (d, 1H, J=7.2 Hz), 7.65- y1)ethy1]thieno[2,3- 7.69 (m, 1H), 7.78 (d, 1H, J=8.0 Hz), 7.98 d]pyridazin—4(5H)- (d, 1H, J=8.4 Hz), 8.06 (d, 1H, J=8.0 Hz), one 8.22 (s, 1H) 73 3-(5-fluoro 432 /2.19 3.48 (t, 2H, J=7.6 Hz), 3.71 (s, 1H), 4.71 (t, methoxyphenyl)-5 - 2H, J=7.6 Hz), 6.87-7.08 (m, 3H), 7.30 (d, ino lin 1H, J=8.8 Hz), 7.47—7.51 (m, 2H), 7.68 (t, y1)ethy1]thieno[2,3- 1H, J=7.6 Hz), 7.77 (d, 1H, J=8.4 Hz), 8.03 d]pyridazin—4(5H)- (t, 2H, J=8.0 Hz), 8.18 (s, 1H). 0116 161 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 74 3-(4-fluoro 432 /2.01, 3.47 (t,2H, J=7.6Hz),3.72 (s, 3H), 4.70 (t, methoxypheny1) 2H, J=7.6 Hz), 6.70-6.72 (m, 2H), 7.18-7.22 [2-(quinolin—2- (m, 1H), 7.44 ,7.49 (t, 1H, J=7.6 Hz), y1)ethy1]thieno[2,3- 7.67 (d, 1H, J=7.6 Hz), 7.77 (d, 1H, J=8.4 d]pyridazin-4(5H)- Hz),7.99 (d, 1H, J=8.4 Hz), 8.02 (t, 2H, one J=8.8 Hz), 8.17 (s,1H) 75 — 444 / 2.20 3.50 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 dimethoxyphenyl)— Hz), 6.50 (t, 1H, J=2.4 Hz), 6.68 (d, 2H, 5-[2-(quinolin J=2.4 Hz), 7.31 (d, 1H, J=7.6 Hz), 7.46- yl)ethy1]thieno[2,3- 7.50 (m, 2H), 7.65-7.69 (m, 1H), 7.76 (d, dazin—4(5H)— 1H, J=8.4 Hz), 8.00-8.05 (m, 2H), 8.19 (5, one 1H) 76 3-(2,5- 420 /2.19 3.49 (t, 2H, J=6.0 Hz), 4.73 (t, 2H, J=6.0 difluoropheny1) Hz), 7.06-7.11 (n1,3H), 7.31 (d, 1H, J=8.4 [2-(quinolin—2- Hz), 7.48 (t,1H, J=8.4 Hz), 7.56 (s, 1H), yl)ethy1]thieno[2,3- 7.64-7.69 (m, 1H), 7.76 (d, 1H, J=8.0 Hz), d]pyridazin—4(5H)— 8.00 (d, 1H, J=8.4 Hz), 8.04 (d, 1H, J=8.4 one Hz), 8.21 (s, 1H) 77 3-(2,3- 444 / 2.15 3.48 (t, 2H, J=7.6 Hz), 3.59 (s, 3H), 3.89 (s, dimethoxyphenyl)- 3H), 4.71 (t, 2H, J=7.6 Hz), 6.86 (dd, 1H, 5-[2-(quinolin J=1.2 Hz, J=7.6 Hz), 6.97 (d, 1H, J=8.0 y1)ethy1]thieno[2,3- Hz), 7.05-7.09 (m, 1H), 7.30 (d, 1H, J=8.0 d]pyridazin—4(5H)- Hz), 7.47—7.51 (m, 2H), 7.67 (t, 1H, J=7.6 0116 Hz), 7.76 (d, 1H, J=8.0 Hz), 8.00-8.04 (m, 2H), 8.20 (s, 1H) 162 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 78 uoro 432 /2.19 3.50 (t, 2H, J=7.6 Hz), 3.93 (s, 3H), 4.74(t, methoxypheny1) 2H, J=7.6 Hz), 6.99 (t, 1H, J=8.4 Hz), 7.23- [2-(quinolin—2- 7.27 (m, 2H), 7.33 (d, 1H, J=8.4 Hz), 7.44 y1)ethy1]thieno[2,3- (s, 1H), 7.49 (t, 1H, J=7.6 Hz), .69 d]pyridazin-4(5H)- (m, 1H), 7.78 (d, 1H, J=8.0 Hz), 8.00 ((1, one 1H, J=8.0 Hz), 8.05 (d, 1H, J=8.4 Hz), 8.19 (s, 1H) 79 3-(2-fluoro—3— 432 / 2.17 3.48 (t, 2H, J=7.6 Hz), 3.92 (s,3H), 4.72 methoxypheny1)-5— (t,2H,J=7.6 Hz), 6.93-7.13 (m, 3H), 7.29 (d, [2-(quinolin—2- 1H, J=8.0 Hz), 7.46-7.50 (m, 1H), 7.55 (s, y1)ethy1]thieno[2,3- 1H), 7.65-7.69 (m, 1H), 7.77 (d, 1H, J=8.0 d]pyridazin—4(5H)— Hz), 8.01-8.04 (m, 2H), 8.19 (s, 1H) one 80 3-(3,5- 420 / 2.24 3.50 (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=7.6 difluoropheny1) Hz), .85 (m, 1H), 7.01-7.03 (m, 2H), [2-(quinolin—2- 7.34 (d,1H, J=8.4 Hz), 7.47-7.51 (m, 2H), y1)ethy1]thieno[2,3- 7.66 (t, 1H, J=8.4 Hz), 7.77 (d, 1H, J=8.4 d]pyridazin—4(5H)— Hz), 7.98 (d, 1H, J=8.4 Hz), 8.06 (d, 1H, one J=8.4 Hz), 8.21 (s, 1H) 81 3-(3-fluoro 432 /2.23 3.51 (t, 2H, J=7.6 Hz), 3.81 (s, 1H), 4.75 (t, methoxyphenyl)-5 - 2H, J=7.6 Hz), 6.63-6.67 (m, 1H), 6.79-6.82 [2-(quinolin—2- (m, 1H), 6.87 (s, 1H), 7.33 (d, 1H, J=8.4 y1)ethy1]thieno[2,3- Hz), 7.47-7.51 (m, 2H), 7.67 (t, 1H, J=7.6 d]pyridazin-4(5H)- Hz), 7.78 (d, 1H, J=8.0 Hz), 8.00-8.07 (m, 0116 2H), 8.20 (s, 1H). 163 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 82 3-(2-methoxy 428/ 1.61 2.21 (s, 3H), 3.59 (s, 3H), 3.78 (t, 2H, J=6.0 phenyl) Hz), 4.66 (t, 2H, J=6.0 Hz), 6.77 (d, 1H, [2-(quinolin—2- J=8.4 Hz), 6.94 (s, 1H), 7.07 (d, 1H, J=8.4 y1)ethy1]thieno[2,3- Hz), 7.37 (s, 1H),7.50 (d, 1H, J=8.4 Hz), d]pyridazin-4(5H)- 7.69 (t, 1H, J=7.6 Hz), .92 (m, 2H), one 8.05 (s,1H), 8.45 (m, 2H) 83 3-(2,5— 452 / 2.09 3.48 (t, 2H, J=7.6 Hz), 4.72 (t, 2H, J=7.6 dichlorophenyl)—5— Hz), 7.29~7.33 (m, 3H), 7.39 (d, 1H, J=8 [2-(quinolin—2- Hz), 7.48 (t, 2H, J=6.4 Hz), 7.66 (t, 1H, y1]thieno[2,3- J=7.2 Hz), 7.76 (d, 1H, J=8 Hz), 7.98~8.04 d]pyridazin—4(5H)- (m, 2H), 8.22 (s, 1H) one 84 3-(naphthalen 434 / 2.11 3.55 (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=7.6 y1)[2-(quinolin- Hz), 7.35 (d, 1H, J=8.4 Hz), 7.48~7.63 (n1, 2- 5H), 7.71 (t, 1H, J=7.2 Hz), 7.79~7.87 (n1, yl)ethy1]thieno[2,3- 4H), 7.94 (s, 1H), 8.09 (t, 2H, J=8.4 Hz), d]pyridazin—4(5H)- 8.23 (s, 1H) one 85 3-pheny1—5-[2- 384 / 2.19 3.50 (t, 2H, J=7.6 Hz), 4.74 (t, 2H, J=7.6 (quinolin Hz), 7.32 (d, 1H, J=8.4 Hz), 7.39~7.53 (n1, y1)ethy1]thieno[2,3- 7 H), 7.67 (t, 1H, J=7.6 Hz), 7.77 (d, 1H, d]pyridazin—4(5H)- J=7.6 Hz), 8.04 (t, 2H, J=8.8 Hz), 8.20 (5, one 1H) 86 3-(1-benzofuran—2- 423 / 1.70 3.75 (t, 2H, J=6.8 Hz),4.79 (t, 2H, J=6.8 y1)[2-(quinolin- Hz), 7.14-7.18 (m,1H), 7.24 (t, 1H,J=7.2 2- Hz), 7.07 (d,1H, J=8.4 Hz), 7.40-7.61 (m, y1)ethy1]thieno[2,3— 4H), 7.77-7.85 (m, 2H), 8.00 (s, 1H), 8.10 d]pyridazin—4(5H)- (s, 2H), 8.30-8.34 (m, 2H) 0116 164 LC-MS: 1H NMR (CDC13) 5: EX. Name m/ e (M+H)+/ Rt [min] 87 3-(1H-indazol—5- 424/200 3.50 (t, 2H, J=7.6 Hz), 4.75 (t, 2H, J=7.6 y1)[2-(quinolin— Hz), 7.32 (d, 1H, J=8.4 Hz), 6.63-6.67 (m, 2- 1H), 6.79-6.82 (m, 1H), 6.87 (s, 1H), 7.33 y1)ethy1]thieno[2,3- (d, 1H, J=8.4 Hz), .51 (m, 2H), 7.67 d]pyridazin-4(5H)- (t, 1H, J=7.6 Hz), 7.78 (d, 1H, J=8.0 Hz), one 8.00-8.07 (m, 2H), 8.20 (s, 1H). 88 3-(1—methy1—1H— 388 / 1.89 3.49 (t, 2H, J=7.6 Hz), 3.56 (s, 3H), 4.73 (t, l—5-y1)—5—[2— 2H, J=7.6 Hz), 6.31 (d, 1H, J=2.0 Hz), 7.34 (quinolin (d, 1H, J=8.0 Hz), 7.47-7.52 (m, 2H), 7.56 yl)ethy1]thieno[2,3- (s, 1H), 7.64-7.69 (m, 1H), 7.77 (d, 1H, d]pyridazin—4(5H)- J=8.0 Hz), 7.96 (d, 1H, J=8.8 Hz), 8.06 ((1, one 1H, J=8.4 Hz), 8.25 (s, 1H) 89 3-(4,5-difluoro 450 / 2.22 3.47 (t, 2H, J=7.6Hz), 3.60 (s, 3H), 4.70 (t, methoxypheny1) 2H, z), 6.75~6.80 (m, 1H), 7.07 (t, [2-(quinolin—2- 1H, J=7.6 Hz), 7.30 (d, 1H, J=8.4 Hz), y1)ethy1]thieno[2,3- 7.45~7.50 (m, 2H), 7.67 (t, 1H, J=7.2 Hz), d]pyridazin—4(5H)- 7.77 (d, 1H, j=8 Hz), 7.99~8.05 (m, 2H), one 8.18 (s, 1H) 90 3-(2-fluoro 416 / 1.60 2.26 (s, 3H), 3.77 (t, 2H, J=6.0 Hz), 4.68 (t, methylpheny1)-5 - 2H, J=6.0 Hz), 6.73 (d, 1H, J=8.4 Hz), 6.83 [2-(quino lin (d, 1H, J=8.0 Hz), 7.05 (t, 1H, J=8.0 Hz), y1)ethy1]thieno[2,3- 7.44(s,1H),7.60 (d, 1H, J=8.4 Hz), 7.75 (t,1 d]pyridazin—4(5H)- H, J=8.0 Hz),7.90-7.99 (m, 2H), 8.14 (s, 0116 1H), 8.28 (d, 1H, J=8.4 Hz), 8.59 (d, 1H, J=8.4 Hz) 165 LC-MS: 1H NMR ) 8: EX. Name m/ e (M+H)+/ Rt [min] 91 3-(2-fluoro 432/218 3.49 (t, 2H, J=7.6 Hz), 3.80 (s, 3H), 4.73 (t, methoxyphenyl)-5 - 2H, J=7.6 Hz), 6.88-6.93 (m, 2H), 7.08 (t, [2-(quinolin—2- 1H, J=8.8 Hz), 7.30 (d, 1H, J=8.4 Hz), 7.46- yl)ethyl]thieno[2,3- 7.51 (m, 1H), 7.65-7.69 (m, 1H), 7.55 (s, d]pyridazin-4(5H)- 1H), 7.65-7.69 (m, 1H), 7.76 (d, 1H, J=8.4 one Hz), 8.03 (t, 2H, J=8.0 Hz), 8.20 (s, 1H). 92 y1—4— {4—oxo— 423 / 1.54 1.99 (s, 3H), 3.84 (t, 2H, J=6.0 Hz), 4.73 5-[2-(quinolin—2— (t,2H,J=6.0 Hz), 7.15 (d, 1H, J=8.0 Hz), yl)ethyl]—4,5- 7.39-7.46 (m, 3H), 7.77 (d, 1H, J=8.4 Hz), dihydrothieno [2 ,3 - 7.87 (t, 1H, J=8.0 Hz), 8.02-8.09 (m, 2H), d]pyridazin—3- 8.32 (s, 1H), 8.36 (d, 1H, J=8.4 Hz), 8.69 yl}benzonitrile (d, 1H, J=8.4 Hz) E 93: 5-[2-(6-F1uoroquinoliny1)ethy1](pyridinyl)thieno[2,3- d]pyridazin—4(5H)-one 93. 1 3-Bromo(2-(6-fluoroquino1in-2—yl)ethyl)thieno[2,3-d]pyridazin—4(5H)-one 2-(6-Fluoroquinolinyl)ethanol (182 mg, 0.952 mmol) from Example a2) and 3- bromo-5H-thieno[2,3-d]pyridazin-4(5H)-one (200 mg, 0.865 mmol) from Example 3.3 were dissolved in THF (10 mL) and stirred for about 10 min. Then Ph3P (342 mg, 1.305 mmol) and DEAD (226 mg, 1.298 mmol) were each added sequentially rapidly to the 10 solution. The reaction mixture was stirred under nitrogen atmosphere overnight. The reaction solution was concentrated and d by TLC (PE/EA=1/ l). The crude product was recrystallized from methanol (130 mg, yield: 37.1%). LC-MS: m/e (M+H)+; Rt : 1.90 min. 166 93.2 5-[2-(6-Fluoroquinolinyl)ethyl]—3—(pyridinyl)thieno[2,3-d]pyridazin-4(5H)- one 3-Bromo(2-(6-fluoroquinolin-2—yl)ethyl)thieno[2,3-d]pyridazin—4(5H)-one (66 mg, 0.163 mmol) was dissolved in dioxane (2.1 mL) and H20 (0.7 mL), nyl boronic acid (20.07 mg, 0.163 mmol), K2CO3 (45.1 mg, 0.327 mmol) and Pd(dppf)C12 (7.23 mg, 9.80 umol) were each added sequentially to the suspension. The suspension was heated in a microwave tube at about 110 °C for 1 h. The crude product was purified by TLC (EA) to give the title compound (45 mg, yield: . 10 LC—MS: m/e (M+H)+: 403.7, Rt: 1.81 min; lH NMR(CDC13, 400 MHz) 8: 8.65 (d, J = 6.0 Hz, 2H), 8.24 (s, 1H), 8.02—7.96 (m, 2H), 7.61 (s, 1H), 7.46-7.42 (m, 3H), 7.41-7.38 (m, 1H), 7.34 (d, J = 8.4 Hz, 1H), 4.75 (t, J = 7.4 Hz, 2H), 3.48 (t, J = 7.4 Hz, 2H). 15 EXAMPLE 94: 5-[2,2-Difluoro(quinolin—2—yl)ethyl](pyridinyl)thieno[2,3- d]pyridazin—4(5H)-one N / \ \ O / / N N I F \ F | N \ S 94.1 3-Bromo(2,2-difluoro-2—(quinolin-2—yl)ethyl)thieno[2,3-d]pyridazin—4(5H)-one A mixture of 3-bromothieno[2,3-d]pyridazin—4(5H)-one (10mg, 0.043 mmol) 20 from Example 3.3, trifluoromethanesulfonic acid 2,2-difluoroquinolinyl ethyl ester (14.77 mg, 0.043 mmol) from Example b1 and CszC03 (28.2 mg, 0.087 mmol) in DMF (0.5 mL) was d at room temperature for 5 h. The solution was purified by Pre-TLC (PE/EA =1/2) to give a dark yellow oil (4 mg, yield 22%).
LC-MS : m/e 422 (M+H)+, Rt: 1.96 min. 25 94.2 5-[2,2—Difluoro—2—(quinolin—2—yl)ethyl]—3—(pyridinyl)thieno[2,3-d]pyridazin— 4(5H)—one A mixture of 3-bromo—5—(2,2—difluoro—2— (quinolinyl)ethyl)thieno[2,3- d]pyridazin—4(5H)-one (0.118 mmol), pyridin—4—yl boronic acid (0.118 mmol), Na2C03 167 (31.4 mg, 0.296 mmol) and PdC12(dppf)—CH2C12 adduct (9.67 mg, 0.012 mmol) in dioxane (1.5 mL) and water (0.5 mL) was stirred at 100 °C for 2h. The solvent was evaporated and the residue was purified by C =1/ 1) to give a crude yellow solid. The solid was dissolved in DCM (1.5 mL) and filtered. The white solid obtained was the title compound (40 mg, yield 80%).
LC-MS (ESI+): m/e 421 (M+H)+, Rt: 2.00 min; 1H NMR (CDCl3, 400MHz) 8: 8.70 (s, 1H), 8.61 (d, J: 8.4 Hz, 1H), 8.50 (dd, J: 4.4 Hz, 1.2Hz, 1H), 8.22 (s, 1H), 8.13 (d, J= 8.4 Hz, 1H), 8.03 (d, J= 8.4 Hz, 1H), 7.88-7.84 (m, 1H), 7.80 (d, J= 8.4 Hz, 1H), 7.76-7.73 (m, 1H), 7.30 (dd, J= 4.4 Hz, 2.0Hz, 2H), 5.15 (t, J= 14 Hz, 2H). 10 EXAMPLE 95: idin—4—yl)—5—[2—(thieno[3,2—b]pyridin—5-yl)ethyl]thieno[2,3- d]pyridazin—4(5H)—one N N \ S 95. l 3-Bromo[2-(thieno [3 ,2-b]pyridin—5—yl)ethyl]thieno [2,3 -d]pyridazin—4(5H)-one 15 To a e of 3-bromothieno[2,3-d]pyridazin-4(5H)-one (100 mg, 0.433 mmol) from Example 3.3 and triphenylphosphine (227 mg, 0.866 mmol) in THF (2 mL) was added (E)-diethyl diazene-l,2-dicarboxy1ate (151 mg, 0.866 mmol) se at 0°C.
After the addition, the mixture was stirred for 1h at 0°C. Then 2-(thieno[3,2-b]pyridin— 5-yl)ethanol from Example a3 (78 mg, 0.433 mmol) in THF(1 mL) was added 20 dropwise. The mixture was stirred at room temperature overnight. The solution was filtered to obtain the title compound (80 mg, yield 47.1 %).
LC-MS (ESI+): m/e 392 (M+H)+, Rt: 1.60 min; 1H NMR (CDC13, 400MHz) 8: 8.67 (d, J: 4.4 Hz, 2H), 8.24 (s, 1H), 8.09 (d, J: 6.4 Hz, 1H), 7.72 (d, J: 4.4 Hz, 1H), 7.60 (s, 1H), 7.49 (d, J= 4.4 Hz, 1H), 7.45 (d, J= 4.8 Hz, 2H), 7.17 (d, J= 6.4 Hz, 1H), 25 4.7 (t, J= 6.4 Hz, 2H), 3.44 (t, J= 6.0 Hz, 2H). 95.2 3-(Pyridin—4—yl)—5—[2—(thieno[3,2—b]pyridin—5—yl)ethyl]thieno[2,3-d]pyridazin- 4(5H)—one 168 A mixture of pyridinylboronic acid (37.6 mg, 0.306 mmol), the compound from Example 95.1 (80 mg, 0.204 mmol), Na2C03 (54.0 mg, 0.510 mmol) and PdClz(dppf)-CH2C12 adduct (16.65 mg, 0.020 mmol) in dioxane (3 mL) and water (1 mL) was stirred at 100 °C in a microwave for 10 min. The solvent was evaporated and the residue was washed with methanol, d, the filtrate was trated and purified by HPLC to afford the title nd (59 mg, yield 74.1 %) as a white solid.
LC-MS (ESI+): m/e 391 (M+H)+, Rt: 1.86 min.
EXAMPLE 96: 5-[2-(Imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)thieno[2,3- 10 d]pyridazin—4(5H)-one N \ , / N O / \ \ N\ S 96. l 2-(2-Chloroethyl)imidazo[l ,2-a]pyridine A mixture of dazo[l,2-a]pyridin—2—yl)ethanol (400mg, 2.466 mmol) from Example a4) and SOClz (2 mL, 27.4 mmol) in DCM (10 mL) was stirred at room 15 temperature for 2 days. The mixture was concentrated in vacuo. The residue was diluted with EtOAc (3>< 100 mL) and washed with ted NaHC03 solution (6 mL><4) and brine (6 mL). The organic layer was dried over NaZSO4, filtered and concentrated in vacuo. The residue was d by Pre—TLC (PE/EA=1/ 1, v/v) to give the title compound as an oil (300 mg, yield 70%). 20 LC-MS (ESI+): m/e 181 (M+H)+, R,: 1.68 min. 96.2 3-Bromo[2-(imidazo[1,2-a]pyridinyl)ethy1]-5H—thieno[2,3-d]pyridazin- 4(5H)-one A mixture of 2-(2-chloroethyl)imidazo[1,2-a]pyridine (90 mg, 0.498 mmol), 3- 25 bromothieno[2,3-d]pyridazin-4(5H)-one from Example 3.4 (115 mg, 0.498 mmol) and CszCO3 (325 mg, 0.996 mmol) in DMF (3 mL) was stirred at 60 °C for 16h. The solvent was evaporated. The residue was purified by Pre-TLC (PE/EA=1/2, V/V) to give the title compound as an oil (140 mg, yield 80%).
LC-MS (ESI+): m/e 374 (M+H)+, Rt: 1.79 min. 169 96.3 5-[2-(Imidazo[ l ,2-a]pyridin—2—yl)ethyl]—3-(pyridinyl)thieno[2,3-d]pyridazin- 4(5H)—one A mixture of pyridinylboronic acid (29.5 mg, 0.240 mmol), the compound from Example 96.2 (60 mg, 0.160 mmol), Na2C03 (42.4 mg, 0.400 mmol) and PdC12(dppf)-CH2C12 adduct (13.06 mg, 0.016 mmol) in e (3 mL) and water (1 mL) was stirred at 100 0C in a microwave for 10 min. The solvent was evaporated and the residue was purified by Pre-TLC (PE/EA=l/4) and purified by HPLC to afford the title product as a white solid (45 mg, yield 75%). 10 LC-MS (ESI+): m/e 374 (M+H)+, Rt: 1.70 min; 1H NMR (DMSO-ds, 400MHz): 8.68 (s, 1H), 8.60 (d, J: 6.0 Hz, 2H), 8.46 (d, J: 6.4 Hz, 1H), 8.21 (s,lH), 7.74 (s, 1H), 7.75 (d, J: 6.0 Hz, 2H), 7.46 (d, J: 9.2 Hz, 1H), 7.19-7.15 (m, 1H), .80 (m, 1H), 4.47 (t, J: 7.6 Hz, 2H), 3.13 (t, J: 7.6 Hz, 2H). 15 EXAMPLE 97: 5-[2-(7-Fluoroimidazo[l ,2—a]pyridin—2-yl)ethyl](pyridin eno[2,3-d]pyridazin—4(5H)-one N , /\ F\\®\AN O / N 'T‘l\ N\ S 97. l 3-Bromo(2-(7-fluoroimidazo[l ,2-a]pyridin—2-yl)ethyl)thieno[2,3-d]pyridazin— 4(5H)—one 20 A mixture of 3-bromothieno[2,3-d]pyridazin-4(5H)-one from Example 3.4 (231 mg, 1 mmol), uoroimidazo[l,2-a]pyridin-2—yl)ethanol from Example a5 (180 mg, 1.00 mmol) and Pth (525 mg, 2.00 mmol) was dissolved in THF (6 mL). Then DEAD (0.317 mL, 2.00 mmol) in THE (1 mL) was added. The resulting mixture was stirred at room temperature under nitrogen overnight. The mixture was concentrated and purified 25 on the ISCO ash system using a 40g C-l8 column using the following gradient: A: Water(0.l%NH4HC03); B: Methanol; 30%B to 80%B over 20 min (160 mg, yield:l7.7%). 170 LC-MS: m/e 393 (M+H)+; Rt:1.74 min; 1H NMR(DMSO-d6) 8: 3.12(t, 2H), 4.44 (t, 2H), 6.88-6.92 (m, 1H), 7.32 (dd, J=10, 2, 1H), 7.74(s, 1H), 8.17 (s, 1H), 8.52-8.56 (m, 1H), 8.62 (s,1H). 97.2 5-[2-(7-Fluoroimidazo[1,2—a]pyridin—2-yl)ethyl]-3—(pyridinyl)thieno[2,3- d]pyridazin—4(5H)-one A mixture of 3-bromo-5—(2-(7—fluoroimidazo[1,2-a]pyridiny1)ethyl)thieno[2,3- dazin-4(5H)-one (80 mg, 0.203 mmol), pyridiny1boronic acid (37.5 mg, 0.305 mmol), dppf)-CH2C12 adduct (16.61 mg, 0.020 mmol) and C82CO3 (133 mg, 10 0.407 mmol) in 1,4—dioxane (3 mL) and water(1.5mL) was heated under ave at about 110 °C for about 15min. The mixture was concentrated and was chromatographed on the ISCO Combiflash system using a 40 g Silicycle SiliaSep Silica gel column C-18 column using the ing gradient: A: Water(0.1%NH4HC03); B: Methanol; 30%B to 80%B over 20 min (15 mg, 18.8%). 15 LC-MS: m/e 392 (M+H); Rt:1.74 min; 1H NMR(CDC13) 8: 3.30 (t, 2H), 4.64 (t, 2H), 6.61-6.65 (m, 1H), 7.15 (dd, J=9.6 Hz, 2.4 Hz, 1H), 7.38 (s, 1H), 7.50 (d, J=6 Hz, 2H), 7.61 (s, 1H), 7.96-7.99 (m, 1H), 8.26 (s, 1H), 8.68 (d, J=6 Hz, 2H). 11.2 ation of compounds of the formula I in which A is Al, X1 is N, R1 is Y1- 20 Cyc1 and x3 is -C(R9)=C(R8)- EXAMPLE 98: 8-(Pyridin—4-yl)—2-[2-(quinolin—2-yl)ethyl]phthalazin-1(2H)-one N |\ / N '1' N\ 25 98.1 N-tert-Buty1chlorobenzamide The mixture of 2-chlorobenzoic acid (20 g, 128 mmol), HOBT (34.6 g, 256 mmol), EDCI (48.8 g, 256 mmol), 2—methylpropan—2—amine (9.3 g, 128 mmol) and TEA (25.9 g, 256 mmol) in THE (600 mL) was stirred at room temperature overnight. After solvent evaporation, the mixture was diluted with ethyl acetate (600 mL) and washed 171 with water (3 x 300 mL). The c layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel, g with EtOAc/PE (1:5) to give the title product as a white solid (23.9 g, yield: 88.5%).
LC-MS: m/e = 212 ; Rt=0.85 min. 98 .2 2-te1t-Butylchloro-3—hydroxy-2,3-dihydroisoindol— 1 -one N—tert-Butylchlorobenzamide (5.4 g, 25.6 mmol) was dissolved in THF (280 mL) and the on was added with TMEDA (12.4 mL, 81.9 mmol) and then added with sec-butyllithium—hexane solution (1.0 mol/L, 82.7 mL, 81.9 mmol) dropwise at 10 -78°C for 40 minutes under argon atmosphere, followed by stirring at the same temperature for 2.5 hours. Then, the mixture was added with DMF (4.36 mL, 56.3 mmol) and warmed from —78°C to room temperature over 2 hours. The reaction mixture was added with water (200 mL), and extracted with ethyl acetate (100 mL*3). The c layer was washed with saturated brine and dried over anhydrous sodium e. 15 The solvent was evaporated under reduced pressure and the residue was purified by crystallization using diisopropyl ether to obtain product 3 (5.0 g, Yield: 81%).
LC-MS: m/e =240 (M+H)+; Rt=l.74 min; 1H NMR (400MHz, DMSO-d6): 5 7.58- 7.54 (m, 1H), 7.49-7.45 (m, 2H), 6.35 (d, J=8.4 Hz, 1H), 5.98 (d, J=8.4 Hz, 1H), 1.52 (s, 9H). 20 98 .3 8-Chloro-2H-phthalazin- 1 —one The compound from Example 98.2 (5 g, 21 mmol) was suspended in 20 mL acetic acid under nitrogen. The resulting thick slurry was heated to 90°C. At approximately 80°C, a homogeneous solution was obtained. Hydrazine monohydrate (64%, 3 .2 mL, 63 25 mmol) was added dropwise (exotherm), keeping the internal temperature between 90 and 93°C over approximately 4 h. The resulting suspension was continued to stir at 90°C as the conversion of starting material was monitored by LC (<1%). Water (40 mL) preheated to 80°C was added, maintaining the mixture at 80-90°C followed by ramping down to 20°C over approximately 3h time. At this point, the resulting suspension was 30 transferred onto a filter. The filter cake was rinsed with water 3). The wet product was air-dried overnight to afford the title product (2.6 g, Yield: 69%). 172 LC-MS: m/e =181 (M+H)+; R.=1.51 min; 1H NMR (400MHz, DMSO-d6) 5: 12.61 (s, 1H), 8.32 (s, 1H), .83 (m, 3H). 98 .4 8-Chloro(2-quinolin-2—yl-ethyl)—2H—phthalazinone To a solution of triphenylphosphine (9.00 g, 34.3 mmol) in THF (100 mL), DEAD (5.44 mL, 34.3 mmol) was added at 0°C. After stirring for 15 min, 2-(quinolin— thanol from Example a1 (2.97 g, 17.17 mmol) was added. After another 15 min, 8-chlorophthalazin-1(2H)-one (3.1 g, 17.17 mmol) was added. The mixture was stirred overnight at room temperature; LC-MS indicated complete conversion to the product. 1 10 N HCl was added (pH= 4). The mixture was extracted with EtOAc (3 x 50 mL), the EtOAc layers were discarded. The aqueous layer was lized by aq.NaHC03 and extracted with ethyl acetate (3 x 500 mL). The combined organic layers were washed with brine, dried over NaZSO4, filtered, and trated under reduced re. The crude t was suspended in EtOAc. The solid was filtered through a Biichner 15 flannel. The purity of the title product was 95%.
LC-MS: m/e = 336 (M+H)+; Rt=1.54 min; 1H NMR (400MHz, DMSO-d6) 5: =8.35 (s, 1H), 8.29-8.26 (d, J=8.8Hz, 1H), 7.94—7.90 (m, 2H), 7.88-7.86 (m, 3H), 7.73- 7.69 (m, 1H), 7.57-7.55 (m, 1H), 7.48-7.46 (m, 1H), .55 (t, J=5.4 Hz, 2H), 3.42- 3.38 (t, J=5.4 Hz, 2H). 20 98 .5 8-(Pyridin—4-yl)[2-(quinolin—2—yl)ethyl]phthalazin-1(2H)-one A reaction tube was charged with pyridin—4-ylboronic acid (0.223 mmol), K2C03 (61.7 mg, 0.447 mmol) under a dry nitrogen atmosphere. A solution of 8-chloro(2- quinolinyl-ethyl)-2H-phthalazin—1-one (50 mg, 0.186 mmol) in 1,4-dioxane (5 mL) 25 along with PdC12(dppf) (5.45 mg, 7.45 umol) was added. After addition of water (1 mL), the resulting mixture was heated at 100°C overnight. After removal of the solvent under reduced pressure, the title compound was obtained as a crude product. It was purified by Prep-HPLC.
LC—MS m/e=379.1 (M+H)+; Rt=1.80 min; 1H NMR (400 MHz, CDC13) 8 ppm 30 8.58 (s, 2 H), 8.18 (s, l H), 8.04 (d, J=8.0 Hz, 1 H), 7.94 (d, J=8.0 Hz, 1H), 7.74-7.78 (m, 3 H), 7.66 (d, J=l.6 Hz, 1 H), 7.46—7.52 (m, 2 H), 7.30 (d, J=8.4 Hz, 1 H), 7.11 (d, J: 5.2 Hz, 2 H), 4.65 (t, J=7.4 Hz, 2 H), 3.45 (t, J=7.4 Hz, 2 H). 173 Examples 99 to 164 were prepared analogously to the method for Example 98.
EX. Name LC-MS: m/ e (M+H)+ / Rt [min] 99 2- [2-(Quinolinyl)ethy1][4-(trifluoro- 446 / 2.34 methy1)phenyl]phthalazin— 1 (2H)—one 100 8-(4-Methylphenyl)—2—[2-(quino lin—2— 392 / 2.3 0 yl)ethy1]phthalazin-1(2H)-one 101 8- [4-(Propanyl)phenyl][2-(quino 1in- 420 / 2 .45 2—yl)ethyl]phthalazin-1(2H)—one 102 8-(4-Ethylphenyl)—2—[2—(quino lin—2— 406 / 2. 15 yl)ethyl]phthalazin— 1 (2H)—one 103 4- {4-Oxo[2-(quino1in—2—yl)ethyl]—3,4— 403 / 1.96 dihydrophthalazin-S-yl}benzonitrile 104 8-(4-Methoxyphenyl)[2—(quinolin—2— 408 / 2. 19 yl)ethyl]phthalazin— 1 (2H)-one 105 8-(4-Fluorophenyl)[2-(quinolin—2- 396 / 2.22 yl]phthalazin— 1 (2H)—one 106 (4- {4-Oxo[2-(quinolin—2-yl)ethy1]-3,4- 417 / 2.11 dihydrophthalazin—5—yl}phenyl)acetonitrile 107 8-(4-Hydroxyphenyl)—2—[2-(quino1in 394 / 1 .99 yl)ethyl]phthalazin— 1 (2H)-one 108 8-(2-chlorophenyl)—2—[2-(quino1in 412 / 2.06 yl)ethyl]phthalazin— 1 (2H)—one 109 ethy1pheny1)—2-[2—(quinolin 392 / 2.08 yl)ethy1]phthalazin— 1 (2H)—one 110 8-(2-Ethylphenyl)[2-(quinolin 406 / 2.35 yl)ethyl]phthalazin-1(2H)-one 1 1 1 8—(2—Fluorophenyl)—2—[2—(quinolin—2— 396 / 2.20 yl]phthalazin— 1 (2H)—one 1 12 8-(2-Methoxyphenyl)—2—[2—(quino lin—2— 408 / 2.20 yl)ethyl]phthalazin— 1 (2H)—one 174 EX. Name LC-MS: m/ e (M+H)+ / Rt [min] 1 13 8-(3-Methoxypheny1)—2—[2-(quino1in 408 / 2.02 y1)ethy1]phthalazin— 1 (2H)-0ne 114 3-{4-Oxo[2-(quinolinyl)ethyl]—3,4- 403 / 1.96 dihydrophthalazin—S—y1 } benzonitrile 1 15 8-(3 -F1u0r0phenyl)—2-[2—(quin0 lin—2— 396 / 2.04 y1)ethy1]phthalazin-1 (2H)—0ne 1 16 8-(3-Hydroxyphenyl)[2-(quino lin 394 / 1 .87 yl)ethy1]phthalazin-1(2H)-one 1 17 N,N—Dimethy1—3- {4-oxo[2-(quinolin 449 / 1.98 y1)ethy1]—3 ydrophthalazin—5— yl } benzamide 1 18 8-(3 -Methy1phenyl)—2—[2—(quino1in—2— 392 / 2. 10 y1)ethy1]phthalazin-1 (2H)—one 1 19 2-[2-(Quinoliny1)ethyl]-8—(thiophen—2- 3 84 / 2. 19 y1)phtha1azin- 1 (2H)-one 120 8-(1-Methy1—1H-indoly1)—2-[2— 431 / 2.26 (quino1iny1)ethy1]phthalazin— 1 (2H)-one 121 8-(3,5-Dimethy1—1H-pyrazol—4-y1)—2-[2- 396 / 1.90 (quino1iny1)ethy1]phthalazin— 1 ne 122 Indol—S-y1)-2—[2—(quin01in—2- 417 / 1.97 yl)ethy1]phthalazin— 1 (2H)-0ne 123 8-(1H-Ind01y1)—2—[2-(quin01in 417 / 1.98 yl)ethy1]phtha1azin— 1 (2H)—0ne 124 8-(Pyrimidin-5 -y1)[2—(quin0 lin—2— 3 80 / 1.82 yl)ethy1]phthalazin— 1 (2H)—0ne 125 8-(2-Methoxypyridinyl)[2-(quino lin- 409 / 2 .07 2-y1)ethy1]phthalazin-1(2H)-one 126 8—(Pyridin—3—y1)—2—[2—(quinolin—2— 379 / 1.81 y1]phthalazin— 1 (2H)—one 127 8-(Furan—3-y1)-2—[2—(quinolin—2— 368 / 1.95 y1)ethy1]phthalazin— 1 (2H)—one 175 EX. Name LC-MS: m/ e (M+H)+ / Rt [min] 128 8-(Quino1iny1)[2—(quin01in—2- 439 y1)ethy1]phtha1azin— 1 (2H)-0ne 129 8-(1H-Ind01y1)-2—[2-(quin01in 417 / 2.12 y1)ethy1]phtha1azin- 1 (2H)—0ne 130 8-(2,3-Dihydr0benzofi1ran—5-yl)—2-[2- 420 / 2.19 (quinoliny1)ethyl]phthalazin— 1 (2H)—one 131 8-(3,4-Dihydro-2H-1,5-benzodioxepin 450 / 2.19 y1) [2-(quino [inyl)ethyl]phthalazin- 1(2H)-one 132 8-(1-Benzofuran—5—y1)—2—[2—(quin0lin—2— 418 / 2.25 y1)ethy1]phtha1azin— 1 (2H)—one 133 8-(6-Methoxypyridin—3—y1)—2—[2—(quinolin- 409 / 2.10 2-y1)ethy1]phtha1azin- 1 (2H)—one 134 -Dihydro-1,4-benzodioxin—6—yl)—2- 436 / 1.99 [2-(quino 1iny1)ethyl]phthalazin— 1 (2H)- one 13 5 8-(2-Methy1pyridin—4-yl)—2-[2-(quino1in 393 / 1 .95 y1)ethy1]phtha1azin— 1 (2H)—one 136 8-(5 -Methoxypyridin—3—y1)—2-[2-(quinolin- 409 / 1 .97 2-y1)ethy1]phtha1azin— 1 (2H)-0ne 137 8-(5 -F1u0ropyridin—3—yl)—2-[2—(quin0lin 397 / 2.00 y1)ethy1]phtha1azin- 1 (2H)—0ne 138 8-(1 ,3-Benzodi0x01—5-y1)—2-[2—(quin0lin—2- 422 / 2.00 y1]phthalazin-1 (2H)—0ne 139 8-(1-Methy1—1H—pyrazol—4—yl)—2-[2- 3 82 / 1.90 (quinolinyl)ethyl]phthalazin- 1 ne 140 2-Methylpropyl)-1H-pyrazoly1] 424 / 2.14 [2—(quino 1111—2—y1)ethyl]phthalazin— 1 (2H)— one 141 tert-Butyl 2- {4—0x0—3—[2—(quinolin—2— 467 / 2.34 y1)ethy1] -3 ,4-dihydrophthalazin—5—yl} — 1 H- 176 EX. Name LC-MS: m/ e (M+H)+ / Rt [min] pyrrolecarboxy1ate 142 8-(3-Chlorofluorophenyl)—2-[2- 430/ 2.32 (quino1iny1)ethy1]phthalazin- 1 (2H)—one 143 8-(2-Chloroflu0rophenyl)—2-[2- 430/ 2.28 (quinoliny1)ethy1]phthalazin— 1 (2H)—one 144 8-(3 ,4-dimethylphenyl)—2—[2-(quin0 lin—2- 406 / 2.3 7 yl)ethy1]phthalazin-1(2H)-one 145 8-(2,4-Dimethoxyphenyl)[2-(quino lin- 43 8 / 2 .20 2—y1)ethy1]phthalazin- 1 (2H)—one 146 8-(2,5—Dimethoxyphenyl)—2—[2—(quin0 lin— 43 8 / 2. 18 2-y1)ethy1]phtha1azin— 1 (2H)—one 147 8-(2,3-Difluorophenyl)—2—[2—(quino lin—2— 414 / 2.23 y1)ethy1]phthalazin- 1 (2H)—one 148 8-(3 ,4-Dimethoxyphenyl)-2—[2—(quin0 1in- 43 8 / 2. 1 1 2-y1)ethy1]phtha1azin- 1 (2H)-one 149 8-(3 u0rophenyl)[2-(quino1in 414 / 2.25 y1)ethy1]phtha1azin— 1 (2H)-one 150 8-(5-F1u0r0methoxyphenyl)—2-[2- 426 / 2.21 (quino1iny1)ethy1]phthalazin— 1 (2H)-one 15 1 8-(4-F1uoromethoxyphenyl)—2-[2- 426 / 2.22 (quino1iny1)ethy1]phthalazin- 1 (2H)—one 152 8-(3 ,5-Fimethoxyphenyl)—2-[2—(quin0 lin—2- 43 8 / 2.21 yl)ethy1]phthalazin- 1 ne 153 8-(2,5-Diflu0r0phenyl)—2-[2—(quin0lin 414 / 2.23 y1]phthalazin-1 (2H)-0ne 154 8-(3-F1uoromethoxyphenyl)[2- 426 / 2.20 (quinolin-Z-yl)ethyl]phthalazin-1(2H)-one 155 8—(2—F1u0ro—3—methoxyphenyl)—2—[2— 426 / 2.19 (quino1111—2—y1)ethy1]phthalazin— 1 (2H)—one 156 8-(3 ,5-Difluorophenyl)—2—[2—(quin0 lin—2— 414 / 2.06 y1]phthalazin— 1 (2H)—one 177 EX. Name LC-MS: m/ e (M+H)+ / Rt [min] 157 8-(3 -Fluoromethoxyphenyl)—2-[2- 426 / 2.25 (quinolinyl)ethyl]phthalazin- 1 (2H)—one l5 8 8-(Naphthalen—2-yl)—2-[2—(quino1in 428 / 2.3 5 yl)ethyl]phthalazin— l (2H)—one 159 8-Phenyl—2-[2-(quinolin—2- 378 / 2.21 yl)ethyl]phthalazin— 1 (2H)—one 160 8-(1-Benzofuranyl)[2-(quinolin 418 / 2 .29 yl)ethyl]phthalazin- l (2H)-one 161 8—( 1 -Methyl— 1 H-pyrazol—5-yl)—2-[2- 3 82 / 1.90 (quinolin—2—yl)ethyl]phthalazin— 1 (2H)—one hydrochloride 162 8-(4,5-Difluoro-2—methoxyphenyl)—2—[2— 434 (quinolinyl)ethyl]phthalazin— l (2H)—one 163 8-(2-Fluoromethylphenyl)—2—[2— 410 / 2.28 (quinolinyl)ethyl]phthalazin— l ne 164 8-(2-Fluoromethoxyphenyl)—2—[2— 442 (quinolinyl)ethyl]phthalazin— 1 (2H)-one EXAMPLE 165 : 2- [2-(Imidazo[1 ,2—a]pyridin—2-yl)ethyl] (pyridin-3 -yl)phthalazin- 1 (2H)-one 5 165. 1 ro(2-imidazo[l ,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one To a solution of triphenylphosphine (5.81 g, 22.15 mmol) in THF (150 mL), DIAD (7.54 mL, 38.8 mmol) was added at 0°C. After stirring for 15 min, 2- (imidazo[l,2-a]pyridin—2-yl)ethanol from Example a4 (2.00 g, 11.07 mmol) was added.
After another 15 min, 8—chlorophthalazin—l(2H)—one from Example 98.3 (1.80 g, 11.07 10 mmol) was added. The mixture was stirred ght at room temperature. LC-MS indicated complete sion to the product. 1 N HCl was added (pH= 4).The mixture 178 was extracted with EtOAc, the EtOAc layers were discarded. The aqueous layer was lized by aq.NaHCO3 and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na2804, filtered, and concentrated under d pressure. The crude title product was recrystallized from ethyl e to give 3.1 g : 86%) of a bright beige solid. 165 .2 2-[2-(Imidazo[1,2-a]pyridinyl)ethyl](pyridin—3—yl)phthalazin-1(2H)-one A reaction tube was charged with 8-chloro(2-imidazo[l ,2-a]pyridin yl)ethyl]phthalazin-l(2H)-one (80 mg, 0.246 mmol) and a mixture of 1.5 mL of ethanol 10 and 1.5 mL of toluene under argon. To this suspension, pyridinylboronic acid (30.3 mg, 0.246 mmol) and Na2C03 (39.2 mg, 0.369 mmol) were added. Then, tetrakis- (triphenylphoshine)palladium (28.5 mg, 0.025 mmol) was added. The reaction mixture was heated in a Biotage microwave at about 130 °C for about 30 min. The reaction was monitored by TLC ethanol = 9:1). After completion of the reaction, EA was 15 added followed by the addition of 1 N HCl. The mixture was extracted twice with EtOAc. The aqueous layer was basified with 2N NaOH and extracted three times with ethyl acetate. The ed organic layers were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was triturated from ethyl acetate to give the title compound as a white solid (12 mg, 13.26%). 20 LC-MS: m/e = 368.1 (M+H)+ The compound of Examples 2 were prepared in analogy to the method described in Example 165.2.
EX. Name LC-MS: m/ e (M+H)+ 166 2-[2-(Imidazo[ l ,2-a]pyridinyl)ethyl](pyridin 368.1 yl)phthalazin- l (2H)-one 167 2—[2—(Imidazo[ l ,2—a]pyridin—2—yl)ethyl]—8—(3— 398.1 methoxypyridin—4—yl)phthalazin— l (2H)—one 168 2- [2-(Imidazo[ l ,2—a]pyridin—2—yl)ethyl]—8—(pyrimidin-5 - 3 68.8 yl)phthalazin— 1 (2H)-one 179 EX. Name LC-MS: m/ e (M+H)+ 169 2-[2-(Imidazo[1,2-a]pyridinyl)ethyl]—8-(1-methyl-1H- 3 71.1 pyrazo l-3 -yl)phthalazin-1 (2H)—one 170 8-(Furan—3-yl)[2-(Imidazo[1,2-a]pyridin 3 5 7.1 yl)ethyl]phthalazin— l (2H)—one 171 2- [2-(Imidazo[ l ,2-a]pyridin—2—yl)ethyl]—8—(2-oxo-2 ,3 - 422.1 dihydro- l H-indo lyl)phthalazin- 1 (2H)-one W a]pyridin—2—yl)ethyl]phthalazin— l (2H)—one EXAMPLE 173: 8-(1,1-Dioxidothiomorpholin—4—yl)—2-[2-(quinolinyl)ethyl]— azin-1(2H)-one ogséo A microwave reaction vial was charged with the 8-chloro(2-(quinolin yl)phthalazin—1(2H)-one from Example 98.4 (100 mg, 0.30 mmol), CS2C03 (194 mg, 0.59 mmol), Pd2(dba)3 (5.45 mg, 5.96 umol) and BINAP (11.13 mg, 0.018 mmol).
The solids were purged with argon for l h. A separate flask was charged with e (993 ul) and thiomorpholin 1,1—dioxide (48.3 mg, 0.36 mmol), degas with argon for 1 h 10 and then transferred to the microwave reaction vial under inert conditions. The resulting reaction mixture was heated on microwave at 100 0C for 20 h. The reaction mixture was poured into water and ted with DCM. The solids were d. The organic layer was washed with water, dried over MgSO4, filtered and concentrated in vacuo. The residue was purified on a silica column (eluent: DCM/methanol) and then recrystallized 15 from EA to afford the title product (87 mg, 67.2%).
LC-MS: m/e = 435.1 (M+H)+ EXAMPLE 174: 2-[2-(Imidazo[l ,2—a]pyridin—2—yl)ethyl](morpholin—4-yl)phthalazin- 1 (2H)-on 180 o A microwave reaction vial was charged with the 8-chloro(2-imidazo[1,2- a]pyridinyl)ethyl]phthalazin—l(2H)—one from Example 165.1 (100 mg, 0.31 mmol), CS2CO3 (201 mg, 0.62 mmol), Pd2(dba)3 (5.64 mg, 6.16 umol) and BINAP (11.50 mg, 0.018 mmol). The solids were purged with argon for l h. A separate flask was charged with toluene (993 pl) and morpholine (32.2 mg, 0.37 mmol), degas with argon for 1 h and then transferred to the microwave reaction Vial under inert conditions. The resulting reaction mixture was heated on microwave at 100 °C for 20 h. The reaction mixture was poured into water and extracted with DCM. The solids were d. The organic layer 10 was washed with water, dried over MgSO4, filtered and concentrated in vacuo. The e was purified on a silica column (eluent: DCM/methanol) and then recrystallized from EA to afford the title product (11 mg, 9.52 %).
LC-MS: m/e = 376.1 (M+H)+ 15 The compound of Examples 175 to 191 were prepared in analogy to the method described in Example 174.
EX. Name LC-MS: m/ e (M+H)+ 175 8-(1,l-Dioxidothiomorpholin—4-yl)—2-[2- 424. l (imidazo [l ,2-a]pyridin—2-yl)ethyl]phthalazin— 1(2H)-one 176 2-[2-(Imidazo[1,2-a]pyridin—2-yl)ethyl] 402.2 (tetrahydro- 1 [3 ,4-c]pyrro l-5(3H)- yl)phthalazin- l (2H)-one hloride 177 8-(5,5-Difluorohexahydrocyclopenta[c]pyrro1- 436.2 2( l H)—yl)—2— [2—(imidazo[l yridin—2— yl)ethyl]phthalazin— l (2H)—one hydrochloride 178 2- [2-(Imidazo[ l yridin—2—yl)ethyl]—8- 375 .2 (piperazin— l -yl)phthalazin— 1 (2H)—one 181 EX. Narne LC-MS: m/ e (M+H)+ hloride 179 8-(4,4-Difluoropiperidinyl)—2—[2-(imidazo[1 ,2- 410.2 a]pyridinyl)ethyl]phthalazin— 1 (2H)—one 180 8- [4-(Chloromethyl)(hydroxymethyl)piperidin- 451.9 l-yl][2-(imidazo[1 ,2-a]pyridin yl]phthalazin- l (2H)-one hydrochloride 181 2— [2—(Imidazo [l ,2—a]pyridin—2—yl)ethyl]—8— 374.2 idin— l —yl)phthalazin— l (2H)—one hydrochloride 182 8-(2,3-Dihydro-4H—l ,4—benzoxazin—4—yl)—2- [2- 424.1 (imidazo[1 ,2-a]pyridin-2—yl)ethyl]phthalazin— 1(2H)-one hydrochloride 183 2- [2-(Irnidazo[ 1 ,2-a]pyridinyl)ethyl] [4- 442.2 (trifluorornethyl)piperidinyl]phthalazin— 1 (2H)- one hydrochloride 184 2- [2-(Irnidazo[ l ,2-a]pyridin—2-yl)ethyl](4- 389.2 rnethylpiperazin- l -yl)phthalazin— l (2H)-one hydrochloride 185 8-(1,3-Dihydro-2H—isoindolyl)—2—[2- 408.2 (irnidazo[1 ,2-a]pyridin—2-yl)ethyl]phthalazin— 1 (2H)-one 186 8-(7-Benzyl—2,7-diazaspiro[4.4]nonyl)—2— [2— 505.3 (imidazo [l ,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one hydrochloride 187 8-( { [(3 aR,4S,6aS)benzyloctahydro- 519.3 cyclopenta[c]pyrrol—4-yl]methyl} amino)—2- [2- (imidazo [l yridin—2—yl)ethyl]phthalazin- 1(2H)-one hydrochloride 182 EX. Name LC-MS: m/ e (M+H)+ 18 8 tert-butyl (3R)-3 -({3—[2—(imidazo[1,2-a]pyridin—2- 475 .2 yl)ethyl] oxo-3 ,4—dihydrophthalazin—5- yl} amino)pyrrolidine— 1 —carboxylate 189 8-(2,6-Dimethylmorpho1inyl)[2— 404.2 (imidazo [1 ,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one hydrochloride 190 2-[2-(Imidazo[1,2-a]pyridinyl)ethyl](1,4- 390.2 oxazepan—4—yl)phthalazin-1(2H)—one hydrochloride 191 tert-Butyl 4— {3 —[2—(imidazo[1,2—a]pyridin—2- 472.2 yl)ethyl] —3 ,4—dihydrophthalazin—5—yl} -3 ,6- opyridine- 1 (2H)—carboxylate 11.3 Preparation of nds ofthe formula I in which A is Al, X1 is N and X3 is -C(R9)=C(R8)- with R9 being Y3—cyc3 5 EXAMPLE 192: 5-(Pyridin—4-yl)—2-[2-(quinolinyl)ethyl]phthalazin-1(2H)-one hydrochloride \ o / N r N \ / / 192. 1 4-Bromohydroxy-3H-isobenzofiaran— 1 -one 10 To a stirred solution of n-butyllithium 1.6 M in s (17.5 mL, 28.1 mmol) was added at —20 °C under argon 2,2,6,6-tetramethylpiperidine (4.7 mL, 28.1 mmol) in anhydrous THF (40 mL). After cooling (—50 °C), 3—bromobenzoic acid (2.54 g, 12.8 mmol) in anhydrous THF (10 mL) was added dropwise and the mixture was stirred for 1 h. The e was then treated with an excess ofDMF (3.7 g, 50.4 mmol). The 15 resulting solution was allowed to warm up to ambient temperature, after which water 183 was added. The aqueous layer was washed with diethyl ether, and then acidified with 4M HCl. The mixture was diluted with diethyl ether and the organic layer was separated and dried with MgSO4. The residue was d by crystallization using with PE to give the crude title product (1.18 g, yield: 41 %).
LC-MS: m/e = 229 (M+H)+; Rt=1.47 min. 192.2 5-Brorno-2H—phthalazin— 1 -one The compound from Example 192.1 (1 g, 4.4 mmol) was suspended in 5 mL acetic acid under nitrogen. The resulting thick slurry was heated to 90°C. At 10 approximately 80°C, a homogeneous solution was obtained. ine monohydrate (64%, 0.66 mL, 13.2 mmol) was added dropwise (exotherm), keeping the internal temperature between 90 and 93°C over approximately 4h. The resulting suspension was continued to stir at 90°C as the conversion of starting material was monitored by LC (<l%). Water (10 mL) preheated to 80°C was added, maintaining the mixture at 80- 15 90°C followed by cooling down to 20°C over imately 2h time. At this point, the resulting suspension was erred onto a filter. The filter cake was rinsed with water (10 mL*3). The wet product was air-dried overnight to afford the title t (570 mg, Yield: 58 %).
LC-MS: m/e =227 (M+H)+; Rt=1.65 min, 1H NMR (400MHz, DMSO-d6): 5 20 12.94 (s, 1H), 8.41 (s, 1H), 8.25-8.19 (m, 2H), 7.77-7.73 (m, 1H). 192.3 5 -Bromo[2-(quinoliny1)ethy1]phthalazin-1(2H)-one To a mixture of PPh3 (699 mg, 2.67 mmol) in THF (50 mL) and DIAD (198 mg, 0.98 mmol), 5-bromo-2H-phthalazinone (300 mg, 1.33 mmol) and then 2-quinolin 25 yl—ethanol from e al (254 mg, 1.46 mmol) were added dropwise at 15°C under en. The mixture was stirred at room temperature overnight. Water was added and the mixture was extracted with EtOAc. The organic phase was washed with HCl (1 N).
The aqueous phase was basified and extracted with DCM. The organic phase was washed with a NaHC03—solution, dried over Na2S04, filtered and concentrated. The 30 crude product was recrystallized from EA and dried to give the title compound as bright beige solid (300 mg, 59.2% yield). 184 192.4 5 dinyl)[2-(quino1inyl)ethyl]phthalazin-1(2H)-one hydrochloride A reaction tube was charged with a solution of 5-bromo[2-(quinolin—2- yl)ethyl]phthalazin—1(2H)-one (70 mg, 0.184 mmol) in 1.5 mL of ethanol and 1.5 mL of toluene under argon. To this suspension, pyridinylboronic acid (22.63 mg, 0.184 mmol) and a 2M solution ofNa2C03 (39.2 mg, 0.369 mmol) were added. Then, is- (triphenylphoshine)palladium (21.27 mg, 0.018 mmol) was added. The reaction mixture was heated in a Biotage microwave at about 130 °C for about 30 min. The on was monitored by TLC (DCM/methanol = 9:1). After completion of the reaction, EA was 10 added followed by the addition of 1 N HCl. The mixture was extracted twice with EtOAc. The organic layer was basified with NaHC03> and ted with brine. The organic layers were dried over MgSO4, filtered, and concentrated under reduced pressure. The crude t was dissolved in isopropanol. Isopropanol containing HCl was added. The precipitate was sucked off and recrystallized from hot isopropanol to 15 give a bright gray solid (55 mg, 72.0%).
LC-MS: m/e = 379.1 (M+H)+ EXAMPLE 193 : 5 -(Pyrimidinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one hydrochloride 20 The title compound was prepared in analogy to the method described in Example 192. LC—MS: m/e = 380.1 (M+H)+ \ O / l N 1 N\ / | NvN EXAMPLE 194: 5-(1-Methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin- 25 1(2H)-one 185 H3C\N \ N The title compound was prepared in analogy to the method bed in Example 192. LC-MS: m/e = 382.2 (M+H)+ EXAMPLE 195: 5-(1,1-Dioxidothiomorpholinyl)[2-(quinolin yl)ethyl]phthalazin- 1 ne \ o / H N 1‘ N\ {N} /s\\ o/ O The title compound was prepared in analogy to the method bed in Example 173 but using 5-bromo[2-(quino1inyl)ethyl]phthalazin-1(2H)-one instead of 8- 10 chloro(2-(quinolinyl)ethyl)phthalazin-1(2H)-one. LC-MS: m/e = 435.1 (M+H)+.
EXAMPLE 196: 2-[2-(1midazo[1,2—a]pyridin—2-yl)ethyl](pyridinyl)phthalazin- 1 (2H)-one 15 196. 1 5-Bromo[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one The title compound was prepared in analogy to the method described in Example 192.3 but using 2—(imidazo[1,2—a]pyridin—2—yl)ethanol from Example a4. Yield: 84%. 196.2 2-[2-(Imidazo[ 1 ,2-a]pyridin—2—yl)ethyl]—5—(pyridin—3 -yl)phthalazin-1(2H)-one 186 The title compound was ed in analogy to the method described in Example 192.4 but using 5-bromo[2-(imidazo[1,2—a]pyridin—2-yl)ethyl]phthalazin—1(2H)-one.
LC-MS: m/e = 368.1 (M+H)+ 5 EXAMPLE 197: 2-[2-(Imidazo[1,2-a]pyridin—2—yl)ethyl](pyrimidin—5- yl)phthalazin1(2H)-on Q,N\Nj\/\'\ll O \ N\ / NVN The title compound was prepared in analogy to the method bed in Example 196 but using 5-(4,4,5,5-tetramethyl-1,3,2-dioxoborolan—2-yl)pyrimidine. Yield: 73.9%. 10 LC—MS: m/e = 368.8 (M+H)+ EXAMPLE 198: 2-[2-(Imidazo[1,2-a]pyridin—2—yl)ethyl](morpholinyl)phthalazin- one hydrochloride Q3mIN O N \ N [O] 15 The title compound was prepared in analogy to the method described in Example 174 but using 5-bromo[2-(imidazo[1,2-a]pyridin—2-y1)ethy1]phthalazin-1(2H)-one from Example 196.1 instead of 8-chloro—2—(2-imidazo[1 ,2-a]pyridin—2- yl)ethyl]phthalazin-1(2H)-one. Yield: 2.1%. LC-MS: m/e = 376.2 (M+H)+ 20 EXAMPLE 199: 2—[2—(Imidazo[1,2—a]pyridin—2—yl)ethyl](tetrahydro- 1 H—furo [3 ,4- c]pyrrol—5(3H)—yl)phthalazin—1(2H)—one hydrochloride 187 QIN\Nj\/\l\lJ O \ N\ N H O The title compound was prepared in analogy to the method described in Example 176 but using 5-bromo[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one from Example 196.1 instead of 8-chloro(2-imidazo[1 ,2-a]pyridin yl)ethyl]phthalazin—1(2H)—one. Yield: 11.8%. LC—MS: m/e = 402.2 (M—I—H)+ 11.4 Preparation of compounds of the formula I in which A is Al, X1 is N, X2 is C-R7 with R7 being z and x3 is -C(R9)=C(R8)- 10 EXAMPLE 200: 2-[2-(Imidazo [1 ,2-a]pyridin—2—yl)ethyl](pyrimidin—5 -yl)phthalazin- 1 (2H)-one QIN\Nj\/\l\ll O \ N\ / I NVN 200. 1 4-Bromo[2-(imidazo[1 yridin—2-yl)ethyl] lazin- l (2H)-one To a solution of triphenylphosphine (1165 mg, 4.44 mmol) in THF (60 mL), 15 DIAD (1.5 mL, 7.78 mmol) was added at 0°C under en. After stirring for 30 min, 4-bromophthalazin—1(2H)one (0.5 g, 2.22 mmol) was added. After further stirring, 2- (2-imidazo[1,2-a]pyridinethanol from Example a4 (396 mg, 2.44 mmol) was added.
The mixture was stirred for 12 h at room temperature. LC-MS ted complete conversion to the product. EA and water were added. The organic phase was washed 20 with 1 N HCl. The EtOAc layers were discarded. The aqueous layer was neutralized by aq.NaHC03 and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The 188 crude title product was tallized from diisopropyl ether/ EA (1 :1) to give 710 mg : 87%) of a white solid. 200.2 2-[2-(Imidazo[1 ,2-a]pyridinyl)ethy1](pyrimidin—5-yl)phthalazin-1(2H)-one The title compound was prepared in analogy to the method described in Example 168 but using 4-bromo[2-(imidazo[1,2-a]pyridiny1)ethy1]—phthalazin-1(2H)-one.
Yield: 35.1%. LC-MS: m/e = 369.1 (M+H)+ E 201: Imidazo[1,2-a]pyridinyl)ethyl](morpho1inyl)phthalazin- 10 1(2H)-one The title compound was prepared in analogy to the method described in Example 198 but using 4-bromo-2—[2—(imidazo[1 ,2—a]pyridin—2—yl)ethyl]-phthalazin-1(2H)-one.
Yield: 48.2%. LC-MS: m/e = 376.2 (M+H)+ 15 11.5 Preparation of compounds ofthe formula I in which A is Al, X1 is CH, R1 is Yl-Cyc1 and X3 is S EXAMPLE 202: 3-(3-Methoxypyridin—4-y1)—5—[2-(quinolinyl)ethyl]thieno[3,2- 20 c]pyridin-4(5H)-one 202.1 (E)—3-(4—Bromothiophen—2—yl)acrylic acid Piperidine (1.036 mL, 10.47 mmol) was added to a mixture of 4-bromothiophene- 2-carbaldehyde (20 g, 105 mmol) and malonic acid (13.07 g, 126 mmol) at 80°C under 25 nitrogen atmosphere. The reaction mixture was stirred for 2 h at 100°C. The reaction 189 mixture was cooled to room temperature and extracted with EA/water. The organic phase was washed with 2N NaOH. The s phase was acidified and a precipitate was formed. The solid was stirred in a mixture of DCM and diisopropyl ether (1 :1). The solid was sucked off and dried under reduced pressure to give a bright beige solid (12.3 g, 50.4%). 202.2 (E)—3-(4-Bromothiophen—2—yl)acryloyl azide To a solution of (4-bromothiopheny1)acry1ic acid (9.30 g, 39.9 mmol) in acetone (100 mL), triethylamine (4.24 g, 41.9 mmol) was added under nitrogen 10 atmosphere. At 0°C, isobutyl chloroforrniate (5.72 g, 41.9 mmol) was slowly added and then, the mixture was stirred for 1 h. 4.67 g (71.8 mmol) of sodium azide dissolved in 10 mL of water was slowly added at 0°C, the mixture was stirred at 0°C for a further hour and then warmed up to room ature ght. The reaction mixture was extracted with EA/water. The organic phase was washed aq. NaHC03 on and then 15 with brine. The organic phase was dried over MgSO4, concentrated to dryness and the residue was purified by trituration with diisopropyl ether to afford a bright beige solid (9.00 g, 34.9 mmol). Yield: 87%. 202.3 3-Bromothieno[3,2-c]pyridin—4(5H)—one 20 100 mL of diphenyl ether were warmed to 210°C and then a solution of (E)(4- bromothiophenyl)acryloyl azide (9.95 g, 38.6 mmol) in 50 mL of diphenyl ether was added under en. The reaction e was held at this temperature for 15 min.
After cooling to room temperature, the reaction mixture was diluted with 100 mL of cyclohexane. The precipitate was sucked off and dried in vaccuo to give 6.6 g (yield: 25 74.4%) of the title compound as brown solid. 202.4 3-Brorno[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one The title compound was prepared in analogy to the method described in Example 192.3 but using 3—bromothieno[3,2—c]pyridin—4(5H)—one. Yield: 26.6%. As a byproduct, 30 3-bromo(2-quinolin—2—yl)ethoxy)thieno[3,2-c]pyridine was obtained (yield: 23.3%).
Using a ion® chromatography system (normal phase, eluent cyclohexane/ethyl acetate), the title compound was obtained as a bright beige solid. 190 202.5 3-(3-Methoxypyridinyl)—5—[2—(quinolinyl)ethyl]thieno[3,2-c]pyridin— one A reaction tube was charged with a on of 3-bromo[2-(quinolin yl]thieno[3,2-c]pyridin-4(5H)—on (100 mg, 0.260 mmol) in 1.5 mL of ethanol and 1.5 mL oftoluene under argon. To this suspension, 3-methoxypyridin—4-y1boronic acid (39.7 mg, 0.260 mmol) and a 2M on ofNa2C03 (41.3 mg, 0.389 mmol) were added. Then, tetrakis(triphenylphoshine)palladium (30.0 mg, 0.026 mmol) was added.
The reaction mixture was heated in a Biotage microwave at about 130 °C for about 30 10 min. The reaction was monitored by TLC (DCM/methanol = 9:1). After completion of the reaction, EA was added followed by the addition of 1 N HCl. The mixture was extracted twice with EtOAc. The c layer was basified with NaHC03 and extracted with brine. The organic layers were dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was dissolved in isopropanol. Isopropanol 15 containing HCl was added. The precipitate was sucked off and recrystallized from hot ethyl acetate to give a white solid (32 mg, 29.8%).
LC—MS: m/e = 414.1 (M+H)+ EXAMPLE 203: 3-(3-Hydroxypyridin—4-yl)—5-[2-(quino1iny1)ethy1]thieno[3,2- 20 c]pyridin-4(5H)-one hydrochloride To 3-(3-methoxypyridinyl)—5-[2-(quinolinyl)ethyl]thieno[3,2—c]pyridin— 4(5H)-one (50.0 mg, 0.121 mmol) in DCM (20 mL) was added 1M BBr3 in DCM (0.363 mmol, 91 mg). The reaction mixture was stirred for 2 h under nitrogen. The 25 reaction mixture was poured onto water and basified with 1N NaOH, extracted with DCM and dried. The organic phase was concentrated to dryness and the residue was purified by column tography (normal phase) on silica using DCM/methanol to give the title compound (13 mg, 0.030 mmol).
LC-MS: m/e = 400.1 (M+H)+ 191 The compounds of Example 204 to 210 were prepared in analogy to the method described above.
EX. Name LC-MS: m/ e (M+H)+ 204 3-( 1 -Methyl- 1 H-pyrazol—5-y1)—5-[2—(quinolin 3 87.1 yl)ethyl]thieno[3 ,2-c]pyridin-4(5)H—one hydrochloride 205 3-(Pyridin—4-yl)[2-(quinolin—2—yl)ethyl]thieno[3,2- 384.1 c]pyridin-4(5H)-one hydrochloride 206 3-(Pyrimidinyl)(2-quinolinyl)ethyl)thieno [3 ,2- 3 85. 1 c]pyridin—4(5H)—one 207 3-(2-Oxoindolin—6—yl)—5—[2—(quinolin—2— 438.1 yl]thieno[3,2—c]pyridin—4(5H)—one 208 3-(3-Hydroxyphenyl)[2—(quinolin—2— 399.1 yl]thieno[3 ,2-c]pyridin-4(5H)—one hydrochloride 209 5-[2-(5-Ethylpyridinyl)ethyl]-3—(pyridin—4— 362.1 yl)thieno[3 ,2-c]pyridin—4(5H)-one hydrochloride 210 5 -[2-(Imidazo[1,2-a]pyridin—2-yl)ethyl]—3—(pyridin 3 73. 1 yl)thieno[3 ,2-c]pyridin-4(5H)—one hydrochloride EXAVIPLE 211: 3-(Morpholinyl)—5—[2—(quinolin—2-yl)ethyl]thieno[3,2-c]pyridin— one hydrochloride \ 05.0) N N l \ \ s 21 1.1 3-Morpholinothieno[3,2-c]pyridin-4(5H)-one 3-Bromothieno[3,2-c]pyridin-4(5H)-one from Example 202.3 (411 mg, 1.786 10 mmol) and line 81501 mg, 17.23 mmol) were stirred in a microwave for 3 h at 220°C. The mixture was poured onto water and extracted with DCM. The organic phase was concentrated and the residue was recrystallized from EA to give 180 mg (yield: 42.6%) of the title compound. 192 21 1.2 3-(Morpholinyl)[2-(quino1in—2-y1)ethyl]thieno[3,2-c]pyridin-4(5H)-one hydrochloride The title compound was prepared in analogy to the method described in e 1.6 but using THF as solvent. Yield: 20.5%. LC—MS: m/e = 392.1 (M+H)+ EXAMPLE 212: tert-Buty1(4-oxo-5—(2-quinolinyl)ethy1)—4,5-dihydrothieno [3 ,2- c]pyridin-3 -yl)-5 ,6-dihydropyridine— 1 (2H) carboxylate The title compound was prepared in analogy to the method described in Example 202.5. 1 0 EXAMPLE 213 : 5 -(2-(Quinolinyl)ethyl)—3—(1,2,3 ,6-tetrahydropyridin—4- yl)thieno[3 ,2-c]pyridin-4(5H)-one hloride Tert-butyl(4-oxo(2-quino1in—2-y1)ethyl)-4,5-dihydrothieno[3,2-c]pyridin—3- 15 yl)-5,6-dihydropyridine-1(2H) carboxylate from e 212 (120 mg, 0.246 mmol) in 1 mL of HCl containing isopropanol was stirred under nitrogen for 12 h at room temperature. The reaction mixture was extracted with DCM, the aqueous phase was basified with 1N NaOH and extracted with DCM. The organic phase was dried, concentrated and the residue was tallized from HCl-isopropanol to give the title 20 compound as hydrochloride salt as yellow solid (90 mg, 86%).
LC-MS: m/e = 388.1 (M+H)+ E 214: tert-Butyl—4—(4—oxo—5—(2—quinolin—2-yl)ethyl)-4,5-dihydrothieno [3 ,2- c]pyridin-3 -yl)-piperidine— 1 carboxylate 193 N \ O / N N I \ \ The title compound was prepared in analogy to the method described in Example 202.5 EXAMPLE 215: 3-(Piperidinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin- 5 4(5H)-one roacetate H / N \ O / N N I \ \ 3 Route a) Under nitrogen, quinolinyl)ethyl)—3-(1,2,3,6-tetrahydropyridin yl)thieno[3,2-c]pyridin—4(5H)-one hydrochloride from Example 213 (80 mg, 0.189 10 mmol) in methanol (15 mL) was hydrogenated at room temperature for 12 h. The title compound was obtained as 2,2,2-trifluoroacetate salt a bright beige solid (4.1 mg, 4.32%) .
LC-MS: m/e = 390.2 (M+H)+ 15 Route b) The title compound was prepared in analogy to the method described in Example 213 starting from tert-butyl—4-(4-oxo(2-quinolinyl)ethy1)—4,5-dihydrothieno[3,2- c]pyridinyl)-piperidine-l carboxylate from Example 214.
LC-MS: m/e = 390.2 (M+H)+ 20 11.6 ation of compounds of the formula I in which A is Al, X1 is CH, R7 is Y2- Cyc2 and X3 is S 194 EXAMPLE 2 l 6: 3-Methyl(pyridin—4—y1)—5-(2-(quino linyl)ethyl)thieno [3 ,2- c]pyridin-4(5H)-one N |\ \ s /| \N 216. l 7-Iodomethyl(2-quino linyl)ethyl)thieno [3 ,2-c]pyridin-4(5)H-one To a mixture of triphenylphosphine (901 mg, 3.44 mmol) and DIAD (1.2 mL, 6.01 mmol) in THF (10 mL), 7—iodo—3—methyl—thieno[3,2-c]pyridin-4(5)H-one (500 mg, 1.72 mmol) was added followed by the addition of 2-(quinolinyl)ethanol from example al (327 mg, 1.89 mmol) at 15°C under en. The mixture was stirred ght at room temperature. The mixture was extracted with EtOAc/HZO, the EtOAc 10 layers were washed with 1N HCl and then discharged. The s layer was lized by aq.NaHC03 and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude title product was recrystallized from ethyl acetate to give the title compound as bright beige solid (yield: ). LC-MS: m/e = 447.0 (M+H)+; 15 Rt: 211 min. 2 l 6.2 3 -Methyl(pyridin—4-yl)—5—(2—(quinolinyl)ethyl)thieno [3 ,2-c]pyridin—4(5H)- one The title compound was prepared in analogy to the method described in Example 20 1652 but using 7-iodomethyl(2-quinoliny1)ethyl)thieno[3,2-c]pyridin-4(5)H- one (0.22 mrnol) and pyridin—4-ylboronic acid (0.22 mmol). The title compound was obtained as white solid (yield: 22.9%).
LC-MS: m/e = 398.1 (M+H)+ 25 II.7 Preparation of compounds of the formula I in which A is Al, X1 is N, R1 is Y1- 1 Cyc 195 EXAMPLE 217: 3-(Pyridin—4-yl)—5—[2—(5,6,7,8-tetrahydroquinolin yl)ethyl]thieno[2,3-d]pyridazin—4(5H)—one hydrochloride The title compound was prepared in analogy to the method described in e 5 1.6 but starting from 3-(pyridinyl)thieno[2,3-d]pyridazin-4(5H)one (obtainable by Suzuki coupling of 3-bromo-5H-thieno[2,3-d]pyridazinone from Example 3.3 and pyridinyl boronic acid as described above) and 2-(5,6,7,8-tetrahydroquinolin—2- yl)ethanol from example a6). The title compound was obtained as white solid.
LC-MS: m/e = 389.1 (M+H)+ 1 0 EXAMPLE 218: (R)-tert-butyl 3-(3—(2—(imidazo[1,2—a]pyridin—2-yl)ethyl)oxo-3,4- dihydrophthalazin—5-ylamino)pyrrolidine— 1 —carboxylate o >70 CH OHN A reaction tube was charged with 8—chloro(2-(imidazo[l ,2-a]pyridin 15 yl)ethyl)phthalazin—1(2H)-one (155 mg, 0.48 mmol), BINAP (17.83 mg, 0.029 mmol), tris(dibenzylideneacetone)dipalladium(0) (8.74 mg, 9.55 umol) and cesium carbonate (311 mg, 0.955 mmol) in 1591 ul oftoluene. The mixture was stirred for l h under argon. To this mixture, (R)-tert—buty1 opyrrolidine-l-carboxylate (89 mg, 0.477 mmol) was added. The reaction e was heated in a microwave at 105 °C for 20 h. 20 After completion of the reaction, the on mixture was ted with water and DCM. The organic layers were washed with water, dried over MgSO4, filtered, and concentrated under d pressure. The crude product was purified by column chromatography using a normal phase Companion® system (4 g gold, gradient elution: DCM to 10% methanol). Yield: 24.73%. LC-MS: m/e = 475.2 (M+H)+. 25 196 EXAMPLE 219: 2-[2-(1midazo[l ,2—a]pyridin—2-yl)ethyl] [(3 R)-pyrrolidin-3 - ylamino]phthalazin— l (2H)-one dihydrochloride H / 13 OHN A solution of (R)-tert-butyl 3-(3-(2-(imidazo[1,2-a]pyridinyl)ethyl)oxo-3,4- 5 dihydrophthalazin—S-ylamino)pyrrolidinecarboxylate from Example 218 (52 mg, 0.110 mmol) in 20 mL ofDCM was mixed with 1 mL ofHCl containing isopropanol and the reaction mixture was stirred under en for 12 h at room temperature. The reaction mixture was mixed with diisopropyl ether and the supernatant was discharged.
Then, the residue was dissolved in isopropanol and diisopropyl ether was added. The 10 supernatant was rged. The precipitate was dried to give 24 mg of a yellow salt as solid foam. LC—MS: m/e = 357.2 (M+H)+ 11.8 Preparation of compounds ofthe formula I in which A is Al, X1 is N, X3 is -C(R9)=C(R8)- with R9 being Y3—cyc3 1 5 EXAMPLE 220: 5-(3-hydroxyphenyl)—2—[2-(quinolin—2-yl)ethyl]phthalazin— l (2H)-one \ o / N N. m D O The title compound was prepared in y to the method described in Example 1 92. 20 11.9 Preparation of compounds of the formula I in which A is A3, X1 is CH and R1 is Yl-Cyc1 EXAMPLE 221 : (E)—8-(Pyridin—4—yl)—2—(2—(quino yl)Vinyl)isoquino lin- l (2H)-one 197 221 . 1 8-(Pyridinyl)isoquinolin-1(2H)—one A reaction tube was charged with 8—bromoisoquinolin-1(2H)—one (1 g, 4.46 mmol) and a mixture of 3 mL of ethanol and 3 mL of toluene under argon. To this, an s sodium bicarbonate solution (2 M, 710 mg, 3.35 mL, 6.69 mmol) was added and then pyridinylboronic acid (549 mg, 4.46 mmol). Then, tetrakis(triphenyl— ne)palladium (516 mg, 4.46 mmol) was added. The on mixture was heated in a Biotage microwave at 130 °C for 30 min. The reaction was monitored by TLC (DCM/methanol = 9: 1). After tion of the reaction, the reaction mixture was 10 mixed with water and dichloromethane. The solid was sucked off. The organic phase was washed with an aqueous NaHC03 solution, brine, dried (MgSO4) and ated.
The residue was taken up with EA and the precipitate was sucked off. The crude product was purified by chromatography (Campanion normal phase, gradient elution, using 3-10% DCM in methanol) to yield 465 mg (46.9%) of the title compound. 15 22 1 .2 (E)—8-(pyridinyl)(2-(quinolinyl)vinyl)isoquino lin- 1 (2H)-one idin—4-yl)isoquinolin—1(2H)—one (40 mg, 0.180 mmol) and (E)—2-(2- bromovinyl)quino line (50.6 mg, 0.216 mmol) were dissolved in 2 mL ofDMF under argon. The reaction mixture was stirred for 10 h at 115°C. Then, water and EA were 20 added. The organic phase was washed with water and brine, dried (MgSO4) and concentrated. The crude product was purified by chromatography (CombiFlash, normal phase, gradient elution, using 3-5% DCM in methanol) to yield the title compound as yellow solid (27 mg, 40.0%). LCMS: 3761; Rt = 1.497. 25 11.10 Preparation of compounds of the formula I in which A is A4, X1 is CH and R1 is Yl-Cyc1 EXAMPLE 222: anti (rac) 8—(Pyridin—4—yl)—2—(2—(quinolinyl)cyclopropyl)iso- quino lin- 1 (2H)-one 198 idin—4-yl)isoquinolin—l(2H)—one (200 mg, 0.900 mmol) and syn(2- bromocyclopropyl)quinoline (223 mg, 0.900 mmol) from Example cl) were dissolved in 10 mL ofDMF under argon. The reaction mixture was stirred for l h at 115°C. Then, an s solution of sodium chloride and DCM were added. The phases were separated. The organic phase was washed with brine, dried (MgSO4) and evaporated.
The crude product was taken up in a small amount of EA. The precipitate formed was sucked off and dried to yield anti (rac) 8-(pyridinyl)(2-(quinolin yl)cyclopropyl)isoquinolin-l(2H)-one as bright beidge solids (248 mg, 70.8%). LCMS: 10 390.2.
Separation of anti (rac) 8-(pyridin—4—yl)—2—(2—(quinolin—2- yl)cyclopropyl)isoquino lin- l (2H)-one 230 mg (0.591 mmol) of anti (rac) idin—4-yl)(2-(quinolin 15 yl)cyclopropyl)isoquinolin-l(2H)—one dissolved in 60 ul of roacetic acid were separated by chromatography (chiral chromatography, Chiralpack AD-H, nheptane /EtOH) to give 76 mg (33.0%) of compound 222a with positive rotation and 62 mg (27%) of compound 222b with negative rotation.
Compound 222a: (+) 8-(pyridin—4-yl)—2—(2-(quinolin 20 yl)cyclopropyl)isoquino lin- l (2H)-one [or] = + 87.80 (methanol, 1 ; yellow solid.
Compound 222b: (-) 8-(pyridin-4—yl)(2—(quinolin—2—yl)cyclopropyl)isoquinolin- 1 (2H)-one [CL] = - 983° (methanol, 1 mg/mL); bright yellow solid. 25 11.11 Preparation of compounds of the formula I in which A is A], X1 is CH and R1 is Yl-Cyc1 EXAMPLE 223: 8-(Pyridin—4—yl)—2—(2—quinolin—2—yl—ethyl)isoquinolin- l (2H)-one 199 A flask was charged with idin—4-yl)isoquinolin-1(2H)-one (0,113 mmol, 50 mg) in NHg/ethanol (2 mol/L) (15 mL, 30,0 mmol). Then, Raney-Nickel in water (5 drops) was added. At room temperature, the en flow was turned on. The hydrogenation reaction was stopped after 4 days. Unter nitrogen, the reaction mixture was filtered over Celite and evaporated to yield 52 mg (80% yield) of the crude title product as soli, yellow oil. The crude product was purified by Combi-flash chromatography (gradient elution using methanol/DCM up to a tration of 10%).
LC-MS: 378.2. 10 11.12 Preparation of compounds of the formula I in which A is A4, X1 is CH and R1 is Yl-Cyc1 EXAMPLE 224: anti (m) 3-(Pyridin—4-yl)—5—(2—(quinolinyl)cyclopropyl)thieno[3,2- 15 c]pyridin-4(5H)-one N\ mN\ o,/ ,\ \ s 224. l 3-(Pyridin—4-yl)thieno [3 yridin—4(5H)—one 3-Bromothieno[3,2-c]pyridin-4(5H)—one (1000 mg, 4.35 mmol) from Example 202.3 was suspended in 2 mL of toluene and 2 mL of ethanol under argon. Then, an 20 aqueous solution of sodiumcarbonate (691 mg, 6.52 mol, 2 M) was added. To the suspension, pyridin—4-boronic acid (534 mg, 4.35 mmol) and then tetrakis(triphenyl phosphine)palladium(0) (502 mg, 0.435 mmol) were added. The reaction e was heated in a Biotage microwave at 130 °C for 30 min. Water and EA were added. The precipitate formed was collected to give 480 mg of the title nd. The filtrate was 25 acidified with 2 M HCl and extracted with EA (twice). The aqueous phase was basified and extracted three times with EA. The organic phases were combined, washed with 200 HCl, dried (MgSO4) and evaporated to give further 43 mg of the title compound. Total yield: 523 mg (52.4%). LC-MS: 229.1 [M+H]+; Rt: 0.427. 224.2 anti (m) 3-(Pyridin—4-yl)(2-(quino1inyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one The title compound was prepared in analogy to the method described in Example 222. Yield: 57.7%, LC-MS: 396.1. 11.13 Preparation of compounds of the formula I in which A is A3, X1 is N and R1 is Y1- 10 Cyc1 EXAMPLE 225: Pyridin—4—yl—2—((2—quinolin—2—yl-vinyl)-2H-phthalazinone / l / N “1‘ N \ The title compound was ed according to the general procedure given above 15 but starting from 8-pyridinyl-2H-phthalazin—1-one. Yield: 5%. LC-MS: 377.1 11.14 Preparation of compounds ofthe formula I in which A is A4, X1 is N and R1 is Y1- Cyc1 20 EXAMPLE 226: anti (rac) 8-Pyridin—4-y1—2-(2-quinolinyl-cyclopropyl)-2H- phthalazinone 2,2,2,—trifluoroacetate N \ l / N 'T‘ N \ Under argon, a flask was charged with (E)(pyridinyl)(2-(quinolin yl)vinyl)phthalazin—l(2H)—on (85 mg, 0.226 mmol) in dichloroethane, extra dry, (2,823 25 mL) at 0°C. Then, diethylzinc (lmol/L in ) (1,129 mL, 1,129 mmol) was added within 5min at -3 to 0°C. The on mixture was stirred for 10 min at 0°C.
Diiodomethane (0,182 mL, 2,258 mmol) was added at 0°C within 2 min. The reaction 201 mixture was warmed to room temperature and stirred overnight at room temperature.
Then, a further portion of diethyl zinc (lmol/L in hexane) (1,129 mL, 1,129 mmol) was added at 0°C followed by a further portion ofdiidomethane (0,182 mL, 2,259 mmol), and the on e was warmed to room temperature. The reaction mixture was stirred for fiirther 3 days at room temperature. The reaction mixture was poured onto an ice-cold aqueous solution ofNaHCO3 (5%). The reaction mixture was extracted with DCM (3 . The organic phase was washed with brine, water, dried over sodium e and evaporated. Purification by prep. HPLC yielded 7.] mg (6.23% yield) of the title compound. LC-MS: 391.1. 10 II. 15 Preparation of compounds of the formula I in which A is Al, X1 is CH, R7 is Y2- Cyc2 and X3 is 0 EXAMPLE 227: 7-(Pyridin—4-yl)—5—[2—(quinolin—2—yl)ethyl]furo[3,2-c]pyridin-4(5H)- 15 one /| g \.
N N |\\ \ o ,f’ \l N 227.1 7-Bromo(2-(quinolin-2—yl)ethyl)furo[3,2-c]pyridin—4(5H)-one DIAD (4.91 mmol, 992 mg) was added dropwise to PPh3 (753 mg, 2,80 mmol) in 20 mL of THF. The mixture was stirred for 30 min. Then 7-bromofi1ro[3,2-c]pyridine- 20 4(5H)—one (300 mg, 1.402 mmol) was added followed by the addition of 2-(quinolin yl)ethanol (243 mg, 1.402 mmol). The reaction e was stirred overnight at room temperature. The reaction mixture was extracted with water/ ethyl acetate. The organic phase was extracted with 1N HCl. The acidic aqueous phase was basified with 1N NaOH and extracted with DCM. The organic phase was extracted with water, dried over 25 MgSO4, filtered, concentrated and d by tography to give the title compound as white solid (119 mg, 23%). 227.2 7-(Pyridinyl)[2—(quinolin—2—yl)ethyl]furo[3,2-c]pyridin—4(5H)-one 202 The title compound was prepared in analogy to the method described in Example 165.2 but using 7-bromo(2-(quino1iny1)ethy1)furo[3,2-c]pyridin-4(5H)-one (61.7 mg, 0.167 mmol) and pyridin-4—ylboronic acid (22.82 mg, 0.167 mmol mmol). The title nd was obtained as yellowish solid (yield: 22 mg, 35.89%).
LC-MS: m/e = 368.1 (M+H)+ 11.16 Preparation of compounds of the formula I in which A is Al, X1 is CH, R1 is Yl-Cyc1 and X3 is 0 10 EXAMPLE 228: 3-(Pyridin—4-yl)—5-(2-(quinoliny1)ethyl)fi1ro[3,2-c]pyridin-4(5H)— one hloride 228.1 (E)(4-Bromofiiranyl)acryloyl chloride To a solution of (4-bromofi1ran—2—yl)acrylic acid (3.8 g, 17.51 mmol) in 35 15 mL oftrichloromethane was added a solution of 2.6 mL of l chloride in 200 uL of DMF. The reaction mixture was refluxed for 1 h and after cooling to room temperature trated to give 4 g (97% yield) ofthe title nd as light brown oil. 228.2 (E)(4-Bromofiiranyl)acryloy1 azide 20 A solution of (E)(4-bromofuran—2—yl)acryloy1 chloride (4.0 g, 16.99 mmol) in 10 mL of 1,4-dioxane was added to a solution of sodium azide (2.21 g, 34.0 mmol) in 10 mL of water and 10 mL of 1,4-dioxane. The reaction mixture was stirred for 2.5 h at room temperature. The reaction mixture was mixed with ethyl acetate and water. Phases were separated and the aqueous phase was extracted with ethyl acetate (twice). The 25 combined organic phases were washed with brine, dried, filtrated and concentrated to give 4 g of the title compound (yield: 97%). 228.3 3-Bromofuro[3,2—c]pyridin-4(5H)—one 203 A on of (E)(4-bromofuran—2-yl)acryloyl azide (4 g, 16.53 mmol) in 20 mL of diphenyl ether was slowly added to 80 mL of diphenyl ether at 230°C. The mixture was stirred for further 15 min. The reaction mixure was allowed to cool down to room temperature. 100 mL of cyclohexane were added and the reaction mixture was stirred overnight. A precipitate was formed, d off and suspended in 50 mL of cyclohexane. The mixture was stirred for 1 h at room temperature. The precipitate was filtered off to give 2.76 g (78%) of the title compound as brownish solid.
LC-MS: m/e 215.9 (M+H]+ 10 228 .4 3 —(Pyridinyl)furo [3 yridin-4(5H)—one A reaction tube was charged with 3-bromofi1ro[3,2-c]pyridin-4(5H)-one (1000 mg, 4.67 mmol) and a mixture of 1 mL of ethanol and 1 mL of toluene under argon. To this mixture, n—4-ylboronic acid (574 mg, 4.67 mmol) and a 2 M on of N32C03 (743 mg, 7.01 mmol) were added followed by the addition of tetrakis- 15 (triphenylphoshine)palladium (540 mg, 0.467 mmol). The on mixture was heated in a Cem microwave at about 130 °C for about 30 min. After completion of the reaction, EA was added followed by the addition of 2 N HCl. The mixture was extracted twice with EtOAc. The s layer was extracted three times with ethyl acetate. The combined organic layers were washed with brine, dried over MgSO4, d, and 20 concentrated under reduced pressure. The crude product was purified by chromatography to give the title compound as dark yellow solid (430 mg, 43.4%). 228.5 3 -(Pyridinyl)-5 -(2-(quino1in-2—y1)ethyl)furo [3 ,2-c]pyridin—4(5H)-one To a solution of triphenylphosphine (247 mg, 0.942 mmol) in THF (10 mL), 25 DIAD (0.321 mL, 1.65 mmol) was added. The mixture was cooled to 15°C. 3-(Pyridin— 4-y1)furo[3,2-c]pyridin—4(5H)-one (100 mg, 0.471 mmol) and 2-(quinolin—2-yl)ethanol from example from example a1 were added. After stirring overnight at room temperature, EA and 2M HCl were added. The phases were separated and extracted with EA. The organic phase was washed with water. The acidic aqueous phase was 30 basified with 2M NaOH and extracted with ethyl acetate. The combined c layers were washed with brine, dried over MgSO4, filtered, and concentrated under reduced 204 re. The residue was purified by tography to give 69 mg ofthe title compound ) as hydrochloride salt as dark yellow solid.
LC-MS: m/e 368.1 (M+H)+ 5 The compounds of examples 229 to 231 were prepared in analogy to the method described in example 228.
EXAMPLE 229: 5-(2-(1H-Benzo[d]imidazolyl)ethyl)(pyridiny1)furo[3,2- c]pyridin-4(5H)-one hydrochloride lO The title compound was obtained as grey solid (Yield: 3.46%).
LC-MS: m/e 357.1 (M+H)+ EXAMPLE 230: 5-(2-(Imidazo[ l ,2-a]pyridin—2—yl)ethyl)(pyridinyl)furo[3 ,2- 15 c]pyridin-4(5H)-one hydrochloride The title compound was obtained as grey solid (Yield: 30.3%) LC-MS: m/e 357.1 (M+H)+ 20 EXAMPLE 231: 3-(Pyrimidinyl)(2-(quinolinyl)ethyl)furo[3,2-c]pyridin-4(5H)- 0116 The title compound was obtained as bright yellow solid (Yield: 28.13%) 205 LC-MS: m/e 369.1 (M+H)+ 11.17 Preparation of compounds ofthe formula I in which A is A4, X1 is CH, R1 is Yl-Cyc1 and X3 is 0 E 232: anti 3-(Pyridazinyl)—5-(2—(quinolin—2—y1)cyclopropyl)furo[3,2- c]pyridin-4(5H)-one 232. 1 anti 3-Bromo—5—(2—(quinolin—2—yl)cyclopropyl)furo [3 yridin—4(5H)-one 10 A microwave reaction tube was charged with 3-bromofiiro[3,2-c]pyridin-4(5H)- one (300 mg, 1.402 mmol) from Example 228.3, syn 2-(2-bromocyclopropyl)quinoline (348 mg, 1.402 mmol) from example cl), cesium carbonate (913 mg, 2.80 mmol) and 5 mL ofDMF. The reaction mixture was stirred for 1.5 h at 110°C. EA and water were added. The phases were separated. The aqueous phase was washed with EA (twice). 15 The organic phases were washed with brine, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was purified by chromatography to give 450 mg (84%) of the title compound as light brown yellow solid 20 232.2 anti 3-(pyridazinyl)(2-(quinolin—2-yl)cyclopropyl)furo[3,2-c]pyridin— 4(5H)—one The compound from Example 232.1 was ded in 1 mL oftoluene and 1 mL of methanol under argon. Then, an aqueous solution of sodium carbonate (41.7 mg, 0.39 mol, 2 M) was added. To the sion, ,5,5-tetramethyl-1,3,2—dioxaborolan—2- 25 yl)pyridazine (54 mg, 0.26 mmol) and then tetrakis(triphenylphosphine)palladium(0) (30.3 mg, 0.026 mmol) were added. The reaction mixture was heated in a Biotage microwave at 130 °C for 30 min. Water and EA were added. The aqueous phase was extracted twice with EA. The organic phases were combined, washed with HCl, dried (MgSO4) and evaporated. The residue was purified by chromatography to give 15 .5 mg 30 (yield: 15.53%) of the title nd as light brownish solid. 206 LC-MS: m/e 380.8 (M+H)+ EXAMPLE 233: anti 3-(pyridin—4—yl)—5-(2—(quinolinyl)cyclopropyl)furo[3,2- c]pyridin-4(5H)-one The title compound was ed according to the method described for example 232.2. The title compound was obtained as yellow solid. LC-MS: m/e 379.9 (M+H)+ 11.18 Preparation of compounds of the formula I in which A is A5, X1 is N, R1 is 10 l and x3 is N=C(R8) EXAMPLE 234: 4-(Pyridin—4-yl)-6—(2—(quino1in—2—yl)allyl)pyrido[2,3-d]pyridazin- 5(6H)-one 15 234.1 (E)—Ethyl 2-((2-acetylhydrazono)methyl)—4-chloronicotinate To 2.1 g (9.83 mmol) of ethyl 4-chloro-2—formylnicotinate in 60 mL of ethanol was added 0.874 g (11.80 mmol) of acetohydrazide at room temperature. Then, the reaction mixture was heated under reflux for 2h. The reaction mixture was extracted with water/DCM. The c phase was washed with brine, dried, filtrated and 20 trated to give 1.95 g (73.6%) of the title compound as white solid. 234.2 4-Chloropyrido[3,2-d]pyridazin-5(6H)-one (E)-ethyl 2-((2-acetylhydrazono)methyl)chloronicotinate (2850 mg, 10.57 mmol) in 15 mL of dioxane was mixed with aq. NaOH (42.3 mg, 1.06 mmol, 2 M). The 25 reaction mixture was stirred in a microwave for 60 min at 145°C. After cooling to room temperature, the solid was filtered off. The solid was dissolved in methanol and the 207 residue was filtered off. The e was concentrated and triturated with EA to give 1.4 g (73.0%) of the title compound as orange brown solid. 234.3 4-(Pyridinyl)pyrido[3,2-d]pyridazin—5(6H)—one A reaction tube was charged with a solution of4—chloropyrido[3,2-d]pyridazin- 5(6H)-one (0.354 mmol) in 1 mL of ethanol and 1 mL oftoluene under argon. To this suspension, a 2M solution ofNa2C03 (0.531 mmol) and pyridinylboronic acid (0.354 mmol) were added. Then, tetrakis(triphenylphoshine)palladium (0.035 mmol) was added. The reaction e was heated in a CEM ave at about 130 °C for about 10 20 min. The reaction was monitored by TLC (DCM/methanol = 9:1). After completion ofthe reaction, EA was added followed by the addition of water. The organic layer was basified with NaHCOg. The organic layers were dried over MgSO4, d, and concentrated under reduced pressure. The crude product was purified by column chromatography (25.2%). 15 234.4 4-(Pyridinyl)(2-(quinolin-2—yl)allyl)pyrido[2,3-d]pyridazin—5(6H)-one A reaction tube was charged with 307 mg (1.369 mmol) of 4-(pyridin yl)pyrido[2,3-d]pyridazin-5(6H)-one, syn(2—bromocyclopropyl)quinoline from example cl and cesium ate (892 mg, 2.74 mmol). The reaction e was 20 heated for 6h at 110°C under argon. After cooling to room temperature, EA was added followed by the addition of water. The organic layer was basified with NaHCOg. The organic layers were dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was purified by column chromatography to give 106 mg of the title compound (yield: ). 25 11.19 Preparation of compounds of the formula I in which A is A4 EXAMPLE 23 5: anti 8-(Oxetan—3-ylamino)(2-(quino lin yl)cyclopropyl)isoquino lin- 1 (2H)—one —O /J %N\ O HN N$6 30 208 235.1 anti 8-bromo(2-(quino1in—2—y1)cyclopropyl)isoquinolin- 1 (2H)-one 8-bromoisoquinolin-l(2H)-one (300 mg, 1.339 mmol) and syn(2- yclopropyl)quinoline (332 mg, 1.339 mmol) from Example cl) were dissolved in 4 mL ofDMF under argon. Cesium carbonate (873 mg, 2.68 mmol) was added. The reaction mixture was stirred for 1 h at 115°C. Then, an aqueous solution of sodium chloride and DCM were added. The phases were separated. The organic phase was washed with brine, dried (MgSO4) and evaporated. The crude t was taken up in a small amount of EA. The precipitate formed was sucked off and dried to yield anti 8- bromo(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one as bright beidge solid 10 (330 mg, 63.0%). LC—MS: 391.0 (M+); Rt: 1.6 min.
Separation of anti 8—bromo—2—(2—(quinolin—2—yl)cyclopropyl)isoquino lin-1(2H)- one 33g of anti 8-bromo(2-(quinolin—2—yl)cyclopropyl)isoquinolin-1(2H)-one were 15 suspended in pyl acetate/heptane 1:4 (5 mL/g). The racemate was separated by chiral chromatography to give 16.1 g of anti (+) 8—bromo(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one (compound 235.1a) R = 4.885 min and 20.25 g of anti (-) 8-bromo(2-(quinoliny1)cyclopropyl)isoquinolin-1(2H)-one (compound 235.1b), R = 9.253 min. 20 235 .2 anti tanylamino)—2—(2-(quinolinyl)cyclopropyl)isoquinolin- 1 (2H)- one A microwave reaction vial was charged with the anti 8-bromo(2-(quinolin lopropyl)isoquinolin-1(2H)—one from Example 235.1 (70 mg, 0.18 mmol), 25 CS2CO3 (175 mg mg, 0.54 mmol), Pd2(dba)3 (3.28 mg mg, 3.58 umol) and BINAP (6.68 mg, 10.73 umol). The solids were purged with argon for 1 h. A separate flask was charged with toluene (2 mL) and amine (19.6 mg, 0.18 mmol), degas with argon for 1 h and then transferred to the microwave reaction vial under inert conditions.
The resulting reaction mixture was heated on microwave at 105 °C for 48 h. The 30 reaction mixture was poured into water and extracted with DCM. The solids were removed. The organic layer was washed with water, dried over MgSO4, filtered and 209 concentrated in vacuo. The residue was purified on a silica column (eluent: DCM/methanol) and then triturated with EA to afford the title product (35 mg, 51.0 %).
LC-MS: m/e = 384.2 (M+H)+.
Separation of anti 8-(oxetan—3-y1amino)(2-(quinolin—2- yl)cyclopropyl)isoquino lin- 1 (2H)—one 484 mg (0.591 mmol) of anti tanylamino)(2—(quino1in—2- yl)cyclopropyl)isoquinolin-1(2H)-one dissolved in 20 mL of ethanol-DCM (1 :1 mixture) and 1 mL of the solution were tographed on a Whelk-O (R,R) 2 x 25 10 cm (10 mkm particles) column in EtOH (containing 0.1% n-propylamine) at 40 mL/min and 254 nm detection. Peaks obtained were concentrated to give first-eluting compound 235a (220 mg, chiral purity 98.6%) and second—eluting compound 235b (230 mg, chiral purity 97.3%), recovery ~93%.
Compound 235a: (+) anti 8-(oxetan—3—ylamino)(2-(quinolin 15 yl)cyclopropyl)isoquino lin- l (2H)-one Compound 23 5b: (-) anti 8-(oxetan—3-ylamino)(2-(quinolin yl)cyclopropyl)isoquino lin- l (2H)—one. 20 EXAMPLE 236: anti (-) 8-(pyridazin—4—y1)(2-(quinolin yl)cyclopropyl)isoquino lin- l (2H)—one The title compound was prepared according to the method described in example 232.2 starting from anti (-) 8-bromo-2—(2-(quinolinyl)cyclopropyl)isoquinolin-l(2H)- one und 235.lb). LC-MS: m/e = 391.2 (M+H)+. 25 E 237: anti (-) 8-(6-fluoropyridin—3—yl)—2-(2—(quino1in—2— yl)cyclopropyl)isoquino lin- 1 (2H)-one The title compound was prepared ing to the method described in example 30 232 starting from anti (—) 8—bromo—2—(2—(quinolin—2—y1)cyclopropyl)isoquinolin-l(2H)- one (compound 235.lb) and 2—fluoro(4,4,5,5-tetramethyl—l,3,2-dioxaborolan—2- yl)pyridine. LC-MS: m/e = 408.2 (M+H)+ 210 [ct] = -lO7° (methanol) EXAMPLE 238: anti (-) 8-(2-fluoropyridiny1)—2-(2-(quinolin yl)cyclopropyl)isoquino lin- l (2H)—one The title compound was ed according to the method bed in example 232 starting from anti (-) 8-bromo-2—(2-(quinolin—2-yl)cyclopropyl)isoquinolin-l(2H)- one und 235.1b) and 2-fluoropyridin—4-ylboronic acid.
LC-MS: m/e = 408.2 (M+H)+ 10 EXAMPLE 239: anti 8—(pyridinyl)(2-(quino1inyl)cyclopropyl)isoquinolin- l(2H)-one hydrochloride The title compound was prepared ing to the method described in example 232 starting from anti 8-bromo(2-(quinolin—2—yl)cyclopropyl)isoquinolin-l(2H)-one 15 and pyridinylboronic acid. LC-MS: m/e = 390.1 (M+H)+ Separation of anti 8-(pyridin-3—y1)—2-(2-(quinolinyl)cyclopropyl)isoquinolin- l (2H)-one: 320 mg of anti 8-(pyridin-3—y1)—2—(2-(quinolinyl)cyclopropyl)isoquinolin- 20 l(2H)-one were subjected to a chromatography on a Chiralpak IC column, eluent: 500 parts of n-heptane, 450 parts ofDCM, 50 parts of methanol and 1 part of triethylamine to give 105 mg of compound 239a (Rt: 4.74 min), 98.8% ee and 120 mg of compound 23% (Rt: 4.68 min), 97.6% ee.
Compound 239a: (+) anti 8-(pyridinyl)(2-(quino1in 25 yl)cyclopropyl)isoquino lin- 1 (2H)-one [CL] = + 108.5° (methanol) nd 23%: (—) anti 8-(pyridin—3-yl)—2-(2-(quinolin—2- yl)cyclopropyl)isoquino lin— l (2H)—one 30 [on] = -94.8° (methanol) 211 EXAMPLE 240: anti 8-(pyrimidin—5—yl)(2-(quinolinyl)cyclopropyl)isoquinolin- l(2H)—one The title compound was ed according to the method described in example 232 starting from anti 8-bromo(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one and pyrimidin-S-ylboronic acid.
LC-MS: m/e = 391.1 (M+H)+ 10 tion of anti 8-(pyrimidin—5—y1)—2—(2—(quinolin—2-yl)cyclopropyl)isoquinolin- l (2H)-one: 330 mg of anti 8-(pyrimidiny1)—2—(2—(quinolinyl)cyclopropyl)isoquinolin- l(2H)-one were subjected to a chromatography on a Chiralpak IC column, eluent: 500 parts of n-heptane, 450 parts of DCM, 50 parts anol and 1 part of triethylamine 15 to give 112 mg of compound 240a (Rt: 980), 95.6% ee and 98 mg of compound 24% (R: 5.52), 81.2% ee.
Compound 240a: anti (+) 8-(pyrimidin—5-yl)(2-(quinolin yl)cyclopropyl)isoquino lin- l (2H)-one [ct] = + 118.30 (methanol) 20 Compound 240b: anti (-) 8-(pyrimidin—5-yl)—2-(2—(quinolin yl)cyclopropyl)isoquino lin- l ne [at] = -l20.4o (methanol) 25 EXAMPLE 241: anti 8-(l-methyl-lH-pyrazolyl)(2-(quinolin yl)cyclopropyl)isoquino lin- l (2H)-one CH3 / —N / 212 The title compound was prepared according to the method described in example 232 starting from anti 8-bromo-2—(2—(quino1inyl)cyclopropyl)isoquinolin-l(2H)-one and l-methyl(4,4,5 ramethyl-l ,3 ,2-dioxaborolan—2-yl)- l H-pyrazo le.
LC-MS: m/e = 393.1 (MJrH)+ EXAMPLE 242: anti 8-(3-fluoropyridinyl)—2-(2-(quino1in yl)cyclopropyl)isoquino lin- l (2H)—one N\ | / \ (IDI F N/ N \ The title compound was ed according to the method described in example 10 232 ng from anti 8-bromo—2—(2—(quinolin—2—yl)cyclopropyl)isoquinolin-l(2H)-one and 3-fluoro(4,4,5 ,5 -tetramethyl—l ,3 ,2—dioxaborolan—2-yl)pyridine.
LC—MS: m/e = 408.1 (M+H)+ EXAMPLE 243: anti 8-(2-fluoropyridin-4—yl)—2—(2-(quinolin 15 yl)cyclopropyl)isoquino lin- l (2H)—one N F \ I / \ o / N N \ The title nd was prepared according to the method described in example 232 ng from anti 8-bromo—2-(2—(quino1in—2-yl)cyclopropyl)isoquinolin-l(2H)-one and 2-fluoropyridinylboronic acid. LC—MS: m/e = 408.1 (M+H)+ 20 EXAMPLE 244: anti 8-((3S)—3-hydroxypiperidin—l-yl)—2—(2-(quinolin—2- y1)cyclopropyl)isoquinolin- 1 (2H)-one (2E)-butenedioate N ,OH \ O / N N \ The title compound was prepared according to the method described in example 25 173 starting from anti 8-bromo—2—(2—(quinolin—2—yl)cyclopropyl)isoquinolin-l(2H)-one 213 and (S)-piperidinol. The title compound was converted into the fumarate salt. LC- MS: m/e=4l2.2 Examples 245 to 253 were prepared in analogy to the method described in 5 example 244.
EXAMPLE 245: anti 8-(3-methoxypiperidin—1—yl)—2-(2—(quinolin—2— yl)cyclopropyl)isoquinolin- 1 (2H)-one (2E)-butenedioate 10 LC-MS: m/e = 426.2 EXAMPLE 246: anti 8-morpholino(2—(quinolin—2-yl)cyclopropyl)isoquinolin-l(2H)- 0116 O \ Org 15 LC-MS: m/e = 398.2 (M+H)+ tion of anti 8-morpholino—2-(2—(quinolin—2-yl)cyclopropyl)isoquinolin- l(2H)—one: 20 246 mg of anti 8-morpholino(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)— one were subjected to a chromatography on a Chiralpak IC column, eluent: 500 parts of n-heptane, 450 parts of DCM, 50 parts of methanol and 1 part of triethylamine to give 95 mg of compound 246 a (R: 8.1), 90.2% ee and 74 mg of compound 246b (R: 9.4), 90.2% ee. 25 Compound 246a: anti (+) 8—morpholino—2—(2—(quinolin—2- yl)cyclopropyl)isoquino lin- l ne [or] = + l4l.4° (methanol) 214 nd 246b: anti (-) 8—morpholino(2-(quinolin y1)cyclopropyl)isoquino1in- 1 (2H)—one [ct] = -142.00 (methanol) Examples 247: anti 1-(1-oxo(2—(quinolin—2—yl)cyclopropy1)—1 ,2-dihydroisoquino 1in- 8-y1)piperidinecarbonitrile CN WE LC-MS: m/e = 421.2 (M+H)+ 10 EXAMPLE 248: anti 8-((3R,4R)—4—fluoro—3—hydroxypiperidin—1-y1)(2-(quinolin y1)cyclopropy1)isoquinolin- 1 (2H)-one F (53m \ O N / N N \ LC—MS: m/e = 430.2 (M+H)+ 15 EXAMPLE 249: anti 8-((3 S)—3-hydroxypyrrolidin— 2-(2-(quino 1in y1)cyclopropy1)isoquino1in- 1 (2H)—one di[(2E)—butenedioate] OH \ Q 0| N/ N \ LC-MS: m/e = 398.2 (M+H)+ 20 EXAMPLE 250: anti 8—((3R)—3—hydroxypyrrolidin—1—y1)(2-(quinolin—2- yl)cyclopropy1)isoquino 1in— 1 (2H)—one (2E)—but—2—enedioate 215 OH \ 0| N N/ N \ LC-MS: m/e = 398.2 (M+H)+ EXAMPLE 251: anti 8-(methyl(0xetanyl)amin0)(2-(quin01in 5 y1)cyclopropyl)isoquino lin- 1 (2H)-one —0 H30\ 4 \ O N N N \ LC-MS: m/e = 398.1 (M+H)+ EXAMPLE 252: anti 8-(4-methoxypiperidin—1—y1)—2—(2-(quin01in 1 0 y1)cyclopropyl)isoquinolin- 1 ne (2E)—but—2—enedi0ate 9H3 0 LC—MS: m/e = 426.2 (MJFH)+ EXAMPLE 253: anti 8-(4-hydroxypiperidin—1-y1)(2-(quin01in 1 5 y1)cyc10propyl)isoquinolin- 1 (2H)-0ne (2E)—but—2—enedioate OH \ Eli? 0| N/ N \ LC-MS: m/e = 412.2 (M+H)+ EXAMPLE 254: anti 8-(1—acetylpiperidin—4—ylamino)—2-(2-quin01in 20 cyclopropy1)isoquino lin- 1 (2H)—one 216 >LCH Q \ OHN / N N \ The title compound was prepared in analogy to the method described in example 235.
EXAMPLE 255: anti 8-(piperidinylamino)(2-(quinolin yl)cyclopropyl)isoquino lin- 1 (2H)-one hloride /H Q \ Ol N N/ N \ The title compound was prepared in analogy to the method described in e 213 but g for 3 h under reflux and the solvent used was ethanol. 10 LC-MS: m/e = 411.2 (M+H)+.
EXAMPLE 256: syn 8-(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one hydrochloride 15 25 6. 1 8-(pyridinyl)(2-(quinolin—2—yl)cyclopropyl)isoquino lin- 1 (2H)-one The title compound was prepared as decribed in example 222.
LC-MS: 390.1 (M+H)+ 256.2 Separation of 8-(pyridin—4-y1)—2—(2-(quinolinyl)cyclopropyl)isoquinolin- 20 1 (2H)-one: 2.8 g of 8-(pyridinyl)—2—(2-(quinolin—2—yl)cyclopropyl)isoquinolin—1(2H)-one were ved in 50 mL ofEA and separated by column chromatography, normal phase, eluent: DCM/methanol to give compound 222 and compound 256..
Compound 222: anti 8—(pyridin—4—yl)—2—(2—(quinolinyl)cyclopropyl)isoquinolin- 25 l(2H)-one, yield: 80%; LC—MS: 390.1 (M+H)+, Rt: 1.128 min 217 Compound 256: syn 8-(pyridin—4—y1)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one, yield: 4.3%; LC-MS: 390.1 (M+H)+, Rt: 1.127 min.
Examples 257 to 275 were prepared in analogy to the methods described above.
EX. Name LC-MS: m/e (M+H)+/ [a] 257 anti 2-(2-(quinolinyl)cyclopropyl)—8- 397.1 (tetrahydro-2H-pyran—4-yl)isoquino lin- 1 (2H)-one 258 anti (-) 2-(2-(quinolinyl)cyclopropyl)(2-oxa- 424.2 / [a] = -194° 6-azaspiro [3 .4]octan—6-yl)isoquino lin- 1 (2H)-one 259 anti (-) 8-(dihydro— 1 [3 ,4—c]pyrrol— 424.2 / [or] = -193° 5 ,6aH)—yl)—2—(2—(quinolin—2— yl)cyclopropyl)isoquinolin— 1 (2H)—one 260 anti (-) -difluoropiperidin—1—yl)—2—(2— 432.2 /[(x] = -162° (quinolinyl)cyclopropyl)isoquinolin— 1 (2H)-one 261 anti 8-morpholino(2-(quinolin-2— 398.1 yl)cyclopropyl)isoquino lin- 1 (2H)-one, dimethanesulfonate 262 anti 8-(3 -(difluoromethyl)pyrrolidin—1-yl)(2- 432.2 (quinolinyl)cyclopropyl)isoquinolin- 1 (2H)-one, trifluoroacetate 263 anti ,5 S)—3 -oxaazabicyclo[3.2.1]octan—8- 424.2 yl)(2-(quino linyl)cyclopropy1)isoquino lin- 1 (2H)-one 264 anti 8-(4-methylpiperazin— 1 -y1)(2—(quino lin-2— 411.2 yl)cyclopropyl)isoquinolin— 1 (2H)-one, 265 anti 8-(3 -(fluoromethyl)pyrrolidinyl)(2- 414.2 (quinolinyl)cyclopropyl)isoquinolin-1(2H)-one, trifluoroacetate 266 8-((1R,5 xa—3—azabicyclo[3 .2. 1 ]octan—3-y1)- 424.2 2-(2-(quinolinyl)cyclopropyl)isoquino lin— 1 (2H)-one, 218 Ex. Name LC-MS: m/e (M+H)+/ [a] 267 anti 8-(4-fluorophenyl)(2—(quinolin 407.2 yl)cyclopropyl)isoquinolin— 1 (2H)—one, 268 anti 8-(fi1ran—3-yl)(2-(quinolin-2— 379.1 yl)cyclopropyl)isoquinolin- 1 (2H)—one 269 anti 8-(4,5-dihydrofuran—3—y1)—2—(2-(quinolin—2- 3 81 .2 yl)cyclopropyl)isoquinolin— 1 (2H)—one 270 anti (-) 8-(4-methoxyphenyl)(2-(quinolin 419.2 / [ct] = -68° yl)cyclopropyl)isoquinolin-1(2H)-one 271 anti 2-(2—(6—fluoroquino lin—2-yl)cyclopropyl)—8- 416.2 morpholinoisoquino lin— l (2H)—one 272 anti 2-((2-(6-fluoroquinolin—2—yl)cyclopropyl)—8- 408.1 in-3 -yl)isoquino lin— 1 (2H)—one 273 anti 2-(2-(6-fluoroquinolin-2—yl)cyclopropyl)—8- 409.1 (pyrimidin-S oquinolin- 1 (2H)—one 274 anti (6-fluoroquinolinyl)cyclopropyl) 408.1 (pyridinyl)isoquino lin- 1 (2H)—one 275 anti 4-fluoro(pyridinyl)—2-(2-(quinolin 408.1 yl)cyclopropyl)isoquino lin- 1 (2H)-one E 276: anti (-)chloro—8—(pyrimidin—5-yl)(2-(quinolin yl)cyclopropyl)isoquino lin- 1 (2H)—one NAN [ / \ o l / N N \ CI Anti (-)(pyrimidin-5 -yl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one hydrochloride from example 24% was mixed with sodium hydrogencarbonate solution and DCM. The phases were separated and the organic phase dried. Purification by column chromatography ethanol) gave the title compound (7.89% yield) as White solid. LC-MS: m/e = 425.1 Example 276a: anti (-)—4—chloro—8—(pyrimidin—5—yl)(2-(quinolin yl)cyclopropyl)isoquino lin- 1 (2H)—one hydrochloride 219 [or] = +217.7o (methanol) EXAMPLE 277: anti 4-(pyridin—4—yl)—6-(2—(quinolinyl)cyclopropyl)pyrido[2,3- d]pyridazin—5(6H)-one 4-(pyridinyl)pyrido[2,3-d]pyridazin-5(6H)-one (19 mg, 0.085 mmol) was suspended in 1 mL of DMF under argon. Then, cesium carbonate carbonate (55.2 mg, 0.169 mmol)) and syn 2—(2—bromocyclopropyl)quinoline (22,08 mg from , 0,089 mmol) e cl were added. The reaction mixture was heated at 110 °C for 3 h min. Water 10 and EA were added. The aqueous phase was extracted twice with EA. The organic phases were combined, washed with HCl, dried (MgSO4) and evaporated. The residue was purified by chromatography to give 12 mg (yield: 36.2%) ofthe title compound as bright beige solid. LC-MS: m/e 392.1 (M+H)+ 15 es 278 to 300 were prepared in y to the methods described above.
Ex. Name LC-MS: m/e (M+H)+/ Rt [min / [0L] 278 anti 2-(2-(6-fluoroquinolin—2—yl)cyclopropyl) 409.1 (pyridinyl)phthalazin- l (2H)—one 279 anti 7-fluoro-3 -(pyridin-4—yl)—5—(2-(quino lin 414. 1 yl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)—one 280 anti 3-(2-fluoropyridinyl)—5-(2—(6-fluoroquinolin—2— 432.1 yl)cyclopropyl)thieno[3,2—c]pyridin—4(5H)—one 281 anti 5-(2-(6-fluoroquinolin—2-yl)cyclopropyl) 414 (pyridinyl)thieno [3 ,2-c]pyridin-4(5H)-one 282 anti imidinyl)(2-(quinolin 397. l yl)cyclopropyl)thieno [3 ,2—c]pyridin—4(5H)—one 282a anti 3-(pyrimidin—5—yl)—5—(2—(quinolin—2— 397. l yl)cyclopropyl)thieno[3,2—c]pyridin—4(5H)—one, 220 EX. Name LC-MS: m/e (M+H)+/ Rt [min/ [or] hydrochloride, enantiorner 1 282b anti 3-(pyrirnidinyl)-5—(2-(quinolin—2— 397.1 yl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)—one, hydrochloride, omer 2 283 anti 3-(6-fluoropyridiny1)(2-(quinolin—2- 414.1 yl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one hydrochloride 284 anti 3-(2—n1ethylpyrimidin—5-yl)(2-(quinolin 41 1.1 yl)cyclopropyl)thieno[3,2—c]pyridin—4(5H)—one 285 anti idazin—4—yl)—5—(2—(quinolin—2— 397.1 lopropyl)thieno[3,2—c]pyridin—4(5H)—one 286 anti 3-(2-fluoropyridinyl)—5—[(2—(quinolin—2- 414.1 yl)cyclopropyl]thieno[3,2-c]pyridin—4(5H)—one 287 anti 3-(n10rpholinyl)-5[2-(quinolin—2— 404.1 yl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one 288 anti 3-(pyridinyl)[2-(thieno[3,2-b]pyridin 401.5 yl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one 289 anti 3-(pyridinyl)[2-(quinolin 396.1 yl)cyclopropyl]thieno[3,2—c]pyridin-4(5H)—one 290 anti 3-(pyrirnidinyl)-5—[2-(quinolin—2— 398.1 yl)cyclopropyl]thieno[2,3—d]pyridazin-4(5H)-one, hydrochloride 291 anti 3-(pyridiny1)[2—(thieno[3,2-b]pyridin yl)cyclopropyl]thieno[2,3—d]pyridazin-4(5H)-one 292 anti3-(pyridinyl)[2-(thieno[3,2-b]pyridin 402.0 yl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one 293 anti 3—(pyridin—4—yl)—5—[2—(quinolin—2— 396.1 yl)cyclopropyl]thieno[3,2—c]pyridin—4(5H)—one 293a anti (+) 3-(pyridin-4—yl)—5—[2—(quinolin—2— 396.1 yl)cyclopropyl]thieno[3,2—c]pyridin—4(5H)—one 221 EX. Name LC-MS:n1/e(M+H)+/ Rt [min/ [0L] 293b anti (-) 3-(pyridinyl)[2-(quinolin 3961/ [0L] = -301.70 yl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)—one (methanol) 294 anti 3-(pyridinyl)[2—(quinolin 397.1/ R = 1.084 yl)cyclopropyl]thieno[2,3—d]pyridazin-4(5H)-one hydrochloride m lopropyl]thieno[2,3-d]pyridazin-4(5H)-one 296 anti 5 —(pyrimidin-5 -y1)—2-[2-(quino lin—2- 3 91.1 yl)cyclopropyl]isoquinolin-1 (2H)—one 297 anti idinyl)—2—[2—(quinolin—2— 390.1 yl)cyclopropyl]isoquinolin— 1 (2H)—one 298 anti 5-(pyridinyl)[2-(quinolin—2— 390.2 yl)cyclopropyl]isoquino lin- 1 (2H)—one 299 anti 5-(morpholinyl)[2-(quinolin—2— 398 .2 yl)cyclopropyl]isoquino lin- 1 ne 300 anti (-) 4-fluoro(pyrimidinyl)(2-(quinolin 396.1 yl)cyclopropyl)isoquino lin- 1 (2H)—one 11.20 Preparation of compounds ofthe formula I in which A is Al, Yl-Cyc1 and X3 is N=C(R9) EXAMPLE 301: 8-(pyridinyl)[2—(quinolinyl)ethyl]pyrido[3,4-d]pyridazin— 1 (2H)-one 301 . 1 Ethyl 3—chloro—5—formylisonicotinate To a solution of l 5—chloropyridine—3,4—dicarboxylate (2.43 g, 9.43 mmol) in 10 100 mL of toluene was added diisobutylaluminiumhydride (2.68 g, 18.86 mL, 1 molar) in hexane at -70°C. The reaction mixture was stirred for 2 h at this temperature. The 222 reaction mixture was poured into an 5% aqueous acetic acid solution followed by the addition ofEA. The phases were separated and the organic phase was washed with aq.
NaHC03 on. The organic phase was dried and trated to give 2.02 g (100%) ofthe title compound as brown oil. 301.2 (E)-ethyl 3-((2-acetylhydrazono)methyl)—5-chloroisonicotinate To ethyl 3-chloroformylisonicotinate (2 g, 9.36 mmol) in 60 mL of ethanol was added acetohydrazide (0.832 g, 11.24 mmol) at room temperature. The reaction mixture was stirred for 2 h. Then, the reaction mixture was mixed with DCM and water. The 10 organic phase was washed with brine, dried and concentrated. The crude title compound was purified by chromatography (eluent: thanol) to give 220 mg (8.7%) of the title compound as orange oil. 3 01 .3 8-chloropyrido [4,3-d]pyridazin—1(2H)-one 15 (E)—ethyl 3-((2-acetylhydrazono)methyl)—5—chloroisonicotinate (220 mg, 0.816 mmol) and aq. NaOH solution (3.26 mg, 0.082 mol, 2 molar) in 5 mL of dioxane were stirred in a ave at 165°C for 60 min. Then the reaction mixture was basified by addition of aq. NaHC03 and extracted with DCM. The organic phase was washed with brine, dried and concentrated. The crude title compound was d by 20 chromatography (eluent: thanol) to give 25 mg (16.88%) of the title compound as beige solid. 3 01 .4 8-chloro(2-(quinolin-2—y1)ethyl)pyrido[4,3-d]pyridazin-1(2H)-one To 69.3 mg (0.264 mmol) in THF was added diisopropylazodicarboxylate (94 mg, 25 0.463 mmol) in THF under ice-cooling and under nitrogen. Then, the resulting solution was stirred for 30 min at room temperature to give a suspension. 8-Chloropyrido[4,3- d]pyridazin-1(2H)-one (24 mg, 0.132 mmol) in THF and then 2-(quinoliny1)ethanol (22.89 mg, 0.132 mmol) in THF were added and the reaction mixture was stirred ght at room temperature. The total amount of THF was 60 mL. Then, the reaction 30 mixture was mixed with DCM and water. The organic phase was washed with 1N HCl and the organic phase was discharged. The aqueous phase was basified by 1N NaOH 223 and extracted with DCM. The organic phase was dried and concentrated to give 34 mg (76%) of the title compound as bright beige solid. LC-MS: m/e 337.1 (M+H)+. 3 01 .5 8-(pyridinyl) [2-(quino1iny1)ethyl]pyrido [3 ,4-d]pyridazin-1(2H)-one ro(2-(quinolinyl)ethyl)pyrido[4,3-d]pyridazin—1(2H)-one (34 mg, 0.101 mmol) was suspended in 2 mL of toluene and 2 mL of methanol under argon.
Then, an aqueous solution of sodium carbonate (16.5 mg, 0.15 mol, 2 M) was added.
To the suspension, pyridinboronic acid (13.79 mg, 0.101 mmol) and then tetrakis(triphenylphosphine)palladium(0) (11.67 mg, 10.10 umol) were added. The 10 on mixture was heated in a CEM microwave at 130 °C for 30 min. Water and EA were added. The organic phase was ted with water, dried (MgSO4) and evaporated. The residue was purified by chromatography (eluent: DCM/methanol) and then recrystallized from sopropyl ether (1 :1) to give 2.6 mg (yield: 6.79%) of the title compound as off-white solid. LC-MS: m/e 380.1 (M+H)+ 15 11.21 Preparation of compounds ofthe formula I in which A is A1 The compounds of example 302 to 409 were ed in analogy to the methods described above.
Ex. Name LC-MS: m/e (M+H)+ / Rt[min] 302 5-[2-(imidazo[1,2-a]pyridinyl)ethy1]—7-(1-methyl- 454.0 / 1.69 1H-imidazolyl)(pyridin-4—yl)thieno[2,3- d]pyridazin—4(5H)-one 303 5 - [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—7—( 1 -methyl- 1H—pyrazolyl)(pyridin—4—yl)thieno[2,3- d]pyridazin—4(5H)-one 304 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridin 451.2 / 1.74 yl)—3—(pyridin—4—yl)thieno[2,3—d]pyridazin—4(5H)-one 305 5-[2-(6-chloroquinolin—2—yl)ethyl]—3—(pyridin—4- 418. 1 yl)thieno [3 ,2-c]pyridin—4(5H)—one 306 5-[2-(3-methquuinolin—2—yl)ethyl]—3—(pyridin—4- 398.1 224 EX. Name LC-MS: rn/e (M+H)+ / Rt[rnin] yl)thieno[3 ,2-c]pyridin—4(5H)—one 307 5-[2-(8-fluoroquinolin-2—yl)ethyl]—3-(pyridin 402. 1 yl)thieno[3 ,2-c]pyridin—4(5H)—one 308 5-[2-(6-fluoroquinolin—2—yl)ethyl]—3—(pyridin 402. 1 yl)thieno[3 ,2-c]pyridin—4(5H)—one hydrochloride 309 5-[2-(6-fluoroquinolinyl)ethyl](pyridin 402. 1 yl)thieno[3 ,2-c]pyridin-4(5H)-one 310 5-[2—(irnidazo[1,2—a]pyridin—2-yl)ethyl](pyridin 373 .3 yl)thieno[3 ,2—c]pyridin—4(5H)—one 3 1 1 3-(pyridin—4-yl)[2—(quinoxalin—Z—yl)ethyl]thieno[2,3- 3 86. 1 d]pyridazin—4(5H)-one hydrochloride 3 12 5-[2-(1,5-naphthyridinyl)ethyl]—3—(pyridin—4- 3 86. 1 yl)thieno[2,3-d]pyridazin-4(5H)-one 3 13 5 - [2-( 1 H-indazol— 1 -yl)ethyl](pyridin—4—yl)thieno [2,3 - 3 74. 1 d]pyridazin—4(5H)-one 3 14 3 -( 1 -methyl— 1 H-pyrazo1yl)[2-(quinolin—2- 3 8 8. 1 yl)ethyl]thieno[2,3-d]pyridazin—4(5H)—one hydrochloride 315 pyrazol—3-yl)[2—(quinolin—2- 374.1 yl)ethyl]thieno[2,3-d]pyridazin-4(5H)—one 3 16 5 -[2-(1H-benzirnidazol—1-yl)ethyl]—3-(pyridin—4- 3 74. 1 yl)thieno[2,3-d]pyridazin—4(5H)—one 3 17 5-[2-(1H—benzimidazol—2—y1)ethyl](pyridin 374. 1 yl)thieno[2,3-d]pyridazin—4(5H)—one hydrochloride 3 18 6-chloroquino lin—2-yl)ethyl](pyridin 419. 1 yl)thieno[2,3-d]pyridazin-4(5H)-one hydrochloride 3 19 3 -(pyridin—3 yr1yl)—5—[2—(quinolin—2— 409. 1 yl]thieno[2,3—d]pyridazin—4(5H)—one hydrochloride 320 3-(pyridin—4-ylethynyl)-5—[2—(quinolin—2— 409.1 225 EX. Name LC-MS: m/e (M+H)+ / Rt[rnin] yl)ethyl]thieno [2,3 -d]pyridazin—4(5H)—one hydrochloride 321 5 - [2-(3 ,5 -dirnethy1pyridin-2—y1)ethy1]-3—(pyridin—4— 3 63. 1 y1)thieno[2,3-d]pyridazin—4(5H)—one 322 5 - [2-(7-fluoroquino 1in-2—yl)ethyl]—3—(pyridin 403. 1 y1)thieno[2,3-d]pyridazin-4(5H)-one 323 5 - [2-(pyrazinyl)ethyl](pyridinyl)thieno[2 ,3 - 3 3 6. 1 dazin—4(5H)—one 324 2-[2-(1,6-naphthyridin—2—yl)ethyl]—8—(pyridin—4- 3 79. 1 yl)isoquino lin- 1 (2H)—one 325 2-[2-(8-fluoroquino lin-2—yl)ethy1]—8—(pyridin—4- 3 96. 1 yl)isoquino lin- 1 ne 326 8-(pyridin—4-yl)[ 1 -(quino1in—2—y1)propan—2- 3 92. 1 yl]isoquinolin—1(2H)-one 327 2-[2-(3-rnethy1quino1in—2-yl)ethyl](pyridin—4- 3 92.2 yl)isoquino lin- 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1](1H-pyrazol— 3 5 7. 1 328 3 -y1)phthalazin-1(2H)-one 8-(rnorpholin—4-y1) [2-(quinolinyl)ethy1]phthalazin- 3 87. 1 329 1(2H)-one hydrochloride 2-[2-(irnidazo[1,2-a]pyridin—2-yl)ethyl]—8—(1-oxa-4,9- 459.2 3 3 0 diazaspiro [5 .6] dodecyl)phthalazin- 1 (2H)-one dihydrochloride 2-[2-(imidazo[1,2-a]pyridin—2—yl)ethyl]—8—(2-oxa 33 1 azaspiro[3.5]non—7-yl)phthalazin—1(2H)-one (2E)—but ate 8- [(3 R)—3—hydroxypiperidin— 1 —yl]—2—[2—(imidazo [1 ,2- 3 90.2 332 a]pyridin—2-y1)ethy1]phthalazin—1(2H)—one ut enedioate (salt) 3 3 3 8- [(3 S)-3 -hydroxypiperidin—1—y1]—2—[2—(imidazo[1 ,2- 3 90.2 226 EX. Name LC-MS: m/e (M+H)+ / Rt[rnin] a]pyridiny1)ethy1]phthalazin- 1 (2H)—one (2E)-but enedioate (salt) 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl]—8-(2-oxa—6— 3 8 8.1 334 azaspiro [3 .3]hepty1)phthalazin— 1 ne 2-[2-(imidazo[1,2-a]pyridin—2-yl)ethyl](1,2- 362.1 3 3 5 oxazolidinyl)phthalazin-1(2H)-one 8-(hexahydrocyclopenta[b][1 ,4]oxazin-4(4aH)-y1)[2- 416.2 3 3 6 (imidazo [1 ,2—a]pyridin—2-yl)ethyl]phthalazin— 1 (2H)-one 2- [2-(imidazo [1 ,2—a]pyridin—2—yl)ethyl]—8— 3 61.2 3 3 7 (tetrahydrofilran-3 —y1)phthalazin— 1 (2H)—one 2- [2-(imidazo [1 ,2-a]pyridin—2—y1)ethy1]—8—(3—oxa 402.2 3 3 8 azabicyclo [3 .2. 1]octy1)phthalazin—1(2H)—one 2- [2-(irnidazo [1 ,2-a]pyridinyl)ethyl]—8—(2—oxa 402.2 3 3 9 azaspiro [3 .4]octy1)phthalazin- 1 (2H)—one 2- [2-(irnidazo [1 ,2-a]pyridinyl)ethy1]—8—(2,2,6,6- 448.1 340 tetrafluorornorpho1iny1)phthalazin— 1 (2H)-one 8-(4-hydroxypiperidiny1)—2—[2-(imidazo[1 ,2- 3 90. 1 341 a]pyridiny1)ethy1]phthalazin— 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1](2- 3 83 .2 342 methylpyrimidin-S-y1)phthalazin- 1 (2H)—one 8-(2-cyclopropy1pyrimidin—5-y1)—2—[2-(imidazo [1 ,2- 409. 1 343 a]pyridiny1)ethy1]phthalazin- 1 (2H)—one 2- [2-(imidazo [1 yridin—2—yl)ethyl]—8—(pyridazin 3 69. 1 344 halazin- 1 (2H)-one 8-(5 -fluoropyridin-3 -yl)[2-(imidazo[1,2-a]pyridin 3 86.2 345 yl)ethy1]phtha1azin- one hydrochloride 8-[2-(3—fluorophenyl)morpholin—4—yl]—2—[2— 470.2 346 (imidazo [1 ,2—a]pyridin—2—y1)ethyl]phthalazin— 1 (2H)-one hloride 347 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1]—8—(2— 3 99.1 227 EX. Name LC-MS: rn/e (M+H)+ / Rt[rnin] methoxypyrirnidin-S-yl)phthalazin- l (2H)—one 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethy1][2- 436.1 348 orornethyl)pyridinyl]phthalazin- 1 (2H)-one 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl](8-oxa-3 - 402.2 349 azabicyclo [3 .2. 1]octyl)phthalazin-1(2H)—one irnidazo[l ,2-a]pyridinyl)ethyl][2- 444.2 3 5 0 (trifluoromethyl)morpholinyl]phthalazin- l (2H)-one hydrochloride 8-(2,2-dimethylmorpholin—4—yl)—2—[2—(imidazo [1 ,2- 404.2 3 5 1 a]pyridinyl)ethyl]phthalazin— l (2H)—one hydrochloride 8- [2-(4-chlorophenyl)morpholin—4—yl]—2—[2— 486.2 352 (imidazo [1 ,2-a]pyridin—2-yl)ethyl]phthalazin— 1 (2H)-one hydrochloride 8- [2-(3 ,4-difluorophenyl)rnorpholin—4-yl]—2-[2- 488 .2 3 5 3 (imidazo [1 ,2-a]pyridin—2-yl)ethyl]phthalazin— 1 (2H)-one 2- nidazo [1 ,2-a]pyridin—2—yl)ethyl](piperidin—4- 3 74.2 354 yl)phthalazin— l (2H)-one hydrochloride 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl](tetrahydro- 375 .2 3 5 5 2H-pyran—4-yl)phthalazin—1(2H)—one 8-(2,6-diazabicyclo [3 .2. 1]octy1)—2-[2—(imidazo [1 ,2- 401.2 3 5 6 a]pyridinyl)ethyl]phthalazin—1(2H)—one dihydrochloride 8-[(l 8,5 S)—3 zabicyclo[3 .2.0]hept-3—yl]—2—[2- 3 87.2 3 5 7 (imidazo [l ,2-a]pyridinyl)ethyl]phthalazin- l (2H)-one hydrochloride 8-(furan—2—yl)—2— [2—(imidazo[l ,2—a]pyridin—2— 3 5 7.1 3 5 8 yl]phthalazin— l (2H)—one hydrochloride 2- [2-(imidazo [l ,2-a]pyridin—2—yl)ethyl]—8—( 1 —methyl- 3 71 .2 3 5 9 1H-pyrazolyl)phthalazin— 1 (2H)—one hydrochloride 228 EX. Narne LC-MS: rn/e (M+H)+ / Rt[rnin] 8-(hexahydropyrrolo[3 ,4—c]pyrrol—2(1H)—yl)[2- 401.2 360 azo [1 ,2-a]pyridin—2-yl)ethy1]phthalazin— 1 (2H)-one dihydrochloride 8-(2,7-diazaspiro[4.4]nony1)—2—[2-(imidazo[1 ,2- 415.2 361 a]pyridinyl)ethyl]phthalazin—1(2H)—one dihydrochloride 8-[(lS,4S)-2,5-diazabicyclo[2.2.1]heptyl][2- 387.2 362 (imidazo [l ,2—a]pyridin—2-yl)ethyl]phthalazin— 1 ne roacetate 8-(2,7-diazaspiro [3 .5 ]nor1—7—yl)—2—[2—(imidazo[1 ,2- 415.2 363 a]pyridinyl)ethyl]phthalazin— 1 (2H)—one hydrochloride 8-(2,6-diazaspiro [3 .5 ]nonyl)—2—[2—(imidazo [1 ,2- 415.2 364 a]pyridinyl)ethyl]phthalazin- 1 (2H)—one hydrochloride 8-(piperidin—4-yl)[2-(5 ,6,7,8-tetrahydroirnidazo[1 ,2- 378.2 365 a]pyridinyl)ethyl]phthalazin— 1 (2H)-one hydrochloride 8-[2-(aminomethyl)chloropyrrolidin—1-yl][2- 423.2 366 (irnidazo [1 ,2-a]pyridin—2—yl)ethy1]phthalazin— 1 (2H)-one trifluoroacetate 2- [2-(irnidazo [1 yridin—2-yl)ethyl]-4,8—di(pyridin— 445.2 367 4-yl)phthalazin— 1 (2H)-one 2-[2-(imidazo[1 ,2-a]pyridin—2—yl)ethyl]—8— 427.2 368 [(3 aR,4S,7R,7aS)-octahydro-1H-4,7-epiminoisoindol—8- yl]phthalazin-1(2H)-one dihydrochloride 8- [5 —(4—chlorophenyl)—2,5—diazabicyclo[2 .2. 1]hept 497.2 369 yl] [2-(imidazo [1 yridin—2—yl)ethyl]phthalazin- 1 (2H)-one 370 4-brorno[2-(imidazo[1 ,2—a]pyridin—2—yl)ethyl] 448 229 EX. Name LC-MS: rn/e (M+H)+ / Rt[rnin] (pyridiny1)phtha1azin— 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2-yl)ethyl][(3aS, 8aS)- 429.2 371 octahydropyrrolo[3,4-c]azepin—2(1H)-yl]phtha1azin— 1(2H)-one dihydrochloride 2-[2-(imidazo[1,2-a]pyridin—2-yl)ethyl][(3aS,8aR)— 429.2 372 octahydropyrrolo[3 ,4-c]azepin—2( 1 H)-yl]phtha1azin- 1(2H)-one dihydrochloride tert—butyl (3 7R,7aS) {3-[2-(imidazo[1 ,2- 527.3 373 a]pyridin-2—y1)ethy1]—4—oxo—3 ,4—dihydrophthalazin—5 - y1} octahydro-2H-4,7—epiminoisoindole—2—carboxylate 8-(hexahydro-5H-furo[2,3—c]pyrrol—5—yl)—2—[2— 402.2 374 (imidazo [1 ,2-a]pyridin—2-yl)ethy1]phthalazin— 1 (2H)-one hydrochloride irnidazo[1 ,2-a]pyridinyl)ethyl]—8—(1,2,3 ,6- 372.2 375 tetrahydropyridiny1)phthalazin- 1 (2H)—one hydrochloride 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1][(3 S)- 376 tetrahydrofiJran-3 -y1arnino]phthalazin— 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1][(3 R)- 377 tetrahydrofilran-3 ino]phthalazin- 1 (2H)-one 8- {[5 -(hydroxyrnethy1)—1,4—dioxan—2-y1]methoxy}[2- 437.1 378 (irnidazo [1 ,2-a]pyridin—2-yl)ethyl]phthalazin-1(2H)—one 2- [2-(imidazo [1 ,2-a]pyridin—2-yl)ethyl](oxetan—3 - 363.1 379 yloxy)phthalazin- 1 (2H)—one imidazo[1 yridinyl)ethyl](pyridin 398.1 380 ylmethoxy)phthalazin— 1 (2H)-one 2-[2-(imidazo[1 ,2—a]pyridin—2—yl)ethyl]—8—(morpholin-4 390.2 381 ylmethy1)phtha1azin— 1 (2H)—one 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—8—(pyridir1—3 - 384.1 382 yloxy)phtha1azin— 1 (2H)-one 230 EX. Narne LC-MS: rn/e (M+H)+ / Rt[rnin] 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1][(oxetan—3 - 3 76.2 3 83 ylrnethyl)amino]phthalazin— 1 (2H)—one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl](tetrahydro- 3 90.2 3 84 2H—pyran—4-ylamino)phthalazin—1(2H)—one 2-[2-(imidazo[l ,2-a]pyridin—2—yl)ethyl]—8—[( 1 - 375 .2 3 85 methylazetidinyl)amino]phthalazin-1(2H)-one 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1,3-oxazol 3 72.3 3 86 no)phthalazin— 1 (2H)—one 2-[2-(imidazo[1,2—a]pyridin—2—yl)ethyl]—8— 376.2 3 87 [methyl(oxetan—3-yl)amino]phthalazin— 1 (2H)—one 2- [2-(imidazo [1 ,2-a]pyridin—2—yl)ethyl]—8—(piperidin—4- 3 89.2 3 8 8 no)phthalazin— 1 (2H)-one dihydrochloride 8-[(1-acetylpiperidin—3 -yl)amino]—2—[2—(imidazo[ 1 ,2- 43 1 .2 3 89 a]pyridinyl)ethyl]phthalazin- 1 (2H)—one hydrochloride acetylpiperidin—4-yl)arnino][2-(irnidazo[l ,2- 43 1 .2 390 a]pyridinyl)ethyl]phthalazin— 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethyl] 3 76.2 3 91 hydrofuran—3 -ylarnino)phthalazin— 1 (2H)-one 2- [2-(irnidazo [1 ,2-a]pyridin—2—yl)ethy1](tetrahydro- 3 90.2 392 2H-pyran—3-ylarnino)phthalazin— 1 (2H)—one hydrochloride 2- idazo [l ,2-a]pyridin—2-yl)ethyl](piperidin—3 - 3 89.2 393 ylamino)phthalazin— 1 (2H)—one dihydrochloride 394 2- [2-(imidazo [1 ,2-a]pyridinyl)ethyl] {methyl [(3 - 404.2 methyloxetanyl)methyl]amino}phthalazin-1(2H)-one 395 2- [2-(imidazo [1 ,2—a]pyridin—2—yl)ethyl]—8—(oxetan-3 - 3 62.2 ylamino)phthalazin— 1 (2H)—one 396 8- {[(3 aS,4S,6aS)-octahydrocyclopenta[c]pyrrol 433 .3 ylrnethyl]amino}[2-(5,6,7,8—tetrahydroimidazo[1,2- 231 EX. Name LC-MS: m/e (M+H)+ / Rt[rnin] diny1)ethy1]phthalazin- 1 (2H)—one hydrochloride 397 5 -(rnorpholin—4-y1) [2-(quinolinyl)ethy1]phthalazin- 3 87.2 1(2H)-one hydrochloride 398 2-[2-(1H—benzimidazo1—2—yl)ethyl](pyridin 3 68. 1 y1)phthalazin-1(2H)-one 399 4-(pyridin-3 -y1) [2-(quino [inyl)ethyl]phtha1azin- 3 79. 1 1 (2H)-one 400 5 -(1,4-dihydropyrimidin—5—y1)—2—[2—(5 ,6,7,8— 375 .2 tetrahydroimidazo [1 ,2—a]pyridin—2—yl)ethyl]phthalazin- 1(2H)-one dihydrochloride 401 2- [2-(irnidazo [1 ,2-a]pyridin-2—y1)ethy1]—5—(pyridin—4- 368 . 1 y1)phtha1azin— 1 (2H)-one 402 5 -(pyridin—3 -y1) [2-(quinoliny1)ethy1]phthalazin- 3 79.2 1 (2H)-one 403 2-[2-(irnidazo[1,2-a]pyridin—2-yl)ethy1]{[(3 - 3 90.2 methyloxetan—3-y1)rnethy1]amino}phthalazin— 1 (2H)-one 404 4-(pyridin—4-y1) [2-(quino1in—2—y1)ethyl]pyrido [2,3 - 3 80. 1 d]pyridazin—S(6H)-one 405 4-(rnorpho 1in—4-y1) [2—(quinolin 3 8 8 .2 y1)ethy1]pyrido[2,3-d]pyridazin—5(6H)—one 406 4-(oxetan—3-y1arnino)-6—[2-(quino1in 3 74.1 y1)ethy1]pyrido[2,3-d]pyridazin-5(6H)—one 407 2- methoxypyridin—2-yl)ethyl](pyridin 3 5 8. 1 quino lin- 1 (2H)-one 408 2-[2-(1,3 thiazo lyl)ethyl](pyridin 3 84.1 quino 1111— 1 (2H)—one 409 2- [2-(5-methy1pyridin—2—y1)ethyl]—8—(pyridin—4— 342. 1 y1)isoquino lin- 1 (2H)—one 232 11.22 Preparation of compounds ofthe formula I in which A is A3 The compounds of e 410 to 415 were prepared in analogy to the s described above.
EX. Name LC-MS: m/e (M+H)+ 410 5 - [(E)(6-methoxyquino linyl)ethenyl] 412.8 (pyridinyl)thieno[3 ,2-c]pyridin-4(5H)-one 41 1 8-(pyridinyl) [(E)(quinazo lin 3 77. 1 yl)ethenyl]isoquino lin- 1 (2H)—one 412 5 - [(E)-2—(6—chloroquino1in—2—yl)ethenyl]—3—(pyridin- 416. l 4-yl)thieno [3 ,2—c]pyridin—4(5H)—one 413 5 - [(E)(3 -methquuino1in—2—y1)ethenyl]—3— 3 96. l (pyridinyl)thieno [3 ,2-c]pyridin—4(5H)—one 414 8-(pyridin—4-yl) [(E)(quino1in—2— 3 76. l yl)ethenyl]isoquino lin- 1 (2H)-one 415 5 - [(E)(l ,3-benzothiazoly1)etheny1](pyridin— 3 8 8 4-yl)thieno [3 ,2-c]pyridin-4(5H)—one 416 3 -(pyridinyl)-5 - [(E)-2—(quino1in 3 82. l yl)ethenyl]thieno[3,2-c]pyridin—4(5H)—one ical Tests a) Measurement ofPDE activity The inant PDE proteins are used in in vitro enzymatic reaction for ement of PDE activity. These recombinant proteins, ing PDElOA (human, rat and mouse PDElO) and isoforms of PDEs 1, 3, 4, and 5, were purchased from commercial vendor BPS Bioscience. The enzymatic activity ofPDEs was determined by cAMP measurement kit from CisBio (IBA) using HTRF technology.
The PDE enzymatic reaction was carried out in assay buffer (20mM Tris-HCl pH7.5, lOmM MgClz, 0.1% bovine serum albumin) containing enzyme and substrate.
The PDE enzymes concentration ranged from 10pM — 250pM, depending on each 233 enzyme’s specific activity. The substrate cyclic nucleotide (cAMP or cGMP) concentration used in the assay was 20nM for PDElO, and 100nM for other PDEs. The inhibitory effect of compound was determined by incubating various concentration of inhibitor in the enzymatic assay. Typically, compound was serial d in DMSO then further diluted in assay buffer. Next, the compound at varying tration was mixed with PDE enzyme. The reaction was ted by addition of cyclic nucleotide substrate, and incubated for 60 minutes at 29C. The reaction was stopped by addition of lysis buffer from assay kit. The cAMP-d2 and anti-CAMP cryptate in the lysis buffer detected the level of CAMP left from the PDE hydrolysis reaction. The PDE activity is reversely 10 correlated with the amount ofCAMP left in the reaction and can be converted to the percent activity of an uninhibited control (100%). Thus, IC50 value of inhibitor can be obtained by plotting inhibitor tration t PDE activity at that concentration.

Claims (80)

We claim:
1. Compound of formula I Q R1 Het A (I) N R2 X1 X2 X3 n Q is O or S; X1 is N or CH; X2 is N or C-R7; X3 is O, S, -X4=C(R8)-, where C(R8) is bound to the carbon atom which carries R2, or -X5=C(R9)-, where X5 is bound to the carbon atom which carries R2; X4 is N or C-R9; X5 is N; Het is selected from i. monocyclic C-bound 6-membered hetaryl having 1 or 2 nitrogen atoms as ring members, which is tituted or may carry 1, 2, 3 or 4 identical or different substituents Rx, ii. fused bicyclic hetaryl having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, benzothienyl or benzofuryl, where bicyclic hetaryl, benzothienyl and benzofuryl are, independently of each other, unsubstituted or may carry 1, 2, 3 or 4 identical or different substituents Rx, and iii. phenyl, which carries a monocyclic l radical having 1 or 2 nitrogen atoms and ally a further heteroatom selected from O, S and N as ring s, which in addition to monocyclic hetaryl, may carry 1, 2 or 3 identical or different substituents Rx, where (11348828_1):GGG 237 Rx is selected from the group ting of H, halogen, C1-C4-alkyl, C1-C4-alkoxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C3-C6- cycloalkyl, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkoxy-C1-C4-alkyl, OH, y-C1-C4-alkyl, O-C3-C6-cycloalkyl, benzyloxy, C(O)O- (C1-C4-alkyl), O-(C1-C4-alkyl)-CO2H, N(Rx1)(Rx2), C(O)N(Rx1)(Rx2), C1-C4-alkyl-N(Rx1)(Rx2), -NRx3-C(O)-N(Rx1)(Rx2), NRx3-C(O)O-(C1-C4-alkyl), -N(Rx3)-SO2-Rx4, phenyl, CN, -SF5, -OSF5, -SO2Rx4, -SRx4 and trimethylsilyl, where Rx1, Rx2, Rx3 and Rx4, independently of each other are ed from the group consisting of hydrogen, C1-C4- alkyl, C1-C4-fluoroalkyl and C3-C6-cycloalkyl or Rx1 and Rx2 form together with the N atom to which they are attached a 3- to 7- membered, nitrogen heterocycle which may have 1, 2 or 3 further ent or identical heteroatoms or heteroatom ning groups selected from the group of O, N, S, SO and SO2 as ring members and which may carry 1, 2, 3, 4, 5 or 6 substituents selected from C1-C4-alkyl; or two radicals Rx which are bound at adjacent carbon atoms may form a fused 5- or 6-membered saturated carbocyclic radical or a fused 5- or 6-membered heterocyclic radical having 1, 2 or 3 heteroatoms as ring s, which are selected from O, S and N; R1 is a moiety Y1-Cyc1; R2 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl, CN and NRx1Rx2; A represents one of the following groups A1, A2, A3, A4 or A5: (11348828_1):GGG 238 R3e R3f R6 R5 R4 A' * * * * * * * * * * R4 R3 R5 R5 R4 R4 R5 (A1 ) (A2 ) (A3 ) (A4 ) (A5 ) where * indicates the points of attachment to Het and to the nitrogen atom, respectively, R3, R4, R5, R6 independently of each other are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl, hylsilyl, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, C3-C6-cycloalkyl, or the radicals together with the carbon atoms to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6-membered heterocycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents selected from ne and methyl or either the radicals R3, R4 or the radicals R5, R6 together with the carbon atom to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6- membered heterocycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents selected from fluorine and ; A' is a O, NR3a, CR3bR3c or linear C2-C3-alkandiyl, where one of the CH2- moieties of C2-C3-alkandiyl may be replaced by oxygen or NR3a, and where 1, 2, 3, or 4 of the hydrogen atoms of C2-C3-alkandiyl may be ed by a radical R3d, where R3a is hydrogen or C1-C4-alkyl, R3b, R3c independently of each other are selected from the group consisting of hydrogen, halogen, alkyl or R3b and R3c together form alkandiyl; R3d is selected from the group consisting of halogen and C1-C4- alkyl; (11348828_1):GGG 239 R3e, R3f independently of each other are selected from the group consisting of hydrogen and C1-C4-alkyl; R7 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1- C4-alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkylsulfanyl-C1-C4- alkoxy, C2-C4-alkenyloxy, fluoroalkyl, C1-C4-fluoroalkoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRx1Rx2, NRx1Rx2-C1-C4-alkoxy and the moiety Y2-Cyc2; R8 is ed from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, nated ropyl, CN and NRx1Rx2; R9 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1- C4-alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkylsulfanyl-C1-C4- alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl, CN, NRx1Rx2, NRx1Rx2-C1-C4-alkoxy and the moiety Y3-Cyc3; Y1, Y2, Y3 independently of each other are selected from a chemical bond, CH2, O, O-CH2, NRy, NRy-CH2, O)2, S, S(O), S(O)2, 1,2-ethandiyl, 1,2- ethendiyl or 1,2-ethyndiyl, where Ry is selected from the group consisting of hydrogen, C1-C4-alkyl, C1-C4-alkylcarbonyl, alkylsulfonyl, C1- C4-fluoroalkylsulfonyl; Cyc1, Cyc2, Cyc3 independently of each other are selected from the group consisting of phenyl, yl, 4- to 8-membered saturated or partially unsaturated heteromonocyclic ls, saturated or partially unsaturated 7- to 10 (11348828_1):GGG 240 membered bicyclic radicals, 5- or 6-membered monocyclic hetaryl, and 8- to 10 membered bicyclic hetaryl, where the saturated or partially unsaturated heteromonocyclic and heterobicyclic radicals have 1, 2, 3 or 4 heteroatoms or heteroatom containing groups as ring members, which are selected from O, S, SO, SO2 and N, and where the 5- or 6-membered monocyclic hetaryl and the 8- to 10-membered bicyclic hetaryl have 1, 2, 3 or 4 heteroatoms as ring members, which are selected from O, S and N, where phenyl, naphthyl, the saturated or partially unsaturated heteromonocyclic and heterobicyclic radicals and the mono- and bicyclic heteroaromatic radicals are unsubstituted or carry 1, 2, 3, 4 or 5 radicals RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4 radicals RC1; where RC1 is selected from hydrogen, halogen, OH, CN, NO2, C1-C4-alkyl, C1- C4-alkoxy, alkylsulfanyl, hydroxy-C1-C4-alkyl, C1-C4- -C1-C4-alkyl, C1-C4-alkoxy-C1-C4-alkoxy, cyano-C1-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C1-C4-alkylsulfonyl, C(O)Ra, Z-C(O)ORb, Z-C(O)NRcRd, S(O)2NRcRd and f, where Ra is selected from the group consisting of C1-C4-alkyl and C1- C4-fluoroalkyl, Rb is selected from the group consisting of hydrogen, C1-C4- alkyl, C2-C4-alkenyl and fluoroalkyl, Rc, Rd is selected from the group consisting of hydrogen, C1-C4- alkyl, C1-C4-fluoroalkyl, C1-C4-alkoxy and C1-C4- fluoroalkoxy, Re, Rf is selected from the group consisting of en, C1-C4- alkyl, fluoroalkyl, C1-C4-alkoxy and C1-C4- fluoroalkoxy, Z is a covalent bond or alkandiyl, or two radicals RC1 which are bound at adjacent carbon atoms may form a fused 5- or 6-membered carbocyclic radical or a fused 5- or 6-membered heterocyclic radical having 1, 2 or 3 atoms as ring members, which are selected from O, S and N; (11348828_1):GGG 241 or two radicals RC1 which are bound at the same carbon atom may form a spiro 5- or 6-membered carbocyclic radical or a spiro 5- or 6-membered heterocyclic radical having 1 or 2 heteroatoms as ring members, which are selected from O, S and N, or two radicals RC1 which are bound at the same carbon atom may form an oxygen atom, where the fused and the spiro radicals are unsubstituted or carry 1, 2, 3 or 4 radicals RC3; Y' is a chemical bond, CH2, O, O-CH2, S(O)2, NRy', NRy'-CH2 or NRy'-S(O)2, where Ry' is selected from the group consisting of hydrogen, C1-C4-alkyl, C1-C4-alkylcarbonyl, C1-C4-alkylsulfonyl, C1-C4-fluoroalkylsulfonyl; RC2 is a carbocyclic or cyclic l selected from the group ting of phenyl, 3- to 7-membered saturated or partially unsaturated monocarbocyclic radicals, 3- to 7-membered saturated or partially unsaturated heteromonocyclic radicals, having 1, 2 or 3 heteroatoms as ring members, which are selected from O, S and N, and 5- or 6-membered aromatic radicals, having 1, 2 or 3 heteroatoms as ring s, which are selected from O, S and N, where the carbocyclic and the heterocyclic radical is unsubstituted or carries 1, 2, 3, 4 or 5 radicals RC3; RC3 is selected from hydrogen, halogen, OH, CN, C1-C4-alkyl, C1-C4- alkoxy, hydroxy-C1-C4-alkyl, C1-C4-alkoxy-C1-C4-alkyl, cyano-C1- C4-alkyl, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C2-C6-alkenyl, , benzyl, Z-C(O)ORb, Z-C(O)NRcRd, S(O)2NRcRd and ZNReRf , where, Z, Ra, Rb, Rc, Rd, Re and Rf are as d above or two ls RC3 which are bound at the same atom may form an oxygen atom; provided that for X3 being X4=C(R8), one or two of the radicals R1, R7 and R9 are a moiety Y1-Cyc1, Y2-Cyc2 or Y3-Cyc3, respectively; (11348828_1):GGG 242 further provided that for X3 being X5=C(R9), one or two of the radicals R1, R7 and R9 are a moiety 1, Y2-Cyc2 or Y3-Cyc3, respectively; and the N-oxides, the prodrugs, the tautomers and the hydrates f, and the pharmaceutically acceptable salts thereof; wherein prodrugs are derivatives of the compounds of formula I carrying an OH or NH2-group, where one of the hydrogen atoms of the OH or NH2-group is substituted by a C1-C4-alkylcarbonyl group, by benzoyl, or by an acyl group derived from an amino acid, which is linked to the oxygen or nitrogen of the OH or NH2-group via the carbonyl group of the amino acid; or wherein prodrugs are alkylcarbonyloxyalkyl ates or carbamates of compounds I carrying an OH- or oup in which one of the hydrogen atoms of the OH- or NH2-group has been replaced by a group of the formula -C(=O)-O-CHRp-O-C(=O)-Rq in which Rp and Rq are independently of one another C1-C4-alkyl; except for the following compounds: 6-Methylphenyl[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, yl[2-(4-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(4-Methylphenyl)[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(2-Furanyl)[1-methyl(3-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(2-Thienyl)[1-methyl(3-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(3,4-Dimethylphenyl)[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-(4-Methylphenyl)[2-(4-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, 5-Phenyl[2-(2-pyridinyl)ethyl]-thieno[2,3-d]pyrimidin(3H)-one, and 5-Phenyl[2-(4-oxophenylthieno[2,3-d]pyrimidin-(3H)yl)ethyl]-thieno[2,3- d]pyrimidin(3H)-one.
2. The compound as claimed in claim 1, wherein X3 is O, S or R8)-, where C(R8) is bound to the carbon atom which carries R2; X4 is N or C-R9; (11348828_1):GGG 243 Het is ed from i. monocyclic d 6-membered hetaryl having 1 or 2 nitrogen atoms as ring members, which is unsubstituted or may carry 1, 2, 3 or 4 identical or different substituents Rx, ii. fused bicyclic hetaryl having 1 or 2 nitrogen atoms and optionally a further heteroatom selected from O, S and N as ring members, benzothienyl or benzofuryl, where bicyclic hetaryl, benzothienyl and benzofuryl are, independently of each other, unsubstituted or may carry 1, 2, 3 or 4 identical or different substituents Rx, and iii. phenyl, which carries a monocyclic hetaryl radical having 1 or 2 nitrogen atoms and optionally a r heteroatom selected from O, S and N as ring members, which in on to clic hetaryl, may carry 1, 2 or 3 identical or different tuents Rx, where Rx is selected from the group consisting of H, halogen, C1-C4-alkyl, C1-C4-alkoxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, C3-C6- cycloalkyl, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkoxy-C1-C4-alkyl, OH, hydroxy-C1-C4-alkyl, O-C3-C6-cycloalkyl, benzyloxy, C(O)O- (C1-C4-alkyl), O-(C1-C4-alkyl)-CO2H, N(Rx1)(Rx2), C(O)N(Rx1)(Rx2), C1-C4-alkyl-N(Rx1)(Rx2), -NRx3-C(O)-N(Rx1)(Rx2), NRx3-C(O)O-(C1-C4-alkyl), -N(Rx3)-SO2-Rx4, phenyl, CN, -SF5, -OSF5, -SO2Rx4, -SRx4 and trimethylsilyl, where Rx1, Rx2, Rx3 and Rx4, ndently of each other are selected from the group consisting of hydrogen, C1-C4- alkyl, C1-C4-fluoroalkyl and C3-C6-cycloalkyl or Rx1 and Rx2 form together with the N atom to which they are attached a 3- to 7- membered, nitrogen heterocycle which may have 1, 2 or 3 r different or identical heteroatoms or heteroatom ning groups selected from the group of O, N, S, SO and SO2 as ring members and which may carry 1, 2, 3, 4, 5 or 6 substituents selected from C1-C4-alkyl; (11348828_1):GGG 244 A represents one of the following groups A1, A2, A3 or A4: R6 R5 R4 A' * * * * * * * * R4 R3 R5 R5 R4 (A1 ) (A2) (A3) (A4) where * indicates the points of attachment to Het and to the nitrogen atom, respectively, R3, R4, R5, R6 independently of each other are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl, trimethylsilyl, fluoroalkyl, C1-C4- alkoxy, C3-C6-cycloalkyl, or the radicals together with the carbon atoms to which they are bound form a saturated 3- to ered carbocycle or a ted 3- to 6-membered heterocycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are tituted or may carry 1, 2, 3 or 4 substituents selected from fluorine and methyl or either the radicals R3, R4 or the radicals R5, R6 together with the carbon atom to which they are bound form a saturated 3- to 6-membered carbocycle or a saturated 3- to 6- membered cycle having 1 or 2 non-adjacent heteroatoms as ring members, where the carbocycle and the heterocycle are unsubstituted or may carry 1, 2, 3 or 4 substituents selected from fluorine and methyl; A' is a O, NR3a, CR3bR3c or linear C2-C3-alkandiyl, where one of the CH2- moieties of C2-C3-alkandiyl may be replaced by oxygen or NR3a, and where 1, 2, 3, or 4 of the hydrogen atoms of C2-C3-alkandiyl may be replaced by a radical R3d, where R3a is hydrogen or C1-C4-alkyl, R3b, R3c ndently of each other are selected from the group consisting of hydrogen, halogen, C1-C4-alkyl or R3b and R3c together form C2-C3-alkandiyl; (11348828_1):GGG 245 R3d is selected from the group consisting of halogen and C1-C4- alkyl; R8 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, alkoxy-C1-C4-alkyl, C1-C4-alkoxy, C1-C4- alkoxy-C1-C4-alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4- fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated ropyl, CN and NRx1Rx2; R9 is selected from the group consisting of hydrogen, halogen, OH, C1-C4- alkyl, trimethylsilyl, C1-C4-alkylsulfanyl, C1-C4-alkoxy-C1-C4-alkyl, C1- C4-alkoxy, C1-C4-alkoxy-C1-C4-alkoxy, C1-C4-alkylsulfanyl-C1-C4- alkoxy, C2-C4-alkenyloxy, C1-C4-fluoroalkyl, C1-C4-fluoroalkoxy, cyclopropyl, ally substituted by 1, 2 or 3 methyl groups, fluorinated ropyl, CN, NRx1Rx2, NRx1Rx2-C1-C4-alkoxy and the moiety Y3-Cyc3.
3. The compound as claimed in claim 1 or 2, where X3 is selected from O, S, -X4=C(R8)-, where C(R8) is bound to the carbon atom which carries R2.
4. The compound as claimed in any one of claims 1 to 3, where A is selected from A1, A2, A3 and A4.
5. The compound as claimed in any of claims 1 to 4, where X2 is C-R7.
6. The compound as claimed in claim 5, where R7 is hydrogen or Y2-Cyc2.
7. The compound as d in any of claims 1 to 6, where X1 is N.
8. The compound as d in claim 1 to 6, where X1 is CH.
9. The compound as claimed in any of claims 1 to 8, where X3 is S. (11348828_1):GGG 246
10. The compound as claimed in any of claims 1 to 8, where X3 is O.
11. The compound as claimed in any one of claims 1 to 8, where X3 is C(R9)=C(R8).
12. The compound as claimed in any one of claims 1 to 8, where X3 is N=(CR9).
13. The compound as claimed in any of claims 11 or 12, where R9 is hydrogen or Y3- Cyc3.
14. The compound as claimed in any of claims 1 to 8, where X3 is N=C(R8).
15. The nd as d in any of claims 11 or 14, where R8 is hydrogen.
16. The compound as claimed in any one of the preceding claims, where R2 is ed from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2-fluoroalkyl, C1- C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, ally substituted by 1, 2 or 3 methyl groups, and fluorinated cyclopropyl.
17. The compound as claimed in claim 16, where R2 is hydrogen.
18. The compound as claimed in any one of the preceding claims, where A is A1.
19. The compound as claimed in claim 18, where R3, R4 are selected from hydrogen and fluorine.
20. The compound as claimed in claim 18 or 19, where R5 and R6 are, independently of each other, selected from the group ting of hydrogen, fluorine and methyl.
21. The compound as claimed in any one of claims 1 to 17, where A is A2.
22. The nd as claimed in any one of claims 1 to 17, where A is A3. (11348828_1):GGG 247
23. The compound as d in claim 22, where R4, R5 are selected from hydrogen and fluorine.
24. The compound as claimed in any one of claims 1 to 17, where A is A4.
25. The compound as claimed in claim 24, where A' is CR3bR3c, where R3b and R3c are independently of each other selected from the group consisting of hydrogen, fluorine and methyl or together form CH2CH2.
26. The compound as d in claim 25, where R3b and R3c are hydrogen.
27. The compound as claimed in any one of claims 1 to 17, where A is A5.
28. The compound as claimed in claim 27, where R3e, R3f, R4 and R5 are each hydrogen.
29. The nd as claimed in any one of the preceding claims, where Het is selected from the group consisting of C-bound 6-membered monocyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members, benzofuryl and C-bound, fused bicyclic hetaryl, which has 1 or 2 nitrogen atoms as ring members and optionally a further heteroatom selected from O, S and N as ring member; where monocyclic hetaryl, uryl and bicyclic hetaryl may be tituted or may carry 1, 2, 3 or 4 substituents Rx.
30. The compound as claimed in claim 29, where Het has at least one imino-nitrogen as ring member, which is located in the position adjacent to the carbon atom which is bound to A.
31. The compound as claimed in claim 30, where Het is selected from the group consisting of 2-pyridyl, 2-pyrimidinyl, midinyl, zinyl, 3-pyridazinyl, 2- quinolinyl, uinolinyl, 2-quinazolinyl, 2-quinoxalinyl, 1,5-naphthyridinyl, 1,8-naphthyridinyl, benzothiazolyl, benzoxazolyl, benzimidazolyl, 1- methylbenzimidazolyl, imidazo[1,2-a]pyridineyl, thieno[3,2-b]pyridineyl, imidazo-[2,1-b]-thiazolyl and 1,2,4-triazolo[1,5-a]pyridineyl, where the aforementioned radicals may carry 1, 2 or 3 radicals ed from fluorine, chlorine, (11348828_1):GGG 248 methyl, fluoromethyl, difluoromethyl, trifluoromethyl, y, fluoromethoxy, difluoromethoxy, trifluoromethoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, and fluorinated cyclopropyl.
32. The compound as claimed in any one of the ing , where Y1, Y2 and Y3, ndently of each other, are selected from the group consisting of a chemical bond, O and NH.
33. The compound as claimed in any one of the preceding claims, where Y1, Y2 and Y3 are each a chemical bond.
34. The compound as claimed in any one of the preceding claims, where Q is O.
35. The compound as claimed in any one of the preceding claims, which is of the formulae I-1.A or I-2.A O R1 Het N (I-1.A) R2 R5 R6 N S R7 O R1 Het N (I-2.A) R2 R5 R6 S R7 where Het, R1, R2, R5, R6 and R7 are as defined in any one of the preceding claims.
36. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 1.B or I-2.B (11348828_1):GGG 249 R3c R3b O R1 Het N (I-1.B) R5 R2 N S R7 R3c R3b O R1 Het N (I-2.B) R5 R2 S R7 where Het, R1, R2, R3b, R3c and R7 are as defined in any one of claims 1 to 35 and R5 is hydrogen.
37. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 1.C or I-2.C R5 O R1 Het N (I-1.C) R2 N S R7 R5 O R1 Het N ) R2 S R7 where Het, R1, R2, R5, and R7 are as defined in any one of claims 1 to 35.
38. The nd as claimed in any of claim 35, 36 or 37, where R1 is a radical Y1-Cyc1 and R7 is selected from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1- (11348828_1):GGG 250 C2-fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, ally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2.
39. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 3.A or I-4.A O R1 Het R2 N (I-3.A) R5 R6 N R8 R7 R9 O R1 Het R2 N (I-4.A) R5 R6 R8 R7 R9 where Het, R1, R2, R5, R6, R7, R8 and R9 are as defined in any one of claims 1 to 35.
40. The nd as claimed in any one of claims 1 to 34, which is of the formulae I- 3.B or I-4.B R3c R3b O R1 Het R2 N (I-3.B) R5 N R8 R7 R9 (11405124_1):JIN 251 R3c R3b O R1 Het R2 N (I-4.B) R5 R8 R7 R9 where Het, R1, R2, R3b, R3c, R7, R8 and R9 are as defined in any one of claims 1 to 35 and R5 is hydrogen..
41. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 3.C or I-4.C R5 O R1 Het R2 N (I-3.C) N R8 R7 R9 R5 O R1 Het R2 N (I-4.C) R8 R7 R9 where Het, R1, R2, R5, R7, R8 and R9 are as defined in any one of claims 1 to 35.
42. The compound as claimed in any one of claims 39, 40 or 41, where R1 is a radical Y1-Cyc1 and R7 and R9 are selected, independently of each other, from the group consisting of hydrogen, fluorine, C1-C4-alkyl, fluoroalkyl, alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally tuted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2 or Y3-Cyc3, respectively.
43. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 5.A or I-6.A (11405124_1):JIN 252 O R1 Het R2 N (I-5.A) R5 R6 N N R8 R7 O R1 Het R2 N (I-6.A) R5 R6 N R8 R7 where Het, R1, R2, R5, R6, R7 and R8 are as defined in any one of claims 1 to 34.
44. The compound as claimed in any one of claims 1 to 34, which is of the ae I- 5.B or I-6.B R3c R3b O R1 R2 Het N (I-5.B) R5 N N R8 R7 R3c R3b O R1 R2 Het N (I-6.B) R5 N R8 R7 where Het, R1, R2, R3b, R3c, R7 and R8 are as defined in any one of claims 1 to 34 and R5 is hydrogen.
45. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 5.C or I-6.C (11405124_1):JIN 253 R5 O R1 R2 Het N (I-5.C) N N R8 R7 R5 O R1 R2 Het N (I-6.C) N R8 R7 where Het, R1, R2, R5, R7 and R8 are as defined in any one of claims 1 to 34.
46. The compound as claimed in any one of claims 43, 44 or 45, where R1 is a radical Y1-Cyc1 and R7 is ed from the group consisting of hydrogen, ne, C1-C4-alkyl, C1-C2-fluoroalkyl, alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2.
47. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 7.A, I-8.A, I-7.B, I-8.B, I-7.C, I-8.C, O R1 O R1 Het Het N N R2 R2 R5 R6 N R5 R6 O O R7 R7 (I-7.A) (I-8.A) R3c R3b O R1 R3c R3b O R1 Het N R5 R2 Het N N O R5 R2 O R7 R7 (I-7.B) (I-8.B) (11405124_1):JIN 254 R5 O R1 R5 O R1 Het N Het N R2 R2 N O O R7 R7 ) (I-8.C) where Het, R1, R2, R3b, R3c, R5, R6, R7 and R8 are as defined in any one of claims 1 to 35.
48. The compound as claimed in claim 47, where R1 is a radical Y1-Cyc1 and R7 is selected from the group consisting of hydrogen, fluorine, C1-C4-alkyl, fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, ropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2.
49. The compound as claimed in any one of claims 1 to 34, which is of formulae O R1 O R1 R3c R3b O R1 Het R2 Het R2 N N Het R2 R5 R6 R5 R6 N N N N R5 N N R7 R9 R7 R9 R7 R9 (I-10.A) (I-9.A) (I-9.B) R3c R3b O R1 R5 O R1 R2 R5 O R1 Het N Het R2 R5 N Het R2 N N N N N R7 R9 R7 R9 R7 R9 (I-10.B) (I-9.C) (I-10.C) wher e Het, R1, R2, R3b, R3c, R5, R6, R7, and R9 are as defined in any one of claims 1 to 34.
50. The compound as claimed in claim 49, where R1 is a radical Y1-Cyc1 and R7 and R9 are selected, independently of each other, from the group ting of hydrogen, fluorine, alkyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2 or Y3-Cyc3, (11405124_1):JIN 255 respectively.
51. The compound as claimed in any one of claims 1 to 34, which is of the formulae I- 1.D, I-2.D, I-3.D, I-4.D, I-5.D, I-6.D, I-7.D, I-8.D, I-9.D, I-10.D O O R1 R1 Het Het N N R2 R2 N S S R3e R3f R3e R3f R7 R7 ) (I-1.D) O R1 O R1 Het R2 Het R2 N N N R8 R3e R3f R8 R3e R3f R7 R9 R7 R9 (I-3.D) (I-4.D) O R1 O R1 Het R2 Het R2 N N N R3e R3f N R8 R3e R3f N R8 R7 R7 (I-5.D) (I-6.D) O R1 O R1 Het Het N N R2 R2 N O O R3e R3f R3e R3f R7 R7 (I-7.D) (I-8.D) (11405124_1):JIN 256 O R1 O R1 Het R2 N Het R2 N N N N R3e R3f R3e R3f R7 R9 R7 R9 (I-9.D) (I-10.D) where Het, R1, R2, R3e, R3f, R7, R8 and R9 are as d in any one of claims 1 to 34.
52. The nd as claimed in claim 51, where R1 is a l 1 and R7 is selected from the group consisting of hydrogen, fluorine, C1-C4-alkyl, C1-C2-fluoroalkyl, C1-C4-alkoxy, C1-C2-fluoroalkoxy, cyclopropyl, optionally substituted by 1, 2 or 3 methyl groups, fluorinated cyclopropyl and Y2-Cyc2.
53. The compound as claimed in any one of claims 35 to 52, where R3b, R3c, R5, R6, if present, are hydrogen.
54. The compound as claimed in any one of the preceding claims, where Cyc1 is selected from the group consisting of saturated 4-, 5-, 6-, 7- or 8-membered heteromonocycles and saturated 7-, 8-, 9- or bered heterobicycles, where the heteromonocycles and the heterobicycles have one nitrogen or oxygen atom as ring member and may have one further heteroatom or heteroatom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5 radicals RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4 radicals RC1, where RC1, RC2 and Y' are as defined in claim 1.
55. The compound as claimed in any one of the preceding claims, where Y1-Cyc1, is selected from the group consisting of 1-piperidinyl, 4,4-difluoropiperidinyl, 4- dinyl, 1-methylpiperidinyl, 1-piperazinyl, ylpiperazinyl, morpholinyl, azepaneyl, azepanyl, hexahydrofuro[3,4-c]pyrrolyl, 2,5- diazabicyclo[2.2.1]heptanyl, 3,8-diazabicyclo[3.2.1]octanyl, thiomorpholinyl, 1- oxothiomorpholinyl, N-(oxetanyl)amino, 1,1-dioxothiomorpholinyl and oxetan ylamino. (11405124_1):JIN 257
56. The compound as claimed in any one of claims 1 to 53, where Cyc1 is selected from the group consisting of phenyl, 5- or 6 membered monocyclic hetaryl, and 9- or 10 ed bicyclic hetaryl, where hetaryl has one heteroatom, selected from O, S and N as ring member and optionally one or two r nitrogen atoms as ring members, where phenyl and the hetaryl radical are unsubstituted or either carry, independently of each other, 1, 2, 3, 4 or 5 radicals RC1.
57. The compound as claimed in claim 56, where Y1 is a chemical bond and Cyc1 is selected from the group consisting of phenyl, 5- or 6-membered monocyclic hetaryl selected from the group consisting of pyridyl, dinyl, furyl, thienyl, pyrrolyl, imidazolyl, lyl, oxazolyl and thiazolyl, 9- or 10-membered bicyclic hetaryl selected from the group consisting of indolyl, quinolinyl, isoquinolinyl, olinyl, benzimidazolyl, benzotriazolyl, yrazolyl and benzofuryl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are ed from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2, or, if Cyc1 is phenyl, two radicals RC1 which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic heterocyclic radical, which is selected from 2,3-dihydrobenzofuranyl, 2,3-dihydrobenzofuranyl, 1,3- dihydroindolonyl, 1,3-dihydroindolonyl, benzo-1,3-dioxolanyl, 1,3- dioxolanyl, benzo-1,4-dioxanyl, benzo-1,4-dioxanyl, benzo-1,5-dioxepanyl and benzo-1,4-dioxepanyl.
58. The compound as claimed in any one of the preceding , where Cyc2 and Cyc3, independently of each other, are selected from the group consisting of saturated 4-, 5-, 6-, 7- or 8-membered heteromonocycles and saturated 7-, 8-, 9- or 10-membered heterobicycles, where the monocycles and the heterobicycles have one nitrogen or oxygen atom as ring member and may have one further atom or atom group as ring member, which is selected from the group consisting of O, S, S(=O), S(=O)2 and N, where the saturated heteromonocycle and the saturated heterobicycle are unsubstituted or carry 1, 2, 3, 4 or 5 radicals RC1 or one radical Y'-RC2 and 0, 1, 2, 3 or 4 radicals RC1, where RC1, RC2 and Y' are as defined in claim 1.
59. The compound as claimed in claim 57, where Y2-Cyc2 and Y3-Cyc3, independently of each other, are is selected from the group consisting of phenyl, 5- or 6-membered (11405124_1):JIN 258 monocyclic hetaryl selected from the group consisting of pyridyl, pyrimidinyl, furyl, thienyl, pyrrolyl, imidazolyl, lyl, oxazolyl and thiazolyl, 9- or 10-membered bicyclic hetaryl selected from the group consisting of indolyl, quinolinyl, isoquinolinyl, quinazolinyl, benzimidazolyl, riazolyl, benzopyrazolyl and benzofuryl, where phenyl and hetaryl are unsubstituted or carry 1, 2 or 3 radicals RC1 which are selected from the group consisting of fluorine, chlorine, CN, methyl, difluoromethyl, trifluoromethyl, methoxy and NH2, or, if Cyc2 or Cyc3 are phenyl, two radicals RC1 which are bound to adjacent carbon atoms, together with the phenyl ring to which they are bound, form a bicyclic heterocyclic l, which is selected from 2,3-dihydrobenzofuranyl, 2,3- obenzofuranyl, 1,3-dihydroindolonyl, 1,3-dihydroindolonyl, benzo- oxolanyl, benzo-1,3-dioxolanyl, benzo-1,4-dioxanyl, benzo-1,4-dioxanyl, benzo-1,5-dioxepanyl and benzo-1,4-dioxepanyl.
60. A compound according to claim 1, which is selected from the group consisting of 3,7-di(pyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(pyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(pyridinyl)[2-(pyridinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3,7-di(pyridinyl)[2-(pyridinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(quinolinyl)ethyl][4-(trifluoromethyl)phenyl]thieno[2,3-d]pyridazin- one; 3-(4-methylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-[4-(propanyl)phenyl][2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(4-ethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 4-{4-oxo[2-(quinolinyl)ethyl]-4,5-dihydrothieno[2,3-d]pyridazin yl}benzonitrile; 3-(4-methoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(4-fluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(4-ethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-[4-(dimethylamino)phenyl][2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; (4-{4-oxo[2-(quinolinyl)ethyl]-4,5-dihydrothieno[2,3-d]pyridazin yl}phenyl)acetonitrile; (11405124_1):JIN 259 3-(4-hydroxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-chlorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-methylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-ethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-fluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-methoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-ethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-hydroxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(quinolinyl)ethyl][2-(trifluoromethyl)phenyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-[3-(methoxymethyl)phenyl][2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(3-methoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; thoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-[3-(dimethylamino)phenyl][2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-[4-Oxo(2-quinolinyl-ethyl)-4,5-dihydro-thieno[2,3-d]pyridazinyl}- benzonitrile; 3-(3-fluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(3-hydroxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; N,N-dimethyl{4-oxo[2-(quinolinyl)ethyl]-4,5-dihydrothieno[2,3- d]pyridazinyl}benzamide; 3-(3-methylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; quinolinyl)ethyl](thiophenyl)thieno[2,3-d]pyridazin-4(5H)-one; 3-(1-methyl-1H-indolyl)(2-quinolinyl-ethyl)-5H-thieno[2,3-d]pyridazin one; 3-(1H-indolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; imidinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2-methoxypyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(pyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(4-methoxypyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(furanyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; (11405124_1):JIN 260 3-(quinolinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(isoquinolinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(isoquinolinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1H-indolyl)(2-quinolinyl-ethyl)-5H-thieno[2,3-d]pyridazinone; 3-(2,3-dihydrobenzofuranyl)(2-quinolinyl-ethyl)-5H-thieno[2,3-d]pyridazin- 4-one; 3-(quinolinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(3,5-dimethyl-1,2-oxazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(6-methoxypyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,3-dihydro-1,4-benzodioxinyl)[2-(quinolinyl)ethyl]thieno[2,3- d]pyridazin-4(5H)-one; ethylpyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(5-methoxypyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-[6-(morpholinyl)pyridinyl][2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(1,3-benzodioxolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(quinolinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- one; 3-[1-(2-methylpropyl)-1H-pyrazolyl][2-(quinolinyl)ethyl]thieno[2,3- d]pyridazin-4(5H)-one; tert-butyl 2-{4-oxo[2-(quinolinyl)ethyl]-4,5-dihydrothieno[2,3-d]pyridazin yl}-1H-pyrrolecarboxylate; 3-(2-methoxypyrimidinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 5-[2-(quinolinyl)ethyl](2,3,4-trifluorophenyl)thieno[2,3-d]pyridazin-4(5H)- one; 3-(4-fluoromethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; (11405124_1):JIN 261 3-(4-fluoromethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(3-chlorofluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2-chlorofluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(3,4-dimethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; -dimethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2,4-difluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2,4-dimethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,5-dimethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,3-difluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(3,4-dimethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(3,4-difluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(5-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(4-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(3,5-dimethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2,5-difluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(2,3-dimethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(3-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(2-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(3,5-difluorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(3-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; (11405124_1):JIN 262 ethoxymethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(2,5-dichlorophenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(naphthalenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-phenyl[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1-benzofuranyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1H-indazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(4,5-difluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-(2-fluoromethylphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 3-(2-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; 3-methyl{4-oxo[2-(quinolinyl)ethyl]-4,5-dihydrothieno[2,3-d]pyridazin yl}benzonitrile; 5-[2-(6-fluoroquinolinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2,2-difluoro(quinolinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)- one; 3-(pyridinyl)[2-(thieno[3,2-b]pyridinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)- one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin- one; 7-fluoroimidazo[1,2-a]pyridinyl)ethyl](pyridinyl)thieno[2,3- d]pyridazin-4(5H)-one; 8-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(quinolinyl)ethyl][4-(trifluoromethyl)phenyl]phthalazin-1(2H)-one; 8-(4-methylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-[4-(propanyl)phenyl][2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(4-ethylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 4-{4-oxo[2-(quinolinyl)ethyl]-3,4-dihydrophthalazinyl}benzonitrile; 8-(4-methoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(4-fluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; (11405124_1):JIN 263 (4-{4-oxo[2-(quinolinyl)ethyl]-3,4-dihydrophthalazinyl}phenyl)acetonitrile; 8-(4-hydroxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-chlorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; ethylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-ethylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-fluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-methoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; ethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 3-{4-oxo[2-(quinolinyl)ethyl]-3,4-dihydrophthalazinyl}benzonitrile; 8-(3-fluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3-hydroxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; N,N-dimethyl{4-oxo[2-(quinolinyl)ethyl]-3,4-dihydrophthalazin yl}benzamide; 8-(3-methylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(quinolinyl)ethyl](thiophenyl)phthalazin-1(2H)-one; 8-(1-methyl-1H-indolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,5-dimethyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1H-indolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1H-indolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(pyrimidinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-methoxypyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(furanyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(quinolinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1H-indolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,3-dihydrobenzofuranyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,4-dihydro-2H-1,5-benzodioxepinyl)[2-(quinolinyl)ethyl]phthalazin- one; 8-(1-benzofuranyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(6-methoxypyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,3-dihydro-1,4-benzodioxinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)- one; 8-(2-methylpyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(5-methoxypyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; (11405124_1):JIN 264 8-(5-fluoropyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1,3-benzodioxolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-[1-(2-methylpropyl)-1H-pyrazolyl][2-(quinolinyl)ethyl]phthalazin-1(2H)- one; utyl 2-{4-oxo[2-(quinolinyl)ethyl]-3,4-dihydrophthalazinyl}-1H- pyrrolecarboxylate; 8-(3-chlorofluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-chlorofluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,4-dimethylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,4-dimethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,5-dimethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,3-difluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,4-dimethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,4-difluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; luoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(4-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,5-dimethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2,5-difluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3,5-difluorophenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(3-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(naphthalenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-phenyl[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(1-benzofuranyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; ethyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(4,5-difluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-fluoromethylphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 8-(2-fluoromethoxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](3-methoxypyridinyl)phthalazin-1(2H)- one; (11405124_1):JIN 265 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyrimidinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1-methyl-1H-pyrazolyl)phthalazin- one; 8-(furanyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-oxo-2,3-dihydro-1H-indol yl)phthalazin-1(2H)-one; 8-(3,4-dihydro-2H-chromenyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-(1,1-dioxidothiomorpholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; imidazo[1,2-a]pyridinyl)ethyl](morpholinyl)phthalazin-1(2H)-one; 8-(1,1-dioxidothiomorpholinyl)[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)- yl)phthalazin-1(2H)-one; 8-(5,5-difluorohexahydrocyclopenta[c]pyrrol-2(1H)-yl)[2-(imidazo[1,2-a]pyridin- 2-yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](piperazinyl)phthalazin-1(2H)-one; 8-(4,4-difluoropiperidinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-[4-(chloromethyl)(hydroxymethyl)piperidinyl][2-(imidazo[1,2-a]pyridin- 2-yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](piperidinyl)phthalazin-1(2H)-one; 8-(2,3-dihydro-4H-1,4-benzoxazinyl)[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][4-(trifluoromethyl)piperidin yl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](4-methylpiperazinyl)phthalazin-1(2H)- one; 8-(1,3-dihydro-2H-isoindolyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-(7-benzyl-2,7-diazaspiro[4.4]nonyl)[2-(imidazo[1,2-a]pyridin yl]phthalazin-1(2H)-one; 8-({[(3aR,4S,6aS)benzyloctahydrocyclopenta[c]pyrrolyl]methyl}amino)[2- (imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; (11405124_1):JIN 266 tert-butyl (3R)({3-[2-(imidazo[1,2-a]pyridinyl)ethyl]oxo-3,4- dihydrophthalazinyl}amino)pyrrolidinecarboxylate; 8-(2,6-dimethylmorpholinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1,4-oxazepanyl)phthalazin-1(2H)-one; tert-butyl 2-(imidazo[1,2-a]pyridinyl)ethyl]oxo-3,4-dihydrophthalazin yl}-3,6-dihydropyridine-1(2H)-carboxylate; 3-(3-methoxypyridinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 3-(3-hydroxypyridinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 3-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5)H- one; 3-(pyridinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 3-(pyrimidinyl)(2-quinolinyl)ethyl)thieno[3,2-c]pyridin-4(5H)-one; 3-(2-oxoindolinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 3-(3-hydroxyphenyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 5-[2-(5-ethylpyridinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)- one; 3-(morpholinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; tert-butyl(4-oxo(2-quinolinyl)ethyl)-4,5-dihydrothieno[3,2-c]pyridinyl)- 5,6-dihydropyridine-1(2H) carboxylate; 5-(2-(quinolinyl)ethyl)(1,2,3,6-tetrahydropyridinyl)thieno[3,2-c]pyridin- 4(5H)-one; tert-butyl(4-oxo(2-quinolinyl)ethyl)-4,5-dihydrothieno[3,2-c]pyridinyl)- piperidine-1 carboxylate; 3-(piperidinyl)[2-(quinolinyl)ethyl]thieno[3,2-c]pyridin-4(5H)-one; 3-(pyridinyl)[2-(5,6,7,8-tetrahydroquinolinyl)ethyl]thieno[2,3-d]pyridazin- 4(5H)-one; (R)-tert-butyl 3-(3-(2-(imidazo[1,2-a]pyridinyl)ethyl)oxo-3,4- dihydrophthalazinylamino)pyrrolidinecarboxylate; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3R)-pyrrolidinylamino]phthalazin- 1(2H)-one; ydroxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; (E)(pyridinyl)(2-(quinolinyl)vinyl)isoquinolin-1(2H)-one; (11405124_1):JIN 267 anti (rac) 8-(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti (+)(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti (-)(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; 8-(pyridinyl)(2-quinolinyl-ethyl)isoquinolin-1(2H)-one; anti (rac) 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one; anti (+) 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; anti (-) 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; (E)pyridinyl(2-quinolinyl-vinyl)-2H-phthalazinone; anti (rac) 8-pyridinyl(2-quinolinyl-cyclopropyl)-2H-phthalazinone; anti (+) 8-pyridinyl(2-quinolinyl-cyclopropyl)-2H-phthalazinone; anti (-) 8-pyridinyl(2-quinolinyl-cyclopropyl)-2H-phthalazinone; and the N-oxides thereof, the prodrugs thereof as defined in claim 1, the ers thereof and the hydrates f, and the pharmaceutically acceptable salts thereof.
61. A compound according to claim 1, which is selected from the group consisting of 3-(pyridinyl)(2-(quinolinyl)ethyl)furo[3,2-c]pyridin-4(5H)-one; 5-(2-(1H-benzo[d]imidazolyl)ethyl)(pyridinyl)furo[3,2-c]pyridin-4(5H)-one; imidazo[1,2-a]pyridinyl)ethyl)(pyridinyl)furo[3,2-c]pyridin-4(5H)-one; 3-(pyrimidinyl)(2-(quinolinyl)ethyl)furo[3,2-c]pyridin-4(5H)-one; 4-(pyridinyl)(2-(quinolinyl)allyl)pyrido[2,3-d]pyridazin-5(6H)-one; syn 8-(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; anti 3-(pyridazinyl)(2-(quinolinyl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one; syn idazinyl)(2-(quinolinyl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one; anti 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one; syn 3-(pyridinyl)(2-(quinolinyl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one; anti 8-(oxetanylamino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(oxetanylamino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(pyridazinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(pyridazinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(6-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; (11405124_1):JIN 268 syn 8-(6-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(2-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(2-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(1-methyl-1H-pyrazolyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(1-methyl-1H-pyrazolyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(3-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(3-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(2-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn luoropyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-((3S)hydroxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one; syn 8-((3S)hydroxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one; anti 8-(3-methoxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(3-methoxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-morpholino(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-morpholino(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 1-(1-oxo(2-(quinolinyl)cyclopropyl)-1,2-dihydroisoquinolinyl)piperidine carbonitrile; syn 1-(1-oxo(2-(quinolinyl)cyclopropyl)-1,2-dihydroisoquinolinyl)piperidine itrile; anti 8-((3R,4R)fluorohydroxypiperidinyl)(2-(quinolin lopropyl)isoquinolin-1(2H)-one; syn 8-((3R,4R)fluorohydroxypiperidinyl)(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-((3S)hydroxypyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one; syn 8-((3S)hydroxypyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one; anti 8-((3R)hydroxypyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- (11405124_1):JIN 269 one; syn 8-((3R)hydroxypyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)- one; anti 8-(methyl(oxetanyl)amino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn hyl(oxetanyl)amino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(4-methoxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4-methoxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(4-hydroxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4-hydroxypiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(1-acetylpiperidinylamino)(2-quinolincyclopropyl)isoquinolin-1(2H)-one; syn 8-(1-acetylpiperidinylamino)(2-quinolincyclopropyl)isoquinolin-1(2H)-one; anti 8-(piperidinylamino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(piperidinylamino)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 2-(2-(quinolinyl)cyclopropyl)(tetrahydro-2H-pyranyl)isoquinolin-1(2H)-one; syn 2-(2-(quinolinyl)cyclopropyl)(tetrahydro-2H-pyranyl)isoquinolin-1(2H)-one; anti 2-(2-(quinolinyl)cyclopropyl)(2-oxaazaspiro[3.4]octanyl)isoquinolin- 1(2H)-one; syn 2-(2-(quinolinyl)cyclopropyl)(2-oxaazaspiro[3.4]octanyl)isoquinolin- 1(2H)-one; anti 8-(dihydro-1H-furo[3,4-c]pyrrol-5(3H,6H,6aH)-yl)(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(dihydro-1H-furo[3,4-c]pyrrol-5(3H,6H,6aH)-yl)(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one; anti -difluoropiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4,4-difluoropiperidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-morpholino(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-morpholino(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(3-(difluoromethyl)pyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one; syn 8-(3-(difluoromethyl)pyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one; anti 8-((1R,5S)oxaazabicyclo[3.2.1]octanyl)(2-(quinolinyl)cyclopropyl )isoquinolin-1(2H)-one; syn 8-((1R,5S)oxaazabicyclo[3.2.1]octanyl)(2-(quinolinyl)cyclo- (11405124_1):JIN 270 )isoquinolin-1(2H)-one; anti 8-(4-methylpiperazinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4-methylpiperazinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(3-(fluoromethyl)pyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one; syn 8-(3-(fluoromethyl)pyrrolidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin- 1(2H)-one; anti 8-((1R,5S)oxaazabicyclo[3.2.1]octanyl)(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-((1R,5S)oxaazabicyclo[3.2.1]octanyl)(2-(quinolin yl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(4-fluorophenyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4-fluorophenyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(furanyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(furanyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(4,5-dihydrofuranyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4,5-dihydrofuranyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 8-(4-methoxyphenyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 8-(4-methoxyphenyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 2-(2-(6-fluoroquinolinyl)cyclopropyl)morpholinoisoquinolin-1(2H)-one; syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)morpholinoisoquinolin-1(2H)-one; anti 6-fluoroquinolinyl)cyclopropyl)(pyridinyl)isoquinolin-1(2H)-one; syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyridinyl)isoquinolin-1(2H)-one; anti 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyrimidinyl)isoquinolin-1(2H)-one; syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyrimidinyl)isoquinolin-1(2H)-one; anti 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyridinyl)isoquinolin-1(2H)-one; syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyridinyl)isoquinolin-1(2H)-one; anti 4-fluoro(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 4-fluoro(pyridinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 4-chloro(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 4-chloro(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; anti 4-(pyridinyl)(2-(quinolinyl)cyclopropyl)pyrido[2,3-d]pyridazin-5(6H)-one; syn4-(pyridinyl)(2-(quinolinyl)cyclopropyl)pyrido[2,3-d]pyridazin-5(6H)-one; anti 6-fluoroquinolinyl)cyclopropyl)(pyridinyl)phthalazin-1(2H)-one; (11405124_1):JIN 271 syn 2-(2-(6-fluoroquinolinyl)cyclopropyl)(pyridinyl)phthalazin-1(2H)-one; anti 7-fluoro(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; syn 7-fluoro(pyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; anti 3-(2-fluoropyridinyl)(2-(6-fluoroquinolinyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one; syn luoropyridinyl)(2-(6-fluoroquinolinyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one; anti 5-(2-(6-fluoroquinolinyl)cyclopropyl)(pyridinyl)thieno[3,2-c]pyridin-4(5H)- one; syn 6-fluoroquinolinyl)cyclopropyl)(pyridinyl)thieno[3,2-c]pyridin-4(5H)- one; anti 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; anti 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; anti 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; anti 3-(6-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; syn 3-(6-fluoropyridinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)- one; anti 3-(2-methylpyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one; syn 3-(2-methylpyrimidinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin- 4(5H)-one; anti 3-(pyridazinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyridazinyl)(2-(quinolinyl)cyclopropyl)thieno[3,2-c]pyridin-4(5H)-one; anti 3-(2-fluoropyridinyl)(2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)- one; syn 3-(2-fluoropyridinyl)(2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)- one; anti 3-(morpholinyl)-5[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; (11405124_1):JIN 272 syn 3-(morpholinyl)-5[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; anti 3-(pyridinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[3,2-c]pyridin- 4(5H)-one; syn 3-(pyridinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[3,2-c]pyridin- 4(5H)-one; anti 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; anti imidinyl)[2-(quinolinyl)cyclopropyl]thieno[2,3-d]pyridazin-4(5H)-one; syn 3-(pyrimidinyl)[2-(quinolinyl)cyclopropyl]thieno[2,3-d]pyridazin-4(5H)-one; anti idinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[2,3-d]pyridazin- 4(5H)-one; syn -(pyridinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[2,3-d]pyridazin- 4(5H)-one; anti 3-(pyridinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[3,2-c]pyridin- 4(5H)-one; syn 3-(pyridinyl)[2-(thieno[3,2-b]pyridinyl)cyclopropyl]thieno[3,2-c]pyridin- 4(5H)-one; anti 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; syn 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[3,2-c]pyridin-4(5H)-one; anti 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[2,3-d]pyridazin-4(5H)-one; syn 3-(pyridinyl)[2-(quinolinyl)cyclopropyl]thieno[2,3-d]pyridazin-4(5H)-one; anti 4-fluoro(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; syn 4-fluoro(pyrimidinyl)(2-(quinolinyl)cyclopropyl)isoquinolin-1(2H)-one; 8-(pyridinyl)[2-(quinolinyl)ethyl]pyrido[3,4-d]pyridazin-1(2H)-one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](1-methyl-1H-imidazolyl)(pyridin yl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](1-methyl-1H-pyrazolyl)(pyridin yl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)(pyridinyl)thieno[2,3- d]pyridazin-4(5H)-one; 5-[2-(6-chloroquinolinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 5-[2-(3-methylquinolinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 8-fluoroquinolinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 5-[2-(6-fluoroquinolinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; (11405124_1):JIN 273 6-fluoroquinolinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 5-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 3-(pyridinyl)[2-(quinoxalinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 1,5-naphthyridinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(1H-indazolyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 3-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(1H-pyrazolyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(1H-benzimidazolyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(1H-benzimidazolyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(6-chloroquinolinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 3-(pyridinylethynyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 3-(pyridinylethynyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(3,5-dimethylpyridinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(7-fluoroquinolinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 5-[2-(pyrazinyl)ethyl](pyridinyl)thieno[2,3-d]pyridazin-4(5H)-one; 2-[2-(1,6-naphthyridinyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 2-[2-(8-fluoroquinolinyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 8-(pyridinyl)[1-(quinolinyl)propanyl]isoquinolin-1(2H)-one; 2-[2-(3-methylquinolinyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1H-pyrazolyl)phthalazin-1(2H)-one; 8-(morpholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1-oxa-4,9-diazaspiro[5.6]dodec yl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-oxaazaspiro[3.5]nonyl)phthalazin- 1(2H)-one; 8-[(3R)hydroxypiperidinyl][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-[(3S)hydroxypiperidinyl][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; imidazo[1,2-a]pyridinyl)ethyl](2-oxaazaspiro[3.3]heptyl)phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1,2-oxazolidinyl)phthalazin-1(2H)-one; 8-(hexahydrocyclopenta[b][1,4]oxazin-4(4aH)-yl)[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; (11405124_1):JIN 274 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydrofuranyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](3-oxaazabicyclo[3.2.1]octyl)phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-oxaazaspiro[3.4]octyl)phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2,2,6,6-tetrafluoromorpholinyl)phthalazin- 1(2H)-one; 8-(4-hydroxypiperidinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-methylpyrimidinyl)phthalazin-1(2H)-one; 8-(2-cyclopropylpyrimidinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridazinyl)phthalazin-1(2H)-one; 8-(5-fluoropyridinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; 8-[2-(3-fluorophenyl)morpholinyl][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](2-methoxypyrimidinyl)phthalazin-1(2H)- one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][2-(trifluoromethyl)pyridinyl]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](8-oxaazabicyclo[3.2.1]octyl)phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][2-(trifluoromethyl)morpholinyl]phthalazin- 1(2H)-one; -dimethylmorpholinyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; 8-[2-(4-chlorophenyl)morpholinyl][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 3,4-difluorophenyl)morpholinyl][2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](piperidinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-2H-pyranyl)phthalazin-1(2H)- one; 8-(2,6-diazabicyclo[3.2.1]octyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; (11405124_1):JIN 275 8-[(1S,5S)-3,6-diazabicyclo[3.2.0]heptyl][2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 8-(furanyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1-methyl-1H-pyrazolyl)phthalazin-1(2H)- one; 8-(hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; -diazaspiro[4.4]nonyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; 8-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptyl][2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 8-(2,7-diazaspiro[3.5]nonyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; 8-(2,6-diazaspiro[3.5]nonyl)[2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)- one; 8-(piperidinyl)[2-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-[2-(aminomethyl)chloropyrrolidinyl][2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl]-4,8-di(pyridinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3aR,4S,7R,7aS)-octahydro-1H-4,7- epiminoisoindolyl]phthalazin-1(2H)-one; 8-[5-(4-chlorophenyl)-2,5-diazabicyclo[2.2.1]heptyl][2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 4-bromo[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3aS,8aS)-octahydropyrrolo[3,4-c]azepin- 2(1H)-yl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3aS,8aR)-octahydropyrrolo[3,4-c]azepin- 2(1H)-yl]phthalazin-1(2H)-one; tert-butyl (3aR,4S,7R,7aS){3-[2-(imidazo[1,2-a]pyridinyl)ethyl]oxo-3,4- ophthalazinyl}octahydro-2H-4,7-epiminoisoindolecarboxylate; ahydro-5H-furo[2,3-c]pyrrolyl)[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1,2,3,6-tetrahydropyridinyl)phthalazin- (11405124_1):JIN 276 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3S)-tetrahydrofuranylamino]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(3R)-tetrahydrofuranylamino]phthalazin- 1(2H)-one; 8-{[5-(hydroxymethyl)-1,4-dioxanyl]methoxy}[2-(imidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](oxetanyloxy)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinylmethoxy)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](morpholinylmethyl)phthalazin-1(2H)-one; imidazo[1,2-a]pyridinyl)ethyl](pyridinyloxy)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][(oxetanylmethyl)amino]phthalazin-1(2H)- one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-2H-pyranylamino)phthalazin- 1(2H)-one; imidazo[1,2-a]pyridinyl)ethyl][(1-methylazetidinyl)amino]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](1,3-oxazolylamino)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl][methyl(oxetanyl)amino]phthalazin-1(2H)- one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](piperidinylamino)phthalazin-1(2H)-one; 8-[(1-acetylpiperidinyl)amino][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 8-[(1-acetylpiperidinyl)amino][2-(imidazo[1,2-a]pyridinyl)ethyl]phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydrofuranylamino)phthalazin-1(2H)- one; imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-2H-pyranylamino)phthalazin- 1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](piperidinylamino)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl]{methyl[(3-methyloxetanyl)methyl]amino}- phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](oxetanylamino)phthalazin-1(2H)-one; 8-{[(3aS,4S,6aS)-octahydrocyclopenta[c]pyrrolylmethyl]amino}[2-(5,6,7,8- (11405124_1):JIN 277 tetrahydroimidazo[1,2-a]pyridinyl)ethyl]phthalazin-1(2H)-one; 5-(morpholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl]{[(3-methyloxetanyl)methyl]amino}- phthalazin-1(2H)-one; 4-(pyridinyl)[2-(quinolinyl)ethyl]pyrido[2,3-d]pyridazin-5(6H)-one; 4-(morpholinyl)[2-(quinolinyl)ethyl]pyrido[2,3-d]pyridazin-5(6H)-one; 4-(oxetanylamino)[2-(quinolinyl)ethyl]pyrido[2,3-d]pyridazin-5(6H)-one; 6-methoxypyridinyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 2-[2-(1,3-benzothiazolyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 5-methylpyridinyl)ethyl](pyridinyl)isoquinolin-1(2H)-one; 5-[(E)(6-methoxyquinolinyl)ethenyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)- one; 8-(pyridinyl)[(E)(quinazolinyl)ethenyl]isoquinolin-1(2H)-one; 5-[(E)(6-chloroquinolinyl)ethenyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 5-[(E)(3-methylquinolinyl)ethenyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 8-(pyridinyl)[(E)(quinolinyl)ethenyl]isoquinolin-1(2H)-one; 5-[(E)(1,3-benzothiazolyl)ethenyl](pyridinyl)thieno[3,2-c]pyridin-4(5H)-one; 3-(pyridinyl)[(E)(quinolinyl)ethenyl]thieno[3,2-c]pyridin-4(5H)-one; and the enantiomers thereof , the N-oxides f, the prodrugs thereof as defined in claim 1, the tautomers thereof and the hydrates thereof, and the pharmaceutically acceptable salts thereof.
62. The compound which is selected from the group consisting of 7-(pyridinyl)[2-(quinolinyl)ethyl]thieno[2,3-d]pyridazin-4(5H)-one; 5-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(pyrimidinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1-methyl-1H-pyrazolyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1,1-dioxidothiomorpholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; imidazo[1,2-a]pyridinyl)ethyl](pyrimidinyl)phthalazin1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](morpholinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)- yl)phthalazin-1(2H)-one; (11405124_1):JIN 278 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](pyrimidinyl)phthalazin-1(2H)-one; 2-[2-(imidazo[1,2-a]pyridinyl)ethyl](morpholinyl)phthalazin-1(2H)-one; 3-methyl(pyridinyl)(2-(quinolinyl)ethyl)thieno[3,2-c]pyridin-4(5H)-one; 5-(3-hydroxyphenyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 7-(pyridinyl)[2-(quinolinyl)ethyl]furo[3,2-c]pyridin-4(5H)-one; anti 5-(pyrimidinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn imidinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(pyridinyl)[2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; anti pholinyl)(2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; syn 5-(morpholinyl)(2-(quinolinyl)cyclopropyl]isoquinolin-1(2H)-one; pholinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 2-[2-(1H-benzimidazolyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 4-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; 5-(1,4-dihydropyrimidinyl)[2-(5,6,7,8-tetrahydroimidazo[1,2-a]pyridin yl)ethyl]phthalazin-1(2H)-one; imidazo[1,2-a]pyridinyl)ethyl](pyridinyl)phthalazin-1(2H)-one; 5-(pyridinyl)[2-(quinolinyl)ethyl]phthalazin-1(2H)-one; and the enantiomers thereof, the N-oxides thereof, the prodrugs thereof, the tautomers thereof and the hydrates thereof, and the pharmaceutically able salts thereof, wherein gs are derivatives of the compounds of formula I carrying an OH or NH2-group, where one of the hydrogen atoms of the OH or NH2-group is substituted by a C1-C4- alkylcarbonyl group, by benzoyl, or by an acyl group derived from an amino acid, which is linked to the oxygen or nitrogen of the OH or NH2-group via the carbonyl group of the amino acid; or wherein prodrugs are alkylcarbonyloxyalkyl carbonates or carbamates of compounds I carrying an OH- or oup in which one of the hydrogen atoms of the OH- or NH2-group has been replaced by a group of the formula -C(=O)-O-CHRp-OC (=O)-Rq in which Rp and Rq are independently of one another C1-C4-alkyl.
63. The compound as claimed in claim 1 being anti (rac) 8-(pyridinyl)(2-(quinolin- 2-yl)cyclopropyl)isoquinolin-1(2H)-one, or an enantiomer thereof, or a N-oxide thereof, or (11405124_1):JIN 279 a prodrug thereof as defined in claim 1, or a tautomer thereof or a hydrate thereof, or a pharmaceutically acceptable salt thereof.
64. The compound as claimed in claim 1 being anti 3-(pyridazinyl)(2-(quinolin yl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one, or an enantiomer thereof, or a N-oxide thereof, or a g thereof as defined in claim 1, or a tautomer thereof or a hydrate thereof, or a pharmaceutically acceptable salt thereof.
65. The compound as claimed in claim 1 being anti 3-(pyridinyl)(2-(quinolin yl)cyclopropyl)furo[3,2-c]pyridin-4(5H)-one, or an enantiomer f, or a e thereof, or a N-oxide thereof, or a prodrug thereof as defined in claim 1, or a tautomer f or a hydrate f, or a pharmaceutically acceptable salt thereof.
66. The compound as claimed in any one of the preceding claims for use in therapy.
67. Pharmaceutical composition which comprises at least one compound as claimed in any one of claims 1 to 65 and at least one excipient.
68. The compounds according to any of claims 1 to 65 for treating a medical disorder, which can be treated by inhibition of phosphodiesterase type 10A.
69. The compounds according to claim 68 where the l disorder is ed from neurological and psychiatric disorders.
70. The compounds according to any of claims 1 to 65 for treating CNS disorders in a mammalian.
71. The compounds according to any of claims 1 to 65 for treating schizophrenia in a mammalian.
72. The compounds according to any of claims 1 to 65 for treating ive dysfunction associated with schizophrenia in a mammalian. (11405124_1):JIN 280
73. The compounds according to any of claims 1 to 65 for ng r disorders in a mammalian.
74. The compounds according to any of claims 1 to 65 for treating depression in a mammalian.
75. The compounds according to any of claims 1 to 65 for treating cognitive dysfunction associated with Alzheimer's disease in a mammalian.
76. The compounds according to any of claims 1 to 65 for treating diet-induced obesity in a mammalian.
77. The compounds according to any of claims 1 to 65 for treating Huntington's disease in a mammalian.
78. The compounds according to any of claims 1 to 65 for treating anxiety in a mammalian.
79. The compounds ing to any of claims 1 to 65 for ng substance-related disorders in a mammalian.
80. Use of at least one compound as d in any of claims 1 to 65 for the manufacture of a medicament for treating a medical disorder, selected from neurological and atric disorders which can be treated by inhibition of phosphodiesterase type 10A. AbbVie Deutschland GmbH & Co. KG AbbVie Inc. By the Attorneys for the Applicant SPRUSON & FERGUSON Per: (11405124_1):JIN
NZ623727A 2011-11-09 2012-11-08 Inhibitors of phosphodiesterase type 10a NZ623727B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161557874P 2011-11-09 2011-11-09
US61/557,874 2011-11-09
PCT/EP2012/072150 WO2013068470A1 (en) 2011-11-09 2012-11-08 Inhibitors of phosphodiesterase type 10a

Publications (2)

Publication Number Publication Date
NZ623727A NZ623727A (en) 2016-06-24
NZ623727B2 true NZ623727B2 (en) 2016-09-27

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012334074A1 (en) Inhibitors of phosphodiesterase type 10A
AU2006210047B2 (en) Heterocyclic substituted phenyl methanones as inhibitors of the glycine transporter 1
US9868749B2 (en) Fused imidazole and pyrazole derivatives as modulators of TNF activity
CA2931586C (en) Imidazopyridine derivatives as modulators of tnf activity
CN103958504B (en) 10A type phosphodiesterase inhibitor compound
JP6571653B2 (en) Fused tricyclic imidazole derivatives as modulators of TNF activity
DK168537B1 (en) 1,2-Benzisoxazol-3-yl and 1,2-benzisothiazol-3-yl derivatives, a process for their preparation and pharmaceutical compositions containing such derivatives
EP3080112B1 (en) Imidazopyridine derivatives as modulators of tnf activity
KR101661383B1 (en) 8-methyl-1-phenyl-imidazol[1,5-a]pyrazine compounds
AU2014230745A1 (en) Novel inhibitor compounds of phosphodiesterase type 10A
JP2016539972A (en) Tetrahydroimidazopyridine derivatives as modulators of TNF activity
CA2905751A1 (en) Novel compounds and compositions for inhibition of fasn
JP2003502272A (en) Bicyclic pyridine and pyrimidine derivatives as neuropeptide Y receptor antagonists
WO2021257614A1 (en) Heterocyclic inhibitors of enpp1
NZ623727B2 (en) Inhibitors of phosphodiesterase type 10a
WO2021092525A1 (en) Wdr5 inhibitors and modulators