HUE033057T2 - 6,7-dihidropirazolo[l,5-a]pirazin-4(5H)-on vegyületek és ezek alkalmazása mint az mGluR2 receptorok negatív alloszterikus modulátorai - Google Patents

6,7-dihidropirazolo[l,5-a]pirazin-4(5H)-on vegyületek és ezek alkalmazása mint az mGluR2 receptorok negatív alloszterikus modulátorai Download PDF

Info

Publication number
HUE033057T2
HUE033057T2 HUE14727824A HUE14727824A HUE033057T2 HU E033057 T2 HUE033057 T2 HU E033057T2 HU E14727824 A HUE14727824 A HU E14727824A HU E14727824 A HUE14727824 A HU E14727824A HU E033057 T2 HUE033057 T2 HU E033057T2
Authority
HU
Hungary
Prior art keywords
methyl
pyrazin
dihydropyrazolo
alkyl
phenyl
Prior art date
Application number
HUE14727824A
Other languages
English (en)
Inventor
Gool Michiel Luc Maria Van
Diego Sergio-Alvar Alonso-De
Jose Maria Cid-Nuòez
Oscar Delgado-Gonzalez
Annelies Marie Antonius Decorte
Gregor James Macdonald
Antonius Adrianus Hendrikus Petrus Megens
Andres Avelino Trabanco-Suarez
Aranzazu Garcia-Molina
Jose Ignacio Andres-Gil
Original Assignee
Janssen Pharmaceutica Nv
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 Janssen Pharmaceutica Nv filed Critical Janssen Pharmaceutica Nv
Publication of HUE033057T2 publication Critical patent/HUE033057T2/hu

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61P25/18Antipsychotics, 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
    • A61P25/20Hypnotics; Sedatives
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Description

(12) EUROPEAN PATENT SPECIFICATION (45) Date of publication and mention (51) Int Cl.: of the grant of the patent: C07D 487104 <200601> A61K 3114985 <200601> 10.05.2017 Bulletin 2017/19 A61P 25100<200601> (21) Application number: 14727824.6 (86) International application number: PCT/EP2014/061478 (22) Date of filing: 03.06.2014 (87) International publication number: WO 2014/195311 (11.12.2014 Gazette 2014/50)
(54) 6,7-DIHYDROPYRAZOLO[1,5-A]PYRAZIN-4(5H)-ONE COMPOUNDS AND THEIR USE AS NEGATIVE ALLOSTERIC MODULATORS OF MGLUR2 RECEPTORS
6,7-DIHYDROPYRAZOLO[1,5-A]PYRAZIN-4(5H)-ON-VERBINDUNGEN UND DEREN VERWENDUNG ALS NEGATIVE ALLOSTERISCHE MODULATOREN VON MGLUR2-REZEPTOREN COMPOSES 6,7-DIHYDROPYRAZOLO[1,5-A]PYRAZIN-4(5H)-ONE ET LEUR UTILISATION EN TANT QUE MODULATEURS ALLOSTERIQUES NEGATIFS DE RECEPTEURS MGLUR2 (84) Designated Contracting States: · DELGADO-GONZALEZ, Oscar AL AT BE BG CH CY CZ DE DK EE ES FI FR GB 45007 Toledo (ES) GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO · DECORTE, Annelies, Marie, Antonius PL PT RO RS SE SI SK SM TR 2340 Beerse (BE)
Designated Extension States: · MACDONALD, Gregor, James BA ME 2340 Beerse (BE) • MEGENS, Antonius, Adrianus, Hendrikus, Petrus (30) Priority: 04.06.2013 EP 13170447 2340 Beerse (BE) 27.06.2013 EP 13173939 · TRABANCO-SUAREZ, Andres, Avelino 29.04.2014 EP 14166450 45007 Toledo (ES) • GARCfA-MOLINA, Aranzazu (43) Date of publication of application: 45007 Toledo (ES) 13.04.2016 Bulletin 2016/15 · ANDRES-GIL, Jose, Ignacio 45007 Toledo (ES) (73) Proprietor: Janssen Pharmaceutics, N.V. 2340 Beerse (BE) (74) Representative: Garcia Prieto, Maria
Johnson &amp; Johnson (72) Inventors: Patent Law Department • VAN GOOL, Michiel, Luc, Maria Turnhoutseweg 30 45007 Toledo (ES) 2340 Beerse (BE) • ALONSO-DE DIEGO, Sergio-Alvar 45007 Toledo (ES) (56) References cited: • CID-NLJNEZ, Jose, Maria WO-A1-2010/130423 WO-A1-2012/083224 45007 Toledo (ES)
Description
Field of the Invention [0001] The present invention relates to novel 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one derivatives as negative allosteric modulators (NAMs) of the metabotropic glutamate receptor subtype 2 ("mGluR2"). The invention is also directed to pharmaceutical compositions comprising such compounds, to processes for preparing such compounds and compositions, and to the use of such compounds and compositions for the prevention or treatment of disorders in which the mGluR2 subtype of metabotropic receptors is involved.
Background of the Invention [0002] The glutamatergic system in the CNS is one of the neurotransmitter systems that play a key role in several brain functions. Metabotropic glutamate receptors (mGluR) belong to the G-protein-coupled family, and eight different subtypes have been identified to date, which are distributed to various brain regions (Ferraguti &amp; Shigemoto, Cell &amp; Tissue Research, 326:483-504, 2006). mGluRs participate in the modulation of synaptic transmission and neuronal excitability in the CNS by the binding of glutamate. This activates the receptor to engage intracellular signaling partners, leading to cellular events (Niswender &amp; Conn, Annual Review of Pharmacology &amp; Toxicology 50:295-322,2010). mGluRs are further divided into three subgroups based on their pharmacological and structural properties: group-l (mGluRI and mGluR5), group-ll (mGluR2 and mGluR3)and group-ill (mGluR4, mGluR6, mGluR7 and mGluR8). Group-11 ligands, both orthosteric and allosteric modulating, are considered to be potentially useful in the treatment of various neurological disorders, including psychosis, mood disorders, Alzheimer disease and cognitive or memory deficiencies. This is consistent with their primary localisation in brain areas such as the cortex, hippocampus and the striatum (Ferraguti &amp; Shigemoto, Cell &amp; Tissue Research 326:483-504, 2006). Particularly antagonists and negative allosteric modulators are reported to hold potential for the treatment of mood disorders and cognitive or memory dysfunction. This is based on findings with group-ll receptor antagonists and negative allosteric modulators tested in laboratory animals subjected to a range of experimental conditions deemed relevant to these clinical syndromes (Goeldner et al, Neuropharmacology 64:337-346, 2013).
[0003] Clinical trials are, for example, underway with mGluR2/3 antagonist R04995819 (F. Hoffmann-La Roche Ltd.) in adjunctive therapy in patients with Major Depressive Disorder having inadequate response to ongoing antidepressant treatment (ClinicalTrials.gov Identifier NCT01457677, retrieved 19 February 2014). WO 2013066736 (Merck Sharp &amp; Dohme Corp.) describes quinoline carboxamide and quinoline carbonitrile compounds as mGluR2 NAMs. WO2013174822 (Domain therapeutics) describes 4H-pyrazolo[1,5-a]quinazolin-5-ones and 4H-pyr-rolo[1,2-a]quinazolin-5-ones and in vitro mGluR2 NAM activity thereof. WO 2014064028 (F. Hoffman-La Roche AG) discloses a selection of mGlu2/3 negative allosteric modulators and their potential use in the treatment of Autistic Spectrum Disorders (ASD).
[0004] The group-ll receptors are mainly located on presynaptic nerve terminals where they exert a negative feedback loop to the release of glutamate into the synapse (Kelmendi et al, Primary Psychiatry 13:80-86,2006). Functional inhibition of these receptors by antagonists or negative allosteric modulators therefore lifts the brake on glutamate release, resulting in enhanced glutamatergic signaling. This effect is believed to underlie the antidepressant-like and procognitive effects observed in preclinical species with inhibitors of the Group-ll receptor. In addition, treatment of mice with group-ll orthosteric antagonists has been shown to enhance signaling by growth factors such as brain derived neurotrophic factor (BDNF) (Koike et al, Behavioural Brain Research 238:48-52, 2013). Since BDNF and other growth factors have been shown to be critically involved mediating synaptic plasticity, this mechanism is likely to contribute to both antidepressant and procognitive properties of these compounds. Inhibition of mGluRs of the group-ll receptor family is therefore considered to represent a potential therapeutic mechanism for neurological disorders, including depression and cognitive or memory dysfunction.
Description of the Invention [0005] The present invention is directed to 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one derivatives of Formula (I) 9 R2 "SA^ V'n/ R3 R4 (I) and stereoisomeric forms thereof, wherein R1 is phenyl or 2-pyridinyl, each optionally substituted with one or more substituents each independently selected from the group of halo, C1.4alkyl, mono- or poly-haloC^alkyl, -0-C1.4alkyl, -C^alkyl-O-C^alkyl, mono- or poly-haloCi_4alkyloxy, -C^alkyl-OH, C-,_4alkylthio, mono- orpoly-haloC^alkylthio, cyano, C3_7cycloalkyl optionally substituted with trifluoromethyl, and -SF5; or is R 2 is selected from yy -6 1 and 1 wherein R5 and R6 are each independently selected from the group of hydrogen, halo, cyano, C^alkyl, -C^alkyl-OH, C3.7cycloalkyl, mono-orpoly-haloC1_4alkyl,-C1^alkyl-0-C1^alkyi,-0-C1^alkyl, mono-orpoly-haloC.|_4alkyloxy, 1-acetylazetidin-3-yl, and NR’R"; wherein R’ is selected from hydrogen and C1_4alkyl; R" is selected from hydrogen and C1_4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a heterocyclic group selected from the group of 1-azetidinyl, 1-pyrrolidinyl, 1-piperidinyl, 1-piperazinyl, and 4-morpholinyl; wherein each of the heterocyclic groups may be optionally substituted with a substituent selected from halo, hydroxyl, C^alkyl, mono- or poly-haloCi_4alkyl, and -(CO)C1.4alkyl; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1_4alkyl, mono- or poly-haloC1_4alkyl, -C1_4alkyl-0-C1_4aikyi, and -C^alkyl-OH; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0006] The present invention also relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier or excipient.
[0007] Additionally, the invention relates to a compound of Formula (I) for use as a medicament, and to a compound of Formula (I) for use in the treatment or in the prevention of central nervous system conditions or diseases selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder.
[0008] The invention also relates to the use of a compound of Formula (I) in combination with an additional pharmaceutical agent for use in the treatment or prevention of central nervous system conditions or diseases selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder.
[0009] Furthermore, the invention relates to a process for preparing a pharmaceutical composition according to the invention, characterized in that a pharmaceutically acceptable carrier is intimately mixed with a therapeutically effective amount of a compound of Formula (I).
[0010] The invention also relates to a method of treating or preventing a central nervous system disorder selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of Formula (I) or a therapeutically effective amount of a pharmaceutical composition according to the invention.
[0011] The invention also relates to a product comprising a compound of Formula (I) and an additional pharmaceutical agent, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of central nervous system conditions or diseases selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder.
Description of the figures [0012]
Figure 1 shows the frequency distributions for distance travelled obtained in historical series of solvent-pretreated control rats. In figure 1, - corresponds to scopolamine + JNJ 42153605; — corresponds to scopolamine alone; and — corresponds to no challenge.
Figure 2 shows the effect of Co. No. 1 (mg/kg p.o., 4 h prior to test) on exploration times of the new vs. the familiar arm by rats in the absence or presence of PCP (0 (= veh) or 0.75 mg/kg s.c., 0.5 h prior to test) in the V-maze. Data are reported as Mean ± SEM, n = 12/group; ANOVA with LSD-post hoc, p vs. familiar arm: *** p<0.001, *<0.05.
Figure 3 shows the interaction of Co. No. 1 with reserpine in rats. Shown are the effects on pupil diameter before reserpine challenge (Fig. 3a) and the reversal of the reserpine-induced ptosis (Fig. 3b), miosis (Fig. 3c) and sedation (Fig. 3d) measured 1 h after s.c. (left panel), 1 h after p.o. (middle panel) and 4 h after p.o. (right panel) administration of Co. No. 1.
Figure 4 illustrates the placement of the hippocampus brain slice in a well of a MEA biochip, with 60 x 3D-tip electrodes (black dots). Traces show the recorded potential at each electrode (Fig. 4a), and the captured fEPSP traces of paired pulses separated by 30 ms (Fig. 4b). The preparation was perfused with artificial cerebrospinal fluid (ACSF). (3D = three-dimensional; fEPSP = field excitatory postsynaptic potentials; MEA = micro-electrode array; ms = milliseconds).
Figure 5 shows how Co. No. 1 restores fEPSP depressed by 1 μΜ LY-354740 in the dentate gyrus of rat hippocampal brain slices.
Figure 5a: fEPSP amplitude (% of baseline) is shown after the application of LY-354740 (1 μΜ), followed by the application of Co. No. 1 (10 μΜ), and then by the application of the mGlu2 antagonist LY-341495, and finally by a washout. At the end of experiments, the AMPA antagonist CNQX (6-cyano-7-nitroquinoxaline-2,3-dione, 50 μΜ) and kynurenic acid (1 mM) were added as controls to block glutamate-mediated fEPSP.
Figure 5b: The same experiments as shown in Figure 5a, but presenting the PPR results. The error bars represent the SEM of 17 independent slices from 4 SD rats. (ACSF = artificial cerebrospinal fluid; fEPSP = field excitatory postsynaptic potential; PPR = paired-pulse ratio; SEM = standard error of the mean).
Figure 6 shows how Co. No. 1 facilitates long-term potentiation in the dentate gyrus of rat hippocampal brain slices. Monitoring of the post-synaptic response for 80 min shows induction of LTP after weak theta stimulation (at 30 min).
Figure 6a: fEPSP in response to the weak theta stimulus under control conditions (vehicle: open circles) and following the application of 10 μΜ Co. No. 1 (solid circles).
Figure 6b: PTP and LTP under control conditions and after application of 10 μΜ Co. No. 1. The error bars represent the SEM of 22 independent slices from 4 SD rats. LTP = long-term potentiation; NAM = negative allosteric modulator; PTP = post-theta potentiation; SEM = standard error of the mean.
Detailed description of the invention [0013] The present invention relates in particular to compounds of Formula (I) as defined hereinabove, and stereoi-someric forms thereof, wherein R1 is phenyl or 2-pyridinyl, each optionally substituted with one or more substituents each independently selected from the group of halo, C^alkyl, mono- or poly-haloC1_4alkyl, -0-C1_4alkyl, -C1.4alkyl-0-C1_4alkyi, mono- or poly-haloC1_4alkyloxy, -C^alkyl-OH, mono- or poly-haloC1_4alkylthio, cyano, and -SF5; or is
R 2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, halo, cyano, C^alkyl, -C^alkyl-OH, C3.7cycloalkyl, mono- or poly-haloC1.4alkyl, -C1.4alkyl-0-C1.4alkyi, -0-C.|_4alkyl, mono- or poly-haloC.|_4alkyloxy, and NR’R"; wherein R’ is selected from hydrogen and C1_4alkyl; R" is selected from hydrogen and C1.4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a heterocyclic group selected from the group of 1-azetidinyl, 1-pyrrolidinyl, and 1-piperidinyl; wherein each of the heterocyclic groups may be optionally substituted with a halo substituent; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1_4alkyl, mono- or poly-haloC^alkyl, -C^alkyl-O-C^^lkyl, and -C^alkyl- OH; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0014] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl optionally substituted with one, two or three substituents each independently selected from the group of halo, C^alkyl, mono- or poly-haloC^alkyl, -O-C^alkyl, -C^alkyl-O-C^alkyl, mono- or poly-haloC.|_4alkyloxy, mono-or poly-haloC1_4alkylthio, cyano, and -SF5; or 2-pyridinyl optionally substituted with one or two substituents each independently selected from the group of halo, C1.4alkyl, mono- or poly-haloC1.4alkyl, and -0-C1_4alkyl; R2 is
wherein R5 and R6 are each independently selected from the group of hydrogen, halo, cyano, C1_4alkyl, C3.7cycloalkyl, mono- or poly-haloC1_4alkyl, -0-C1_4alkyl, mono- or poly-haloC^alkyloxy, and NR’R"; wherein R’ is selected from hydrogen and C^alkyl; R" is selected from hydrogen and C1_4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a heterocyclic group selected from the group of 1-azetidinyl, 1-pyrrolidinyl, and 1-piperidinyl; wherein each of the heterocyclic groups may be optionally substituted with a halo substituent; r3 js selected from hydrogen and C1_4alkyl; R4 is selected from the group of C1_4alkyl, mono- or poly-haloC^^lkyl, and -C1_4alkyl-0-C1_4alkyi; in particular >CR3R4 is selected from the group of >CH(CH3), >CH(CH2CH3), >CH(CH2F), and >CH(CH2OCH3); and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0015] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl optionally substituted with one, two or three substituents each independently selected from the group of halo, C1_4alkyl, poly-haloC14alkyl, -0-C14alkyl, -C1.4alkyl-0-C1_4alkyl, poly-haloC.|_4alkyloxy, poly-haloC1_4alkylthio, cy-ano, and -SF5; or 2-pyridinyl optionally substituted with one or two substituents each independently selected from the group of halo, C1_4alkyl, mono- or poly-haloC1.4alkyl, and -0-C1.4alkyl; R2 is
wherein R5 and R6 are each independently selected from the group of hydrogen, cyano, C^alkyl, poly-haloC14alkyl, -0-C1_4alkyl, and NR’R"; wherein R’ is selected from hydrogen and C14alkyl; R" is C1_4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a 1-azetidinyl; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of C1_4alkyl, mono- or poly-haloC^alkyl, and -C1_4alkyl-0-C1_4alkyl; in particular >CR3R4 is selected from the group of >CFI(CFI3), >CH(CH2CH3), >CFI(CH2F), and >CH(CH2OCH3); and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0016] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is selected from (a) a substituted phenyl selected from the group of and
or (b) a substituted 2-pyridinyl selected from the group of
R2 is selected from the group of i
R3 is selected from hydrogen and C1.4alkyl; R4 is selected from the group of C^alkyl, mono- or poly-haloC.|_4alkyl, and -C^alkyl-O-C^alkyl; in particular >CR3R4 is selected from the group of >CH(CH3), >CH(CH2CH3), >CH(CH2F), and >CH(CH2OCH3); and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0017] In an additional embodiment, the present invention relates to compounds selected from the group of (7S)-7- 1 Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)- 5-(4-Chlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Methoxy- 4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Me- thyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo [1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-meth- ylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophe- nyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[6-Ethoxy-5-(trifluorome-thyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S) ^2 methylpyridin-4-yl)-5-[4-(pentafluoro-X6-sulfanyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H) one, e y 3-(2-methylpyridin-4-yl)-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-Chloro-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; 5-[(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(tri-fluoromethyl)benzonitrile; (7S)-3-(2-Methoxypyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Ethylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2,6-dimethylpyridin-4-yl)-7-methyl-6,7-dihydropyra-zolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2,6-dimethylpyridin-4-yl)-7-methyl-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Ethylamino)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyra-zolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethoxy)phenyl]-6,7-dihydropyrazo-lo[1,5-'a]pyrazin-4(5H)-one; (7S)-3-[2-(Dimethylamino)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihy-dropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-{4-[(trifluoromethyl)sulfanyl]phenyl}-6,7-di-hydropyrazolo[1,5-a]pyrazin-4(5H)-one; 4-{(7S)-7-Methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-4,5,6,7-tetrahydropyrazo-lo[1,5-a]pyrazin-3-yl}pyridine-2-carbonitrile; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(meth-ylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-7-methyl- 5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[4-(trifluoromethyl)phenyl]-3-[2-(trifluoromethyl)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 4-{(7S)-5-[3-Chloro-4-(trifluorome-thyl)phenyl]-7-methyl-4-oxo-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}-6-(methylamino)pyridine-2-carbonitrile; (7S)-3-(2-Methoxypyridin-4-yl)-5-[3-methoxy-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4(5H)-one; and (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof. [0018] In an additional embodiment, the present invention relates to compounds selected from the group of (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, or a hydrochloride salt, or a sulfate salt, or a methanesulfonate salt, or a maleate salt thereof; (7S)-5-[3-Ethoxy-4-(trifluorome-thyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-5-[3-
Methoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-7-Methyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-5-[6-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-di-hydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(pentafluoro- X6-sulfanyl)phenyl]- 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[3-methyl-4-(trifluorome- thyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-3-(2,6-Dimethylpyridin-4- yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one;(7S)-5-[3-Chloro-4-(trifluorome- thyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Fluoro-4-(trif-luoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-Chloro-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-[(7S)-7-
Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(trifluoromethyl)benzonitrile; (7S)- 3-(2-Methoxypyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Ethylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2,6-dimethylpyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2,6-dimethylpyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Ethylamino)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethoxy)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Dimethylamino)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(5-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-{4-[(trifluoromethyl)sulfanyl]phenyl}-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; 4-{(7S)-7-Methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-4,5,6,7- tetrahydropyrazolo[1,5-a]pyrazin-3-yl}pyridine-2-carbonitrile; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2-methox-ypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-me-thyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[4-(trifluorome-thyl)phenyl]-3-[2-(trifluoromethyl)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 4-{(7S)-5-[3-Chloro-4-(trif-luoromethyl)phenyl]-7-methyl-4-oxo-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}-6-(methylamino)pyridine-2-carboni-trile; (7S)-3-(2-Methoxypyridin-4-yl)-5-[3-methoxy-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-[3-Fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-di-hydropyrazolo[1,5-a]pyrazin-4(5H)-one.
[0019] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, optionally substituted with one, two or three substituents each independently selected from the group of halo, C^alkyl, poly-haloC1.4alkyl, -O-C^alkyl, -C^alkyl-OH and cyano; or 2-pyridinyl, substituted with one or two substituents each independently selected from the group of halo, poly-haloC^alkyl, and -O-C^alkyl; R2 is
wherein R5a is selected from hydrogen and C^alkyl, and R6a is selected from the group of hydrogen, C^alkyl, -C^alkyl- 0-C1_4alkyl, -OC1_4alkyl, and -C^^lkyl-OH; or
wherein one of R5b and R6b is hydrogen, and the other R5b or R6b is C1_4alkyl; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1_4alkyl, mono- or poly-haloC^alkyl, and -C^alkyl-O-C^alkyl; in particular >CR3R4 is selected from the group of >CH2, >CH(CH3), >CH(CH2CH3), >CH(CH2F), >CH(CH2OCH3) and >C(CH3)2; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0020] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, optionally substituted with one, two or three substituents each independently selected from the group of fluoro, chloro, methyl, CF3, -0-CH3, -0-CH2CH3, cyano, -CH(CH3)(OH); or 2-pyridinyl, substituted with one or two substituents each independently selected from the group of fluoro, chloro, CF3, and -0-CH2CH3; R2 is
wherein R5a is selected from hydrogen and methyl, and R6a is selected from the group of hydrogen, methyl, -CH2-0-CH3, -0-CH3, and -CH2-OH; or
wherein one of R5b and R6b is hydrogen, and the other R5b or R6b is methyl; R3 is selected from hydrogen and C1.4alkyl; R4is selected from the group of hydrogen, C^alkyl, mono- orpoly-haloC^alkyl, and -C1_4alkyl-0-C1.4alkyl; in particular >CR3R4 is selected from the group of >CH2, >CH(CH3), >CH(CH2CH3), >CH(CH2F), >CH(CH2OCH3) and >C(CH3)2; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0021] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is (a) a phenyl substituent selected from the group of
or (b) a 2-pyridinyl substituent selected from the group of
R2 is
wherein R5a is selected from hydrogen and methyl, and R6a is selected from the group of hydrogen, methyl, -CH2-0-CH3,-O-CHg, and-CH2-OH; or
wherein one of R5b and R6b is hydrogen, and the other R5b or R6b is methyl; R3 js selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1.4alkyl, mono- or poly-haloC^alkyl, and -C^alkyl-O-C^^lkyl; in particular >CR3R4 is selected from the group of >CH2, >CH(CH3), >CH(CH2CH3), >CH(CH2F), >CH(CH2OCH3) and >C(CH3)2; and the N-oxides, and the pharmaceutically acceptable salts and the solvates thereof.
[0022] In an additional embodiment, the present invention relates to compounds selected from the group of (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Me-thyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Methoxyme-thyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S)-7-Methyl- 3-(5-methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7R)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-[3-Ethoxy-4-(trif-luoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, 5-[3-Ethoxy-4-(trif-luoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-M ethoxy-3,5-dimethylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S) 7 Methyl-3-(6- methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7 ethoxyphe nyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 3-(2-Methylpyridin-4-yl)-5 [ π luoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dimethoxyphenyl)-7-methyl-3-(2 me ylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Methoxypyridin-4-yl)-7-methyl-5-[4-(tri luorome thyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-(4-methylphenyl)-3-(2-methylpyridin y) 6.7- dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[3-(1-methylethoxy)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 4-[(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazoo 1,5-a]pyrazin-5(4H)-yl]benzonitrile; (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,5-Difluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Difluoro-5-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Fluoro-5-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-phenyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(1-Hydroxye-thyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-(3,4-Dichlorophe-nyl)-7-(fluoromethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-(3,4-Dichlorophe- nyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-(3,4-Dichlo-rophenyl)-3-[2-(hydroxymethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-7-Ethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S)-7-Methyl 3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-(1-
Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chlo- ro-2-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(2,4-
Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 7,7-Dimethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S)-5-(4 Chloro 2 methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2- methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Chloropy- ridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-pyridin-4-yl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Ethoxy-6-fluoropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Chloro-6-ethoxypyrid-in-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-[6-Ethoxy-5-(trif-luoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and the N-ox-ides, and the pharmaceutically acceptable salts and the solvates thereof.
[0023] In an additional embodiment, the present invention relates to compounds selected from the group of (7S)-7-
Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, or a hydrochloride salt, or a sulfate salt, or a methanesulfonate salt, or a maleate salt thereof; (7S)-7-Methyl-3 pyridin 4 yl 5 [4 (tri fluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Methoxymethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-7-Methyl- 3-(5-methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-[3-Ethoxy-4-(trif- luoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-[3-Ethoxy-4-(trif-luoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-M ethoxy-3,5- dimethylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(6- methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Methoxyphe- nyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 3-(2-Methylpyridin-4-yl)-5-[4-(trif-luoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dimethoxyphenyl)-7-methyl-3-(2-meth-ylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Methoxypyridin-4-yl)-7-methyl-5-[4-(trifluorome-thyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-(4-methylphenyl)-3-(2-methylpyridin-4-yl)- 6.7- dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[3-(1-methylethoxy)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; 4-[(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4- oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]benzonitrile; (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4- yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-5-(3,5-Difluorophenyl)-7-methyl- 3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Ethoxyphenyl)-7-methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Difluoro-5-methoxyphenyl)-7-methyl-3-(2-meth-ylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Fluoro-5-methoxyphenyl)-7-methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-phenyl-6,7-dihy-dropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydro-pyrazolo[1,5-a] pyrazi n-4(5H )-o n e; (7R)-5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-(3,4-Dichlorophenyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihy-dropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-(3,4-Dichlorophenyl)-3-[2-(hydroxymethyl)pyridin-4-yl]-7-methyl-6,7- dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-7-Ethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihy-dropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-di-hydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-2-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-di-hydropyrazolo[l!5-a]pyrazin-4(5H)-one; (7S)-5-(2,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4(5H)-one; 7,7-Dimethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyra-zolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-2-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazo-l0[i 5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazo-lo[l!5-a]pyrazin-4(5H)-one; (7S)-5-(5-Chloropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-pyridin-4-yl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Ethoxy-6-fluoropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(3-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-[6-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihy- dropyrazolo[1,5-a]pyrazin-4(5H)-one; and the pharmaceutically acceptable salts and the solvates thereof.
[0024] In an additional embodiment, the present invention relates to compounds selected from the group of (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, or a hydrochloride salt, or a sulfate salt, or a methanesulfonate salt, or a maleate salt thereof; (7S)-7-Methyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Methoxymethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one or a hydrochloride salt thereof; (7R)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Methoxy-3,5-dimethylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(6-methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 3- (2-Methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; ^73)-5-(3,4-Dimethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Methoxypyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (73)_7-Methyl-5-[3-(1-methylethoxy)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; 4- [(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]benzonitrile; (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one or a hydrochloride salt thereof; (7S)-5-(3,5-Difluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Difluoro-5-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Fluoro-5-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7R)-5-(3,4-Dichlorophenyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-(3,4-Dichlorophenyl)-3-[2-(hydroxymethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-Chloro-2-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(2,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; 7,7-Dimethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-2-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(5-Chloropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-pyridin-4-yl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Ethoxy-6-fluoropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; and (7S)-5-[6-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one.
[0025] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein r1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C1_4alkyl, mono- or poly-haloC1.4alkyl, -O-C^alkyl, mono- or poly-haloC^alkyloxy, cyano and -SF5; or is
R 2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl, mono- or poly-haloC^alkyl, -C1_4alkyl-0-C1_4alkyl, -O-C^alkyl, and NR’R"; wherein R’ is selected from hydrogen and C^alkyl; R" is selected from hydrogen and C1_4alkyl; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1_4alkyl, mono- or poly-haloC1_4alkyl, -C1^alkyl-0-C1_4alkyi, and -C^alkyl-OH; and the pharmaceutically acceptable salts and the solvates thereof.
[0026] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C^alkyl, mono- or poly-haloC^alkyl, -O-C^alkyl, mono- or poly-haloC^alkyloxy, cyano and -SF5; or is in particular
R2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl, -C^alkyl-O-C^alkyl, -O-C1.4alkyl, and NR’R"; wherein R’ is hydrogen; R" is hydrogen; R3 is selected from hydrogen and C1.4alkyl; R4 is selected from the group of hydrogen and C1_4alkyl; and the pharmaceutically acceptable salts and the solvates thereof.
[0027] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C1_4alkyl, poly-haloC1_4alkyl and -SF5; R2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C1_4alkyl and -O-C^alkyl; R3 is selected from hydrogen and C1.4alkyl; R4 is hydrogen; and the pharmaceutically acceptable salts and the solvates thereof.
[0028] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C1.4alkyl, poly-haloC1_4alkyl and -SF5; R2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl and -O-C^alkyl; R3 is hydrogen; R4 is selected from hydrogen and C1_4alkyl; and the pharmaceutically acceptable salts and the solvates thereof.
[0029] In an additional embodiment, the present invention relates to compounds of Formula (I) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, substituted with one, two or three substituents each independently selected from the group of halo, and poly-haloCi_4alkyl; R2 is
wherein R5 and R6 are each independently selected from the group of hydrogen, C1.4alkyl, -0-C1_4alkyl, and NR’R"; wherein R’ is hydrogen; R" is C^alkyl; R3 is hydrogen; R4 is hydrogen or C^alkyl; in particular R4 is C1.4alkyl; and the pharmaceutically acceptable salts and the solvates thereof.
[0030] In an additional embodiment, R1 is selected from the group of
and the rest of variables are as defined in Formula (I) herein.
[0031] In an additional embodiment, R1 is selected from the group of
and
and the rest of variables are as defined in Formula (I) herein; and the pharmaceutically acceptable salts and the solvates thereof.
[0032] In a further embodiment, the present invention relates to compounds of Formula (I) as defined herein wherein R3 is hydrogen and R4 is a substituent different from hydrogen having a configuration as depicted in the Formula (Γ) below, wherein the 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one core, R1 and R2 are in the plane of the drawing and R4 is projected above the plane of the drawing (bond shown with a bold wedge), and the rest of variables are as defined in Formula (I) herein
[0033] In a yet further embodiment, the present invention relates to compounds of Formula (I) as defined herein wherein R4 is hydrogen and R3 is a substituent different from hydrogen, for example a Chalky I substituent having a configuration as depicted in the Formula (I") below, wherein the 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one core, R1 and R2 are in the plane of the drawing and R3 is projected above the plane of the drawing (bond shown with a bold wedge), and the rest of variables are as defined in Formula (I) herein
[0034] In an additional embodiment, the present invention relates to compounds of Formula (I’) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, substituted with one or two substituents each independently selected from the group of halo, and poly-haloCi_4alkyl; R2 is
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl, -0-C1_4alkyl, and NR’R"; wherein R’ is hydrogen; R" is C1_4alkyl; R4 is hydrogen or C1_4alkyl; in particular R4 is C1_4alkyl, more in particular methyl; and the pharmaceutically acceptable salts and the solvates thereof.
[0035] In an additional embodiment, the present invention relates to compounds of Formula (Γ) as defined hereinabove, and stereoisomeric forms thereof, wherein R1 is phenyl, substituted with one or two substituents each independently selected from the group of halo, and poly-haloC1.4alkyl; R2 is
wherein one of R5 and R6 is hydrogen or methyl, in particular hydrogen; and the other one of R5 or R6 is selected from the group of C1.4alkyl, -O-C^alkyl, and NR’R"; wherein R’ is hydrogen; R" is C^alkyl; R4 is hydrogen or C1_4alkyl; in particular R4 is C^alkyl, more in particular methyl; and the pharmaceutically acceptable salts and the solvates thereof.
[0036] Specific compounds according to the invention include: (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(pentafluoro-X6-sulfanyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-3-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[4-Chloro-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-Fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Methoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 5-[(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(trifluorome- thyl)benzonitrile; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-Chlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Chlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Chloro-4-ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-3-ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-3-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Chloro-4-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Fluoro-3-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Methoxymethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[3-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(5-methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S)-5-(3,4-Difluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7R)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7S)-5-[4-(Difluoromethoxy)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Fluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(5,6-Dimethylpyridin-3-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-Chloro-2-fluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one, (7R)-5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-pyridin-3-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-[3-Ethoxy-4-(trifluoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Methoxy-3,5-dimethylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Fluoro-4-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(6-methylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(6-Aminopyridin-3-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-[3-Fluoro-4-(trifluoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 3- (2-Methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5FI)-one; (7S)-5-(3,4-Dimethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(2,3-Dihydro-1,4-benzodioxin-6-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-, 4(5H)-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-3-(2,6-dimethyl-4-pyridyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4- one; (7S)-3-[2-(ethylamino)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2-methoxy-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2-ethyl-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(3,4-dichlorophenyl)-3-(2,6-dimethyl-4-pyridyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-[3-(2-Fluoroethoxy)-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-3-(2,6-dimethyl-4-pyridyl)-5-[3-ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-, 4-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2,6-dimethyl-4-pyridyl)-5-[3-methoxy-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; 7-(fluoromethyl)-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2,6-dimethyl-4-pyridyl)-7-methyl-5-[4-(pentafluoro- X6-sulfanyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4-one; (7S)-5-[4-chloro-3-(trifluoromethyl)phenyl]-3-(2,6-dimethyl-4-pyridyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4- one; (7S)-5-[3-ethoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; , (7S)-7-methyl-5-[4-(pentafluoro-X6-sulfanyl)phenyl]-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(3,4-dichlorophenyl)-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-methyl-3-(2-methyl-4-pyridyl)-5-[3-(trifluoromethoxy)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; 5- [(7S)-3-(2,6-dimethyl-4-pyridyl)-7-methyl-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5-yl]-2-(trifluoromethyl)ben-zonitrile; (7S)-5-[3-fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2,6-dimethyl-4-pyridyl)-5-[3-fluoro-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4- one; (7S)-5-(4-Chloro-3-fluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-chloro-5-fluoro-phenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; , (7S)-5-[3-methoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(4-isopropylphenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-methyl-3-(2-methyl-4-pyridyl)-5-(4-propylphenyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-[4-Fluoro-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-chloro-3-(trifluoromethyl)phenyl]-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-[3-(difluoromethoxy)-5-fluoro-phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; 7-ethyl-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2-Aminopyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; , (7S)-5-(4-chlorophenyl)-3-(2,6-dimethyl-4-pyridyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(4-chlorophenyl)-7-methyl-3-(4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; 5- [3-Ethoxy-4-(trifluoromethyl)phenyl]-7-(hydroxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 2-Fluoro-4-[(7S)-7-methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]benzonitrile; (7S)-5-(3-Fluoro-4-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(2-fluoroethoxy)-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-Methyl-5-(4-methylphenyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[3-(1-methylethoxy)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 4- [(7S)-7-Methyl-3-(2-methylpyridin-4-yl)-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]benzonitrile; (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,5-Difluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-difluoro-5-methoxy-phenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(3-fluoro-5-methoxy-phenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-phenyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 7-(Hydroxymethyl)-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; I 5-[(7S)-3-(2-Methoxypyridin-4-yl)-7-methyl-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(trifluorome- thyl)benzonitrile; (7S)-5-(4-Chlorophenyl)-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5- (3,4-Dichlorophenyl)-3-[2-(dimethylamino)pyridin-4-yl]-7-(fluoromethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); ; 5-(3,4-Dichlorophenyl)-3-[2-(dimethylamino)pyridin-4-yl]-7-(fluoromethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one (7S or 7R); (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-[2-(3-fluoroazetidin-1-yl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazo- lo[1,5-a]pyrazin-4(5H)-one; 5- (3,4-Dichlorophenyl)-7-(fluoromethyl)-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- , 4(5H)-one; (7S)-3-[2-(3-Fluoroazetidin-1-yl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; 4-{(7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-4-oxo-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}- 6- (methylamino)pyridine-2-carbonitrile; . (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-5-[3-chloro-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-5-(3,4-dichlorophenyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-(Fluoromethyl)-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5FI)-one; ( (7S)-7-Methyl-5-[4-(trifluoromethyl)phenyl]-3-[2-(trifluoromethyl)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 4-{(7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-4-oxo-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}pyrid- ine-2-carbonitrile; (7S)-3-[2-(3-Hydroxyazetidin-1-yl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-. a]pyrazin-4(5H)-one; (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(2-pyrrolidin-1-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-3-[2-(4-Acetylpiperazin-1-yl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-piperidin-1-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; ^g).7-Methyl-3-(2-morpholin-4-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 4-[(7S)-5-(3,4-Dichlorophenyl)-7-methyl-4-oxo-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl]pyridine-2-carboni-trile; ¢75)-5-(3,4-Dichlorophenyl)-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-[2-(1-hydroxyethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Fluoromethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; ^S)_7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(2,2,2-trifluoro-1-methylethyl)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (73)-5-(3,4-Dichlorophenyl)-3-[2-(difluoromethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one;
5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or 7R);
5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); (7S)-5-(3,4-Dichlorophenyl)-3-[2-(fluoromethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 5-(3,4-Dichlorophenyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or 7R); 5-(3,4-Dichlorophenyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); 7-(Methoxymethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); 7-(Methoxymethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or7R); (7S)-3-(2-Cyclopropylpyridin-4-yl)-5-(3,4-dichlorophenyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Ethoxypyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-[2-(1-methylethyl)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-[2-(hydroxymethyl)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-[2-(1-methylethyl)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- . 4(5H)-one; 4- {(7S)-7-Methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}pyridine-2-car- bonitrile; (7S)-3-[2-(1-Hydroxyethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7R)-7-Ethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Ethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Difluoromethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3,5-Difluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-ethoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-ethylpyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-[2-(Hydroxymethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-{4-[1-(trifluoromethyl)cyclopropyl]phenyl}-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-[2-(1-methylethoxy)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(1-methylethoxy)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-Bromophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methyl-1-oxidopyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-tert-Butylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-methoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5- (3,4-Dichlorophenyl)-7-(methoxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 7-(Methoxymethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Methoxy-6-methylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-methoxy-6-methylpyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(6-Methoxypyridin-3-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-{(7S)-7-Methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-3-yl}pyridine-2-car-bonitrile; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethoxy)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-3-pyridin-4-yl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-5-[3-(1-methylethoxy)-4-(trifluoromethyl)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; 5-[(7S)-7-Methyl-4-oxo-3-pyridin-4-yl-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(trifluoromethyl)benzonitrile; (7S)-5-(4-Cyclopropylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-(Methoxymethyl)-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(3,5-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(2,2,2-trifluoroethoxy)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-2-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-(Difluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-Chloro-3-(difluoromethoxy)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-3-(2-fluoropyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(2,2,2-trifluoro-1-methylethoxy)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-3-[2-(Dimethylamino)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(2,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(difluoromethoxy)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[4-Chloro-3-(trifluoromethoxy)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-(2-methy 1-1-oxidopyridin-4-yl )-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 7,7-Dimethyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; , (7S)-5-(4-Chloro-2-methylphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Cyclopropylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[3-Chloro-4-(difluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; . (7S)-7-Methyl-3-(2-piperazin-1-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-7-Methyl-3-(6-piperazin-1-ylpyridin-3-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-[2-(2-fluoroethoxy)pyridin-4-yl]-7-methyl-6,7-dihydropyrazolo[1,5-, a]pyrazin-4(5H)-one; (7S)-5-(4-Chlorophenyl)-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-[(7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-5(4H)-yl]-2-(trifluorome- thyl)benzonitrile; (7S)-3-(2-Methoxypyridin-4-yl)-5-[3-methoxy-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazolo[1,5-j a]pyrazin-4(5H)-one; 7-(Difluoromethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-Fluoro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-[3-Methoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; 5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); 5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or7R); (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Chloropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[6-Chloro-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-[4-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-pyridin-4-yl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Ethoxy-6-fluoropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[6-methyl-5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-methyl-5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; ¢75)-3-(2,6-Dimethylpyridin-4-yl)-7-methyl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(3-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- . 4(5H)-one; (7S)-5-(5-Chloro-6-methylpyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Fluoropyridin-4-yl)-7-methyl-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-Chloro-5-methylpyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)_5-[6-Ethoxy-3-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-[6-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-5-(5-Chloro-6-ethoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5,6-Dichloropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4,5-Dichloropyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-Chloro-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (75)-3-(2,6-Dimethylpyridin-4-yl)-5-[6-ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(5-Chloro-6-methoxypyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[6-Methoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[6-Ethoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-pyridin-4-yl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-3-(2-Azetidin-1-ylpyridin-4-yl)-5-[6-methoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)pyridin-4-yl]-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; 3-(2-Methylpyridin-4-yl)-5-[5-(trifluoromethyl)pyridin-2-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[4-lodo-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4-Chloro-5-iodopyridin-2-yl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-7-Methyl-3-[2-(methylamino)-1-oxidopyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4(5H)-one; (7S)-3-(2-Chloropyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 7_(1-Hydroxyethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (1R or 1S); 7_(1-Hydroxyethyl)-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (1S or 1R); (7S)-3-(2-Chloropyridin-4-yl)-5-(3,4-dichlorophenyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5.(3,4-Dichlorophenyl)-7-(hydroxymethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(4,5-Dichloro-2-iodophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-(3,4-Dichloro-2-iodophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-(3,4-Dichlorophenyl)-7-(fluoromethyl)-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Bromopyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-(3,4-Dichlorophenyl)-3-(2-fluoropyridin-4-yl)-7-(hydroxymethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-lodopyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-(3,4-Dichlorophenyl)-3-(2-fluoropyridin-4-yl)-7-(hydroxymethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or 7R); 5-(3,4-Dichlorophenyl)-3-(2-fluoropyridin-4-yl)-7-(hydroxymethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one(7R or7S); (7S)-5-[3-Chloro-4-(trifluoromethyl)phenyl]-3-(2-fluoropyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-Bromo-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-(4-lodophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; 5-(3,4-Dichlorophenyl)-3-[2-(dimethylamino)pyridin-4-yl]-7-(hydroxymethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7S or 7R); 5-(3,4-Dichlorophenyl)-3-[2-(dimethylamino)pyridin-4-yl]-7-(hydroxymethyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (7R or 7S); 5-(3,4-Dichlorophenyl)-7-(hydroxymethyl)-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-3-(2-Chloro-6-methoxypyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-3-[6-(1-Acetylazetidin-3-yl)pyridin-3-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5- I ' a]pyrazin-4(5H)-one (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-{4-[(trifluoromethyl)sulfanyl]phenyl}-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one; (7S)-5-[6-Methoxy-5-(trifluoromethyl)pyridin-2-yl]-7-methyl-3-[2-(methylamino)pyridin-4-yl]-6,7-dihydropyrazo-| |o[1,5-a]pyrazin-4(5H)-one; 3-(2-Methylpyridin-4-yl)-7-(trifluoromethyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one; (7S)-5-[3-(hydroxymethyl)-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-(2-methoxy-4-pyridyl)-5-[6-methoxy-5-(trifluoromethyl)-2-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; (7S)-7-methyl-3-[2-methyl-6-(methylamino)-4-pyridyl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-[2-methoxy-6-(methylamino)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-, a]pyrazin-4-one; (7S)-3-[2-fluoro-6-(methylamino)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; 5-(2,4-dichlorophenyl)-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-[2-(dimethylamino)-4-pyridyl]-5-[3-(hydroxymethyl)-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4-one; (7S)-3-[2-(dimethylamino)-4-pyridyl]-5-[3-(fluoromethyl)-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4-one; (7S)-3-(2-fluoro-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-(3,4-dichlorophenyl)-3-[2-(dimethylamino)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-[2-(dimethylamino)-4-pyridyl]-5-[3-(2-fluoroethoxy)-4-(trifluoromethyl)phenyl]-7-methyl-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4-one; (7S)-3-[2-(dimethylamino)-4-pyridyl]-5-[6-methoxy-5-(trifluoromethyl)-2-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-methyl-3-[2-(methylamino)-4-pyridyl]-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methyl-1-oxido-pyridin-1-ium-4-yl)-6,7-dihydropyrazo-lo[1,5-a]pyrazin-4-one; 1 (7S)-3-[2-[ethyl(methyl)amino]-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-3-[2-[ethyl(methyl)amino]-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-3-[2-(ethylamino)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5-’ a]pyrazin-4-one; (7S)-3-[2-(ethylamino)-4-pyridyl]-7-methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; (7S)-5-(3,4-dichlorophenyl)-3-[2-(ethylamino)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-7-methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-3-[2-(propylamino)-4-pyridyl]-6,7-dihydropyrazolo[1,5-1 a]pyrazin-4-one; (7S)-7-methyl-3-[2-(propylamino)-4-pyridyl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-[2-(azetidin-1-yl)-4-pyridyl]-7-methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-3-[2-(isopropylamino)-4-pyridyl]-7-methyl-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-’ a]pyrazin-4-one; (7S)-3-[2-(isopropylamino)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4- one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-3-[2-(fluoromethyl)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; 1 (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-7-methyl-3-[2-(propylamino)-4-pyridyl]-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; (7S)-5-(3,4-dichlorophenyl)-7-methyl-3-[2-(propylamino)-4-pyridyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; (7S)-5-[3-chloro-4-(trifluoromethyl)phenyl]-3-[2-(isopropylamino)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5- a]pyrazin-4-one; ’ (7S)-5-(3,4-dichlorophenyl)-3-[2-(isopropylamino)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one; and the pharmaceutically acceptable salts and solvates of such compounds.
[0037] In another embodiment, specific compounds according to the invention include: > (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one sulfate salt; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one i methane sulfonate salt; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one maleate salt; (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[3-methyl-4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; i (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydro chloride salt; (7S)-5-[3-Methoxy-4-(trifluoromethyl)phenyl]-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyraz'n' 4(5H)-one hydrochloride salt; (7S)-5-(4-Chlorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochlo-. ride salt; (7S)-5-(3-Chloro-4-ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(3-Chloro-4-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one hydrochloride salt; (7S)-3-[2-(Methoxymethyl)pyridin-4-yl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one hydrochloride salt; (7S)-5-(3,4-Difluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(4-Fluorophenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(3-Fluoro-4-methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin- 4(5H)-one hydrochloride salt; (7S)-5-(4-Methoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-7-methyl-3-(2-methyl-4-pyridyl)-5-[3-(trifluoromethoxy)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt; (7S)-5-(3-chloro-5-fluoro-phenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt; (7S)-5-(4-isopropylphenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt; (7S)-7-methyl-3-(2-methyl-4-pyridyl)-5-(4-propylphenyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt; (7S)-5-[3-(difluoromethoxy)-5-fluoro-phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4- one hydrochloride salt; (7S)-7-Methyl-5-[3-(1-methylethoxy)phenyl]-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methylpyridin-4-yl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-7-Methyl-3-(2-piperidin-1-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-3-(2-Cyclopropylpyridin-4-yl)-5-(3,4-dichlorophenyl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(3,4-Dichlorophenyl)-3-(2-ethoxypyridin-4-yl)-7-methyl-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-5-(3,4-Dichlorophenyl)-7-methyl-3-[2-(1-methylethoxy)pyridin-4-yl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-7-Methyl-3-[2-(1-methylethoxy)pyridin-4-yl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-3-(2-Cyclopropylpyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt; (7S)-7-Methyl-3-(2-piperazin-1-ylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt, and (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-{4-[(trifluoromethyl)sulfanyl]phenyl}-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one hydrochloride salt.
[0038] The names of the compounds of the present invention were generated according to the nomenclature rules agreed upon by the Chemical Abstracts Service (C.A.S.) using Advanced Chemical Development, Inc., software (ACD/Name product version 10.01.0.14105, October 2006). In case of tautomeric forms, the name of the depicted tautomeric form of the structure was generated. However it should be clear that the other non-depicted tautomeric form is also included within the scope of the present invention.
Definitions [0039] The notation "C1.4alkyl" as used herein alone or as part of another group, defines a saturated, straight or branched, hydrocarbon radical having, unless otherwise stated, from 1 to 4 carbon atoms, such as methyl, ethyl, 1-propyl, 1-methylethyl, butyl, 1-methyl-propyl, 2-methyl-1-propyl, 1,1-dimethylethyl and the like. The notation "-C^alkyl-OH" as used herein alone or as part of another group, refers to C1.4alkyl as defined before, substituted with one OH group at any available carbon atom.
[0040] The notation "halogen" or "halo" as used herein alone or as part of another group, refers to fluoro, chloro, bromo or iodo, with fluoro or chloro being preferred.
[0041] The notation "mono- and polyhaloC-Malkyl" as used herein alone or as part of another group, refers to C1_4alkyl as defined before, substituted with 1,2, 3 or where possible with more halo atoms as defined before.
[0042] The notation "C3_7cycloalkyl" as used herein refers to a saturated, cyclic hydrocarbon radical having from 3 to 7 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. A particular C3.7cycloalkyl group is cyclopropyl.
[0043] The N-oxide forms of the compounds according to Formula (I) are meant to comprise those compounds of Formula (I) wherein one or several nitrogen atoms are oxidized to the so called N-oxide, particularly those N-oxides wherein a nitrogen atom in a pyridinyl radical is oxidized. N-oxides can be formed following procedures known to the skilled person. The N-oxidation reaction may generally be carried out by reacting the starting material of Formula (I) with an appropriate organic or inorganic peroxide. Appropriate inorganic peroxides comprise, for example, hydrogen peroxide, alkali metal or alkaline metal peroxides, e.g. sodium peroxide, potassium peroxide/ appropriate organic peroxides may comprise peroxy acids such as, for example, benzenecarboperoxoic acid or halo substituted benzenecarboperoxoic acid, e.g. 3-chloroperoxybenzoic acid (or 3-chloroperbenzoic acid), peroxoalkanoic acids, e.g. peroxoacetic acid, alkyl-hydroperoxides, e.g. tert-butyl hydroperoxide. Suitable solvents, e.g are for example, water, lower alkanols, e.g. ethanol and the like, hydrocarbons, e.g. toluene, ketones, e.g. 2-butanone, halogenated hydrocarbons, e.g. dichloromethane, and mixtures of such solvents.
In a particular embodiment, the invention relates to a compound of Formula (I) wherein R2 is
and the rest of variables are as defined herein.
[0044] Whenever the term "substituted" is used in the present invention, it is meant, unless otherwise is indicated or is clear from the context, to indicate that one or more hydrogens, preferably from 1 to 3 hydrogens, more preferably from 1 to 2 hydrogens, more preferably 1 hydrogen, on the atom or radical indicated in the expression using "substituted" are replaced with a selection from the indicated group, provided that the normal valency is not exceeded, and that the substitution results in a chemically stable compound, i.e. a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into a therapeutic agent.
[0045] The term "subject" as used herein, refers to an animal, preferably a mammal, most preferably a human, who is or has been the object of treatment, observation or experiment.
[0046] The term "therapeutically effective amount" as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.
[0047] As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.
[0048] It will be appreciated that some of the compounds of Formula (I) and their pharmaceutically acceptable addition salts and solvates thereof may contain one or more centres of chirality and exist as stereoisomeric forms.
[0049] The term "compounds of the invention" as used herein, is meant to include the compounds of Formula (I), and the salts and solvates thereof.
[0050] As used herein, any chemical formula with bonds shown only as solid lines and not as solid wedged or hashed wedged bonds, or otherwise indicated as having a particular configuration (e.g. R, S) around one or more atoms, contemplates each possible stereoisomer, or mixture of two or more stereoisomers.
[0051] Hereinbefore and hereinafter, the term "compound of Formula (I)" is meant to include the stereoisomers thereof and the tautomeric forms thereof.
[0052] The terms "stereoisomers", "stereoisomeric forms" or "stereochemically isomeric forms" hereinbefore or hereinafter are used interchangeably.
[0053] The invention includes all stereoisomers of the compounds of the invention either as a pure stereoisomer or as a mixture of two or more stereoisomers.
[0054] Enantiomers are stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a racemate or racemic mixture.
[0055] Diastereomers (or diastereoisomers) are stereoisomers that are not enantiomers, i.e. they are not related as mirror images. If a compound contains a double bond, the substituents may be in the E or the Z configuration.
[0056] Substituents on bivalent cyclic (partially) saturated radicals may have either the cis- or trans-configuration; for example if a compound contains a disubstituted cycloalkyl group, the substituents may be in the cis or trans configuration.
[0057] Therefore, the invention includes enantiomers, diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers and mixtures thereof, whenever chemically possible.
[0058] The meaning of all those terms, i.e. enantiomers, diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers and mixtures thereof are known to the skilled person.
[0059] The absolute configuration is specified according to the Cahn-Ingold-Prelog system. The configuration at an asymmetric atom is specified by either R or S. Resolved stereoisomers whose absolute configuration is not known can be designated by (+) or (-) depending on the direction in which they rotate plane polarized light. For instance, resolved enantiomers whose absolute configuration is not known can be designated by (+) or (-) depending on the direction in which they rotate plane polarized light.
[0060] When a specific stereoisomer is identified, this means that said stereoisomer is substantially free, i.e. associated with less than 50%, preferably less than 20%, more preferably less than 10%, even more preferably less than 5%, in particular less than 2% and most preferably less than 1%, of the other isomers. Thus, when a compound of Formula (I) is for instance specified as (R), this means that the compound is substantially free of the (S) isomer; when a compound of Formula (I) is for instance specified as E, this means that the compound is substantially free of the Z isomer; when a compound of Formula (I) is for instance specified as cis, this means that the compound is substantially free of the trans isomer.
[0061] Some of the compounds according to Formula (I) may also exist in their tautomeric form. Such forms in so far as they may exist, although not explicitly indicated in the above formula are intended to be included within the scope of the present invention.
[0062] It follows that a single compound may exist in both stereisomeric and tautomeric forms.
[0063] For therapeutic use, salts of the compounds of Formula (I) are those wherein the counterion is pharmaceutically acceptable. However, salts of acids and bases which arenon-pharmaceutically acceptable may also find use, for exam pie, in the preparation or purification of a pharmaceutically acceptable compound. All salts, whether pharmaceutically acceptable or not, are included within the ambit of the present invention.
[0064] The pharmaceutically acceptable acid and base addition salts as mentioned hereinabove or hereinafter are meant to comprise the therapeutically active non-toxic acid and base addition salt forms which the compounds of Formula (I) are able to form. The pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid. Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e. ethanedioic), malonic, succinic (i.e. butanedioicacid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoicand the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form.
[0065] The compounds of Formula (I) containing an acidic proton may also be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases. Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. primary, secondary and tertiary aliphatic and aromatic amines such as methylamine, ethylamine, propylamine, isopropylamine, the four butylamine isomers, dimeth-ylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-n-butylamine, pyrrolidine, piperidine, morpholine, trimethylamine, triethylamine, tripropylamine, quinuclidine, pyridine, quinoline and isoquinoline; the benzathine, W-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely the salt form can be converted by treatment with acid into the free acid form.
[0066] The term solvate comprises the solvent addition forms as well as the salts thereof, which the compounds of Formula (I) are able to form. Examples of such solvent addition forms are e.g. hydrates, alcoholates and the like.
[0067] In the framework of this application, an element, in particular when mentioned in relation to a compound according to Formula (I), comprises all isotopes and isotopic mixtures of this element, either naturally occurring or synthetically produced, either with natural abundance or in an isotopically enriched form, for example 2H. Radiolabelled compounds of Formula (I) may comprise a radioactive isotope selected from the group of 3H, 11C, 14C, 18F, 122l, 123l, 125l, 131l, 75Br, 76Br, 77Br and 82Br. Preferably, the radioactive isotope is selected from the group of 3H, 11C and 18F.
Preparation [0068] The compounds according to the invention can generally be prepared by a succession of steps, each of which is known to the skilled person. In particular, the compounds can be prepared according to the following synthesis methods.
[0069] The general preparation of some typical examples of the compounds of Formula (I) is described hereunder and in the specific examples, and are generally prepared from starting materials which are either commercially available or prepared by standard synthetic processes commonly used by those skilled in the art. The following schemes are only meant to represent examples of the invention and are in no way meant to be a limit of the invention.
[0070] Alternatively, compounds of the present invention may also be prepared by analogous reaction protocols as described in the general schemes below, combined with standard synthetic processes commonly used by those skilled in the art of organic chemistry.
[0071] The compounds of Formula (I) may be synthesized in the form of racemic mixtures of enantiomers which can be separated from one another following art-known resolution procedures. The racemic compounds of Formula (I) may be converted into the corresponding diastereomeric salt forms by reaction with a suitable chiral acid. Said diastereomeric salt forms are subsequently separated, for example, by selective or fractional crystallization and the enantiomers are liberated therefrom by alkali. An alternative manner of separating the enantiomeric forms of the compounds of Formula (I) involves liquid chromatography using a chiral stationary phase or chiral supercritical fluid chromatography (SFC). Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically.
The absolute configuration of compounds of the invention reported herein was determined by analysis of the racemic mixture by supercritical fluid chromatography (SFC) followed by SFC comparison of the separate enantiomer(s) which were obtained by asymmetric synthesis or by chiral separation of mixtures, followed by vibrational circular dichroism (VCD) analysis of the particular enantiomer(s). A. Preparation of the final compounds
Experimental procedure 1 [0072] Final compounds according to Formula (I) can be prepared by a Goldberg coupling reaction of a compound of Formula (II) with an appropriate aryl halide of Formula (III) where X is halo, in particular bromo or iodo, according to conditions known to the skilled person. Such conditions include for example using a suitable copper(l) catalyst such as copper(l) iodide, in the presence of a ligand, such as Λ/,Λ/’-dimethylethylenediamine, in the presence of a base, such as inorganic carbonates, for example sodium carbonate (Na2C03) or potassium carbonate (K2C03), in a suitable solvent, such as toluene or a mixture of toluene and N,N- dimethylformamide (DMF), under suitable reaction conditions, such as at a convenient temperature, typically ranging between 100 °C and 140 °C, in particular 110 °C, for a period of time to ensure the completion of the reaction. A compound of Formula (III) can be obtained commercially or made according to procedures known in the art. In Reaction Scheme 1, all variables are defined as in Formula (I).
Reaction Scheme 1
Experimental procedure 2
[0073] Alternatively, final compounds according to Formula (I) can be prepared by a Suzuki type coupling reaction of a compound of Formula (IVa) with a suitable boron species or a compound of Formula (IVb), wherein R7a and R8a may be each independently selected from H, C^alkyl or R7a and R8a are taken together to form for example a bivalent radical of formula -CH2CH2-, -CH2CH2CH2- or-C(CH3)2C(CH3)2-, with a suitable heteroaryl halide or aryl halide derivative in the presence of a palladium catalyst, according to reaction conditions known to the skilled person. Such reaction conditions include the use of a palladium catalyst, such as tetrakis(triphenyiphosphine)pailadium(0) or an alternative catalyst system prepared in situ from Pd(OAc)2 and PPh3, a suitable base, such as Na2C03, K2COs, NaOAc, NaHC03 or K3P04, and in a suitable solvent, such as 1,4-dioxane, ora mixture of dimethoxyethane (DME) and water. Degassing the reaction mixture with an inert gas, such as N2 or argon, and heating the reaction mixture to high temperatures, such as reflux temperature under classical heating or microwave irradiation, in particular 80 °C, may enhance the reaction outcome. In Reaction Schemes 2a and 2b, all variables are defined as in Formula (I).
Reaction Scheme 2a
Reaction Scheme 2b
The suitable boron species may be selected for example from a boronic acid or a boronate ester, which may be conveniently represented as a compound of Formula (Ilia)
wherein R2 is as defined in Formula (I) herein and R7a and R8a may be each independently selected from H, C^alkyl or R7a and R8a are taken together to form for example a bivalent radical of formula -CH2CH2-, -CH2CH2CH2- or -C(CH3)2C(CH3)2-. A skilled person can envisage that the reaction under Reaction Scheme 2a can also be performed under similar conditions, when the compound of Formula (IVa) bears a bromo group in place of an iodo group. Such a reaction can be represented as in Reaction Scheme 2c, wherein the compound of Formula (IV), wherein R2a is halo, in particular bromo or iodo and all other variables are as defined in Formula (I), undergoes a Suzuki type coupling as described hereinbefore.
Reaction Scheme 2c
Experimental procedure 3 [0074] Alternatively, final compounds according to Formula (I), wherein R2 is an optionally substituted 4-pyridinyl or 3-pyridinyl, hereby referred to as compounds of Formula (la) or Formula (lb), respectively, can be prepared by a reaction of deprotection of a compound of Formula (la-1) or a compound of Formula (la-2) following art known procedures. A compound of Formula (la) or a compound Formula (lb) can be obtained by removal of the protecting group such as for example a Boc protecting group in the compound of Formula (la-1) or compound of Formula (la-2), in the presence of acidic media, such as trifluoroacetic acid, in an inert solvent such as dichloromethane (DCM), under suitable reaction conditions, such as at a convenient temperature, typically rt, fora period of time to ensure the completion of the reaction.
In Reaction Schemes 3a and 3b, all variables are defined as in Formula (I) and R5a and R6a’ include the residues indicated in the scope as R5 and R6 as well as their protected forms.
Reaction Scheme 3a
Reaction Scheme 3b
Experimental procedure 4 [0075] Alternatively, final compounds according to Formula (la) and Formula (lb) can be prepared by manipulation of a suitable precursor of Formula (lb-1) and of Formula (lb-2) respectively, bearing one or more functional groups which can be converted to the residues R5 and R6 by means of simple reactions known to the person skilled in the art, such as for example introduction of an alcohol or an amine in presence of a base and a suitable solvent or acylation with an acyl chloride in the presence of a base and a suitable solvent or reduction for example by using a suitable reducing agent such as sodium borohydride, in a suitable solvent or by means of cross coupling reactions known to the person skilled in the art, such as for example the Suzuki reaction with a suitable boron species or the Stille reaction with a suitable tin species. In Reaction Schemes 4a and 4b, all variables are defined as in Formula (I) and R5b and R6b include the residues indicated in the scope as R5 and R6 as well as their possible precursors. The person skilled in the art will recognize that suitable reaction conditions should be chosen for different R5b and R6b combinations, to avoid undesired side reactions.
Reaction Scheme 4a
Reaction Scheme 4b
Experimental procedure 5 [0076] Alternatively, final compounds according to Formula (la) and Formula (lb) can be prepared respectively by a reaction of fluorination of a compound of Formula (lc-1) and of Formula (lc-2) wherein Y is N, CH or CR9c and one of r5c, r6c r3c r4ci r9c ancj r10c may be each independently selected from C^alkyl-OH or C1_3alkyl-CHO and herein referred to as a compound of Formula (lc-1) and compound of Formula (lc-2) respectively. A compound of Formula (Ιοί) or compound of Formula (lc-2) can be treated in the presence of afluorinating agent such as for example ©Deoxofluor ([Bis(2-methoxyethyl)amino]sulfur trifluoride) or (diethylamino)sulfur trifluoride in a suitable solvent such as, for example DCM, and stirring the reaction mixture at rt. In Reaction Schemes 5a and 5b, all variables are defined as in Formula (I) and R5c, R6c, R3c, and R4c include the residues indicated in the scope of Formula (I) as R5, R6, R3 ,R4 as well as their possible precursors, and R9c and R10c, when present, include the residues indicated in the scope of Formula (I) as substituents in R1 as well as their possible precursors, wherein one of R5c, R6c, R3c, R4c, and R9cand R10cwhen present, is C^alkyl-OH or C1.3alkyl-CHO and in (R10d)n, n = 0-4.
Experimental procedure 6 [0077] Alternatively, final compounds according to Formula (la) and Formula (lb) can be prepared by manipulation of a suitable precursor of Formula (Id-1) or of Formula (Id-2) respectively, wherein Y is N, CH orCR9d, bearing one or more functional groups, R5d, R6a, R9a and R10d, which can be converted to the residues R5, R6 and the substituents of R1 as defined in Formula (I) by means of simple reactions known to the person skilled in the art, such as for example by reduction of a double bond to the corresponding saturated form, for example by means of catalytic hydrogenation. In Reaction Schemes 6a and 6b, all variables are defined as in Formula (I) and R5d and R6d, include the residues indicated in the scope as R5, R6 as well as their possible precursors, and R9d and R10d when present include the substituents of R1 as well as their possible precursors. The person skilled in the art will recognize that suitable reaction conditions should be chosen for different R5d, R6d, R9a and R10d combinations to avoid undesired side reactions and in (R10d)n, n = 0-4.
Reaction Scheme 6a
R part inn Srhnmn fib
Experimental procedure 7 [0078] Alternatively, final compounds according to Formula (Id) can be prepared by means of an oxidation reaction of a compound of Formula (I) in the presence of an oxidant, such as for example 3-chloroperoxybenzoic acid and in a suitable solvent. In Reaction Scheme 7, all variables are defined as in Formula (I).
Reaction Scheme 7
Experimental procedure 8 [0079] Alternatively, final compounds according to Formula (I) can be prepared by intramolecular amidation starting from a compound of Formula (VI). Typically, amidation conditions can be applied, such as stirring the starting materials, dissolved in a suitable solvent, such as DMF, in the presence of a coupling agent, such as HATU (2-(7-aza-1/-/-benzo-triazole-1 -yl)-1,1,3,3-tetramethyluronium hexafluorophosphate) and in the presence of a base, such as TEA (triethyl-amine). In Reaction Scheme 8, all variables are defined as in Formula (I).
Reaction Scheme 8
Experimental procedure 9 [0080] Alternatively, final compounds according to Formula (I) can be prepared in one pot starting from a compound of Formula (II). First, a reaction of nucleophilic substitution of a compound of Formula (II) with an appropriate (hetero)aryl halide of Formula (III) where X is halo in the presence of a base such as for example sodium hydride in a suitable solvent such as for example DMF, followed by an intramolecular peptide type coupling of compound of Formula (VI) applying typical peptide type coupling conditions. Typically, peptide coupling conditions can be applied, such as stirring the starting materials, dissolved in a suitable solvent, such as DMF, in the presence of a peptide coupling agent, such as HATU and in the presence of a base, such as TEA. In Reaction Scheme 9, all variables are defined as in Formula (I).
Reaction
Scheme 9 [0081] Alternatively, final compounds according to Formula (I) can be prepared in one pot starting from a compound of Formula (II). First by a coupling reaction of a compound of Formula (I I) with an appropriate heteroaryl halide of Formula (III) where X is halo in the presence of a palladium catalyst, such as tetrakis(triphenyiphosphine)pailadium(0), in the presence of a ligand, such as 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene, in the presence of a base, such as Cs2C03 and in a suitable solvent, such as 1,4-dioxane, under suitable reaction conditions, such as at a convenient temperature, typically ranging between 100 °C and 140 °C, fora period of time to ensure the completion of the reaction, followed by an intramolecular peptide type coupling of compound of Formula (VI) applying typical peptide type coupling conditions. Typically, peptide coupling conditions can be applied, such as stirring the starting materials, dissolved in a suitable solvent, such as DMF, in the presence of a peptide coupling agent, such as HATU and in the presence of a base, such as TEA. In Reaction Scheme 9, all variables are defined as in Formula (I). B. Preparation of the intermediate compounds
Experimental procedure 10 [0082] Intermediate compound according to Formula (II) (Reaction Scheme 10a) can be prepared following art known procedures, such as by subjecting an intermediate compound of Formula (Va) to a Suzuki type coupling reaction under conditions known to a skilled person. Such conditions include for example, reacting the intermediate compound of Formula (Va) with a suitable boron species, such as for example a boronic acid or a boronate ester, for example as described in Experimental procedure 2 hereinbefore, in the presence of a palladium catalyst, such as tetrakis(triphenyl- phosphine)palladium(O) or an alternative catalyst system prepared in situ from Pd(OAc)2 and PPh3, a suitable base, such as Na2C03, K2COs, NaHC03 and K3P04, and in a suitable solvent, such as 1,4-dioxane, or a mixture of DME and water. Degassing the reaction mixture with an inert gas, such as N2 or argon, and heating the reaction mixture to high temperatures, such as reflux temperature, in particular 80 °C, may enhance the reaction outcome. In Reaction Scheme 10a, all variables are defined as in Formula (I).
Reaction Scheme 10a
[0083] A skilled person can envisage that the reaction under Reaction Scheme 10a can also be performed under similar conditions, when the compound of Formula (Va) bears a bromo group in place of a iodo group. Such a reaction can be represented as in Reaction Scheme 10b, wherein the compound of Formula (V), wherein R2a is halo, in particular bromo or iodo and all other variables are as defined in Formula (I), undergoes a Suzuki type coupling as described hereinbefore.
Reaction Scheme 10b
In particular aspect, the invention therefore relates to an intermediate compound of Formula (V), wherein R2a is halo, in particular Br or I
In a particular embodiment, the invention relates to an intermediate compound of Formula (V’), wherein R2a is halo, in particular, Br (referred to herein as compound (1-13a) or I (referred to herein as compound (1-13))
Experimental procedure 11 [0084] Intermediate compound of Formula (Va) or of Formula (V) can be prepared by removal of the protecting group, for example a Boc group, in an intermediate of Formula (Via) or of Formula (Vlb), respectively, for example in the presence of acidic media, such as hydrochloric acid, in an inert solvent such as 1,4-dioxane or acetonitrile or ethyl acetate (EtOAc), under suitable reaction conditions, such as at a convenient temperature, such as from 15 to 80 °C, typically 80 °C or from 15-30 °C depending on the solvent system, for a period of time to ensure the completion of the reaction followed by treatment with a base such as Na2C03, K2C03 or NaHC03, under suitable reaction conditions, such as at a convenient temperature, typically ranging between 0 °C and 40 °C, in particular from 15 to 30 °C, for a period of time to ensure the completion of the reaction. In Reaction Schemes 11a and 11b, R2a is halo, in particular bromo or iodo, R7 is C^alkyl, PG is a protecting group, for example Boc, and all other variables are defined as in Formula (I).
Reaction Scheme 1 la
Reaction Scheme lib
Experimental procedure 12 [0085] Intermediate compound of Formula (Via) or (Vlb) wherein R7 is C^alkyl and PG is a protecting group, for example Boc, can be prepared by a Mitsunobu type reaction between an intermediate compound of Formula (Vila) or (VII) respectively, and an appropriate alcohol of Formula (VIII), in the presence of a suitable triarylphosphine, such as triphenylphosphine typically 1.5 equivalents, or a suitable trialkylphosphine, and a suitable dialkyl azodicarboxylate reagent, such as di-ferf-butyl azodicarboxylate or diethyl azodicarboxylate typically 1.5 equivalents, in a suitable inert solvent, such as tetrahydrofuran (THF), under suitable reaction conditions, such as at a convenient temperature, typically ranging 0 °C and rt, e.g. 20 °C, for a period of time to ensure the completion of the reaction. An intermediate compound of Formula (VIII) can be obtained commercially or synthesized according to literature procedures.
[0086] Intermediate compound of Formula (Vila) wherein R7 is C1.4alkyl, can be prepared via a reaction of halogenation of intermediate of Formula (IX) with a halogenating reagent such as N-iodosuccinimide, in an inert solvent such as dichloromethane, under suitable reaction conditions, such as at a convenient temperature, typically rt, for a period of time to ensure the completion of the reaction. Intermediate compound of Formula (VII), wherein R7 is methyl and R2a is bromo, can be obtained commercially and is a particularly preferred material for use in the synthesis, including large scale, of a variety of final compounds of Formula (I) according to the general procedures described herein. An intermediate compound of Formula (IX) can be obtained commercially or synthesized according to literature procedures.
[0087] In Reaction Scheme 12a and 12b, R2a is halo, in particular bromo or iodo, R7 is C^alkyl, PG is a protecting group, such as for example Boc, and all other variables are defined as in Formula (I).
Reaction Scheme 12a
Reaction Scheme 12b
I
Experimental procedure 13 [0088] Intermediate compound of Formula (IVb) can be prepared via a reaction of boronic ester or boronic acid formation starting from an intermediate of Formula (IVa) with a trans metallating agent such as for example BuLi or a Grignard reagent, a particular example of reagents includes isopropylmagnesium chloride lithium chloride complex solution and a boron species such as 2-/'sopropoxy-4,4,5,5- tetramethyl-1,3,2-dioxaborolane, in an inert solvent such as anhydrous TFIF, under suitable reaction conditions, such as at a convenient temperature, typically -25°C, for a period of time to ensure the completion of the reaction. Depending on reaction conditions, boronic ester or boronic acid are obtained. In Reaction Scheme 13, R7a and R8a are FI or 01-4 alkyl or R7a and R8a are taken together to form for example a bivalent 1 radical of formula-CF^CF^-, -CFI2CFI2CFI2- or -C(CFi3)2C(CFl3)2-, and all other variables are defined as in Formula (I).
Reaction Scheme 13
Experimental procedure 14 [0089] Intermediate compound of Formula (IVa) can be prepared via a reaction of halogenation of an intermediate of Formula (X) with a halogenating reagent such as iodine, in the presence of ammonium cerium(IV) nitrate and in an inert solvent such as acetonitrile, under suitable reaction conditions, such as at a convenient temperature, typically 70 °C, fora period of time to ensure the completion of the reaction. In an analogous manner, intermediate compound of Formula (Va) can be prepared from intermediate of Formula (XI). In Reaction Schemes 14a and 14b, all variables are defined as in Formula (I).
Reaction Scheme 14a
Rftar.tinn Srhemp 14h
Experimental procedure 15 [0090] Intermediate compound of Formula (X) can be prepared by a coupling reaction of an intermediate compound of Formula (XI) with an appropriate aryl/heteroaryl halide of Formula (III) where X is halo with a suitable copper(l) catalyst such as copper(l) iodide, in the presence of a ligand, such as Λ/,Λ/’-dimethylethylenediamine, in the presence of a base, such as Na2C03, in a suitable solvent, such as toluene, under suitable reaction conditions, such as at a convenient temperature, typically ranging between 100 °C and 140 °C, fora period of time to ensure the completion of the reaction. In an analogous manner, intermediate compound of Formula (IV) can be prepared from intermediate of Formula (V). An intermediate compound of Formula (III) can be obtained commercially. In Reaction Schemes 15a and 15b, all variables are defined as in Formula (I) and R2a is halo, in particular bromo or iodo.
Reaction Scheme 15a
Reaction Scheme 15b
Experimental procedure 16 [0091] Intermediate compound of Formula (XI) can be prepared by removal of the protecting group in an intermediate of Formula (XII), for example in the presence of acidic media, such as hydrochloric acid, in an inert solvent such as 1,4-dioxane, under suitable reaction conditions, such as at a convenient temperature, typically 80 °C, for a period of time to ensure the completion of the reaction followed by treatment with a base, such as Na2COs or NaHC03, under suitable reaction conditions, such as at a convenient temperature, typically ranging between 0 °C and 40 °C, for a period of time to ensure the completion of the reaction. In an analogous manner, intermediate compound of Formula (V) can be prepared from intermediate of Formula (Vlb). In Reaction Schemes 16a and 16b, R2a is halo, in particular bromo or iodo, R7 is C1.4alkyl, PG is a protecting group and all other variables are defined as in Formula (I).
Reaction Scheme 16a
Reaction Scheme 16b
Experimental procedure 17 [0092] Intermediate compound of Formula (XII) wherein R7 is C^alkyl and PG is a protecting group, can be prepared by a Mitsunobu type reaction between a compound of Formula (IX) and an appropriate alcohol of Formula (VIII), in the presence of a suitable triarylphosphine, such as triphenylphosphine, or a suitable trialkylphosphine, and a suitable dialkyl azodicarboxylate reagent, such as di-fert-butyl azodicarboxylate, in a suitable inert solvent, such as THF, under suitable reaction conditions, such as at a convenient temperature, typically rt, for a period of time to ensure the completion of the reaction. Intermediate compounds of Formula (IX) and of Formula (VIII) can be obtained commercially or synthesized according to literature procedures. In Reaction Scheme 17, R7isC1.4alkyl, PG is a protecting group and all other variables are defined as in Formula (I).
Reaction Scheme 17
Experimental procedure 18 [0093] Intermediate compound of Formula (IX) wherein R7 is C1_4alkyl can be obtained by esterification of the commercially available intermediate compound of Formula (XIII), by methods known to the person skilled in the art, or may be commercially available. The reaction can be performed for example in the presence of an acidic agent, such as sulfuric acid, and an alcohol, such as EtOH, in a suitable solvent, such as EtOH, under suitable reaction conditions, such as at a convenient temperature, typically between 80 °C and 100 °C, for a period of time to ensure the completion of the reaction. In Reaction Scheme 18, R7 is C^alkyl.
Reaction Scheme 18
Experimental procedure 19 [0094] Intermediate compound of Formula (XI) wherein R3 is H and R4 is CF3 herein referred to as compounds of Formula (Xla) can be prepared by hydrogenation of an intermediate of Formula (XIV) followed by one pot intramolecular cyclization, in the presence of a hydrogenation catalyst, such as Pd/C (palladium on carbon), under hydrogen atmosphere generated by using ammonium formate, in an inert solvent such as MeOH, under suitable reaction conditions, such as at a convenient temperature, typically 70 °C, for a period of time to ensure the completion of the reaction. In Reaction Scheme 19, R7 is C^alkyl.
Reaction Scheme 19
Experimental procedure 20 [0095] Intermediate compound of Formula (XIV) wherein R7 is C1.4alkyl, can be prepared by an intermolecular reaction between an appropriate hydrazine of Formula (XV), in the presence of a suitable ketoester of Formula (XVI), such as ethyl pyruvate, in a suitable inert solvent, such as EtOH, under suitable reaction conditions, such as at a convenient temperature, typically rt, fora period of time to ensure the completion of the reaction. Intermediate compound of Formula (XVI) can be obtained commercially or synthesized according to literature procedures. In Reaction Scheme 20, R7 is C1_4alkyl.
Reaction
Scheme 20
Experimental procedure 21 [0096] Intermediate compound of Formula (XV) can be prepared by a reaction of deprotection of a compound of Formula (XVI) following art known procedures. A compound of Formula (XV) can be obtained by removal of the protecting group such as for example a Boc protecting group in the compound of Formula (XVI), in the presence of acidic media, such as hydrochloric acid, in an inert solvent such as MeOH, under suitable reaction conditions, such as at a convenient temperature, typically rt, for a period of time to ensure the completion of the reaction.
[0097] Intermediate compound of Formula (XVI) can be obtained by addition of a protected hydrazine of Formula (XVIII) to 3,3,3-trifluoro-1-nitro-prop-1-ene (XVII) (prepared as described in J. Fluorine Chem. 2008,767-774). In Reaction Scheme 21, PG is a protecting group, for example BOC.
Reaction Scheme 21
[0098] In order to obtain the HCI salt forms of the compounds, several procedures known to those skilled in the art can be used. In a typical procedure, for example, the free base can be dissolved in DIPE or Et20 and subsequently, a 6N HCI solution in 2-propanol, 4N HCI solution in dioxane, or a 1N HCI solution in Et20 can be added dropwise. The mixture typically is stirred for 10 minutes after which the product can be filtered off. The HCI salt usually is dried in vacuo. It will be appreciated by those skilled in the art that in the processes described above the functional groups of intermediate compounds may need to be blocked by protecting groups. In case the functional groups of intermediate compounds were blocked by protecting groups, they can be deprotected after a reaction step.
Pharmacology [0099] The compounds provided in this invention are negative allosteric modulators (NAMs) of metabotropic glutamate receptors, in particular they are negative allosteric modulators of mGluR2. The compounds of the present invention do not appear to bind to the glutamate recognition site, the orthosteric ligand site, but instead to an allosteric site within the seven transmembrane region of the receptor. In the presence of glutamate, the compounds of this invention decrease the mGluR2 response. The compounds provided in this invention are expected to have their effect at mGluR2 by virtue of their ability to decrease the response of such receptors to glutamate, attenuating the response of the receptor.
[0100] As used herein, the term "treatment" is intended to refer to all processes, wherein there may be a slowing, interrupting, arresting or stopping of the progression of a disease or an alleviation of symptoms, but does not necessarily indicate a total elimination of all symptoms.
[0101] Hence, the present invention relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof for use as a medicament.
[0102] The invention also relates to the use of a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, or a pharmaceutical composition according to the invention for the manufacture of a medicament.
[0103] The invention also relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, or a pharmaceutical composition according to the invention for use in the treatment or prevention of, in particular treatment of, a condition in a mammal, including a human, the treatment or prevention of which is affected or facilitated by the neuromodulatory effect of allosteric modulators of mGluR2, in particular negative allosteric modulators thereof.
[0104] The present invention also relates to the use of a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, or a pharmaceutical composition according to the invention for the manufacture of a medicament for the treatment or prevention of, in particular treatment of, a condition in a mammal, including a human, the treatment or prevention of which is affected or facilitated by the neuromodulatory effect of allosteric modulators of mGluR2, in particular negative allosteric modulators thereof.
[0105] The present invention also relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, or a pharmaceutical composition according to the invention for use in the treatment, prevention, amelioration, control or reduction of the risk of various neurological and psychiatric disorders associated with glutamate dysfunction in a mammal, including a human, the treatment or prevention of which is affected or facilitated by the neuromodulatory effect of negative allosteric modulators of mGluR2.
[0106] Also, the present invention relates to the use of a compound according to the general Formula (I), or a stere-oisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, ora pharmaceutical composition according to the invention for the manufacture of a medicament for treating, preventing, ameliorating, controlling or reducing the risk of various neurological and psychiatric disorders associated with glutamate dysfunction in a mammal, including a human, the treatment or prevention of which is affected or facilitated by the neuromodulatory effect of negative allosteric modulators of mGluR2.
[0107] In particular, the neurological and psychiatric disorders associated with glutamate dysfunction, include one or more of the following central nervous system conditions or diseases: mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder.
[0108] In particular, the central nervous system disorder is a psychotic disorder selected from the group of schizophrenia (in particular in antipsychotic-stabilized patients), schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, and substance-induced psychotic disorder.
[0109] In particular, the central nervous system disorder is a substance-related disorder selected from the group of alcohol dependence, alcohol abuse, amphetamine dependence, amphetamine abuse, caffeine dependence, caffeine abuse, cannabis dependence, cannabis abuse, cocaine dependence, cocaine abuse, hallucinogen dependence, hallucinogen abuse, nicotine dependence, nicotine abuse, opioid dependence, opioid abuse, phencyclidine dependence, and phencyclidine abuse.
[0110] In particular, the central nervous system disorder is a mood disorder selected from the group of majordepressive disorder, depression, treatment resistant depression, dysthymic disorder, cyclothymic disorder, and substance-induced mood disorder.
[0111] In particular, the central nervous system disorder is a disorder usually first diagnosed in infancy, childhood, or adolescence selected from mental retardation, learning disorder, motorskills disorder, communication disorder, attention-deficit and disruptive behaviour disorders (such as Attention-Deficit/Hyperactivity Disorder (ADHD)). An additional disorder usually first diagnosed in infancy, childhood, or adolescence is autistic disorder.
[0112] In particular, the central nervous system disorder is a cognitive disorder selected from the group of dementia, in particular, dementia of the Alzheimer’s type, vascular dementia, dementia due to HIV disease, dementia due to head trauma, dementia due to Parkinson’s disease, dementia due to Huntington’s disease, dementia due to Pick’s disease, dementia due to Creutzfeldt-Jakob disease, and substance-induced persisting dementia.
[0113] In particular, the central nervous system disorder is an amnestic disorder, such as substance-induced persisting amnestic disorder.
[0114] As already mentioned hereinabove, the term "treatment" does not necessarily indicate a total elimination of all symptoms, but may also refer to symptomatic treatment in any of the disorders mentioned above. In particular, symptoms that may be treated include but are not limited to, memory impairment in particular in dementia or in major depressive disorder, age-related cognitive decline, mild cognitive impairment, and depressive symptoms.
[0115] Of the disorders mentioned above, the treatment of dementia, majordepressive disorder, depression, treatment resistant depression, attention-deficit/hyperactivity disorder and schizophrenia, in particular in antipsychotic-stabilized patients, are of particular importance.
[0116] The fourth edition of the Diagnostic &amp; Statistical Manual of Mental Disorders (DSM-IV) of the American Psychiatric Association provides a diagnostic tool for the identification of the disorders described herein. The person skilled in the art will recognize that alternative nomenclatures, nosologies, and classification systems for neurological and psychiatric disorders described herein exist, and that these evolve with medical and scientific progresses.
[0117] A skilled person will be familiar with alternative nomenclatures, nosologies, and classification systems for the diseases or conditions referred to herein. For example, the "American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric Association, 2013" (DSM-5™) utilizes terms such as depressive disorders, in particular, majordepressive disorder, persistent depressive disorder (dysthymia), substance-medication-induced depressive disorder; neurocognitive disorders (NCDs) (both major and mild), in particular, neurocognitive disorders due to Alzheimer’s disease, vascular NCD (such as vascular NCD present with multiple infarctions), NCD due to HIV infection, NCD due to traumatic brain injury (TBI), NCD due to Parkinson’s disease, NCD due to Huntington’s disease, frontotemporal NCD, NCD due to prion disease, and substance/medication-induced NCD; neurodevelopmental disorders, in particular, intellectual disability, specific learning disorder, neurodevelopmental motor disorder, communication disorder, and attention-deficit/hyperactivity disorder (ADHD); substance-related disorders and addictive disorders, in particular, alcohol use disorder, amphetamine use disorder, cannabis use disorder, cocaine use disorder, other hallucinogen use disorder, tobacco use disorder, opiod use disorder, and phencyclidine use disorder; schizophrenia spectrum and other psychotic disorders, in particular, schizophrenia, schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, substance/medication-induced psychotic disorder; somatic symptom disorders; hypersomnolence disorder; and cyclothymic disorder (which under DSM-5™ falls under the bipolar and related disorders category). Such terms may be used by the skilled person as an alternative nomenclature for some of the diseases or conditions referred to herein. An additional neurodevelopmental disorder includes autism spectrum disorder (ASD), which encompasses according to the DSM-5™, disorders previously known by the terms early infantile autism, childhood autism, Kanner’s autism, high-functioning autism, atypical autism, pervasive developmental disorder not otherwise specified, childhood disintegrative disorder, and Asperger’s disorder. In particular, the disorder is autism. Specifiers associated with ASD include those where the individual has a genetic disorder, such as in Rett syndrome or Fragile X syndrome.
[0118] Therefore, the invention also relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, for use in the treatment of any one of the diseases mentioned hereinbefore.
[0119] The invention also relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, for use in treating any one of the diseases mentioned hereinbefore.
[0120] The invention also relates to a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, for the treatment or prevention, in particular treatment, of any one of the diseases mentioned hereinbefore.
[0121] The invention also relates to the use of a compound according to the general Formula (I), or a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or a solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, for the manufacture of a medicamentforthe treatment or prevention of any one of the disease conditions mentioned hereinbefore.
[0122] The compounds of the present invention can be administered to mammals, preferably humans, for the treatment or prevention of any one of the diseases mentioned hereinbefore.
[0123] In view of the utility of the compounds of Formula (I), there is provided a method of treating warm-blooded animals, including humans, suffering from any one of the diseases mentioned hereinbefore, and a method of preventing in warm-blooded animals, including humans, any one of the diseases mentioned hereinbefore.
[0124] Said methods comprise the administration, i.e. the systemic or topical administration, preferably oral administration, of a therapeutically effective amount of a compound of Formula (I), a stereoisomeric form thereof, or an N-oxide thereof, or a pharmaceutically acceptable salt or solvate thereof, in particular, a compound of Formula (I) or a stereoisomeric form thereof, or a pharmaceutically acceptable salt or a solvate thereof, to warm-blooded animals, including humans.
[0125] Therefore, the invention also relates to a method for the prevention and/or treatment of any one of the diseases mentioned hereinbefore comprising administering a therapeutically effective amount of a compound according to the invention to a subject in need thereof.
[0126] One skilled in the art will recognize that a therapeutically effective amount of the NAMs of the present invention is the amount sufficient to modulate the activity of the mGluR2 and that this amount varies inter alia, depending on the type of disease, the concentration of the compound in the therapeutic formulation, and the condition of the patient. Generally, an amount of NAM to be administered as a therapeutic agent for treating diseases in which modulation of the mGluR2 is beneficial, such as the disorders described herein, will be determined on a case by case by an attending physician.
[0127] Generally, a suitable dose is one that results in a concentration of the NAM at the treatment site in the range of 0.5 nM to 200 μΜ, and more usually 5 nM to 50 μΜ. To obtain these treatment concentrations, a patient in need of treatment likely will be administered an effective therapeutic daily amount of about 0.01 mg/kg to about 50 mg/kg body weight, preferably from about 0.01 mg/kg to about 25 mg/kg body weight, more preferably from about 0.01 mg/kg to about 10 mg/kg body weight, more preferably from about 0.01 mg/kg to about 2.5 mg/kg body weight, even more preferably from about 0.05 mg/kg to about 1 mg/kg body weight, more preferably from about 0.1 to about 0.5 mg/kg body weight. The amount of a compound according to the present invention, also referred to here as the active ingredient, which is required to achieve a therapeutically effect will, of course vary on case-by-case basis, vary with the particular compound, the route of administration, the age and condition of the recipient, and the particular disorder or disease being treated. A method of treatment may also include administering the active ingredient on a regimen of between one and four intakes per day. In these methods of treatment the compounds according to the invention are preferably formulated prior to admission. As described herein below, suitable pharmaceutical formulations are prepared by known procedures using well known and readily available ingredients.
[0128] The compounds of the present invention may be utilized in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of Formula (I) or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone. Examples of such combinations include the compounds of the invention in combination with antipsychotic(s), NMDA receptor antagonists (e.g. memantine), NR2B antagonists, acetylcholinesterase inhibitors (e.g. donepezil, galan-tamine, physostigmine and rivastigmine) and/or antidepressant neurotransmitter reuptake inhibitors. Particular combinations include the compounds of the invention in combination with antipsychotics, or the compounds of the invention in combination with memantine and/or NR2B antagonists.
Pharmaceutical compositions [0129] The present invention also provides compositions for preventing or treating diseases in which modulation of the mGluR2 receptor is beneficial, such as the disorders described herein. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical composition. Accordingly, the present invention also relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier or diluent and, as active ingredient, a therapeutically effective amount of a compound according to the invention, in particular a compound according to Formula (I), an N-oxide, a pharmaceutically acceptable salt thereof, a solvate thereof or a stereochemically isomeric form thereof, more in particular, a compound according to Formula (I), a pharmaceutically acceptable salt thereof, a solvate thereof ora stereochemically isomeric form thereof. The carrier or diluent must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
[0130] The compounds according to the invention, in particular the compounds according to Formula (I), the N-oxides thereof, the pharmaceutically acceptable salts thereof, the solvates and the stereochemically isomeric forms thereof, more in particular the compounds according to Formula (I), the pharmaceutically acceptable salts thereof, the solvates and the stereochemically isomeric forms thereof, or any subgroup or combination thereof may be formulated into various pharmaceutical forms for administration purposes. As appropriate compositions there may be cited all compositions usually employed for systemically administering drugs.
[0131] The pharmaceutical compositions of this invention may be prepared by any methods well known in the art of pharmacy, for example, using methods such as those described in Gennaro etal. Remington’s Pharmaceutical Sciences (18th ed., Mack Publishing Company, 1990, see especially Part 8: Pharmaceutical preparations and their Manufacture). To prepare the pharmaceutical compositions of this invention, a therapeutically effective amount of the particular compound, optionally in salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier or diluent, which carrier or diluent may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirable in unitary dosage form suitable, in particular, for oral, topical, rectal or percutaneous administration, by parenteral injection or by inhalation. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as, for example, suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as, for example, starches, sugars, kaolin, diluents, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets. Because of the ease in administration, oral administration is preferred, and tablets and capsules represent the most advantageous oral dosage unit forms in which case solid pharmaceutical carriers are obviously employed. For parenteral compositions, the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, surfactants, to aid solubility, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution ora mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations that are intended to be converted, shortly before use, to liquid form preparations. In the compositions suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin. Said additives may facilitate the administration to the skin and/or may be helpful for preparing the desired compositions. These compositions may be administered in various ways, e.g., as a transdermal patch, as a spot-on, as an ointment.
[0132] It is especially advantageous to formulate the aforementioned pharmaceutical compositions in unit dosage form for ease of administration and uniformity of dosage. Unit dosage form as used herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such unit dosage forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, suppositories, injectable solutions or suspensions and the like, teaspoonfuls, tablespoonfuls, and segregated multiples thereof.
[0133] Since the compounds according to the invention are orally administrable compounds, pharmaceutical compositions comprising aid compounds for oral administration are especially advantageous.
[0134] In order to enhance the solubility and/or the stability of the compounds of Formula (I) in pharmaceutical compositions, it can be advantageous to employ α-, β- or γ-cyclodextrins or their derivatives, in particular hydroxyalkyl substituted cyclodextrins, e.g. 2-hydroxypropyl-p-cyclodextrin or sulfobutyl-p-cyclodextrin. Also co-solvents such as al cohols may improve the solubility and/or the stability of the compounds according to the invention in pharmaceutical compositions.
[0135] The exact dosage and frequency of administration depends on the particular compound of formula (I) used, the particular condition being treated, the severity of the condition being treated, the age, weight, sex, extent of disorder and general physical condition of the particular patient as well as other medication the individual may be taking, as is well known to those skilled in the art. Furthermore, it is evident that said effective daily amount may be lowered or increased depending on the response of the treated subject and/or depending on the evaluation of the physician prescribing the compounds of the instant invention.
[0136] Depending on the mode of administration, the pharmaceutical composition will comprise from 0.05 to 99 % by weight, preferably from 0.1 to 70 % by weight, more preferably from 0.1 to 50 % by weight of the active ingredient, and, from 1 to 99.95 % by weight, preferably from 30 to 99.9 % by weight, more preferably from 50 to 99.9 % by weight of a pharmaceutically acceptable carrier, all percentages being based on the total weight of the composition.
[0137] The amount of a compound of Formula (I) that can be combined with a carrier material to produce a single dosage form will vary depending upon the disease treated, the mammalian species, and the particular mode of administration. Flowever, as a general guide, suitable unit doses for the compounds of the present invention can, for example, preferably contain between 0.1 mg to about 1000 mg of the active compound. A preferred unit dose is between 1 mg to about 500 mg. A more preferred unit dose is between 1 mg to about 300 mg. Even more preferred unit dose is between 1 mg to about 100 mg. Such unit doses can be administered more than once a day, for example, 2, 3, 4, 5 or 6 times a day, but preferably 1 or 2 times per day, so that the total dosage for a 70 kg adult is in the range of 0.001 to about 15 mg per kg weight of subject per administration. A preferred dosage is 0.01 to about 1.5 mg per kg weight of subject per administration, and such therapy can extend for a number of weeks or months, and in some cases, years. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs that have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those of skill in the area.
[0138] A typical dosage can be one 1 mg to about 100 mg tablet or 1 mg to about 300 mg taken once a day, or, multiple times per day, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient. The time-release effect can be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
[0139] It can be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art. Further, it is noted that the clinician or treating physician will know how and when to start, interrupt, adjust, or terminate therapy in conjunction with individual patient response.
[0140] As already mentioned, the invention also relates to a pharmaceutical composition comprising the compounds according to the invention and one or more other drugs for use as a medicament or for use in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of Formula (I) or the other drugs may have utility. The use of such a composition for the manufacture of a medicament as well as the use of such a composition for the manufacture of a medicament in the treatment, prevention, control, amelioration or reduction of risk of diseases or conditions for which compounds of Formula (I) or the other drugs may have utility are also contemplated. The present invention also relates to a combination of a compound according to the present invention and an additional drug selected from the group of antipsychotics; NMDA receptor antagonists (e.g. memantine); NR2B antagonists; acetylcholinesterase inhibitors (e.g. donepezil, galantamine, physostigmine and rivastigmine) and/or antidepressant neurotransmitter reuptake inhibitors. In particular, the present invention also relates to a combination of a compound according to the present invention and antipsychotic(s), or to a combination of a compound according to the present invention and memantine and/or an NR2B antagonist. The present invention also relates to such a combination for use as a medicine. The present invention also relates to a product comprising (a) a compound according to the present invention, an N-oxide thereof, a pharmaceutically acceptable salt thereof or a solvate thereof, in particular, a pharmaceutically acceptable salt thereof or a solvate thereof, and (b) an additional component selected from antipsychotics, NMDA receptor antagonists (e.g. memantine), NR2B antagonists, acetylcholinesterase inhibitors and/or antidepressant neurotransmitter reuptake inhibitor(s), as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of a condition in a mammal, including a human, the treatment or prevention of which is affected or facilitated by the neuromodulatory effect of mGluR2 allosteric modulators, in particular negative mGluR2 allosteric modulators. More in particular the additional component (b) is selected from antipsychotic(s) or memantine and/or an NR2B antagonist. The different drugs of such a combination or product may be combined in a single preparation together with pharmaceutically acceptable carriers or diluents, or they may each be present in a separate preparation together with pharmaceutically acceptable carriers or diluents.
[0141] The following examples are intended to illustrate but not to limit the scope of the present invention.
Chemistry [0142] Several methods for preparing the compounds of this invention are illustrated in the following Examples. Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification.
[0143] Hereinafter, "Boc" or"BOC" means fert-Butyloxycarbonyl; "Cl" means chemical ionisation; "DAD" means diode-array detector; "THF" means tetrahydrofuran; "TEA" means triethylamine; "DIPE" meansdiisopropylether; "DMF" means /\/,/\/-dimethylformamide; "Et20" means diethylether; "EtOAc" means ethyl acetate; "DCM" means dichloromethane; "DMSO" means dimethylsulfoxide; "L" means liter; "LRMS" means low-resolution mass spectrometry/spectra; "HATU" means 2-(7-aza-1/-/-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate; "HPLC" means high performance liquid chromatography; "HRMS" means high-resolution mass spectrometry/spectra; "mL" or "ml" means milliliter; "NH4Ac" means ammonium acetate; "EtOH" means ethanol; "ES" means electrospray; "iPrOH" means isopropanol; "iPrNH2" means isopropylamine; "MeOH" means methanol; "eq" means equivalent(s); "RP" means Reverse Phase; "rt" means room temperature; "M.p." means melting point; "min" means minutes; "h" means hour(s); "s" means second(s); "TOF" means time of flight; "QTOF" means Quadrupole-Time of Flight; "sat." means saturated; "SFC" means supercritical fluid chromatography; "sol." means solution.
[0144] Microwave assisted reactions were performed in a single-mode reactor: Initiator™ Sixty EXP microwave reactor (Biotage AB), or in a multimode reactor: MicroSYNTH Labstation (Milestone, Inc.).
[0145] Thin layer chromatography (TLC) was carried out on silica gel 60 F254 plates (Merck) using reagent grade solvents. Open column chromatography was performed on silica gel, particle size 60 A, mesh = 230-400 (Merck) using standard techniques.
[0146] Automated flash column chromatography was performed using ready-to-connect cartridges , on irregular silica gel, particle size 15-40 μηι (normal phase disposable flash columns) on different flash systems: either a SPOT or LAFLASH systems from Armen Instrument, or PuriFlash® 430evo systems from Interchim, or 971-FP systems from Agilent, or Isolera 1 SV systems from Biotage.
[0147] Nuclear Magnetic Resonance (NMR): Fora number of compounds, 1H N MR spectra were recorded either on a BrukerAvance III, on a BrukerDPX-400oron a Bruker AV-500 spectrometer with standard pulse sequences, operating at 400 MHz and 500 MHz, respectively. Chemical shifts (δ) are reported in parts per million (ppm) downfield from tetramethylsilane (TMS), which was used as internal standard.
Synthesis of Intermediate Compounds
Intermediate 1 (1-1) 2H-Pyrazole-3-carboxylic acid ethyl ester (1-1) [0148]
[0149] Sulfuric acid (10 mL, 187.6 mmol) was added to a solution of 1-H-pyrazole-3-carboxylic acid (1.93 g, 17.22 mmol) in EtOH (20 mL). The mixture was stirred at 90 °C for 15 h. Then it was allowed to cool to rt and the solvents were evaporated in vacuo. The residue was poured into water and the solution basified with K2C03 and extracted with EtOAc. The organic layerwas separated, dried (MgS04), filtered and the solvent evaporated in vacuo to yield intermediate compound 1-1 as a white solid (2.28 g, 93 % purity, 94%) which was used in the following step without further purification.
Intermediate 2 (I-2) 4-lodo-2H-pyrazole-3-carboxylic acid ethyl ester (I-2) [0150]
[0151] Intermediate 1-1 (100 g, 0.68 mol), N-iodosuccinimide (213.5 g, 0.95 mol) were dissolved in DCM (2 L). The mixture was stirred at rt for 24 h. The mixture was treated with a sat. sol. of Na2S203 and a sat. sol. of Na2C03 and extracted with DCM. The organic layer was separated, dried (MgS04), filtered and the solvent evaporated in vacuo to yield intermediate compound I-2 as a white solid (160 g, 85%).
Intermediate 3 (I-3) (2R-Hydroxy-propyl)-carbamic acid ferf-butyl ester (I-3) [0152]
[0153] Di-ferf-butyl dicarbonate (58.1 g, 266.3 mmol) in DCM (50 mL) was added to a stirred solution of (R)-(-)-1-amino-2-propanol in DCM (50 mL) at 0 °C under nitrogen. The mixture was stirred at rt for 2 h. The mixture was diluted with cooled water and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo to yield intermediate I-3 as a colorless oil (47 g, quant.). The product was used in the next step without further purification.
Intermediate 4 (I-4) (2S-Hydroxy-propyl)-carbamic acid tert-butyl ester (I-4) [0154]
[0155] Intermediate compound I-4 was synthesized following a similar approach described for intermediate I-3. Starting from (S)-(-)-1-amino-2-propanol (3 mL, 38.1 mmol), intermediate compound I-4 was obtained as a colorless oil (6.13 g, 89% purity, 82%), that solidified upon standing at rt.
Intermediate 5 (I-5) (2-Hydroxy-propyl)-carbamic acid ferf-butyl ester (I-5) [0156]
[0157] Intermediate compound I-5 was synthesized following a similar approach described for intermediate I-3. Starting from 1-amino-2-propanol (3 mL, 38.1 mmol), intermediate compound I-5 was obtained as a colorless oil (6.69 g, 98%).
Intermediate 6 (I-6) 2-(2-ferf-Butoxycarbonylamino-1S-methyl-ethyl)-4-iodo-2H-pyrazole-3-carboxylic acid ethyl ester (I-6) [0158]
[0159] Di-ferf-butyl azodicarboxylate (4.67 g, 20.3 mmol) was added to a stirred solution of intermediate I-2 (3 g, 11.28 mmol), intermediate I-3 (4.44 g, 22.55 mmol) and triphenylphosphine (5.32 g, 20.3 mmol) in THF (56 mL) under nitrogen. The mixture was stirred at rt for 5 h. The solvent was evaporated in vacuo and the crude product was triturated with DIPE. The solid was filtered and the filtrate was evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 0/100 to 30/70). The desired fractions were collected and the solvents evaporated in vacuo to give intermediate compound I-6 as a colorless oil (4.9 g, 91 % purity, 93%).
Intermediate 7 (I-7) 2-(2-ferf-Butoxycarbonylamino-1R-methyl-ethyl)-4-iodo-2H-pyrazole-3-carboxylic acid ethyl ester (I-7) [0160]
[0161] Intermediate compound I-7 was synthesized following a similar approach described for intermediate I-6. Starting from intermediate I-2 (1.18 g, 4.44 mmol) and intermediate I-4 (1.75 g, 8.87 mmol), intermediate compound I-7 was obtained as a white solid as two fractions (790 mg, 41 %) and (900 mg, 86% purity, 41 %).
Intermediate 8 (I-8) 2-(2-ferf-Butoxycarbonylamino-1-methyl-ethyl)-4-iodo-2H-pyrazole-3-carboxylic acid ethyl ester (I-8) [0162]
[0163] Intermediate compound I-8 was synthesized following a similar approach described for intermediate I-6. Starting from intermediate I-2 (2.87 g, 10.79 mmol) and intermediate I-5 (3.78 g, 21.6 mmol), intermediate compound I-8 was obtained as a colorless oil (3.46 g, 75%).
Intermediate 9 (I-9) 2-(2-ferf-Butoxycarbonylamino-ethyl)-4-iodo-2H-pyrazole-3-carboxylic acid ethyl ester (I-9) [0164]
[0165] Intermediate compound I-9 was synthesized following a similar approach described for intermediate I-6. Starting from intermediate I-2 (3.18 g, 11.95 mmol) and /\/-(ferf-butoxycarbonyl)ethanolamine (3.78 g, 23.9 mmol), intermediate compound I-9 was obtained as a colorless oil (3.46 g, 75%).
Intermediate 10 (1-10) 2-(2-ferf-Butoxycarbonylamino-1 S-methyl-ethyl)-2H-pyrazole-3-carboxylic acid ethyl ester (1-10) [0166]
[0167] Intermediate compound 1-10 was synthesized following a similar approach described for intermediate i-6. Starting from intermediate 1-1 (25.82 g, 184.25 mmol) and intermediate I-3 (47.16 g, 239.5 mmol), intermediate compound 1- 10 was obtained as a yellow oil (123 g, quant) which was used in the following step without further purification.
Intermediate 11 (1-11) 2- (2-Amino-1 S-methyl-ethyl)-4-iodo-2H-pyrazole-3-carboxylic acid ethyl ester hydrochloride salt (1-11) [0168]
[0169] A 4M solution of HCI in 1,4-dioxane (10 mL, 40 mmol) was added to a solution of intermediate I-6 (4.2 g, 9.63 mmol) in acetonitrile (20 mL). The mixture was stirred at 80 °C for 2 h. The solvent was evaporated in vacuo to yield intermediate compound 1-11 (3.5 g, 97%).
Intermediate 12 (1-12) 2- (2-Amino-1 S-methyl-ethyl)-2H-pyrazole-3-carboxylic acid ethyl ester hydrochloride salt (1-12) [0170]
[0171] Intermediate compound 1-12 was synthesized following a similar approach described for intermediate 1-11. Starting from intermediate 1-10 (54.79 g, 184.25 mmol) and a 4M solution of HCI in 1,4-dioxane (415 mL, 1.66 mol), intermediate compound 1-12 was obtained as a white solid (32.5 g, 82% purity, 75%) which was used in the following step without further purification.
Intermediate 13 (1-13) 3- lodo-7S-methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-13) [0172]
[0173] Intermediate 1-11 as HCI salt (180 g, 350.4 mmol) was dissolved in a sat. sol. of NaHC03 (2 L). The mixture was stirred at rtfor 12 h. The mixture was diluted with water and extracted with DCM. The organic layers were separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. Then the residue was washed with ferf-butyl methyl ether to yield intermediate compound 1-13 (92 g, 90%), mp 182.6-186.1°C. 1H NMR (500 MHz, DMSO-d6) δ ppm 1.42 (d, J=6.65 Hz, 3 H) 3.26 - 3.35 (m, 1 H) 3.57 - 3.71 (m, 1 H) 4.44 - 4.60 (m, 1 H) 7.68 (s, 1 H) 8.26 (br. s., 1 H). LC-HRMS (ESI+) Calculated for C7H8IN30 (M+H)+: 277.9790, Found: miz 277.9796 (+0.6mDa), Rt = 0.76 min (Method 13, see table 2). [a] = +11.7° (589 nm, c 1.00 w/v%, CH3OH, 25 °C).
Intermediate 13a (1-13a) (7S)-3-Bromo-7-methyl-6,7-dihydro-5/-/-pyrazolo[1,5-a]pyrazin-4-one (1-13a) [0174]
[0175] Intermediate 13a was prepared in 71% yield according to the following general description of a synthesis performed at a large scale: A mixture of methyl 4-bromo-1/-/-pyrazole-5-carboxylate (referred to as "pyrazole SM" herein) (1 eq.), triphenyl phosphine (1.2 eq.), I-3 (1.2 eq.) and anhydrous THF (15 mL/g pyrazole SM) under nitrogen was cooled to 5-10 °C. Di-tert-butyl azodicarboxylate (1.2 eq.) was added in portions at 5-15 °C under nitrogen. The solution was heated to 20-30 °C and stirred at 20-30 °C for 2-3 h. The obtained solution was concentrated and co-evaporated with isopropyl acetate to remove THF to afford a solution of crude 4-bromo-1-[(1S)-1-[[(1,1-dimethylethoxy)carbonyl]ami-no]ethyl]-1 /-/-pyrazole-5-carboxylic acid methyl ester l-6a in isopropyl acetate (20 mL/g pyrazole SM). To the solution of l-6a was bubbled HCI gas at 15-30 °C until cleavage of the Boc protecting group was completed. The suspension was bubbled with nitrogen gas to remove most of the HCI gas. The suspension was concentrated to a volume of about 5 mL/g pyrazole SM below 50 °C, and then isopropyl acetate (15 mL/g pyrazole SM) was added to the residue. Water (10 mL/g pyrazole SM) was added at 10-20 °C. The mixture was stirred at 10-20 °C for 20-30 min. The mixture was filtered and the aqueous layer was separated. The organic layer was extracted with water (2 mL/g pyrazole SM). The combined aqueous layers were washed with isopropyl acetate (2x10 mL/g pyrazole SM) to remove residual triphenylphosphine oxide, l-lla was obtained as an aqueous solution (6.25 mL/g pyrazole SM). To the aqueous solution of 1-11a was added potassium carbonate (~1 g/g pyrazole SM) to adjust to pH=8-9 at 10-25 °C. The mixture was stirred at 10-25 °C for 5-6 h and solid l-13a precipitated. The suspension was cooled to 5-10 °C and stirred at 5-10 °C for 2-3 h, it was then filtered and washed with water (1 mL/g pyrazole SM) and heptanes (1 mL/g pyrazole SM), then dried in vacuo at 40-45 °C to afford l-13a as a white solid, mp. 196.12 °C. 1H NMR (500 MHz, CDCIg) δ ppm 1.61 (d, J=6.36 Hz, 3 H) 3.48 (ddd, J=12.72, 7.22, 2.60 Hz, 1 H) 3.75 - 3.84 (m, 1 H) 4.49 -4.59 (m, 1 H) 6.54 (br. s„ 1 H)7.56(s, 1 H). LC-HRMS (ESI+) Calculated for C7H8BrN30 (M+H)+: 229.9929, Found: m/z 229.9931 (+0.2mDa), Rt = 0.62 min (Method 13, see table 2). [a] =+25.2° (589 nm, c 0.53 w/v %, DMF, 20 °C).
Intermediate 14 (1-14) 7S-Methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-14) [0176]
[0177] Intermediate compound 1-14 was synthesized following a similar approach described for intermediate 1-13.
Starting from intermediate 1-12 (32.5 g, 139.1 mmol), intermediate compound 1-14 was obtained as a solid (14.8 g, 70%).
Intermediate 15 (1-15) 3-lodo-7R-methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-15) [0178]
[0179] A 4M solution of HCI in 1,4-dioxane (2.3 mL, 9.2 mmol) was added to a solution of intermediate I-7 (0.78 g, 1.84 mmol) in CH3CN (8.3 mL). The mixture was stirred at 80 °C for 7 h. After Boc deprotection was complete, part of the solvent was evaporated in vacuo and the solution was basified with a sat. sol. of NaHC03. The mixture was stirred for 16 h at rt. Then the mixture was diluted with water and extracted with DCM. The organic layers were separated, dried (MgS04), filtered and the solvents evaporated in vacuo. The solid was triturated with DIPE to yield intermediate compound 1-15 as a white solid (0.42 g, 82%).
Intermediate 16 (1-16) 3-lodo-7-methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-16) [0180]
[0181] Intermediate compound 1-16 was synthesized following a similar approach described for intermediate 1-15. Starting from intermediate I-8 (3.46 g, 8.17 mmol), intermediate compound 1-16 was obtained as a white solid (1.87 g, 82%).
Intermediate 17 (1-17) 3-lodo-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-17) [0182]
[0183] Intermediate compound 1-17 was synthesized following a similar approach described for intermediate 1-15. Starting from intermediate I-9 (4.89 g, 11.95 mmol), intermediate compound 1-17 was obtained as a white solid (1.87 g, 59%).
Intermediate 18 (1-18) 7S-Methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-18) [0184]
[0185] Pd(PPh3)4 (0.33 g, 0.29 mmol) was added to a stirred suspension of intermediate 1-13 (1.6 g, 5.77 mmol) and 2-picoline-4-boronic acid (0.95 g, 6.93 mmol) in 1,4-dioxane (8 mL) and a sat. sol. of NaHC03 (4 mL) in a sealed tube under nitrogen. The mixture was stirred at 100 °C for 16 h. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 6/94). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound 1-18 as a white solid (1 g, 71%), mp 173.20 °C. 1H NMR (500 MHz, CDCI3) δ ppm 1.67 (d, J=6.65 Hz, 3 H) 2.60 (s, 3 H) 3.52 (ddd, J=12.79, 7.15, 2.89 Hz, 1 H) 3.84 (dt, J=12.72, 4.00 Hz, 1 H) 4.57 - 4.66 (m, 1 H) 6.10 (br. s., 1 H) 7.51 (dd, J=5.20, 1.44 Hz, 1 H) 7.55 (s, 1 H) 7.78 (s, 1 H) 8.50 (d, J= 5.20 Hz, 1 H). LC-HRMS (ESI+) Calculated for C13H14IN40 (M+H)+: 243.1246, Found: m/z 243.1250 (+0.4mDa), Rt = 0.82 min (Method 13, see table 2). [a] = +32.8 ° (589 nm, c 0.52 w/v %, DMF, 20 °C).
[0186] Intermediate 1-18 was alternatively prepared in 70% yield according to the following general description of a synthesis performed at a large scale: A mixture of l-13a (1 eq.), 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-pyridine (1.1 eq.), anhydrous potassium phosphate (2 eq.), DME (7.5 mL/g 1-13a) and purified water (2.5 mL/g 1-13a) was evacuated and backfilled with nitrogen 3 times. Triphenyl phosphine (0.261 eq.) and palladium (II) acetate (0.131 eq.) were added in one portion under nitrogen. The mixture was evacuated and backfilled with nitrogen 3 times again, it was heated to 75-80 °C and stirred at 75-80 °C for 12-15 h under nitrogen. The aqueous layer was separated at 60-70 °C and discarded, and then water (8 mL/g 1-13a) was added to the organic layer. DME was removed by concentration below 40 °C. Isopropyl acetate (15 mL/g 1-13a) was added, the pH of the mixture was adjusted to 1-2 with cone. HCI. The mixture was filtered and the filter cake was washed with water (1 mL/g 1-13a), the aqueous layer was separated and the organic layer was extracted with water (2 mL/g 1-13a). The combined aqueous layers were washed with Isopropyl acetate (2x15 mL/g 1-13a). The aqueous layer was concentrated to remove residual DME and isopropyl acetate. MTBE (2 mL/g 1-13a) was added and the mixture was cooled to 0-5 °C, stirred at 0-5 °C for 2-3 h. 1-18 was filtered, washed with cooled water (1 mL/g 1-13a), and dried in vacuum at 45-50 °C to afford 1-18 as an off-white solid.
Intermediate 19 (1-19) 7R-Methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydro-SH-pyrazolo[1,5-a]pyrazin-4-one (1-19) [0187]
[0188] Intermediate compound 1-19 was synthesized following a similar approach described for intermediate 1-18. Starting from intermediate 1-15 (0.62 g, 2.24 mmol), intermediate compound 1-19 was obtained as a white solid (0.38 g, 70%).
Intermediate 20 (1-20) 7-Methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-20) [0189]
[0190] Intermediate compound I-20 was synthesized following a similar approach described for intermediate 1-18. Starting from intermediate 1-16 (846 mg, 3.05 mmol), intermediate compound I-20 was obtained as a yellowish solid (585 mg, 79%).
Intermediate 18 (1-18) and intermediate 19 (1-19) 7S-Methyl-3-(2-methyl-pyridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-18) and 7R-Methyl-3-(2-methyl-py-ridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-19).
[0191]
[0192] Intermediate I-20 (975 mg, 4.02 mmol) was purified by RP HPLC (Stationary phase: irregular bare silica 40 g), Mobile phase: 0.5% NH4OH, 95% DCM, 5% MeOH) then by chiral SFC ((Stationary phase: CHIRALCEL® OD-H 5 μηι 250x20mm), (Mobile phase: 75% C02, 25% iPrOH (0.3% iPrNH2)) to yield intermediate compound 1-18 (390 mg) and intermediate compound 1-19 (395 mg).
Intermediate 21 (1-21) 3-(2-Methyl-pyridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-21) [0193]
[0194] Intermediate compound 1-21 was synthesized following a similar approach described for intermediate 1-18. Starting from intermediate 1-17 (908 mg, 3.45 mmol), intermediate compound 1-21 was obtained as a white solid (0.5 g, 63%).
Intermediate 22 (1-22) 7S-Methyl-5-(4-trifluoromethyl-phenyl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-22) [0195]
[0196] A mixture of intermediate 1-14 (5 g, 33.01 mmol), copper(l) iodide (3.78 g, 19.85 mmol) and K2C03 (9.14 g, 66.15 mmol) in toluene (150 mL) was nitrogen flushed for a few min. Then 4-bromobenzotrifluoride (9.3 mL, 66.1 mmol) and Λ/,Λ/’-dimethylethylenediamine (2.1 mL, 19.8 mmol) were added. The mixture was stirred under nitrogen at rtfor 10 min and then stirred at 100 °C for 16 h. Then, DMF (20 mL) was added and the mixture was stirred at 100 °C for 8 h. Then water, a cone. sol. of ammonia and DCM were added. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 50/50). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound I-22 as a pale yellow oil (9.6 g, 98%).
Intermediate 23 (I-23) 3-lodo-7S-methyl-5-(4-trifluoromethyl-phenyl)-6,7-dihydro-5H-pyrazolo [1,5-a]pyrazin-4-one (I-23) [0197]
[0198] Iodine (11.55 g, 45.5 mmol) was added to a solution of intermediate I-22 (19.2 g, 65.0 mmol) and ammonium cerium(IV) nitrate (24.95 g, 45.5 mmol) in acetonitrile (350 mL). The mixture was stirred at 70 °C for 1 h. Then the mixture was diluted with EtOAc and washed with a sat. sol. of Na2S2Os and brine. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The residue was precipitated with DIPE and then was purified by short column chromatography (silica, DCM) then by flash column chromatography (silica; DCM in heptane 50/50 to 100/0). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound I-23 as a solid (24.8 g, 90%).
Intermediate 24 (I-24) 2-Amino-pyridine-4-boronic acid (I-24) [0199]
[0200] 2-Amino-4-chloropyridine (3g, 23.34 mmol) was added to a mixture of bis(pinacolato)diboron (17.78 g, 70.01 mmol), 2-dicyclohexylphosphino-2’,6’-dimethoxybiphenyl (0.38 g, 0.93 mmol), tris(dibenzylideneacetone)dipalladium(0) (0.21 g, 0.23 mmol) and potassium acetate (3.89 g, 39.67 mmol) in 1,4-dioxane (78 mL) under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for 3 h. The hot reaction mixture was filtered through diatomaceous earth and washed with EtOAc. The organic layer was evaporated in vacuo. The residue was precipitated with DIPE to yield intermediate compound I-24 as a white solid (5.84 g, quant.) that was used in the next reaction step without further purification.
Intermediate 25 (I-25) 3-(2-Methoxy-pyridin-4-yl)-7S-methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-25) [0201]
[0202] Pd(PPh3)4 (0.42 g, 0.36 mmol) was added to a stirred suspension of intermediate 1-13 (2 g, 7.22 mmol) and 2-methoxypyridine-4-boronic acid (1.77 g, 11.55 mmol) in 1,4-dioxane (16 mL) and a sat. sol. of NaHC03 (8 mL) in a sealed tube under nitrogen atmosphere. The mixture was stirred at 100 °C for 3 days. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 6/94). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound I-25 as a pale brown solid (1.6 g, 86%).
Intermediate 26 and final compound 215 (I-26 and Co. No. 215) 3-(2-Chloro-pyridin-4-yl)-7S-methyl-5-(4-trifluoromethyl-phenyl)-6,7-dihydro-SH-pyrazolo[1,5-a]pyrazin-4-one(l-26and Co. No. 215) [0203]
[0204] This reaction was divided in four batches to a combined total amount indicated herein and combined for workup and purification. Pd(PPh3)4 (401 mg, 0.35 mmol) was added to a stirred suspension of intermediate I-23 (2.92 g, 6.94 mmol) and 2-chloropyridine-4-boronic acid (1.42 g, 9.02 mmol) in 1,4-dioxane (39 mL) and a sat. sol. of NaHC03 (19.5 mL). The mixture was stirred at 150 °C for 10 min under microwave irradiation. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 20/80). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound I-26 as a yellow solid (1.84 g, 65%).
Intermediate 27 (I-27) 7S-Methyl-5-(4-trifluoromethyl-phenyl)-3-(2-vinyl-pyridin-4-yl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-27) [0205]
[0206] Pd(PPh3)4 (51 mg, 0.044 mmol) was added to a stirred suspension of intermediate I-26 (600 mg, 1.48 mmol) and vinylboronic acid pinacolester (0.325 mL, 1.92 mmol) in 1,4-dioxane (10 mL) and a sat. sol. of NaHC03 (5 mL). The mixture was stirred at 150 °C for 15 min under microwave irradiation. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 30/70). The desired fractions were collected and the solvents evaporated in vacuo to yield intermediate compound I-27 as a yellow oil (0.53 g, 90%).
Intermediate 28 (I-28)
Ethyl 2-[1-[(ferf-butoxycarbonylamino)methyl]-2-[tert-butyl(dimethyl)silyl]oxy-ethyl]-4-iodo-pyrazole-3-carboxylate (1-28) [0207]
[0208] Di-ferf-butyl azodicarboxylate (1.97 g, 8.53 mmol) was added to a stirred solution of 4-iodo-1 H-pyrazole-3-carboxylic acid ethyl ester (1.26 g, 4.74 mmol), [3-(ferf-butyldimethylsilanyloxy)-2-hydroxypropyl]carbamic acid tert-butyl ester (2.90 g, 9.48 mmol) and triphenylphosphine (2.24 g, 8.53 mmol) in THF (23.6 mL) under nitrogen atmosphere. The mixture was stirred at rt for 2.5 h. The solvent was evaporated and the residue was treated with DIPE. The solid (Ph3PO) was filtered off and the filtrate was evaporated in vacuo. The crude product was purified by flash column chromatography (silica; DCM in Heptane 50/50 to 100/0 then EtOAc in DCM 0/100 to 3/97). The desired fractions were collected and concentrated in vacuo to yield intermediate compound I-28 (2.57 g, 98%) as a colorless oil.
Intermediate 29 (l-29)_
Ethyl 2-[1-(aminomethyl)-2-hydroxy-ethyl]-4-iodo-pyrazole-3-carboxylate hydrochloride salt (I-29) [0209]
[0210] Hydrochloric acid (4 M in 1,4-dioxane, 5.80 mL, 23.21 mmol) was added to a stirred solution of intermediate I-28 (2.57 g, 4.64 mmol) in CH3CN (21 mL). The mixture was stirred at rt for 1 h. The mixture was concentrated in vacuo to yield intermediate compound I-29 (1.69 g) as a beige solid which was used in the next step without any further purification.
Intermediate 30 (I-30) 7-(Hydroxymethyl)-3-iodo-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-30) [0211]
[0212] TEA (1.38 mL, 9.93 mmol) was added to a stirred solution of intermediate 1-29 (1.68 g, 4.48 mmol) in DMF (16.8 mL). The mixture was stirred at rt for 3 h. The mixture was treated with a sat. sol. of NaHCOs and EtOAc and filtered. The filtrate was partitioned between water and EtOAc and extracted with EtOAc and EtOAc/THF. The organic layer was separated, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 20/80). The desired fractions were collected and concentrated in vacuo to yield intermediate compound i-30 (0.88 g, 67%) as a white solid.
Intermediate 31 (1-31) 7-(Hydroxymethyl)-3-(2-methyl-4-pyridyl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (1-31) [0213]
[0214] Pd(PPh3)4 (134 mg, 0.12 mmol) was added to a stirred suspension of intermediate I-30 (0.88 g, 3.00 mmol) and 2-picoline-4-boronic acid (658 mg, 3.00 mmol) in 1,4-dioxane (15.4 mL) and a sat. aq. NaHCOs (10 mL) under nitrogen atmosphere. The mixture was stirred at 90 °C for 16 h. Then additional 2-picoline-4-boronic acid (263 mg, 1.20 mmol) and Pd(PPh3)4 (35 mg, 0.03 mmol) were added at rt and under nitrogen. The mixture was stirred at 100 °C for 7 h. Then the mixture was diluted with water and extracted with EtOAc. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica;
MeOH in DCM 0/100 to 10/90). The desired fractions were collected and concentrated in vacuo to yield intermediate compound 1-31 (347 mg, 45%) as pale orange solid.
Intermediate 32 (I-32) 7-(Hydroxymethyl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-32) [0215]
[0216] Palladium 10% on charcoal (907 mg, 0.0.853 mmol) was added to a solution of intermediate I-30 (2.5 g, 8.53 mmol) and TEA (4.74 mL, 34.12 mmol) in DMF (125 mL) under nitrogen atmosphere. The mixture was hydrogenated (at atmospheric pressure) at rt for 16 h. The mixture was filtered through a pad of diatomaceous earth and the residue was washed with MeOH and 7M solution of ammonia in MeOH. The filtrate was concentrated in vacuo and the residue was treated with a small amount of water and extracted with EtOAc/THF. The organic layer was separated, dried (Na2S04), filtered and concentrated in vacuo to yield intermediate compound I-32 (1.4 g, quant.) as a brown oil.
Intermediate 33a (l-33a) and intermediate 33b (l-33b) (7S)-7-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (l-33a) and [(7S)-7-methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo-[1,5-a]pyrazin-3-yl]boronic acid (l-33b) [0217]
[0218] Isopropylmagnesium chloride lithium chloride complex (1,3M solution, 32.9 mL, 42.7 mmol) was added dropwise to a stirred solution of intermediate I-23 (10 g, 23.7 mmol) and 2-/'sopropoxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (9.7 mL, 47.5 mmol) in anhydrous THF (100 mL) at -25 °C under nitrogen atmosphere. The mixture was stirred for 30 min at -25 °C. Then the reaction was quenched with a 10% NH4CI aq sol. and extracted with EtOAc. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 3/97). The desired fractions were collected and the solvents evaporated in vacuo. The crude product was triturated with DIPE, filtered and dried to yield intermediate compound I-33a (6.4 g, 64%) as a white solid. The solution and impure fractions from the column purification were combined and repurified by flash column chromatography (silica, EtOAc in Heptane 30/70 to 70/30). The desired fractions were collected and the solvents evaporated in vacuo. The product was triturated in DIPE/Heptane, filtered and dried to yield intermediate compound l-33b (1 g, 10%) as a white solid.
Intermediates I-34 to I-37 [0219] The following intermediates were synthesized by following an analogous synthetic procedure as reported for intermediate 22.
Intermediate 38 and final compound 170 (7S)-5-(3,4-dichlorophenyl)-7-methyl-3-(2-methyl-1-oxido-pyridin-1-ium-4-yl)-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (I-38 and Co. No. 170) [0220]
[0221] 3-Chloroperoxybenzoic acid (2.03 g, 11.77 mmol) was added to a stirred solution of intermediate 1-34 (2.28 g, 5.88 mmol) in DCM (37 mL) at 0 °C. The mixture was allowed to reach rt and stirred at rt for 2 h. The mixture was treated with a sat sol. of Na2C03 and diluted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo to yield intermediate compound 1-38 (1.84 g, 77%) that was used in the next step without any further purification.
Intermediate 39 (1-39) 4-[(7S)-5-(3,4-dichlorophenyl)-7-methyl-4-oxo-6,7-dihydropyrazolo[1,5-a]pyrazin-3-yl]pyridine-2-carbaldehyde (I-39) [0222]
[0223] Manganese dioxide (2.39 g, 27.57 mmol) was added to a solution of final compound 125 (E-14) (1.11 g, 2.75 mmol) in chloroform (11.7 mL). The mixture was stirred at rt for 16 h, at 60 °C for 5 h and then at rt for 16 h. Then, the mixture was filtered through a pad of diatomaceous earth and washed with DCM. The solvent was evaporated In vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 50/50). The desired fractions were collected and evaporated in vacuo to yield intermediate compound I-39 (537 mg, 48%) as a pale yellow solid.
Intermediate 40 (I-40) [0224] The following intermediate was synthesized by following an analogous synthetic procedure as reported for intermediate 39.
Intermediate 41 (1-41) ferf-Butyl 4-(5-chloro-2-pyridyl)piperazine-1-carboxylate (1-41) [0225]
[0226] A mixture of 2-bromo-5-chloropyridine (1.5 g, 7.79 mmol), 1-BOC-piperazine (2.18 g, 11.69 mmol), sodium ferf-butoxide (1.49 g, 15.59 mmol), 9,9-dimethyl-4,5-bis(diphenylphosphino)xanthene (0.451 g, 0.78 mmol) and tris(dibenzylideneacetone)dipalladium(0) (0.357 g, 0.390 mmol) in toluene (25 mL) was stirred at 120 °C for 16 h. The mixture was poured into water, and extracted with EtOAc. The mixture was filtered through a short pad of diatomaceous earth. The organic layer was separated, washed with water and brine, dried (MgS04) and evaporated in vacuo. The crude product was purified by flash column chromatography (silica, EtOAc in DCM 0/100 to 20/80). The desired fractions were collected and evaporated in vacuo to yield intermediate compound 1-41 (0.888 g, 38%) as an orange solid.
Intermediate 42 (1-42) tert-Butyl 4-[5-[(7S)-7-methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-3-yl]-2-pyridyl]pip-erazine-1-carboxylate (I-42) [0227]
[0228] A suspension of intermediate 1-41 (478 mg, 1.60 mmol), intermediate l-33a (653 mg, 1.55 mmol), palladium(ll) acetate (7 mg, 0.032 mmol), 2-dicyclohexylphosphino-2’,6’-dimethoxybiphenyl (26 mg, 0.064 mmol) and K2COs (554 mg, 4.013 mmol) in CH3CN (1.6 mL) and H20 (2.5 mL) was flushed with nitrogen for a few minutes and the mixture was stirred at 80 °C for 24 h. Then the mixture was diluted with H20 and extracted with EtOAc. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 50/50). The desired fractions were collected and evaporated in vacuo to yield intermediate compound I-42 (663 mg, 74%) as a yellow oil.
Intermediate 43 and final compound 188 (7S)-5-[6-chloro-5-(trifluoromethyl)-2-pyridyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (I-43 and Co. No. 188) [0229]
[0230] Pd(PPh3)4 (47 mg, 0.041 mmol) was added to a stirred suspension of intermediate 1-18 (100 mg, 0.413 mmol), 2,6-dichloro-3-(trifluoromethyl)pyridine (86 μΙ-, 0.620 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (47 mg, 0.082 mmol), Cs2C03 (269 mg, 0.825 mmol) in 1,4-dioxane (3 mL) in a sealed tube and under nitrogen. The mixture was stirred at 120 °C for 4 h. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The filtrate was evaporated in vacuo. The crude product was purified by flash column chromatography (silica, EtOAc in DCM 0/100 to 30/70). The desired fractions were collected and concentrated in vacuo. Then the product was triturated with Et20 and filtered to yield intermediate compound I-43 (71 mg, 40%) as a white solid.
Intermediate 44 (1-44) 7-(Difluoromethyl)-5-[4-(trifluoromethyl)phenyl]pyrazolo[1,5-a]pyrazin-4-one (I-44) [0231]
[0232] Diethylaminosulfur trifluoride (0.373 mL, 3.866 mmol) was added to a stirred solution of intermediate I-40 (297 mg, 0.966 mmol) in DCM (33 mL) at 0 °C. The mixture was allowed to warm to rt and stirred for 5 h. Then additional diethylaminosulfur trifluoride (0.355 mL, 2.9 mmol) was added at 0 °C and the mixture was stirred at rt for 20 h. The mixture was treated with water and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (silica; DCM). The desired fractions were collected and concentrated in vacuo to yield intermediate compound I-44 (258 mg, 81 %) as a colorless oil.
Intermediate 45 (I-45) [0233] The following intermediate was synthesized by following an analogous synthetic procedure as reported for intermediate 23.
Intermediate 46 (I-46)
[0234] The following intermediate was synthesized by following an analogous synthetic procedure as reported for intermediate 18.
Intermediates 47 and 48 (I-47 and I-48) [0235]
i) NH3(28% in water, 54 mL) was added over 2-[1-[[(1,1-dimethylethyl)dimethylsilyl]oxy]ethyl]-oxirane (4.73 g, 23.373 mmol) and the mixture was stirred at 120 °C for 40 min under microwave irradiation. The solvent was then concentrated in vacuo to yield intermediate compound I-47 as an orange oil (3.298 g, 64%). ii) Intermediate I-48 was synthesized following an analogous synthetic procedure as reported for intermediate I-3. Starting from intermediate I-47 (3.269 g, 14.9 mmol), intermediate compound I-48 was obtained as a colorless oil (4.642 g, 97.5%).
Intermediates 49-52 (I-49 to I-52) [0236] The following intermediates were synthesized by following an analogous synthetic procedure as reported for intermediate 6.
Intermediate 53 (1-53) [0237] The following intermediate was synthesized following the procedure for the synthesis of intermediate 1-29, followed by the procedure for the synthesis of intermediate 1-30.
Intermediate 54 (1-54)
Ethyl 2-[1-(aminomethyl)-2-methoxy-ethyl]-4-iodo-pyrazole-3-carboxylate (I-54) [0238]
[0239] HCI (4 M in dioxane, 2.2 mL, 8.82 mmol) was added to a solution of intermediate 1-52(0.8 g, 1.765 mmol) in CH3CN (8 mL). The mixture was stirred at rtfor 1 h and then the solvent was concentrated in vacuo to give intermediate compound I-54 (700 mg, 87%) as a cream solid.
Intermediate 55 (I-55) 3-lodo-7-(methoxymethyl)-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-55) [0240]
[0241] Et3N (0.55 mL, 3.98 mmol) was added to a solution of intermediate I-54 (0.7 g, 1.80 mmol) in DMF (6.7 mL). The mixture was stirred at rt for 3 h then neutralized with a sat. sol. of NaHC03 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo to give intermediate compound I-55 (440 mg, 80%) as a white solid.
Intermediates 56 and 57 (I-56 and I-57) [0242] The following intermediates were synthesized by following an analogous synthetic procedure as that reported for intermediate 15.
Intermediates 58-61 (I-58 to 1-61) [0243] The following intermediates were synthesized by following an analogous synthetic procedure as reported for intermediate 25.
(continued)
Intermediate 62 (1-62) 2-[(1S)-2-(3,4-dichloroanilino)-1-methyl-ethyl]-4-(2-methyl-4-pyridyl)pyrazole-3-carboxylic acid (I-62) [0244]
[0245] Sodium hydride (60% dispersion in mineral oil, 20 mg, 0.344 mmol) was added to a solution of compound Co. No. 6a (200 mg, 0.516 mmol) in DMF (4 mL) and the mixture was stirred at 60 °C for 24 h. Then more sodium hydride (60% dispersion in mineral oil, 11 mg, 0.172 mmol) was added and the mixture was stirred at 60 °C for 3 h. Then, the mixture was quenched with a NH4CI sat. sol. and extracted with EtOAc. The organic layer was separated, dried (MgS04), filtered and the solvents evaporated In vacuo to give intermediate compound I-62 (230 mg, quantitative) as a solid which was used in the next step without further purification.
Intermediate 63 (I-63) 2-[(1 S)-2-(4,5-dichloro-2-iodo-anilino)-1-methyl-ethyl]-4-(2-methyl-4-pyridyl)pyrazole-3-carboxylic acid (l-63a) and 2-[(1S)-2-(3,4-dichloro-2-iodo-anilino)-1-methyl-ethyl]-4-(2-methyl-4-pyridyl)pyrazole-3-carboxylic acid (l-63b) [0246]
(1-0.3 UJ
[0247] A/-iodosuccimide(124 mg, 0.552 mmol) was added to a solution of intermediate compound I-62 (224 mg, 0.5523 mmol) in chloroform (5 mL) and the mixture was stirred at rtfor 3 h. Then more N-iodosuccimide (62 mg, 0.277 mmol) was added and the mixture was stirred at rt for 18 h. Then the reaction was quenched with a sat. sol. of Na2S03 and extracted with DCM. The organic layer was separated, dried (MgSQ4), filtered and the solvent evaporated in vacuo to give a mixture of intermediates compounds l-63a and l-63b (240 mg, 41 %) which was used in next step without further purification.
Intermediate 64 (I-64) ferf-Butyl /\/-[[2,2,2-trifluoro-1-(nitromethyl)ethyl]amino]carbamate (1-64) [0248]
[0249] ferf-Butyl carbazate (281 mg, 2.13 mmol) was added to a stirred solution of 3,3,3-trifluoro-1-nitro-prop-1-ene (prepared as described in J. Fluorine Chem. 2008, 767-774) (73 μΙ_, 0.709 mmol) in MeOH (3.1 mL) at rt. The mixture was stirred for 1 h and the solvents evaporated In vacuo. The crude product was purified by flash column chromatography (silica, EtOAc in Heptane 40/60 to 60/40) to yield intermediate compound I-64 (200 mg, quant.)
Intermediate 65 (I-65) [2,2,2-Trifluoro-1-(nitromethyl)ethyl]hydrazine hydrochloride salt (I-65) [0250]
[0251] HCI (6M in 1,4-dioxane, 10.5 mL, 42 mmol) was added to a solution of intermediate 1-64 (1.15 g, 4.2 mmol) in MeOH (10 mL) at rt. The mixture was stirred for 2 h and the solvents were evaporated in vacuo to yield intermediate compound I-65 (880 mg, quant.) that was used in the next step without further purification.
Intermediate 66 (I-66)
Ethyl 2-[2,2,2-trifluoro-1-(nitromethyl)ethyl]pyrazole-3-carboxylate (l-66a) and ethyl 1-[2,2,2-trifluoro-1-(nitrome-thyl)ethyl]pyrazole-3-carboxylate (l-66b) [0252]
[0253] Ethyl pyruvate (77 μι, 0.692 mmol) and W,/V-dimethylformamide dimethyl acetal (92 μί, 0.692 mmol) were stirred at rt for 16 h. The dark red/brown solution was added to a solution of intermediate i-65 (145 mg, 0.692 mmol) in EtOH (2 mL). The mixture was stirred at 85 °C for 3 h. The solvent was concentrated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 30/70 to 60/40) to yield intermediate compounds I- 66a (78 mg, 40%) and l-66b (54 mg, 28%).
Intermediate 67 (1-67) 7-Methyl-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazin-4-one (I-67) [0254]
[0255] Pd (10% on charcoal, 100 mg, 0.094 mmol) and ammonium formate (112 mg, 1.78 mmol) were added to a stirred solution of intermediate I-66 (100 mg, 0.355 mmol) in MeOH (3.3 mL). The mixture was stirred in a sealed tube at 70 °C for 2 h. The solvent was concentrated in vacuo to yield intermediate compound I-67 (70 mg, 96%) that was used in the following step without further purification.
Intermediate 68 (I-68) 3-lodo-7-(trifluoromethyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (I-68) [0256]
[0257] Intermediate compound i-68 was synthesized by following the sequence of an analogous synthetic procedure as reported for intermediate I-22 starting from intermediate I-67 and 4-bromobenzotrifluoride, followed by the procedure for intermediate I-23.
Final Compounds
Example 1 (7S)-7-Methyl-3-(2-methylpyridin-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (E-1, Co. No. 1) [0258]
Procedure A: Copper(l) iodide (872 mg, 4.58 mmol) was added to a stirred suspension of intermediate 1-18 (1.85 g, 7.64 mmol), 4-bromobenzotrifluoride (2.14 mL, 15.27 mmol), K2C03 (2.11 g, 15.27 mmol) and Λ/,Λ/’-dimethyleth-ylenediamine (0.492 mL, 4.58 mmol) in toluene (70 mL) in a sealed tube and under nitrogen. The mixture was stirred at 100 °C for 16 h. Then DMF (10 mL) was added and the mixture was stirred at 100 °C for additional 8 h. The mixture was filtered through diatomaceous earth and washed with EtOAc. The organic layer was washed with diluted NH4OH sol, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 20/80 to 50/50). The desired fractions were collected and the solvents evaporated in vacuo. The product was precipitated with heptane, filtered and dried in vacuo to yield final product compound 1 as a white solid (2.32 g, 78%). 1H NMR (500 MHz, CDCI3) δ ppm 1.75 (d, J=6.4 Hz, 3 H), 2.57 (s, 3 H), 4.02 (dd, J=12.7, 7.2 Hz, 1 H), 4.30 (dd, J=12.6, 4.2 Hz, 1 H), 4.75 - 4.84 (m, 1 H), 7.44 (d, J= 5.2 Hz, 1 H), 7.49 (d, J=3.8 Hz, 2 H), 7.51 (s, 1 H), 7.71 (d, J=8.4 Hz, 2 H), 7.80 (s, 1 H), 8.48 (d, J=5.2 Hz, 1 H).
Procedure B: Copper(l) iodide (94 mg, 0.495 mmol) was added to a stirred suspension of intermediate 1-18 (200 mg, 0.825 mmol), 4-bromobenzotrifluoride (0.231 mL, 1.651 mmol), K2C03 (228 mg, 1.65 mmol) and Λ/,Λ/’-dimeth-ylethylenediamine (53 μί) in toluene (7.5 mL) in a sealed tube and under nitrogen. The mixture was stirred at 100 °C overnight. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The organic layer was separated, dried (MgS04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica, EtOAc in Heptane 0/100 to 70/30). The desired fractions were collected and concentrated in vacuo to yield compound 1 (283 mg, 89%) as a pinkish solid.
Procedure C: Pd(PPh3)4 (384 mg, 0.332 mmol) was added to a stirred suspension of intermediate I-23 (2 g, 4.74 mmol) and 2-methylpyridine-4-boronic acid pinacol ester (1.66 g, 7.60 mmol) in 1,4-dioxane (10 mL) and a sat. sol. of Na2COs (5 mL) in a sealed tube under nitrogen. The mixture was stirred at 100 °Cfor 16 h. Then the mixture was diluted with H20 and extracted with DCM and DCM with a small amount of EtOH. The organic layer was dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; 7M solution of ammonia in MeOH in DCM 0/100 to 3/97 then EtOAc in Heptane 0/100 to 100/0). The desired fractions were collected and evaporated in vacuo to yield compound 1 as a white solid (480 mg, 26%). (1.31 g of starting material was recovered).
Procedure D; general description of a synthesis performed at a large scale by which Co. No. 1 was isolated in 90% yield before purification: A mixture of 1-18 (1 eq.), potassium carbonate (2 eq.), copper(l) iodide (0.3 eq.), 4-bromobenzotrifluoride (1.3 eq.), Ν,Ν’-Dimethyl ethylenediamine (0.35 eq.), DMF (5 mL/g 1-18) and toluene (8 mL/g 1-18) was evacuated and backfilled with nitrogen 3 times. It was heated to 100-110 °C and stirred at 100-110 °C for 7-8 h under nitrogen. The reaction solution was concentrated to remove toluene below 50 °C. Isopropyl acetate (15 mL/g 1-18) was added. The mixture was washed with 5% NH4OH aqueous solution (3x7 mL/g 1-18), and then 5% N-acetyl-L-cysteine and 5% K2C03 aqueous solution (2x7 mL/g 1-18) at 10-25 °C. Finally, it was washed with 5% NaCI aqueous solution (5 mL/g 1-18). The obtained solution was concentrated and co-evaporated with MTBE to remove isopropyl acetate. The resulting solid was filtered and dried in vacuo at 45-50 °C. Co. No. 1 was obtained as an off-white solid which was further purified as follows:
Co. No. 1 was dissolved in a solvent mixture of IPA (4 mL/g Co. No. 1) and water (1 mL/g Co. No. 1) at 48-55 °C. The solution was filtered and cooled to 0-5 °C. An IPA/water mixture (0.5 mL/g Co. No. 1,4/1 v/v) was used to rinse. Water (650 μί/g Co. No. 1) was added drop-wise and seeding with Co. No. 1 was performed. The mixture was stirred at 0-5 °C for 3-4 h. Water (14 mL/g Co. No. 1) was added drop-wise at 0-5 °C for 3-4 h, and then the suspension was stirred at 0-5 °C for 5-6 h. The wet product was filtered and rinsed with water (2 mL/g Co. No. 1), then dried in vacuo at 45-50 °C for 16 h to afford Co. No. 1 as a white solid.
[0259] For compound 1 (DSC mp = 155.35 °C), the hydrochloride salt (.HCI) (DSC mp = decomposes above 200 °C); the sulfate salt (.H2S04) (DSC mp = decomposes above 200 °C); the methane sulfonate salt (.CH3S03H) (DSC mp = 252 °C); and the maleate salt (.H02CCH=CHC02H-c/'s) (DSC mp = 163 °C); wherein the mp were determined by DSC (Mettler Toledo Q2000 MDSC, heating from 25 to 350 °C at 10 °C /min) were obtained following the procedure described below:
Compound 1 (1.5 g) in 9 mL of IPA or acetone (hydrochloride and sulfate salts were generated in acetone; meth-anesulfonate and maleate salts were generated in IPA) were stirred at 50 °C until all the solid was dissolved. The acid (1.1 mol equivalents) was added to the solution and the reaction mixture was further stirred for 2 h at 50 °C, then cooled to 20 °C in 1 h and further stirred for 30 h at 20 °C. The suspension was filtered and the solids were dried at 50 °C in a vacuum oven overnight.
Example 2 (7S)-7-Methyl-3-pyridin-4-yl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo [1,5-a]pyrazin-4(5H)-one (E-2, Co. No. 2) [0260]
[0261] Pd(PPh3)4 (41 mg, 0.036 mmol) was added to a stirred suspension of intermediate I-23 (300 mg, 0.71 mmol) and pyridine-4-boronicacid (114 mg, 0.93 mmol) in 1,4-dioxane (3.3 mL) and a sat. sol. of NaHC03 (1.5 mL). The mixture was stirred at 150 °C for 10 min under microwave irradiation. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 6/94). The desired fractions were collected and the solvents evaporated in vacuo. The residue was purified by ion exchange chromatography using an ISOLUTE® SCX2 cartridge eluting first with MeOH and then with 7M solution of ammonia in MeOH. The desired fractions contained in the 7M solution of ammonia in MeOH were collected and the solvents evaporated in vacuo. The residue was precipitated with DIPE to yield compound 2 as a white solid (215 mg, 81%). 1H NMR (400 MHz, CDCI3) δ ppm 1.76 (d, J=6.5 Hz, 3 H), 4.03 (dd, J=12.7, 7.2 Hz, 1 H), 4.31 (dd, J=12.7, 4.2 Hz, 1 H), 4.81 (qdd, J= 6.7, 6.7, 6.7, 6.5, 4.3 Hz, 1 H), 7.51 (d, J=8.1 Hz, 2 H), 7.65 (dd, J=4.4, 1.6 Hz, 2 H), 7.71 (d, J= 8.3 Hz, 2 H), 7.83 (s, 1H), 8.60 (dd, J=4.6, 1.8 Hz, 2 H).
Example 3 (7S)-3-(2-Aminopyridin-4-yl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one (E-3, Co. No. 71) [0262]
[0263] Pd(PPh3)4 (96 mg, 0.083 mmol) was added to a stirred suspension of intermediate I-23 (700 mg, 1.66 mmol) and intermediate I-24 (458 mg, 3.32 mmol) in 1,4-dioxane (10 mL) and a sat. sol. of NaHCOs (5 mL). The mixture was stirred at 150 °C for 10 min under microwave irradiation. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 10/90). The desired fractions were collected and the solvents evaporated in vacuo and the residue was purified by RP HPLC (RP C18 XBridge™ 30 x 100 mm 5 urn), mobile phase (gradient from 67% 0.1% NH4C03H/NH40H pH 9 solution in Water, 33% CH3CN to 50% 0.1% NH4C03H/NH40H pH 9 solution in Water, 50% CH3CN). The residue was purified by ion exchange chromatography using an ISOLUTE® SCX2 cartridge eluting first with MeOH and then with 7M solution of ammonia in MeOH. The desired fractions contained in the 7M solution of ammonia in MeOH were collected and the solvents evaporated in vacuo to yield final product compound 71 as a white solid (163 mg, 25%). 1H NMR (500 MHz, CDCI3) δ ppm 1.74 (d, J=6.4 Hz, 3 H) 4.01 (dd, J=12.6, 7.1 Hz, 1 H) 4.29 (dd, J=12.6, 4.2 Hz, 1 H) 4.43 (br. s„ 2 H) 4.78 (quind, J= 6.6, 4.3 Hz, 1 H) 6.94 (dd, J=5.5,1.4 Hz, 1 H) 6.98 (s, 1 H) 7.51 (br. d, J=8.4 Hz, 2 H) 7.71 (br. d, J=8.4 Hz, 2 H) 7.79 (s, 1 H) 8.06 (d, J=4.9 Hz, 1 H). Example 4 (7S)-3-[2-(Ethylamino)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-4, Co. No. 44) [0264]
[0265] Sodium triacetoxyborohydride (246 mg, 1.16 mmol) was added to a stirred solution of compound 71 (300 mg, 0.77 mmol) and acetaldehyde (0.048 mL, 0.85 mmol) in 1,2-dichloroethane (3 mL). The mixture was stirred at rt for 16 h. Then the mixture was diluted with a sat. sol. of NaHC03 and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in heptane 0/100 to 40/60). The desired fractions were collected and the solvents evaporated in vacuo. The residue was purified by ion exchange chromatography using an ISOLUTE® SCX2 cartridge eluting first with MeOH and then with 7M solution of ammonia in MeOH. The desired fractions contained in the 7M solution of ammonia in MeOH were collected and the solvents evaporated in vacuo and the residue was purified by RP HPLC (RP C18 XBridge™ 30 x 100 mm 5 um), mobile phase (gradient from 60% 0.1% NH4C03H/NH40H pH 9 solution in Water, 40% CH3CN to 43% 0.1% NH4C03H/NH40H pH 9 solution in Water, 57% CH3CN). The residue was precipitated with DIPE to yield compound 44 as a white solid (28 mg, 9%). 1H NMR (500 MHz, CDCI3) δ ppm 1.24 (t, J= 7.2 Hz, 3 H) 1.74 (d, J=6.4 Hz, 3 H) 3.29 - 3.37 (m, 2 H) 4.00 (dd, J=12.6, 7.1 Hz, 1 H) 4.29 (dd, J=12.6, 4.2 Hz, 1 H) 4.42 (br. t, J=4.6 Hz, 1 H) 4.74 - 4.82 (m, 1 H) 6.83 (s, 1 H) 6.84 (dd, J= 5.3, 1.3 Hz, 1 H) 7.51 (br. d, J=8.7 Hz, 2 H) 7.70 (br. d, J= 8.7 Hz, 2 H) 7.79 (s, 1 H) 8.07 (d, J= 5.5 Hz, 1 H).
Example 5 (7S)-3-(2-Methoxy-4-pyridyl)-7-methyl-5-[4-(tritluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-5, Co. No. 45) [0266]
[0267] Copper(l) iodide (66 mg, 0.348 mmol) was added to a stirred suspension of intermediate I-25 (150 mg, 0.58 mmol), 4-bromobenzotrifluoride (209 mg, 0.93 mmol), K2COs (161 mg, 1.16 mmol) and Λ/,Λ/’-dimethylethylenediamine (0.037 mL, 0.348 mmol) in toluene (3.75 mL) in a sealed tube and under nitrogen. The mixture was stirred at 100 °C for 24 h. Then, more 4-bromobenzotrifluoride (131 mg, 0.58 mmol) was added and the mixture was stirred at 100 °C for additional 16 h. The mixture was filtered through diatomaceous earth and washed with DCM. The organic layer was evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 0/100 to 20/80). The desired fractions were collected and the solvents evaporated in vacuo. The product was precipitated with Et20. The solid was purified by RP HPLC (RP C18 XBridge™ 30 x 100 mm 5 um), mobile phase (gradient from 60% 0.1% NH4C03H/NH40H pH 9 solution in Water, 40% CH3CN to 43% 0.1% NH4C03H/NH40H pH 9 solution in Water, 57% CH3CN) to yield compound 45 as a white solid (130 mg, 56%). %). 1H NMR (500 MHz, CDCI3) δ ppm 1.75 (d, J= 6.6 Hz, 3 H) 3.94 (s, 3 H) 4.02 (dd, J=12.7, 7.2 Hz, 1 H) 4.30 (dd, J=12.6, 4.2 Hz, 1 H) 4.75 - 4.83 (m, 1 H) 7.09 (s, 1 H) 7.23 (dd, J= 5.5,1.2 Hz, 1 H) 7.50 (br. d, J= 8.7 Hz, 2 H) 7.70 (br. d, J= 8.7 Hz, 2 H) 7.79 (s, 1 H) 8.14 (d, J= 5.5 Hz, 1 H).
Example 6 (7S)-3-(2-Ethyl-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-6, Co. No. 46) [0268]
[0269] A solution of intermediate I-27 (114 mg, 0.29 mmol) in EtOH (5.7 mL) was hydrogenated in a Η-Cube® reactor (1 mL/min, 30 mm Pd(OH)2/C 20% cartridge, full H2 mode, rt, 1 cycle). Then, the solvent was evaporated in vacuo. The crude product was purified by RP HPLC (RP C18XBridge™ 30 x 100 mm 5 um), mobile phase (gradient from 60% 0.1% NH4C03H/NH40H pH 9 solution in Water, 40% CH3CN to 43% 0.1% NH4C03H/NH40H pH 9 solution in Water, 57% CH3CN) to yield compound 46 as a white solid (84 mg, 73%). 1H NMR (400 MHz, CDCI3) δ ppm 1.32 (t, J= 7.6 Hz, 3 H) 1.75 (d, J= 6.7 Hz, 3 H) 2.85 (q, J= 7.6 Hz, 2 H) 4.02 (dd, J=12.7, 7.2 Hz, 1 H) 4.31 (dd, J=12.7, 4.2 Hz, 1 H) 4.80 (quind, J= 6.7, 4.2 Hz, 1 H) 7.46 (dd, J=5.1, 1.6 Hz, 1 H) 7.48 (br. s, 1 H) 7.51 (br. d, J=8.3 Hz, 2 H) 7.71 (br. d, J=8.3 Hz, 2 H) 7.81 (s, 1 H) 8.51 (dd, J=5.3, 0.7 Hz, 1 H).
Example 7 7-(Hydroxymethyl)-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-7, Co. No. 87) [0270]
[0271] Copper(l) iodide (0.135 g, 0.709 mmol) was added to a stirred suspension of intermediate 1-31 (305 mg, 1.18 mmol), 4-bromobenzotrifluoride (298 μΙ_, 2.12 mmol), K2C03 (330 mg, 2.36 mmol) and Λ/,Λ/’-dimethylethylenediamine (76 μί, 0.71 mmol) in toluene (7.63 mL) in a sealed tube and under nitrogen. The mixture was stirred at 100 °C for 18 h. Then additional K2COs (160 mg, 1.18 mmol), copper(l) iodide (0.067 g, 0.354 mmol), Λ/,Λ/’-dimethylethylenediamine (38 μί, 0.35 mmol) and 4-bromobenzotrifluoride (132 μί, 0.95 mmol) were added under nitrogen and the mixture was stirred at 100 °C for 5 h. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The organic layer was separated, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (silica; methanol in DCM 0/100 to 7/93). The desired fractions were collected and concentrated in vacuo to yield compound 87 (321 mg, 68%) as yellow oil that precipitated upon standing at rt.
Example 8 7-(Fluoromethyl)-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-8, Co. No. 52) [0272]
[0273] (Diethylamino)sulfur trifluoride (23 μι, 0.185 mmol) was added to a stirred solution of compound 87 (50 mg, 0.124 mmol) in DCM (2.4 mL) at -10 °C. The mixture was allowed to warm to rt and stirred for 18 h. The mixture was treated with water and extracted with DCM. The organic layer was separated, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 100/0 and MeOH in EtOAc 0/100 to 1/99). The desired fractions were collected and concentrated in vacuo. Then the compound was triturated with DIPE to yield compound 52 (14.5 mg, 29%) as a white solid. 1H NMR (500 MHz, CDCI3) δ ppm 2.58 (s, 3 H) 4.31 (dd, «7=13.1,4.8 Hz, 1 H) 4.47 - 4.53 (m, 1 H) 4.86 - 5.07 (m, 3 H) 7.45 (br. d, 7=4.6 Hz, 1 H) 7.51 (br. d, 7= 8.7 Hz, 2 H) 7.50 (s, 1 H) 7.72 (br. d, 7=8.7 Hz, 2 H) 7.85 (s, 1 H) 8.49 (d, 7= 5.2 Hz, 1 H).
Example 9 (7S)-5-[4-Fluoro-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one(E-9, Co. No. 67) [0274]
[0275] Compound 67 was obtained starting from intermediate 1-18 (160 mg, 0.66 mmol), 5-bromo-2-fluorobenzotrif-luoride (149 μΙ_, 1.06 mmol), Λ/,Λ/’-dimethylethylenediamine (42 μι, 0.396 mmol), copper(l) iodide (75 mg, 0.396 mmol), K2C03 (182mg, 1.32 mmol) in toluene (4.27 mL), following a procedure similar to that described in E-1, yielding compound 67 (224 mg, 84%) as a pale yellow solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.76 (d, 7=6.4 Hz, 3 H) 2.58 (s, 3 H) 3.98 (dd, 7=12.7, 7.2 Hz, 1 H) 4.25 (dd, 7=12.6, 4.2 Hz, 1 H) 4.80 (quind, 7=6.6, 4.3 Hz, 1 H) 7.29 (d, 7=9.5 Hz, 1 H) 7.43 (dd, 7=5.2, 1.2 Hz, 1 H) 7.48 (s, 1 H) 7.54 - 7.61 (m, 2 H) 7.80 (s, 1 H) 8.49 (d, 7=5.2 Hz, 1 H).
Example 10 (7S)-5-[4-(2-Fluoroethoxy)-3-(trifluoromethyl)phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-10, Co. No. 77) [0276]
[0277] Sodium hydride (60% dispersion in mineral oil, 22 mg, 0.544 mmol) was added to a solution of 2-fluoroethanol (453 μι, 0.495 mmol) in DMF (4.5 mL) at 0 °C and the mixture was stirred at rt for 10 minutes. Then compound 67 (200 mg, 0.495 mmol) was added. The mixture was stirred at 110 °C for 23 h. The reaction mixture was cooled to rt and a solution of 2-fluoroethanol (227 μι, 0.247 mmol) and Sodium hydride (60% dispersion in mineral oil, 12 mg, 0.297 mmol) in DMF (0.5 ml) was added. The resulting mixture was stirred at 110 °C for 16 h. The mixture was quenched with water and extracted with EtOAc. The organic layer was separated, dried (Na2S04) and the solvents were evaporated In vacuo. The crude product was purified by flash column chromatography (silica; 7M solution of ammonia in MeOH in DCM/ DCM 0/100 to 2/98). The desired fractions were collected and the solvents evaporated in vacuo to afford 164 mg of compound 77, which was further purified by RP HPLC (Stationary phase: C18 XBridge™ 30 x 100 mm 5 urn), mobile phase: Gradient from 67% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 33% CH3CN to 50% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 50% CH3CN), yielding 125 mg of compound 77, which was further purified by RP HPLC (Stationary phase: C18 XBridge™ 30 x 100 mm 5 urn), mobile phase: Gradient from 67% 0.1% NH4C03H/NH40H pH 9 solution in Water, 33% CH3CN to 50% 0.1% NH4C03H/NH4OH pH 9 solution in Water, 50% CH3CN), yielding 117 mg of compound 77 which was further purified by RP HPLC (Stationary phase: C18 XBridge™ 30 x 100 mm 5 μΓη), mobile phase: Gradient from 47% 0.1% NH4C03H/NH4OH pH 9 solution in Water, 53% MeOH to 30% 0.1% NH4C03H/NH40H pH 9 solution in Water, 70% MeOH), yielding compound 77 (39 mg, 18%), also recovering 38 mg of starting material, compound 67. For compound 77: 1H NMR (400 MHz, CDCI3) δ ppm 1.75 (d, J=6.5 Hz, 3 H) 2.57 (s, 3 H) 3.96 (dd, J=12.8, 7.3 Hz, 1 H) 4.24 (dd, J=12.7, 4.4 Hz, 1 H) 4.28 - 4.38 (m, 2 H) 4.70 - 4.87 (m, 2 H) 4.75 - 4.83 (m, 1 H) 7.08 (d, J=8.6 Hz, 1 H) 7.44 (dd, J=5.2,1.3 Hz, 1 H) 7.48 - 7.57 (m, 3 H) 7.79 (s, 1 H) 8.47 (dd, J=5.3, 0.5 Hz, 1 H).
Example 11 (7S)-5-(4-Ethoxyphenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt (E-11, Co. No. 81) [0278]
[0279] Copper(l) iodide (47 mg, 0.247 mmol) was added to a stirred suspension of intermediate 1-18 (0.1 g, 0.413 mmol), 4-iodophenetole (0.164 g, 0.661 mmol), K2C03 (114 mg, 0.825 mmol) and Λ/,Λ/’-dimethylethylenediamine (26 μί, 0.211 mmol) in toluene (6 mL) in a sealed tube and under nitrogen. The mixture was stirred at 100 °C for 24 h. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The organic layer was evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 40/60). The desired fractions were collected and concentrated in vacuo to yield compound 81 as an oil. The residue was dissolved in EtOAc and HCI (4N) (103 μΙ_, 0.413 mmol) was added. The residue was triturated from DIPE, filtered and dried in vacuo to yield compound 81 (163 mg, 99%) as a white solid. Free base: 1H NMR (300 MHz, CDCI3) δ ppm 1.35 (t, J= 6.9 Hz, 3 H) 1.65 (d, J=6.5 Hz, 3 H) 2.50 (s, 3 H) 3.84 (dd, J=12.9, 7.0 Hz, 1 H) 3.97 (q, J= 7.0 Hz, 2 H) 4.16 (dd, J=12.9, 4.3 Hz, 1 H) 4.60 - 4.76 (m, 1 H) 6.87 (br. d, J=8.8 Hz, 2 H) 7.18 (br. d, J= 8.7 Hz, 2 H) 7.43 (br. d, J=4.8 Hz, 1 H) 7.48 (br. s, 1 H) 7.72 (s, 1 H) 8.39 (br. d, J=4.3 Hz, 1 H); HCI salt: 1H NMR (300 MHz, DMSO-d6) δ ppm 1.34 (t, J= 6.9 Hz, 3 H) 1.60 (d, J=6.3 Hz, 3 H) 2.68 (s, 3 H) 3.38 (br. s., 1 H) 3.90 - 4.14 (m, 3 H) 4.28 (dd, J=13.0, 4.1 Hz, 1 H) 4.78 - 4.94 (m, 1 H) 7.00 (br. d, J= 8.9 Hz, 2 H) 7.35 (br. d, J= 8.8 Hz, 2 H) 8.23 - 8.42 (m, 3 H) 8.69 (d, J=6.3 Hz, 1 H).
Example 12 4-[(7S)-7-Methyl-4-oxo-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-3-yl]pyridine-2-carbonitrile (E-12, Co. No. 127) [0280]
[0281] Pd(PPh3)4 (42 mg, 0.036 mmol) was added to a stirred suspension of intermediate l-33a (250 mg, 0.593 mmol) and 4-bromopyridine-2-carbonitrile (162 mg, 0.884 mmol) in 1,4-dioxane (4 mL) and a sat. sol. of Na2C03 (2 mL). The mixture was stirred at 150 °C for 10 min under microwave irradiation. Then the mixture was diluted with H20 and extracted with DCM. The organic layer was dried (Na2S04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 50/50). The desired fractions were collected and evaporated in vacuo. The residue was precipitated with DIPE. The solid was filtered to yield compound 127 as a white solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.77 (d, J=6.4 Hz, 3 H) 4.05 (dd, J=12.9, 7.4 Hz, 1 H) 4.32 (dd, J=12.7, 4.0 Hz, 1 H) 4.79 - 4.88 (m, 1 H) 7.51 (br. d, J=8.4 Hz, 2 H) 7.74 (br. d, J=8.4 Hz, 2 H) 7.86 (s, 1 H) 7.92 (dd, J=5.2, 1.7 Hz, 1 H) 8.04 - 8.14 (m, 1 H) 8.67 (d, J=5.2 Hz, 1 H).
Example 13 (7S)-3-(2-lsopropyl-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one(E-13,Co. No. 126) [0282]
[0283] Pd(PPh3)4 (26 mg, 0.022 mmol) was added to a stirred suspension of intermediate l-33b (150 mg, 0.442 mmol) and 4-bromo-2-isopropyl-pyridine (prepared as described in W02009/118292) (97 mg, 0.486 mmol) in a sat. aq. sol. NaHC03 (1 mL) and 1,4-dioxane (1 mL). The mixture was stirred at 120 °C for 10 min under microwave irradiation. The mixture was filtered through diatomaceous earth and washed with DCM. The organic layer was washed with water, separated, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography ((silica; 7N solution of ammonia in MeOH in DCM 0/100 to 10/90) then (silica, EtOAc in DCM 0/100 to 30/70)) then by RP HPLC (Stationary phase: C18 XSelect™ 19x100mm5 μίτι, Mobile phase: Gradient from 80% 0.1 % NH4CO3H/NH4OH pH 9 solution in Water, 20% CH3CN to 0% 0.1% NH4C03H/NH40H pH 9 solution in Water, 100% CH3CN)). The desired fractions were collected and evaporated in vacuo to yield compound 126 as a white solid. 1H NMR (400 MHz, CDCI3) δ ppm 1.32 (d, J=6.9 Hz, 6 H) 1.75 (d, J= 6.7 Hz, 3 H) 3.08 (spt, J= 6.9 Hz, 1 H) 4.02 (dd, J=12.7, 7.2 Hz, 1 H) 4.31 (dd, J=12.6, 4.0 Hz, 1 H) 4.80 (quind, J= 6.7, 4.3 Hz, 1 H) 7.45 - 7.48 (m, 2 H) 7.51 (br. d, J= 8.3 Hz, 2 H) 7.71 (br. d, J= 8.6 Hz, 2 H) 7.81 (s, 1 H) 8.50 - 8.55 (m, 1 H)
Example 14 (7S)-5-(3,4-Dichlorophenyl)-3-[2-(hydroxymethyl)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-14, Co. No. 125) [0284]
[0285] A suspension of intermediate I-34 (1.09 g, 4.56 mmol) in acetic anhydride (8 mL) was stirred at 100 °Cfor2 h. The mixture was cooled to rt, and poured into water (15 mL) and EtOAc (30 mL). The organic layer was separated, washed with a sat. NaHCOs sol., dried (Na2S04), filtered and concentrated in vacuo. The resultant oil was stirred with lithium hydroxide (259 mg, 10.81 mmol) in MeOH (5.45 mL) and H20 (2.72 mL) at rt for 30 min. Then, EtOAc was added and the organic layer was washed with water, brine, dried (Na2S04), filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (silica; 7M solution of ammonia in MeOH in DCM 0/100 to 3/97). The desired fractions were collected and evaporated in vacuo to yield compound 125 (670 mg, 61 %).
Crude compound 125 (100 mg) was purified by RP HPLC (Stationary phase: C18 XBridge 30 x 100 mm 5 urn), Mobile phase: Gradientfrom 54% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 46% CH3CN to 64% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 36% CH3CN), yielding 72 mg compound 125.1H NMR (500 MHz, CDCI3) δ ppm 1.74 (d, J=6.4 Hz, 3 H) 3.73 (br. s„ 1 H) 3.96 (dd, J=12.7, 7.2 Hz, 1 H) 4.24 (dd, J=12.7, 4.3 Hz, 1 H) 4.72 - 4.83 (m, 3 H) 7.23 (dd, J=8.7, 2.3 Hz, 1 H) 7.49 (d, J=2.3 Hz, 1 H) 7.51 (d, J=8.7 Hz, 1 H) 7.55 (d, J=4.9 Hz, 1 H) 7.59 (s, 1 H) 7.82 (s, 1 H) 8.54 (d, J=4.9 Hz, 1 H).
Example 15 (7S)-5-(3,4-Dichlorophenyl)-3-[2-(1-hydroxyethyl)-4-pyridyl]-7-methyl-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-15, Co. No. 111) [0286]
[0287] Methylmagnesium chloride 3M in THF (249 μΙ_, 0.747 mmol) was added dropwise to a solution of intermediate 1-39 (150 mg, 0.374 mmol) in THF (1.24 mL) at -78 °C and under nitrogen. The mixture was stirred at -78 °C for 2 h. Then, more methylmagnesium chloride 3M in THF (125 μΙ_, 0.374 mmol) was added and the mixture was stirred at -78 °C for 1 h. Then, it was quenched at -78 °C with a sat. NH4CI sol. and allowed to reach rt. The mixture was extracted with EtOAc. The organic layer was separated, dried (MgS04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 5/95). The desired fractions were collected and evaporated in vacuo. The residue was precipitated with Ether/Heptane to yield compound 111 as a pale yellow solid. 1H NMR (400 MHz, CDCI3) δ ppm 1.52 (dd, J= 6.6, 0.8 Hz, 3 H) 1.74 (dd, J=6.5, 2.3 Hz, 3 H) 3.91 -4.02 (m, 1 H) 4.13 - 4.31 (m, 2 H) 4.72 - 4.84 (m, 1 H) 4.92 (q, J= 6.5 Hz, 1 H) 7.23 (dd, J= 8.6, 2.5 Hz, 1 H) 7.49 (d, J=2.3 Hz, 1 H) 7.51 (d, J= 8.6 Hz, 1 H) 7.56 (br. d, J=5.3 Hz, 1 H) 7.59 - 7.63 (m, 1 H) 7.82 (s, 1 H) 8.52 (dd, J=5.1,0.7 Hz, 1 H)
Example 16 (7S)-7-Methyl-3-(2-methyl-1-oxido-pyridin-1-ium-4-yl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-16, Co. No. 140) [0288]
[0289] 3-Chloroperoxybenzoic acid (2.96 g, 17.18 mmol) was added to a stirred solution of final compound E-1 (3.32 g, 8.59 mmol) in DCM (133 mL) at 0 °C. The mixture was allowed to reach rt and stirred at rt for 3 h. The mixture was treated with Na2C03 sat sol. and diluted with DCM. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo to yield compound 140 (3.4 g, 98%) as a pale yellow solid.
[0290] A small fraction (350 mg) was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 5/95). The desired fractions were collected and evaporated in vacuo. The residue was precipitated with Et20 and filtered to yield pure compound 140 (290 mg, 8%). 1H NMR (500 MHz, CDCI3) δ ppm 1.75 (d, J=6.4 Hz, 3 H) 2.53 (s, 3 H) 4.02 (dd, J=12.7, 7.2 Hz, 1 H) 4.30 (dd, J=12.7, 4.0 Hz, 1 H) 4.75 - 4.85 (m, 1 H) 7.50 (d, J=8.4 Hz, 2 H) 7.63 (dd, J=6.8, 2.5 Hz, 1 H) 7.67 - 7.77 (m, 3 H) 7.81 (s, 1 H) 8.21 (d, J= 6.6 Hz, 1 H).
Example 17 (7S)-5-[4-(1-Hydroxyethyl)phenyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-17, Co. No. 149) [0291]
[0292] Sodium borohydride (6 mg, 0.166 mmol) was added to a stirred solution of intermediate 1-35 (60 mg, 0.166 mmol) in MeOH (5 mL) at 0 °C. The mixture was stirred at rtfor 16 h. The solvent was concentrated in vacuo. The residue was dissolved with DCM and washed with a sat. Na2COs sol. The organic layer was separated, dried (MgS04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 100/0). The desired fractions were collected and concentrated in vacuo to yield compound 149 (40 mg, 66%). 1H NMR (300 MHz, CDCI3) δ ppm 1.52 (d, J=6.5 Hz, 3 H) 1.73 (d, J= 6.5 Hz, 3 H) 2.31 (br. s„ 1 H) 2.57 (s, 3 H) 3.97 (dd, J=12.8, 6.9 Hz, 1 H) 4.16 - 4.39 (m, 1 H) 4.60 - 4.86 (m, 1 H) 4.95 (q, J=6.4 Hz, 1 H) 7.35 (br. d, J= 8.2 Hz, 2 H) 7.42 - 7.59 (m, 4 H) 7.81 (s, 1 H) 8.37 - 8.49 (m, 1 H).
Example 18 (7S)-5-(4-Cyclopropylphenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[l,5-a]pyrazin-4-one (E-18, Co. No. 156) [0293]
[0294] Pd(PPh3)4 (37 mg, 0.032 mmol) was added to a stirred suspension of intermediate I-36 (255 mg, 0.642 mmol), cyclopropylboronic acid (165 mg, 1.92 mmol) and K2COs (177 mg, 1.28 mmol) in CH3CN (5 mL) and H20 (2 mL). The mixture was stirred at 150 °Cfor 10 min under microwave irradiation. Then more cyclopropylboronic acid (165 mg, 1.92 mmol) and Pd(PPh3)4 (37 mg, 0.032 mmol) were added. The mixture was stirred at 150 °C for another 10 min under microwave irradiation.Then the mixture was diluted with H20 and extracted with DCM. The organic layer was dried (MgS04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 0/100 to 75/25) and by RP HPLC (Stationary phase: C18XBridge™ 30 x 100 mm 5 urn, Mobile phase: Gradient from 67% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 33% CH3CN to 50% 0.1 % NH4C03H/NH40H pH 9 solution in Water, 50% CH3CN). The desired fractions were collected and concentrated in vacuo to yield compound 156 (80 mg, 24%) as a solid. 1H NMR (400 MHz, CDCI3) δ ppm 0.62-0.77 (m, 2 H) 0.91 -1.06 (m, 2 H) 1.72(d, J=6.7 Hz, 3 H) 1.91 (tt, J= 8.5, 5.1 Hz, 1 H) 2.56 (s, 3 H) 3.93 (dd, J=12.9, 6.9 Hz, 1 H) 4.25 (dd, J=12.9, 4.2 Hz, 1 H) 4.75 (quind, J= 6.6, 4.4 Hz, 1 H) 7.09 - 7.18 (m, 2 H) 7.18 - 7.25 (m, 2 H) 7.48 (dd, J=5.1, 1.2 Hz, 1 H) 7.53 (s, 1 H) 7.79 (s, 1 H) 8.45 (d, J=5.1 Hz, 1 H)
Example 19 (7S)-7-Methyl-3-(6-piperazin-1-yl-3-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-19, Co. No. 176) [0295]
[0296] Trifluoroacetic acid (0.911 mL, 11.91 mmol) was added to a stirred solution of intermediate I-42 (663 mg, 1.19 mmol) in DCM (1.9 mL). The mixture was stirred at rtfor 1 h. The solvent was concentrated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 20/80). The desired fractions were collected and evaporated In vacuo. The residue was precipitated with Et20 and filtrated a solid that was purified by RP HPLC (Stationary phase: C18 XBridge™ 50 x 100 5 μΓη, Mobile phase: Gradient from 53% 0.1% NH4C03H/NH40H pH 9 solution in Water, 43% CH3CN to 40% 0.1% NH4C03H/NH40H pH 9 solution in Water, 60% CH3CN), to yield compound 176 (151 mg, 28%) as a solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.73 (d, J=6.6 Hz, 3 H) 1.76 (br. s„ 1 H) 2.92 - 3.02 (m, 4 H) 3.50 - 3.57 (m, 4 H) 4.00 (dd, J=12.4, 7.2 Hz, 1 H) 4.27 (dd, J=12.4, 4.0 Hz, 1 H) 4.70 - 4.82 (m, 1 H) 6.63 (d, J= 8.7 Hz, 1 H) 7.50 (br. d, J= 8.7 Hz, 2 H) 7.68 (br. d, J=8.7 Hz, 2 H) 7.71 (s, 1 H) 7.96 (dd, J= 9.0, 2.3 Hz, 1 H) 8.42 (d, J=2.3 Hz, 1 H)
Example 20 (7S)-7-Methyl-3-(2-methyl-4-pyridyl)-5-[6-(trifluoromethyl)-3-pyridyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-20, Co. No. 186) [0297]
[0298] Pd(PPh3)4(155 mg, 0.134 mmol) was added to a stirred suspension of intermediate 1-18(325 mg, 1.341 mmol), 2-chloro-5-(trifluoromethyl)pyridine (365 mg, 2.012 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (155 mg, 0.268 mmol), Cs2C03 (874 mg, 2.683 mmol) in 1,4-dioxane (10 mL) in a sealed tube and under nitrogen. The mixture was stirred at 120 °C for 7 h. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The filtrate was evaporated in vacuo and the crude product was purified by flash column chromatography (silica, EtOAc in DCM 0/100 to 30/70). The desired fractions were collected and concentrated in vacuo. The residue was purified by ion exchange chromatography using an (ISOLUTE® SCX2 cartridge) eluting first with MeOH then with 7M solution of ammonia in MeOH. The desired fractions contained in the 7M solution of ammonia in MeOH were collected and the solvents evaporated in vacuo. The residue was triturated with Et20 to yield compound 186 (415 mg, 80%) as a white solid. 1H NMR (500 MHz,, CDCI3) δ ppm 1.73 (d, J=6.4 Hz, 3 H) 2.60 (s, 3 H) 4.35 - 4.44 (m, 1 H) 4.68 - 4.80 (m, 2 H) 7.41 (dd, J=5.2, 1.2 Hz, 1 H) 7.45 (s, 1 H) 7.78 (s, 1 H) 7.94 (dd, J=8.8, 2.2 Hz, 1 H) 8.24 (d, J= 9.0 Hz, 1 H) 8.52 (d, J= 5.2 Hz, 1 H) 8.69 - 8.73 (m, 1 H).
Example 21 (7S)-7-Methyl-3-(2-methyl-4-pyridyl)-5-[6-methyl-5-(trifluoromethyl)-2-pyridyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-21, Co. No. 192) [0299]
[0300] Tetramethyltin (32 μι, 0.231 mmol) was added to a mixture of intermediate I-43 (65 mg, 0.154 mmol), lithium chloride (13 mg, 0.308 mmol) and bis(triphenylphosphine)palladium(ll) dichloride (6 mg, 0.007 mmol) in degassed DMF (2.4 mL), in a sealed tube under nitrogen. The mixture was stirred at 110 °C for 5 h. The mixture was diluted with a sat. sol. of NaHCQ3 and extracted with EtOAc. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 30/70). The desired fractions were collected and concentrated in vacuo. Then the product was triturated with Et20 to yield compound 192 (26 mg, 42%) as a white solid. 1H NMR (400 MHz, CDCI3) δ ppm 1.73 (d, J= 6.5 Hz, 3 H) 2.60 (s, 3 H) 2.69 (br. d, J= 1.6 Hz, 3 H) 4.34 - 4.46 (m, 1 H) 4.66 - 4.80 (m, 2 H) 7.41 (dd, J=5.1, 1.2 Hz, 1 H) 7.44 (s, 1 H) 7.78 (s, 1 H) 7.90 (d, J=8.8 Hz, 1 H) 8.02 (d, J=8.6 Hz, 1 H) 8.52 (d, J=4.9 Hz, 1 H).
Example 22a (7S)-5-[6-Ethoxy-5-(trifluoromethyl)-2-pyridyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-22a, Co. No. 189) [0301]
[0302] EtOH (114 μΙ_, 1.95 mmol) was added to a stirred suspension of sodium hydride (60% dispersion in mineral oil) (78 mg, 1.95 mmol) in DMF (5 mL) at 0 °C. The mixture was stirred at rt for 10 min. Then, a solution of 2-chloro-4-iodo-5-(trifluoromethyl)pyridine (0.5 g, 1.62 mmol) in DMF (5 mL) was added at 0 °C and the mixture was stirred at rt for 18 h. Then, more sodium hydride (60% dispersion in mineral oil) (26 mg, 0.65 mmol) and EtOH (38 μί, 0.65 mmol) were added at 0 °C and the mixture was stirred at rt for 2 h. Intermediate 1-18 (157 mg, 0.65 mmol) was then added and the mixture was cooled to 0 °C. More sodium hydride (60% dispersion in mineral oil) (52 mg, 1.301 mmol) was added and the mixture was stirred at rtfor 1 h and at 80 °C for 16 h. Then more sodium hydride (60% dispersion in mineral oil) (13 mg, 0.325 mmol) was added at rt and the mixture was stirred at 80 °C for2 h more. The mixture was treated with a 10% NH4CI sol. and extracted with EtOAc/THF. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The residue was dissolved in DMF (10 mL). TEA (0.226 mL, 1.626 mmol) and HATU (0.247 g, 0.605 mmol) were added. The mixture was stirred at rtfor 1 h. The mixture was treated with a sat. NaHCOs sol./brine at 0 °C and extracted with EtOAc. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 100/0). The desired fractions were collected and concentrated in vacuo. Then the residue was repurified by RP HPLC (Stationary phase: C18 XBridge™ 30 x 100 mm 5 urn, Mobile phase: Gradient from 54% 0.1% NH4C03H/NH40H pH 9 solution in Water, 46% CH3CN to 64% 0.1% NH4C03H/NH40H pH 9 solution in Water, 36% CH3CN) to yield compound 189 (27 mg, 4%) as a beige solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.46 (t, J= 6.9 Hz, 3 H) 1.71 (d, J=6.6 Hz, 3 H) 2.60 (s, 3 H) 4.15 - 4.27 (m, 2 H) 4.39 (dd, J=13.3, 7.2 Hz, 1 H) 4.65 - 4.80 (m, 2 H) 7.40 (dd, J= 5.2, 1.2 Hz, 1 H) 7.43 (s, 1 H) 7.77 (s, 1 H) 7.82 (s, 1 H) 8.47 (s, 1 H) 8.52 (d, J= 5.2 Hz, 1 H).
Example 22b (7S)-5-[4-Chloro-5-(trifluoromethyl)-2-pyridyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-22b, Co. No. 204) [0303]
[0304] Pd(PPh3)4 (47 mg, 0.041 mmol) and 2,4-dichloro-5-(trifluoromethyl)pyridine (134 mg, 0.619 mmol) were added to a stirred suspension of intermediate 1-18 (100 mg, 0.413 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (48 mg, 0.082 mmol) and Cs2C03 (269 mg, 0.082 mmol) in 1,4-dioxane (2.5 mL) in a sealed tube and under nitrogen. The mixture was stirred at 110 °C for 4 h and at 100 °C for 2 days. The mixture was filtered through a pad of diatomaceous earth and washed with DCM. The filtrate was evaporated in vacuo. The residue was dissolved in DMF (7 mL) and TEA (57 μί, 0.413 mmol) then HATU (157 mg, 0.413 mmol) were added. The mixture was stirred at rtfor 2 h. The mixture was treated at 0 °C with a sat. sol. NaHC03/brine then EtOAc was added. The mixture was filtered through a pad of diatomaceous earth. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 100/0). The desired fractions were collected and concentrated in vacuo. The residue was repurified by RP HPLC (Stationary phase: C18 XBridge™ 30 x 100 mm 5 urn, Mobile phase: Gradient from 54% 0.1% NH4C03H/NH40H pH 9 solution in Water, 46% CH3CN to 64% 0.1% NH4C03H/NH4OH pH 9 solution in Water, 36% CH3CN) to yield compound 204 (27 mg, 15%) as a white solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.72 (d, J=6.4 Hz, 3 H) 2.61 (s, 3 H) 4.36 - 4.45 (m, 1 H) 4.68 - 4.79 (m, 2 H) 7.39 (dd, J=5.2, 1.7 Hz, 1 H) 7.43 (s, 1 H) 7.78 (s, 1 H) 8.42 (s, 1 H) 8.54 (d, J= 5.2 Hz, 1 H) 8.68 (s, 1 H).
Example 23 (7S)-3-(2-lodo-4-pyridyl)-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one(E-23,Co. No. 225) [0305]
[0306] Acetyl chloride (84 μί, 1.18 mmol) was added to a stirred suspension of intermediate 1-26 (320 mg, 0.786 mmol) and Nal (1.18 g, 7.866 mmol) in CH3CN (12.8 mL) at rt. The mixture was stirred at 120 °C for 30 min under MW irradiation. Then the mixture was diluted with EtOAc and washed with a sat. sol. of Na2S203 and brine. The organic layer was separated, dried (Na2S04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 0/100 to 60/40). The desired fractions were collected and evaporated in vacuo to yield compound 225 (289 mg, 74%). 1H NMR (400 MHz, CDCI3) δ ppm 1.75 (d, J= 6.5 Hz, 3 H) 4.02 (dd, J=12.8, 7.3 Hz, 1 H) 4.30 (dd, J=12.7, 4.2 Hz, 1 H) 4.80 (quind, J= 6.7, 4.2 Hz, 1 H) 7.50 (br. d, J=8.3 Hz, 2 H) 7.67 (dd, J=5.1, 1.6 Hz, 1 H) 7.72 (br. d, J= 8.3 Hz, 2 H) 7.80 (s, 1 H) 8.03 - 8.05 (m, 1 H) 8.32 (dd, J= 5.2, 0.6 Hz, 1 H)
Example 24 (7S)-7-Methyl-3-(2-piperazin-1-yl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one hydrochloride salt (E-24, Co. No. 175) [0307]
HC1 [0308] Compound 175 was obtained starting from intermediate l-33a (200 mg, 0.474 mmol), 1-(4-bromo-2-pyridyl)pip-erazine (CAS: 1201643-59-5, 157 mg, 0.649 mmol, 1.06 mmol), Pd(PPh3)4 (34 mg, 0.029 mmol) in 1,4-dioxane (4 mL) and a sat. sol. of Na2C03 (2 mL), following a procedure similar to that described in E-12, then treatment with a solution of HCI 5N in /PrOH, yielded compound 175 (224 mg, 84%) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.59 (d, J= 6.5 Hz, 3 H) 3.22 (br. s„ 4 H) 3.83 (br. s., 4 H) 4.10 (dd, J=12.9, 7.4 Hz, 1 H) 4.39 (dd, J=12.9, 4.2 Hz, 1 H) 4.81 - 4.92 (m, 1 H) 7.29 (br. d, J=4.4 Hz, 1 H) 7.56 (br. s„ 1 H) 7.69 (br. d, J=8.6 Hz, 2 H) 7.84 (br. d, J=8.6 Hz, 2 H) 8.11 (d, J= 5.8 Hz, 1 H) 8.19 (br. s, 1 H) 9.20 (br. s., 2 H).
Example 25 (7S)-3-[2-(4-Acetylpiperazin-1-yl)-4-pyridyl]-7-methyl-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-25, Co. No. 106) [0309]
[0310] Acetyl chloride (4 μί, 0.060 mmol) was added to a solution of compound 175 (25 mg, 0.054 mmol) and TEA (16 μι, 0.115 mmol) in DCM (1 mL) under nitrogen. The mixture was stirred at rt for 5 h. Then the mixture was diluted with HCI 0.1 N and extracted with DCM. The organic layer was separated, dried (MgS04), filtered and the solvent evaporated in vacuo. The crude product was purified by flash column chromatography (silica; MeOH in DCM 0/100 to 05/95). The desired fractions were collected and concentrated in vacuo to yield compound 106 (17 mg, 62%) as a solid. 1H NMR (400 MHz, CDCI3) δ ppm 1.75 (d, J=6.5 Hz, 3 H)2.13(s, 3 H) 3.49-3.55 (m, 2 H) 3.55-3.61 (m, 2 H) 3.61 -3.67 (m, 2 H) 3.70 - 3.77 (m, 2 H) 4.01 (dd, J=12.7, 6.9 Hz, 1 H) 4.30 (dd, J=12.7, 4.2 Hz, 1 H) 4.79 (quind, J=6.6, 4.4 Hz, 1 H) 6.94 (dd, J=5.2, 1.3 Hz, 1 H)7.17(br. s, 1 H) 7.46 - 7.55 (m, 2 H) 7.66 - 7.76 (m, 2 H) 7.80 (s, 1 H) 8.19 (dd, J= 5.2, 0.6 Hz, 1 H).
Example 26 7-(Difluoromethyl)-3-(2-methyl-4-pyridyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-26, Co. No. 181) [0311]
[0312] Compound 181 was obtained starting from intermediate I-46 (71 mg, 0.169 mmol) using Pd/C 10% (36 mg, 0.033 mmol) in EtOH (3 mL) under H2 atmospheric pressure, following a procedure similar to that described in E-6, yielding compound 181 (13 mg, 19%) as a white solid. 1H NMR (400 MHz, CDCI3) δ ppm 2.58 (s, 3 H) 4.36 (dd, J=13.6, 3.2 Hz, 1 H) 4.61 (ddd, J=13.6, 5.0, 1.3 Hz, 1 H) 4.88 - 4.99 (m, 1 H) 6.21 -6.55 (m, 1 H) 7.45 (dd, J=5.2, 1.3 Hz, 1 H) 7.47 - 7.54 (m, 3 H) 7.73 (br. d, J= 8.3 Hz, 2 H) 7.89 (s, 1 H) 8.50 (d, J=5.1 Hz, 1 H).
Example 27 (7S)-7-Methyl-3-[2-(methylamino)-4-pyridyl]-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-27, Co. No. 147) [0313]
[0314] Compound 147 was obtained starting from intermediate l-33a (1.5 g, 3.561 mmol), 4-bromo-/\/-methyl-pyridin-2-amine (799 mg, 4.273 mmol, 1.06 mmol), Pd(PPh3)4 (206 mg, 0.178 mmol) in 1,4-dioxane (8.1 mL) and a sat. sol. of Na2C03 (8.2 mL), following a procedure similar to that described in E-12, yielding compound 147 (1.14 g, 80%) as a white solid. 1H NMR (500 MHz, CDCI3) δ ppm 1.74 (d, J=6.4 Hz, 3 H) 2.93 (d, J= 5.2 Hz, 3 H) 4.00 (dd, J=12.6, 7.1 Hz, 1 H) 4.29 (dd, J=12.7, 4.0 Hz, 1 H) 4.54 (br. d, J=3.2 Hz, 1 H) 4.73 - 4.82 (m, 1 H) 6.84 (s, 1 H) 6.86 (d, J=5.2 Hz, 1 H) 7.50 (br. d, J=8.4 Hz, 2 H) 7.70 (br. d, J=8.4 Hz, 2 H) 7.79 (s, 1 H) 8.09 (d, J= 5.2 Hz, 1 H).
Example 28 (7S)-5-[4-lodo-5-(trifluoromethyl)-2-pyridyl]-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (E-28, Co. No. 212) [0315]
[0316] Intermediate 1-18 (320 mg, 1.32 mmol) was added portionwise to a stirred suspension of sodium hydride (60% dispersion in mineral oil, 78 mg, 1.98 mmol) in DMF (5 mL) at rt. The mixture was stirred at rt for 15 min and a solution of 2-chloro-4-iodo-5-(trifluoromethyl)pyridine (446 mg, 1.453 mmol) in DMF (5 mL) was added at rt. The mixture was stirred at 80 °C for 16 h. Then more sodium hydride (60% dispersion in mineral oil, 27 mg, 0.66 mmol) was added at rt and the mixture was stirred at 80 °C for 1 h. The mixture was treated with 10% NH4CI sol./brine and extracted with EtOAc. The organic layer was separated, washed with brine, dried (Na2S04), filtered and the solvents evaporated In vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in DCM 0/100 to 100/0). The desired fractions were collected and the solvents concentrated in vacuo to yield compound 212 (220 mg, 32%) as a white foam.
Example 29 (7S)-5-(3,4-Dichloro-2-iodo-phenyl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (Co. No. 220) and (7S)-5-(3,4-dichloro-6-iodo-1-methyl-cyclohexa-1,3,5-trien-1-yl)-7-methyl-3-(2-methyl-4-pyridyl)-6,7-dihydro-pyrazolo[1,5-a]pyrazin-4-one (Co. No. 221) (E-29, Co. No. 220 and Co. No. 221) [0317]
[0318] HATU (89 mg, 0.235 mmol) was added to a stirred solution of mixture intermediate compounds l-63a and I-63b (250 mg, 0.235 mmol) and TEA (65 μι, 0.471 mmol) in DMF (3 mL). The mixture was stirred at rt for 2 h. The mixture was treated with a sat. sol. of NH4CI and a sat. sol. of NaHC03 and extracted with EtOAc. The organic layer was separated, dried (MgS04), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (silica; EtOAc in Heptane 0/100 to 90/10). The desired fractions were collected and the solvents concentrated in vacuo to give two fractions that were triturated with DCM/Heptane to yield compound 220 (55 mg, 45%) and compound 221 (20 mg, 16%) as solids. Compound 220: 1H NMR (500 MHz, CDCI3) δ ppm 1.78 (d, J=6.4 Hz, 2 H) 1.85 (d, J=6.6 Hz, 1 H) 2.58 (s, 3 H) 3.82 (dd, J=12.7, 9.2 Hz, 0.65 H) 3.97 (dd, J=12.7, 7.2 Hz, 0.35 H) 4.00 - 4.07 (m, 1 H) 4.76 - 4.84 (m, 0.35 H) 4.92 - 5.00 (m, 0.65 H) 7.39 (s, 0.35 H) 7.40 (s, 0.65 H) 7.48 (br. d, J= 5.2 Hz, 1 H) 7.52 - 7.56 (m, 1 H) 7.82 (s, 1 H) 8.03 (s, 0.35 H) 8.03 (s, 0.65 H) 8.48 (d, J=5.2 Hz, 1 H); compound 221: 1H NMR (500 MHz, CDCI3) δ ppm 1.78 (d, J= 6.6 Hz, 2 H) 1.85 (d, J=6.6 Hz, 1 H) 2.57 (s, 3 H) 3.81 (dd, J=12.7, 9.0 Hz, 0.65 H) 3.98 (dd, J=12.7, 4.6 Hz, 0.35 H) 4.02 - 4.11 (m, 1 H) 4.76 - 4.84 (m, 0.35 H) 4.95 - 5.04 (m, 0.65 H) 7.16 (d, J=8.7 Hz, 0.35 H) 7.17 (d, J=8.7 Hz, 0.65 H) 7.49 (br. d, 5.2 Hz, 1 H) 7.52 - 7.61 (m, 2 H) 7.83 (s, 0.65 H) 7.83 (s, 0.35 H) 8.47 (d, J= 5.2 Hz, 1 H).
[0319] The following final compounds were synthesized by following an analogous synthetic procedure as reported for compound 1 (E-1) followed by the procedure for intermediates 1-18 and 1-19 when needed.
(continued)
I
I
I
I
[0320] The following compound was synthesized by following the sequence of an analogous synthetic procedure as reported for intermediate 1-22 starting from intermediate 1-14 and 1-bromo-3,4-dichlorobenzene, followed by the procedure for intermediates I-23 then following an analogous synthetic procedure as reported for compound 2 (E-2) using 2-, methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine.
Example 30 3-(2-Methyl-4-pyridyl)-7-(trifluoromethyl)-5-[4-(trifluoromethyl)phenyl]-6,7-dihydropyrazolo[1,5-a]pyrazin-4-one (Co.
No. 238) [0321]
[0322] Compound 238 (E-30) was obtained starting from intermediate I-68 (20 mg, 0.042 mmol), 2-picoline-4-boronic acid (8 mg, 0.059 mmol), Pd(PPh3)4 (2 mg, 0.002 mmol) in 1,4-dioxane (0.4 mL) and a sat. sol. of NaHC03 (0.4 mL) following a procedure similar to that described in E-12 and purified by RP HPLC ((Stationary phase: C18 XBridge 30 x 100 5 urn), (Mobile phase: Gradient from 60% 0.1% NH4C03H/NH40H pH 9 solution in Water, 40% CH3CN to 43% 0.1% NH4C03H/NH40H pH 9 solution in Water, 57% CH3CN)), yielding compound 238 (14 mg, 75%). 1H NMR (500 MHz, CDCI3) δ ppm 2.53 (s, 3 H) 4.23 (dd, J=14.0, 1.3 Hz, 1 H) 4.67 - 4.74 (m, 1 H) 5.07 - 5.14 (m, 1 H) 7.38 - 7.43 (m, 3 H) 7.47 (s, 1 H) 7.67 (br. d, J=8.4 Hz, 2 H) 7.87 (s, 1 H) 8.44 (d, J=5.2 Hz, 1 H).
[0323] Tables 1 a (compounds 1 -87) and 1 b (with an alternative representation for compounds 1 -87) below list additional compounds of Formula (I).
[0324] Tables 1a and 1b. The following compounds were prepared following the methods exemplified in the Experimental Part (Ex. No.). Compounds exemplified and described in the experimental part are marked with an asterisk *. The work-up for compounds synthesized by an analogous procedure to E1 can be performed either by filtration through a pad of diatomaceous earth or by extraction with an organic solvent, washing with aqueous ammonia. The coupling agent used in the synthesis of compounds synthesized by an analogous procedure to E2 was either a boronic acid or a boronic ester. For some compounds the stereochemical configuration has been designated as *R or *S when the absolute stereochemistry is undetermined although the compound itself has been isolated as a single stereoisomer and is enantiomerically pure.
Table 1a
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
Table 1b
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
(continued)
[0325] The values of salt stoichiometry or acid content in the compounds as provided herein, are those obtained experimentally and may vary when using different analytical methods. The content of hydrochloric acid reported herein was determined by 1H NMR integration and/or elemental analysis. For compound 1 the salt stoichiometry was determined by ion chromatography (hydrochloride and sulfate salts) and by NMR (methanesulfonate and maleate salts).
Analytical part
Melting points [0326] Values are peak values, and are obtained with experimental uncertainties that are commonly associated with this analytical method.
Mettler FP 62 (A): For a number of compounds, melting points were determined in open capillary tubes on a Mettler FP62 apparatus. Melting points were measured with a temperature gradient of 3 or 10 °C/minute. Maximum temperature was 300 °C. The melting point was read from a digital display.
Mettler FP 62 (A1): Melting points (m.p.) were determined in open capillary tubes on a Mettler FP62 apparatus. Melting points were measured with a temperature ranging from 50°C to 300°C , using a gradient of 10 °C/minute. The melting point value was read from a digital display.
Mettler FP 81HT / FP90 (B): For a number of compounds, melting points were determined in open capillary tubes on a FP 81 HT / FP90 apparatus (Mettler-Toledo). Melting points were measured with a temperature gradient of 1, 3, 5 or 10 °C/minute. Maximum temperature was 300 °C. The melting point was read from a digital display. Mettler Toledo MP50 (C): For a number of compounds, melting points were determined in open capillary tubes on a Mettler Toledo MP50. Melting points were measured with a temperature gradient of 10 °C/minute. Maximum temperature was 300 °C. The melting point data was read from a digital display and checked from a video recording system. DSC823e (D): For a number of compounds, melting points (m.p.) were determined with a DSC823e (Mettler-Toledo). Melting points were measured with a temperature gradient of 30 °C/minute. Maximum temperature was 400 °C. Peak values were recorded.
LCMS
General procedure [0327] The High Performance Liquid Chromatography (HPLC) measurement was performed using a LC pump, a diode-array (DAD) or a UV detector and a column as specified in the respective methods. If necessary, additional detectors were included (see table of methods below).
Flow from the column was brought to the Mass Spectrometer (MS) which was configured with an atmospheric pressure ion source. It is within the knowledge of the skilled person to set the tune parameters (e.g. scanning range, dwell time...) in order to obtain ions allowing the identification of the compound’s nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate software. Compounds are described by their experimental retention times (Rt) and ions. If not specified differently in the table of data, the reported molecular ion corresponds to the [M+H]+ (protonated molecule) and/or [M-H]- (deprotonated molecule). In case the compound was not directly ionizable the type of adduct is specified (i.e. [M+NH4]+, [M+HCOO]-, [M+CH3COO]" etc...). For molecules with multiple isotopic patterns (Br, Cl..), the reported value is the one obtained for the lowest isotope mass. All results were obtained with experimental uncertainties that are commonly associated with the method used.
[0328] Hereinafter, "LCT" means LC-Time of Flight, "SQD" Single Quadrupole Detector, "MSD" Mass Selective Detector, "QTOF" Quadrupole-Time of Flight, "RT" room temperature, "BEH" bridged ethylsiloxane/silica hybrid, "DAD" Diode Array Detector.
Table 2. LC-MS Methods (Flow expressed in mL/min; column temperature (T) in °C; Run time in minutes).
(continued)
(continued)
Table 3a. Analytical data - melting point (M.p.) and LCMS: [M+H]+ means the protonated mass of the free base of the compound, [M-H]- means the deprotonated mass of the free base of the compound or the type of adduct specified [M+CH3COO]·). Rt means retention time (in min). For some compounds, exact mass was determined.
(continued)
(continued)
(continued)
(continued)
(continued)
Table 3b. Analytical data - melting point (M.p.) and LCMS: [M+H]+ means the protonated mass of the free base of the compound, Rt means retention time (in min), method refers to the method used for LCMS.
(continued)
(continued)
(continued)
SFC-MS
General procedure [0329] The SFC measurement was performed using Analytical system from Berger instrument comprising a FCM-1200 dual pump fluid control module for delivering carbon dioxide (C02) and modifier, a CTC Analytics automatic liquid sampler, a TCM-20000 thermal control module for column heating from room temperature to 80°C. An Agilent 1100 UV photodiode array detector equipped with a high-pressure flow cell standing up to 400 bars was used. Flow from the column was split to a MS spectrometer. The MS detector was configured with an atmospheric pressure ionization source.The following ionization parameters for the Waters ZQ mass spectrophotometer are: corona: 9μθ, source temp: 140°C, cone: 30 V, probe temp 450°C, extractor 3 V, desolvatation gas 400L/hr, cone gas 70 L/hr. Nitrogen was used as the nebulizer gas. Data acquisition was performed with a Waters-Micromass MassLynx-Openlynx data system.
Table 4. Analytical SFC-MS Methods (Flow expressed in mL/min; column temperature (T) in °C; Pressure in Mpa).
Table 5. Analytical SFC data - Rt means retention time (in minutes), [M+H]+ means the protonated mass of the compound, method refers to the method used for SFC/MS analysis of enantiomerically pure compounds. The measurement was compared against the mixture.
(continued)
Optical Rotations [0330] Optical rotations were measured on a Perkin-Elmer341 polarimeter with a sodium lamp and reported as follows: [α]° (λ, c g/100ml, solvent, T°C).
[α]λτ = (100a)/(/xc) : where/is the path length in dm and c is the concentration ing/100 ml for a sample at a temperature T (°C) and a wavelength λ (in nm). If the wavelength of light used is 589 nm (the sodium D line), then the symbol D might be used instead. The sign of the rotation (+ or-) should always be given. When using this equation the concentration and solvent are always provided in parentheses after the rotation. The rotation is reported using degrees and no units of concentration are given (it is assumed to be g/100 ml).
Table 6. Optical Rotation data.
(continued)
(continued)
(continued)
(continued)
(continued)
NMR
[0331] Co. No. 237: 1H NMR (500 MHz, CDCI3) δ ppm 1.72 (d, J= 6.6 Hz, 3 H) 2.96 (d, J= 5.2 Hz, 3 H) 4.05 (s, 3 H) 4.42 (dd, J=13.7, 7.1 Hz, 1 H) 4.53 - 4.59 (m, 1 H) 4.64 (dd, J=13.6, 4.0 Hz, 1 H) 4.70 - 4.78 (m, 1 H) 6.76 (s, 1 H) 6.84 (d, J= 5.2 Hz, 1 H) 7.78 (d, J= 8.7 Hz, 1 H) 7.77 (s, 1 H) 7.88 (d, J=8.4 Hz, 1 H) 8.13 (d, J=5.2 Hz, 1 H)
Pharmacological examples [0332] The compounds provided in the present invention are negative allosteric modulators of mGluR2. These compounds appear to inhibit glutamate responses by binding to an allosteric site other than the glutamate binding site. The response of mGluR2 to a concentration of glutamate is decreased when compounds of Formula (I) are present. Compounds of Formula (I) are expected to have their effect substantially at mGluR2 by virtue of their ability to reduce the function of the receptor. The effects of negative allosteric modulators tested at mGluR2 using the [35S]GTPyS binding assay method described below and which is suitable for the identification of such compounds, and more particularly the compounds according to Formula (I), are shown in Table 7. 1) [35S]GTPyS binding assay [0333] The [35S]GTPyS binding assay is a functional membrane-based assay used to study G-protein coupled receptor (GPCR) function whereby incorporation of a non-hydrolysable form of GTP, [35S]GTPyS (guanosine 5’-triphosphate, labelled with gamma-emitting 35S), is measured. The G-protein a subunit catalyzes the exchange of guanosine 5’-diphosphate (GDP) by guanosine triphosphate (GTP) and on activation of the GPCR by an agonist, [35S]GTPyS, becomes incorporated and cannot be cleaved to continue the exchange cycle (Harper (1998) Current Protocols in Pharmacology 2.6.1-10, John Wiley &amp; Sons, Inc.). The amount of radioactive [35S]GTPyS incorporation is a direct measure of the activity of the G-protein and hence the activity of the antagonist can be determined. mGlu2 receptors are shown to be preferentially coupled to Gai-protein, a preferential coupling for this method, and hence it is widely used to study receptor activation of mGlu2 receptors both in recombinant cell lines and in tissues. Here we describe the use of the [35S]GTPyS binding assay using membranes from cells transfected with the human mGlu2 receptor and adapted from Schaffhauser et al. (Molecular Pharmacology, 2003, 4:798-810) for the detection of the negative allosteric modulation (NAM) properties of the compounds of this invention.
Membrane preparation [0334] CHO-cells were cultured to pre-confluence and stimulated with 5 mM butyrate for 24 h. Cells were then collected by scraping in PBS and cell suspension was centrifuged (10 min at 4000 RPM in benchtop centrifuge). Supernatant was discarded and pellet gently resuspended in 50 mM Tris-HCI, pH 7.4 by mixing with an Ultra Turrax homogenizer. The suspension was centrifuged at 12,400 RPM (Sorvall F14S-6x250Y) for 10 minutes and the supernatant discarded. The pellet was homogenized in 5 mM Tris-HCI, pH 7.4 using an Ultra Turrax homogenizer and centrifuged again (13,000 RPM, 20 min, 4 °C). The final pellet was resuspended in 50 mM Tris-HCI, pH 7.4 and stored at -80 °C in appropriate aliquots before use. Protein concentration was determined by the Bradford method (Bio-Rad, USA) with bovine serum albumin as standard. P5S] GTPyS binding assay [0335] Measurement of mGluR2 negative allosteric modulatory activity of test compounds was performed as follows. Test compounds and glutamate were diluted in assay buffer containing 10 mM HEPES acid, 10 mM HEPES salt, pH 7.4, 100 mM NaCI, 3 mM MgCI2 and 10 μΜ GDP. Human mGlu2 receptor-containing membranes were thawed on ice and diluted in assay buffer supplemented with 18 μg/ml saponin. Membranes were pre-incubated with compound together with a predefined (~EC80) concentration of glutamate (60 μΜ) for 30 min at 30 °C. After addition of [35S]GTPyS (f.c. 0.1 nM), assay mixtures were shaken briefly and further incubated to allow [35S]GTPyS incorporation on activation (30 minutes, 30 °C). Final assay mixtures contained 7 μς of membrane protein in 10 mM HEPES acid, 10 mM HEPES salt, pH 7.4, 100 mM NaCI, 3 mM MgCI2, 10 μΜ GDP and 10 μg/ml saponin. Total reaction volume was 200 μΙ. Reactions were terminated by rapid filtration through Unifilter-96 GF/B plates (Perkin Elmer, Massachusetts, USA) using a 96-well filtermate universal harvester. Filters were washed 6 times with ice-cold 10 mM NaH2PO4/10 mM Na2HP04, pH 7.4. Filters were then air-dried, and 30 μΙ of liquid scintillation cocktail (Microscint-O) was added to each well. Membrane-bound radioactivity was counted in a Topcount.
Data analysis [0336] The concentration-response curves of representative compounds of the present invention were generated using the Lexis software interface (developed at J&amp;J). Data were calculated as % of the control glutamate response, defined as the response that is generated upon addition of an EC80-equivalent concentration of glutamate. Sigmoid concentration-response curves plotting these percentages versus the log concentration of the test compound were analyzed using non-linear regression analysis. The concentration producing half-maximal inhibition was calculated as the IC50.
The plC50 values were calculated as the -log IC50, when the IC50 is expressed in M. Emax is defined as the relative maximal effect (i.e. maximal % inhibition relative to the control glutamate response).
Table 7. Pharmacological data for compounds according to the invention.
n.t. means not tested
2) Reversal of the effect of the mGluR2 PAM JNJ-42153605 on scopolamine-induced hyperlocomotion
Apparatus [0337] Motor activity was measured in microprocessor-based motor activity arenas (closed gray PVC cylinders with a height of 39 cm and a diameter of 31 cm). Each arena was placed on an infrared LED (8x8 LEDs) lit box (white PVC squared box; 40 x 40 cm2; height 12.5 cm. An infrared-sensitive tube camera and a white light source were mounted to the ceiling above the observation chamber to track the animal. The total distance traveled (cm) was recorded and analyzed using the Noldus Ethovision XT Video Tracking System (Version 7.0.418; Noldus, Wageningen, The Netherlands). The intensity of the light within the activity cages (measured in the centre at the level of the floor) ranged between 4 and 8 LUX.
General Procedure [0338] The rats were pretreated with test compound or vehicle at 60 min before the start of the activity recordings and placed into individual cages. The rats were challenged with JNJ-42153605 (3-(cyclopropylmethyl)-7-(4-phenylpiperidin-1-yl)-8-(trifluoromethyl)[1,2,4]triazolo[4,3-a]pyridine; WO2010/130424; Cid et al. J. Med. Chem. 2012, 55, 8770-8789) (20 mg/kg, i.v.) 30 min before the start of the activity recording combined with scopolamine (0.16 mg/kg, i.v.) just before the start of the activity measurements. Immediately after the injection of scopolamine, the rats were placed into the activity monitors and total distance travelled over the first 30 min was measured.
Solvent-pretreated control rats.
[0339] Frequency distributions obtained in historical series of solvent-pretreated control rats are given in figure 1 and Table 8 below. Animals receiving the combination of JNJ-42153605 and scopolamine (n = 433) almost always travelled a distance of less than 1500 cm (< 1500 cm) (only 2.5% of the control rats travelled a distance of more than 1500 cm (> 1500 cm)). On the other hand, animals challenged with scopolamine alone (n = 215) always travelled a total distance of more than 1500 cm (> 1500 cm) and almost always (in 95.8% of the rats) a distance of more than 4400 cm (> 4400 cm). Rats that did not receive any challenge travelled almost always a distance of more than 1500 cm (> 1500 cm) (in 93.3% of the rats) and less than 4400 cm (< 4400 cm) (in 98.9% of the rats). For reversal of the inhibitory effect of JNJ-42153605 on the scopolamine-induced hyperlocomotion, two all-or-none criteria were adopted: (1) reversal: total distance > 1500 cm; (2) normalization: total distance > 4400 cm.
The results on the reversal of the effect of JNJ-42153605 are shown in table 9 below.
Table 8. Frequency distributions obtained in historical series of solvent-pretreated control rats. Ntested means number of animals tested.
Table 9. Reversal of the effect of JNJ 42153605 on scopolamine-induced hyperlocomotion. ED50 means effective dose; PO means oral route; SC means subcutaneous route.
_
3) V-maze test [0340] The V-maze-test is a two-trial shortterm visual-spatial working memory task based on spontaneous exploration of a new and a familiar arm in a 2-arm maze (Embrechts et al. (2013) "Longitudinal characterization of the TauPS2APP mouse model of Alzheimer’s disease in a two trial discrimination taskofvisuo-spatial recognition memory", 45th European Brain and Behaviour Society Meeting 6-9 September 2013, Munich, Abstract P202). Performance in this task can be disrupted by a low dose of PCP, such that the animals do not discriminate anymore between the new and a familiar arm.
Method [0341] Male Long Evans rats (Janvier, France, body weight 280 to 295 g) were group housed in enriched individually ventilated cages and habituated to environmental conditions for 5 days. After acclimatization, animals were single housed for 4 days until testing. During this period animals were handled for 2 min per day and received sham dosing once a day for 3 days prior to the test. The V-maze consisted of two arms (LxWxH: 70x10x30 cm) at a 90° angle to each other to form a V-shaped maze connected by guillotine doors to a center zone. The walls of each arm were of a different context displaying horizontally black and white striped in one arm vs. uniform black walls in the other. Background infrared illumination was provided via the bottom of the maze and a top view video camera above the platform was used for video recording of the experiments. The animal’s exploration of each arm was automatically quantified using Ethovision XT 7.0 (Noldus, The Netherlands). Animals were treated with Co. No. 1 or its vehicle (20% ΗΡ-β-CD + 1 eq. HCI) administered p.o. 4 h before the start of the test. PCP (0.75 mg/kgs.c.) or its vehicle (0.9% NaCI solution) was administered 30 min prior to the test. The test consisted of 2 sessions of 5 min each: in the first session (exploration) the animal was placed in the center zone and given access to one of both arms (=familiar). After 5 min, the animal was taken out of the maze, the door of the other arm (new) was also opened, and the animal was put back in the center zone for a second session (choice). The time spent in the familiar and new arm respectively during the choice session was recorded for 5 min. Results [0342] Co. No. 1 was evaluated in rats in a series of dose-response studies evaluating doses from 0.16 to 10 mg/kg. While control animals (treated with vehicle of the test compound and the vehicle of PCP) displayed a strong preference for exploration of the new vs. the familiar arm in the second session, the PCP-treated rats did not discriminate anymore between both arms in each of these studies. PCP-challenged rats that were pretreated with Co. No. 1 at doses from 0.32 mg/kg onwards showed again a clear preference for the new arm (Figure 2). This reversal effect against PCP was observed up to the highest dose tested (10 mg/kg). 4) Reserpine interaction test in rats [0343] Some exemplified compounds were observed to induce mydriasis in rats.
It was investigated to what extent the mydriatic action of test compounds was sufficient to counteract the miosis induced by the monoamines-depleting agent reserpine (10 mg/kg; SC) in Wiga rats. Test compounds induced mydriasis before the reserpine challenge (time = -1 h unless otherwise stated; Table 10).
For instance, Co. No. 1 induced mydriasis before the reserpine challenge (ED50:1.78 mg/kg s.c.; 1.55 mg/kg p.o., -1 h; 0.89 mg/kg, p.o., -4 h) and reversed the reserpine-induced ptosis (ED50: 1.03 mg/kg s.c.; 0.78 mg/kg p.o., -1 h; 0.78 mg/kg, p.o., -4 h), miosis (ED50:4.1 mg/kg s.c.; 9.4 mg/kg p.o., -1 h; 9.4 mg/kg, p.o., -4 h) and sedation (ED50: 9.4 mg/kg s.c.; 7.1 mg/kg p.o., -1 h; 14 mg/kg, p.o., -4 h). The efFects are illustrated in Figure 3. Co. No. 1 did not affect the tail-pinch response before reserpine nor the reserpine-induced blockade of the tail-pinch response and did not induce scratching or hyperemia after reserpine. The reference mGlu2 NAMs RO-4995819 (40 mg/kg, p.o.), RO-4491533 (40 mg/kg, p.o.; 10 mg/kg, s.c.) and [CAS 579473-69-1] (40 mg/kg, s.c.) were devoid of these interactions with reserpine. Without wishing to be bound by theory, the observed effect may be mediated by a mechanism different from, and additional to, mGluR2 NAM activity.
Table 10. Reversal of reserpine-induced ptosis (reserpine; 10 mg/kg; SC; -1 h) in Wiga rats.
(continued)
5) Ro-4-1284 interaction test in rats [0344] The relative ability of Co. No. 1 to increase palpebral opening was also studied in rats challenged with another monoamines depleting agent, viz. Ro-4-1284 (1.25 mg/kg, s.c.). Effects on body temperature immediately before the injection of Ro-4-1284 were also measured. The cumulative palpebral opening score (every 5 min over a 1-h period) was used for evaluation. The median cumulative palpebral opening in solvent-pretreated control animals (n = 70) was 18; a scores > 25 occurred in only 1.4% of these control animals and was adopted as all-or-none criterion for drug-induced reversal of the Ro-4-1284-induced palpebral ptose. Co. No. 1 increased palpebral opening to scores > 25 (ED50: 0.51 mg/kg, p.o.) without affecting body temperature (> 10 mg/kg, p.o.). 6) Reversal of LY-404039-induced decrease of palpebral opening in apomorphine-challenged rats.
[0345] Male Wiga Wistar rats (Cr1 :WI; Charles River Germany; 220 ± 40 g) were housed under standard laboratory conditions (21 ± 2 °C; 50-65% relative humidity; light-dark cycle set at 12 h; lights on at 6.00 h) and fasted overnight prior to the start of the experiments (tap water remained available ad libitum). During the test period, they were housed in individual cages. The local Ethical Committee approved all studies in compliance with the Declaration of Helsinki. Palpebral opening was scored every 5 min over the first hour after injection of apomorphine (1.0 mg/kg, i.v.) in animals either pretreated or not pretreated with LY-404039 (2.5 mg/kg, s.c.) at 1 h prior to the apomorphine injection. The animals were also pretreated with test compound or solvent at a predefined interval before apomorphine challenge. The score system was: (5) exophthalmos, (4) wide open, (3) open for three-quarters, (2) half open, (1) open for one-quarter, (0) closed. The scores for palpebral opening were cumulated over the 60-min observation period. A cumulative palpebral opening score > 26 was selected for drug-induced reversal of the LY-404039-induced decrease of palpebral opening (occurrence in 3.2% of control animals pretreated with LY-404039 (n = 154) versus in 99.5% of control rats not pretreated with LY-404039 (n = 6335)).
Table 11a shows the palpebral opening score in control animals receiving apomorphine alone and in animals receiving apomorphine and LY-404039. In animals receiving apomorphine alone the median palpebral opening is 43 whereas in animals receiving apomorphine and LY-404039, the median palpebral opening is 17. In animals treated with apomorphine alone, the palpebral opening score is almost always (in 95.5% of the rats) greater than 34, whereas in animals treated with the combination (apomorphine + LY-404039) only 3.2% of the animals show palpebral opening greater than 26.
Table 11a. Palpebral opening score in control animals.
Table 11b. Reversal of LY-404039-induced decrease of palpebral opening in apomorphine challenged rats.
7) Reversal of mGluR2-agonism in hippocampal brain slices
Introduction [0346] Electrophysiology recordings of field excitatory postsynaptic potentials (fEPSPs) in acute hippocampal brain slices represent a model fortesting synaptic transmission and plasticity. The effect of Co. No. 1 on synaptic transmission and plasticity in dentate gyrus synapses was investigated using this model. This region was chosen because of the high expression of mGluR 2 (Shigemoto et al., The Journal of Neuroscience, October 1, 1997, 17(19), 7503-7522).
Methods [0347] Recordings of fEPSPs were made from hippocampal brain slices using a multi-electrode array (MEA) biochip, and 3-dimensional- (3D) tip electrodes, according to a standard protocol. These recordings were used to monitor glutamate-mediated synaptic transmission (Figure 4).
Results [0348] Superfusion of rat hippocampal brain slices with the mGlu2/3-specific agonist LY-354740 (1 μΜ) depressed fEPSP by 50% within 15 min of application (Figure 5) and was associated with an increase of the paired-pulse ratio (PPR), indicating a presynaptic mechanism. Fifteen min after the application of 10 μΜ Co. No. 1 the depression of fEPSP had recovered by 80%. This was associated with a decrease of the PPR, indicating an increase in neurotransmitter release (Figure 5, n = 17 slices from 4 rats).
Subsequently, the effects of Co. No. 1 on synaptic function using long-term potentiation (LTP) protocols in the dentate gyrus (Goeldner et. al., Neuropharmacology 2013, 64, 337-346) were evaluated.
Small magnitude LTP (110%) was induced using isolated glutamatergic-mediated fEPSP: trains of theta-burst stimulation that are known to induce LTP at these particular synapses were applied (Dinklo et al., J. Pharmacol. Exp. Ther. 2011, 336(2), 560-574). In the presence of 10 μΜ Co. No. 1, the magnitude of LTP was enhanced by 150% compared to baseline (p=0.005). Also noteworthy is the finding that the post-theta potentiation (PTP) in the presence of 10 μΜ Co. No. 1 was significantly different from vehicle treatment: 160% vs. 120% respectively (p=0.01) (Figure 6, 22 slices from 4 SD rats). At the end of the experiments, addition of 1 mM kynurenic acid to block glutamatergic neurotransmission, confirmed that the post-synaptic response is mediated by glutamate neurotransmission.
Discussion [0349] LY-354740 stimulates presynaptic mGlu2/3 receptors to limit the release of glutamate. Furthermore, the effects of Group II mGluR agonists and antagonists in rodent models of cognition are totally absent in mGluR2 knock-out mice (Higgins etal. Neuropharmacology, 2004,46,907-917). Co. No. 1 reversed synaptic depression evoked by the mGlu2/3-agonist LY-354740. These data illustrate that Co. No. 1 is able to restore depressed synaptic transmission in rat hippocampal slices in vitro. The increase in network excitability, as a result of enhanced excitatory neurotransmission, affected the threshold of LTP induction. Thus, LTP was efficiently induced by weak theta stimulation, but only when Co. No. 1 was pre-applied. Thus, the compound might act as a cognitive enhancer via an ability to elevate the synaptic strength in glutamatergic synapses and by priming the system for enhanced LTP.
Prophetic composition examples [0350] "Active ingredient" as used throughout these examples relates to a final compound of Formula (I), the pharmaceutically acceptable salts thereof, the solvates and the stereochemically isomeric forms and the tautomers thereof. Typical examples of recipes for the formulation of the invention are as follows: 1. Tablets [0351]
Active ingredient 5 to 50 mg
Di-calcium phosphate 20 mg
Lactose 30 mg
Talcum 10 mg
Magnesium stearate 5 mg
Potato starch ad 200 mg [0352] In this Example, active ingredient can be replaced with the same amount of any of the compounds according to the present invention, in particular by the same amount of any of the exemplified compounds. 2. Suspension [0353] An aqueous suspension is prepared for oral administration so that each 1 milliliter contains 1 to 5 mg of one of the active compounds, 50 mg of sodium carboxymethyl cellulose, 1 mg of sodium benzoate, 500 mg of sorbitol and water ad 1 ml. 3 . Injectable [0354] A parenteral composition is prepared by stirring 1.5 % by weight of active ingredient of the invention in 10% by volume propylene glycol in water. 4. Ointment [0355]
Active ingredient 5 to 1000 mg Stearyl alcohol 3 g
Lanoline 5 g
White petroleum 15 g
Water ad 100 g [0356] In this Example, active ingredient can be replaced with the same amount of any of the compounds according to the present invention, in particular by the same amount of any of the exemplified compounds.
Claims 1. A compound of Formula (I)
or a stereoisomeric form thereof, wherein R1 is phenyl or 2-pyridinyl, each optionally substituted with one or more substituents each independently selected from the group of halo, C1.4alkyl, mono- or poly-haloC^alkyl, -O-C^alkyl, -C1^aikyl-0-C1.4alkyl, mono- or poly-haloC^alkyloxy, -C^alkyl-OH, C1_4alkylthio, mono- or poly-haloC^alkylthio, cyano, C3.7cycloalkyl optionally substituted with trifluoromethyl, and -SF5; or is R 2 is selected from
wherein R5 and R6 are each independently selected from thegroupofhydrogen,halo,cyano,C.Malkyl,-C.Malkyl-OH, C3_7cycloalkyl, mono- or poly-haloC^alkyl, -C^alkyl-O-C^alkyl, -O-C^alkyl, mono- or poly-haloC^alkyloxy, 1-acetylazetidin-3-yl, and NR’R"; wherein R’ is selected from hydrogen and C1.4alkyl; R" is selected from hydrogen and C1_4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a heterocyclic group selected from the group of 1-azetidinyl, 1-pyrrolidinyl, 1-piperidinyl, 1-piperazinyl, and 4-morpholinyl; wherein each of the heterocyclic groups may be optionally substituted with a substituent selected from halo, hydroxyl, C^alkyl, mono- or poly-haloC-^alkyl, and -(CO)C1.4alkyl; R3 is selected from hydrogen and C^alkyl; R4 is selected from the group of hydrogen, C^alkyl, mono- or poly-haloC.|_4alkyl, -C^alkyl-O-C^alkyl, and -C^alkyl-OH; or a N-oxide, or a pharmaceutically acceptable salt or a solvate thereof. 2. A compound according to claim 1, or a stereoisomeric form thereof, wherein R1 is phenyl or 2-pyridinyl, each optionally substituted with one or more substituents each independently selected from the group of halo, C1_4alkyl, mono- or poly-haloC^alkyl, -O-C^alkyl, -C^alkyl-O-C^alkyl, mono- or poly-haloC^alkyloxy, -C^alkyl-OH, mono- or poly-haloC.|_4alkylthio, cyano, and -SF5; or is R 2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, halo, cyano, C1_4alkyl, -C^alkyl-OH, C3_7cycloalkyl, mono-orpoly-haloC1_4alkyl,-C1^aikyl-0-C1^alkyl,-0-C1^alkyl, mono-orpoly-haloC^alkyloxy, and NR’R"; wherein R’ is selected from hydrogen and C1_4alkyl; R" is selected from hydrogen and C1_4alkyl; or R’ and R" together with the Nitrogen atom to which they are attached form a heterocyclic group selected from the group of 1-azetidinyl, 1-pyrrolidinyl, and 1-piperidinyl; wherein each of the heterocyclic groups may be optionally substituted with a halo substituent; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C^alkyl, mono- or poly-haloC^alkyl, -C^alkyl-O-C^alkyl, and -C^alkyl-OH; or a N-oxide, or a pharmaceutically acceptable salt or a solvate thereof. 3. A compound according to claim 1 or 2, or a stereoisomeric form thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C^alkyl, mono- or poly-haloC1_4alkyl, -O-C^alkyl, mono- or poly-haloC^alkyloxy, cyano and -SF5; or is
R 2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl, mono- or poly-haloC1.4alkyl, -C1_4alkyl-0-C1_4alkyl, -0-C1_4alkyl, and NR’R"; wherein R’ is selected from hydrogen and C1_4alkyl; R" is selected from hydrogen and C1_4alkyl; R3 is selected from hydrogen and C1_4alkyl; R4 is selected from the group of hydrogen, C1.4alkyl, mono- or poly-haloC^alkyl, -C^alkyl-O-C^alkyl, and -C^alkyl-OH; or a pharmaceutically acceptable salt or a solvate thereof. 4. The compound according to any one of claims 1 to 3, or a stereoisomeric form thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C^alkyl, mono- or poly-haloC^alkyl, -O-C^alkyl, mono- or poly-haloC^alkyloxy, cyano and -SF5; or is R 2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl, -C^alkyl-O-C^alkyl, -0-C1.4alkyl, and NR’R"; wherein R’ is hydrogen; R" is hydrogen; R3 is selected from hydrogen and C^alkyl; R4 is selected from the group of hydrogen and C^alkyl; or a pharmaceutically acceptable salt or a solvate thereof. 5. The compound according to any one of claims 1 to 4, or a stereoisomeric form thereof, wherein R1 is phenyl, optionally substituted with one or more substituents each independently selected from the group of halo, C^alkyl, poly-haloC^alkyl and -SF5; R2 is selected from
wherein R5 and R6 are each independently selected from the group of hydrogen, C^alkyl and -0-C1.4alkyl; R3 is selected from hydrogen and C1_4alkyl; R4 is hydrogen; or a pharmaceutically acceptable salt or a solvate thereof. 6. A compound according to any one of claims 1 to 4, or a stereoisomeric form thereof, wherein R1 is selected from the group of
and the rest of variables are as defined in any one of claims 1 to 4. 7. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1 to 6 and a pharmaceutically acceptable carrier or excipient. 8. A compound according to any one of claims 1 to 6 or a pharmaceutical composition according to claim 7 for use as a medicament. 9. A compound according to any one of claims 1 to 6 or a pharmaceutical composition according to claim 7 for use in the treatment or in the prevention of central nervous system conditions or a diseases selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder. 10. A compound according to any one of claims 1 to 6 or a pharmaceutical composition according to claim 7 for use in the treatment or prevention of central nervous system disorders or conditions selected from depressive disorders; neurocognitive disorders; neurodevelopmental disorders; substance-related and addictive disorders; schizophrenia spectrum and other psychotic disorders; somatic symptom and related disorders; and hypersomnolence disorder. 11. The compound or the pharmaceutical composition for use according to claim 9 or 10, wherein the central nervous system conditions or diseases are selected from dementia or neurocognitive disorder, major depressive disorder, depression, treatment resistant depression, attention-deficit/hyperactivity disorder and schizophrenia. 12. A process for preparing the pharmaceutical composition according to claim 7, characterized in that a pharmaceu- tically acceptable carrier is intimately mixed with a therapeutically effective amount of a compound according to any one of claims 1 to 6. 13. A product comprising a compound according to any one of claims 1 to 6 and an additional pharmaceutical agent, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of central nervous system conditions or diseases selected from mood disorders; delirium, dementia, amnestic and other cognitive disorders; disorders usually first diagnosed in infancy, childhood or adolescence; substance-related disorders; schizophrenia and other psychotic disorders; somatoform disorders; and hypersomnic sleep disorder. 14. A product comprising a compound according to any one of claims 1 to 6 and an additional pharmaceutical agent, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of central nervous system conditions or diseases selected from depressive disorders; neurocognitive disorders; neurodevelopmental disorders; substance-related and addictive disorders; schizophrenia spectrum and other psychotic disorders; somatic symptom and related disorders; and hypersomnolence disorder. 15. A compound of Formula (V)
wherein R2a is halo, and R3 and R4 are as defined in any one of claims 1 to 6. 16. A compound according to claim 15, having the Formula (V’)
17. A compound according to claim 15 or 16, having the Formula (1-13) or (1-13a)
or a salt thereof.
18. A compound of Formula (II)
wherein R2, R3 and R4 are as defined in any one of claims 1 to 6. 19. A process for the preparation of a compound according to Formula (I) as defined in any one of claims 1 to 6, comprising steps a) and b) a) reacting a compound of Formula (V), wherein R2a is halo, with a suitable boron species, in the presence of a palladium catalyst and a suitable base and solvent, to form a compound of Formula (II)
b) reacting the compound of Formula (II) obtained in step a) with a compound of Formula (III) where X is halo, in the presence of a suitable coupling agent or a catalyst and ligand, in the presence of a base in a suitable solvent
20. A compound according to claim 1 or 2, wherein the compound is
or a N-oxide, or a pharmaceutically acceptable salt or a solvate thereof. 21. The compound according to claim 20 wherein the compound is the free base, the hydrochloride salt, the sulfate salt, the methane sulfonate salt or the maleate salt. 22. The compound according to claim 20, wherein the compound is
or a pharmaceutically acceptable salt or a solvate thereof. 23. A compound according to claim 1 or 2, wherein the compound is
or a pharmaceutically acc=HLQ^,= oQ,l α u.=. <»..
Patentansprüche 1. Verbindung der Formel (I)
oder eine stereoisomere Form davon, wobei R1 für Phenyl oder 2-Pyridinyl steht, jeweils gegebenenfalls substituiert durch einen oder mehrere jeweils unabhängig voneinander aus der aus Halogen, C^-Alkyl, Mono- oder Polyhalogen-C-^-alkyl, -O-C^-Alkyl, -C^-Alkyl-O-C^-alkyl, Mono- oder Polyhalogen-C1_4-alkyloxy, -C1.4-Alkyl-OH, C1.4-Alkylthio, Mono- oder Po-lyhalogen-C^-alkylthio, Cyano, gegebenenfalls durch Trifluormethyl substituiertem C3_7-Cycloalkyl und -SF5 bestehenden Gruppe ausgewählte Substituenten, oder für
steht, R2 aus
ausgewählt ist, wobei R5 und R6 jeweils unabhängig voneinander aus der aus Wasserstoff, Halogen, Cyano, C1_4-Alkyl, -Ci_4-Alkyl-OH, C3_7-Cycloalkyl, Mono- oder Polyhalogen-C1.4-alkyl,-C1_4-Alkyl-0-C1_4-alkyl, -O-Ci_4-Alkyl, Mono- oder Polyhalogen-C1.4-alkyloxy, 1-Acetylazetidin-3-yl und NR’R" bestehenden Gruppe ausgewählt sind, wobei R’ aus Wasserstoff und C^-Alkyl ausgewählt ist, R” aus Wasserstoff und C^-Alkyl ausgewählt ist oder R’ und R" zusammen mit dem Stickstoffatom, an das sie gebunden sind, eine aus der aus 1-Azetidinyl, 1-
Pyrrolidinyl, 1-Piperidinyl, 1-Piperazinyl und 4-Morpholinyl bestehenden Gruppe ausgewählte heterocyclische Gruppe bilden, wobei die heterocyclischen Gruppen jeweils gegebenenfalls durch einen aus Halogen, Hydroxy, Ci_4-Alkyl, Mono- oder Polyhalogen-C^-alkyl und -(COj-C^-Alkyl ausgewählten Substituenten substituiert sein können, R3 aus Wasserstoff und C^-Alkyl ausgewählt ist, R4 aus der aus WasserstofF, C^-Alkyl, Mono- oder Polyhalogen-C.M-alkyl,-C1_4-Alkyl-0-C1_4-alkyl und -C-^-AI-kyl-OH bestehenden Gruppe ausgewählt ist, oder ein N-Oxid oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 2. Verbindung nach Anspruch 1 oder eine stereoisomere Form davon, wobei R1 für Phenyl oder2-Pyridinyl steht, jeweils gegebenenfalls substituiert durch einen oder mehrerejeweils unabhängig voneinander aus der aus Halogen, C^-Alkyl, Mono- oder Polyhalogen-C^-alkyl, -O-C^-Alkyl, -C^-Alkyl-O-C1_4-alkyl, Mono- oder Polyhalogen-C^-alkyloxy, -C^-Alkyl-OH, Mono- oder Polyhalogen-C^-alkylthio, Cyano und -SF5 bestehenden Gruppe ausgewählte Substituenten, oder für
steht, R2 aus
ausgewählt ist, wobei R5 und R6 jeweils unabhängig voneinander aus der aus Wasserstoff, Halogen, Cyano, C^-Alkyl, -C^-Alkyl-OH, C3.7-Cycloalkyl, Mono- oder Polyhalogen-C^-alkyl, -C^-Alkyl-O-C^-alkyl, -O-C^-Alkyl, Mono- oder Poly-halogen-C^-alkyloxy und NR’R" bestehenden Gruppe ausgewählt sind, wobei R’ aus Wasserstoff und C^-Alkyl ausgewählt ist, R" aus Wasserstoff und C^-Alkyl ausgewählt ist oder R’ und R" zusammen mit dem Stickstoffatom, an das sie gebunden sind, eine aus der aus 1-Azetidinyl, 1-Pyrrolidinyl und 1-Piperidinyl bestehenden Gruppe ausgewählte heterocyclische Gruppe bilden, wobei die heterocyclischen Gruppen jeweils gegebenenfalls durch einen Halogensubstituenten substituiert sein können, R3 aus Wasserstoff und C^-Alkyl ausgewählt ist, R4 aus der aus Wasserstoff, C^-Alkyl, Mono- oder Polyhalogen-C^-alkyl, -C^^Alkyl-O-C^-alkyl und -C^-Alkyl-OH bestehenden Gruppe ausgewählt ist, oder ein N-Oxid oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 3. Verbindung nach Anspruch 1 oder 2 oder eine stereoisomere Form davon, wobei R1 für Phenyl steht, gegebenenfalls substituiert durch einen oder mehrere jeweils unabhängig voneinander aus der aus Halogen, C^-Alkyl, Mono- oder Polyhalogen-C^-alkyl, -O-C^-Alkyl, Mono- oder Polyhalogen-Ci_4-alkyloxy, Cyano und -SF5 bestehenden Gruppe ausgewählte Substituenten, oder für
steht, R2 aus
ausgewählt ist, wobei R5 und R6 jeweils unabhängig voneinander aus der aus Wasserstoff, C^-Alkyl, Mono- oder Polyhalogen-C^-alkyl, -C^-Alkyl-O-C^-alkyl, -0-Ci.4-Alkyl und NR’R” bestehenden Gruppe ausgewählt sind, wobei R’ aus Wasserstoff und C^-Alkyl ausgewählt ist, R” aus Wasserstoff und C^-Alkyl ausgewählt ist, R3 aus Wasserstoff und C^-Alkyl ausgewählt ist, R4 aus der aus Wasserstoff, C1_4-Alkyl, Mono- oder Polyhalogen-C^-alkyl, -C^-Alkyl- o-cM -alkyl und -G^^-Alkyl-OH bestehenden Gruppe ausgewählt ist, oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 4. Verbindung nach einem der Ansprüche 1 bis 3 oder eine stereoisomere Form davon, wobei R1 für Phenyl steht, gegebenenfalls substituiert durch einen oder mehrere jeweils unabhängig voneinander aus der aus Halogen, C^-Alkyl, Mono- oder Polyhalogen-C14-alkyl, -O-C^-Alkyl, Mono- oder Polyhalogen-C^-alkyloxy, Cyano und -SF5 bestehenden Gruppe ausgewählte Substituenten, oder für steht,
R2 aus
ausgewählt ist, wobei R5 und R6 jeweils unabhängig voneinander aus der aus Wasserstoff, C^-Alkyl, -C^-Alkyl-O-C^-alkyl, -O-C.|_4-Alkyl und NR’R” bestehenden Gruppe ausgewählt sind, wobei R’ für Wasserstoff steht, R” für Wasserstoff steht, R3 aus Wasserstoff und C^-Alkyl ausgewählt ist, R4 aus der aus Wasserstoff und C^-Alkyl bestehenden Gruppe ausgewählt ist, oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 5. Verbindung nach einem der Ansprüche 1 bis 4 oder eine stereoisomere Form davon, wobei R1 für Phenyl steht, jeweils gegebenenfalls substituiert durch einen oder mehrere jeweils unabhängig voneinander aus der aus Halogen, C^-Alkyl, Polyhalogen-C^-alkyl und -SF5 bestehenden Gruppe ausgewählte Substituenten, R2 aus
n6 ausgewählt ist, wobei R5und R6jeweils unabhängig voneinander aus der aus Wasserstoff, C^-Alkyl und -O-C^-Alkyl bestehenden Gruppe ausgewählt sind, R3 aus Wasserstoff und C^-Alkyl ausgewählt ist, R4 für Wasserstoff steht, oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 6. Verbindung nach einem der Ansprüche 1 bis 4 oder eine stereoisomere Form davon, wobei R1 aus der aus
bestehenden Gruppe ausgewählt ist, und die restlichen Variablen wie in einem der Ansprüche 1 bis 4 definiert sind. 7. Pharmazeutische Zusammensetzung, umfassend eine therapeutisch wirksame Menge einer Verbindung nach einem der Ansprüche 1 bis 6 und einen pharmazeutisch unbedenklichen Träger oder Exzipienten. 8. Verbindung nach einem der Ansprüche 1 bis 6 oder pharmazeutische Zusammensetzung nach Anspruch 7 zur Verwendung als Medikament. 9. Verbindung nach einem der Ansprüche 1 bis 6 oder pharmazeutische Zusammensetzung nach Anspruch 7 zur Verwendung bei der Behandlung oder Prävention von Leiden bzw. Krankheiten des zentralen Nervensystems ausgewählt aus affektiven Störungen, Delirium, Demenz, amnesischen und anderen kognitiven Störungen, Störungen, die gewöhnlich zuerst bei Kleinkindern, Kindern oder Jugendlichen diagnostiziert werden, substanzbedingten Störungen, Schizophrenie und anderen psychotischen Störungen, somatoformen Störungen und hypersomnischer Schlafstörung. 10. Verbindung nach einem der Ansprüche 1 bis 6 oder pharmazeutische Zusammensetzung nach Anspruch 7 zur Verwendung bei der Behandlung oder Prävention von Störungen bzw. Leiden des zentralen Nervensystems ausgewählt aus depressiven Störungen, neurokognitiven Störungen, Störungen der Entwicklung des Nervensystems, substanzbedingten Störungen und Suchtstörungen, Störungen des schizophrenen Spektrums und anderen psychotischen Störungen, somatischem Symptom und verwandten Störungen und Hypersomnolenzstörung. 11. Verbindung oder pharmazeutische Zusammensetzung zur Verwendung nach Anspruch 9 oder 10, wobei die Leiden bzw. Krankheiten des zentralen Nervensystems ausgewählt sind aus Demenz und neurokognitiver Störung, schwerer Depression, Depression, behandlungsresistenter Depression, Aufmerksamkeitsdefizit-Hyperaktivitätsstörung und Schizophrenie. 12. Verfahren zur Herstellung der pharmazeutischen Zusammensetzung nach Anspruch 7, dadurch gekennzeichnet, dass man einen pharmazeutisch unbedenklichen Träger innig mit einer therapeutisch wirksamen Menge einer Verbindung nach einem der Ansprüche 1 bis 6 mischt. 13. Produkt, umfassend eine Verbindung nach einem der Ansprüche 1 bis 6 und ein zusätzliches Pharmazeutikum als Kombinationspräparat zur gleichzeitigen, getrennten oder aufeinanderfolgenden Anwendung bei der Behandlung oder Prävention von Leiden bzw. Krankheiten des zentralen Nervensystems ausgewählt aus affektiven Störungen, Delirium, Demenz, amnesischen und anderen kognitiven Störungen, Störungen, die gewöhnlich zuerst bei Kleinkindern, Kindern oder Jugendlichen diagnostiziert werden, substanzbedingten Störungen, Schizophrenie und anderen psychotischen Störungen, somatoformen Störungen und hypersomnischer Schlafstörung. 14. Produkt umfassend eine Verbindung nach einem der Ansprüche 1 bis 6 und ein zusätzliches Pharmazeutikum als Kombinationspräparat zur gleichzeitigen, getrennten oder aufeinanderfolgenden Anwendung bei der Behandlung oder Prävention von Leiden bzw. Krankheiten des zentralen Nervensystems ausgewählt aus depressiven Störungen, neurokognitiven Störungen, Störungen der Entwicklung des Nervensystems, substanzbedingten Störungen und Suchtstörungen, Störungen des schizophrenen Spektrums und anderen psychotischen Störungen, somatischem Symptom und verwandten Störungen und Hypersomnolenzstörung. 15. Verbindung der Formel (V)
wobei R2a für Halogen steht und R3 und R4 wie in einem der Ansprüche 1 bis 6 definiert sind. 16. Verbindung nach Anspruch 15 mit der Formel (V’)
17. Verbindung nach Anspruch 15 oder 16 mit der Formel (1-13) oder (1-13a)
oder ein Salz davon. 18. Verbindung der Formel (II)
wobei R2, R3 und R4wie in einem der Ansprüche 1 bis 6 definiert sind. 19. Verfahren zur Herstellung einer wie in einem der Ansprüche 1 bis 6 definierten Verbindung gemäß Formel (I), welches die Schritte a) und b) umfasst: a) die Umsetzung einer Verbindung der Formel (V), in welcher R2a für Halogen steht, mit einer geeigneten Borspezies in Gegenwart eines Palladiumkatalysators und einer geeigneten Base und eines geeigneten Lösungsmittels unter Bildung einer Verbindung der Formel (II)
b) die Umsetzung der in Schritt a) erhaltenen Verbindung der Formel (II) mit einer Verbindung der Formel (III), wobei X für Halogen steht, in Gegenwart eines geeigneten Kupplungsmittels oder eines Katalysators und Liganden in Gegenwart einer Base in einem geeigneten Lösungsmittel
20. Verbindung nach Anspruch 1 oder 2, wobei es sich bei der Verbindung um
handelt, oder ein N-Oxid oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 1 21. Verbindung nach Anspruch 20, wobei es sich bei der Verbindung um die freie Base, das Hydrochloridsalz, das
Sulfatsalz, das Methansulfonatsalz oder das Maleatsalz handelt. 22. Verbindung nach Anspruch 20, wobei es sich bei der Verbindung um
I
' handelt, oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon. 23. Verbindung nach Anspruch 1 oder 2, wobei es sich bei der Verbindung um
handelt, oder ein pharmazeutisch unbedenkliches Salz oder ein Solvat davon.
Revendications 1. Composé de Formule (I)
ou forme stéréoisomère de celui-ci, où R1 représente un groupement phényle ou 2-pyridinyle, chacun étant éventuellement substitué par un ou plusieurs substituants, chacun étant indépendamment choisi dans le groupe constitué par les groupements halogéno, alkyle en C.^, mono- ou poly-halogénoalkyle en Ci_4, -0-(alkyle en C^), -(alkyle en C1_4)-0-(alkyle en C^), mono- ou poly-halogénoalkoxy en Cf.4, -(alkyle en C^-OH, (alkyle en C^-thio, mono- ou poly-halogéno-(alkyle en C1.4)-thio, cyano, cycloalkyle en C3_7 éventuellement substitué par trifluorométhyle et -SF5; ou représente
R2 est choisi parmi
où chacun des radicaux R5 et R6 est indépendamment choisi dans le groupe constitué par l’atome d’hydrogène et les groupements halogéno, cyano, alkyle en C^,-(alkyle en C1.4)-OH, cycloalkyle en C3_7, mono- ou poly-halogénoalkyle en C-|_4, -(alkyle en C-^-CKalkyle en C14), -0-(alkyleen C^alkyl, mono-ou poly-halogénoalkoxy en C.|_4, 1-acétylazétidin-3-yle, et NR’R" ; où R’ est choisi parmi l’atome d’hydrogène et les groupements alkyle en C^_4 ; R" est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 ; ou R’ et R" forment ensemble et avec l’atome d’azote auquel ils sont liés forment un groupement hétérocyclique choisi dans le groupe constitué par les groupements 1-azétidinyle, 1-pyrrolidinyle, 1-pipéridinyle, 1-pipérazinyle, et4-morpholinyle ; où chacun des groupements hétérocycliques est éventuellement substitué par un substituant choisi parmi les groupements halogéno, hydroxyle, alkyle en C1_4, mono- ou poly-halogénoalkyle en C.|_4, et -(CO)-(alkyle en C^) ; R3 est choisi parmi l’atome d’hydrogène et les groupements alkyle en C^_4 ; R4 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C^, mono- ou poly-halogénoalkyle en C-|_4, -(alkyle en C.|_4)-0-(alkyle en C^) et -(alkyle en C-^-OH ; ou N-oxyde de celui-ci, ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 2. Composé selon la revendication 1, ou forme stéréoisomère de celui-ci, où R1 représente un groupement phényle ou 2-pyridinyle, chacun étant éventuellement substitué par un ou plusieurs substituants, chacun étant indépendamment choisi dans le groupe constitué par les groupements halogéno, alkyle en C-|_4, mono- ou poly-halogénoalkyle en C^, -0-(alkyle en C^), -(alkyle en C.|^)-0-(alkyle en C^), mono- ou poly-halogénoalkoxy en C^, -(alkyle en C-^-OH, mono- ou poly-halogéno- (alkyle en C1_4)-thio, cyano et -SF5; ou représente
R2 est choisi parmi
R6 où chacun des radicaux R5 et R6 est indépendamment choisi dans le groupe constitué par l’atome d’hydrogène et les groupements halogéno, cyano, alkyle en 01-4,-(alkyle en C^-OH, cycloalkyle en C3.7, mono- ou poly-halogénoalkyle en 01-4, -(alkyle en C1 ^)-0-(81 kyle en C^), -0-(alkyle en C^), mono- ou poly-halogénoalkoxy en C1-4 et NR’R" ; où R’ est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 ; R" est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 ; ou R’ et R" forment ensemble et avec l’atome d’azote auquel ils sont liés forment un groupement hétérocyclique choisi dans le groupe constitué par les groupements 1-azétidinyle, 1-pyrrolidinyle et 1-pipéridinyle ; où chacun des groupements hétérocycliques est éventuellement substitué par un substituant halogéno ; R3 est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 ; R4 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C-|.4, mono- ou poly-halogénoalkyle en 01-4, -(alkyle en C1_4)-0-(alkyle en 01-4) et -(alkyle en C^J-OH ; ou N-oxyde de celui-ci, ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 3. Composé selon la revendication 1 ou 2, ou forme stéréoisomère de celui-ci, où R1 représente un groupement phényle, éventuellement substitué par un ou plusieurs substituants, chacun étant indépendamment choisi dans le groupe constitué par les groupements halogéno, alkyle en C·^, mono- ou poly-halogénoalkyle en C1-4, -0-(alkyle en 01-4), mono- ou poly-halogénoalkoxy en C1.4, cyano et -SF5 ; ou représente R2 est choisi parmi
où chacun des radicaux R5 et R6 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C^, mono- ou poly-halogénoalkyle en C1_4, -(alkyle en C-^-CKalkyle en C^), -(alkyle en C-j^-OH et NR’R" ; où R’ est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1_4 ; R" est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1_4 ;
R3 est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 I R4 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C^, mono- ou poly-halogénoalkyle en C-|_4, -(alkyle en C-^-CKalkyle en C^) et -(alkyle en C^-OH ; ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 4. Composé selon l’une quelconque des revendications 1 à 3, ou forme stéréoisomère de celui-ci, où R1 représente un groupement phényle, éventuellement substitué par un ou plusieurs substituants, chacun étant indépendamment choisi dans le groupe constitué par les groupements halogéno, alkyle en C^, mono- ou poly-halogénoalkyle en C^, -0-(alkyle en C^), mono- ou poly-halogénoalkoxy en C^, cyano et -SF5 ; ou représente
R2 est choisi parmi
où chacun des radicaux R5 et R6 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en Ci_4, - (alkyle en C-^-O-ialkyle en C^), -0-(alkyle en C^) et NR’R" ; où R’ représente un atome d’hydrogène ; R" représente un atome d’hydrogène ; R3 est choisi parmi l’atome d’hydrogène et les groupements alkyle en C.|_4 ; R4 est choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C1-4 ; ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 5. Composé selon l’une quelconque des revendications 1 à 4, ou forme stéréoisomère de celui-ci, où R1 représente un groupement phényle, éventuellement substitué par un ou plusieurs substituants, chacun étant indépendamment choisi dans le groupe constitué par les groupements halogéno, alkyle en C.|_4, poly-halogénoalkyle en C1-4 et -SF5 ; R2 est choisi parmi
où chacun des radicaux R5 et R6 est indépendamment choisi dans le groupe constitué par l’atome d’hydrogène et les groupements alkyle en C1 ^ et -0-(alkyle en 0^4) ; R3 est choisi parmi l’atome d’hydrogène et les groupements alkyle en C1-4 I R4 représente un atome d’hydrogène ; ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 6. Composé selon l’une quelconque des revendications 1 à 4, ou forme stéréoisomère de celui-ci, où R1 est choisi dans le groupe comprenant
et le reste des variables est tel que défini dans l’une quelconque des revendications 1 à 4. 7. Composition pharmaceutique comprenant une quantité thérapeutiquement active d’un composé selon l’une quelconque des revendications 1 à 6 ainsi qu’un vecteur ou excipient pharmaceutiquement acceptable. 8. Composé selon l’une quelconque des revendications 1 à 6 ou composition pharmaceutique selon la revendication 7, pour utilisation en tant que médicament. 9. Composé selon l’une quelconque des revendications 1 à 6 ou composition pharmaceutique selon la revendication 7, pour utilisation dans le traitement prophylactique ou thérapeutique des états pathologiques ou des maladies du système nerveux central choisis parmi les suivantes : troubles de l’humeur ; délire, démence, troubles amnésiques ou autres troubles cognitifs ; troubles généralement diagnostiqués pour la première fois dans la petite enfance, l’enfance ou l’adolescence ; troubles liés à des substances ; schizophrénie et autres troubles psychotiques ; troubles somatoformes ; et trouble du sommeil de type hypersomnie. 10. Composé selon l’une quelconque des revendications 1 à 6 ou composition pharmaceutique selon la revendication 7 pour utilisation dans le traitement prophylactique ou thérapeutique des troubles ou états pathologiques du système nerveux central choisis parmi les suivants : troubles dépressifs ; troubles neurocognitifs ; troubles neurodéveloppementaux ; troubles liés aux substances et troubles d’addiction ; troubles du spectre schizophrénique et troubles apparentés ; symptôme somatique et troubles apparentés ; et trouble d’hypersomnolence. 11. Composé ou composition pharmaceutique pour utilisation selon la revendication 9 ou 10, où les états pathologiques ou maladies du système nerveux central sont choisis parmi les suivants : démence ou trouble neurocognitif, trouble dépressif majeur, dépression, dépression résistant au traitement, trouble de déficit d’attention/hyperactivité et schizophrénie. 12. Procédé d’élaboration d’une composition pharmaceutique selon la revendication 7, caractérisé en ce qu’un vecteur pharmaceutiquement acceptable est intimement mélangé à une quantité thérapeutiquement active d’un composé selon l’une quelconque des revendications 1 à 6. 13. Produit comprenant un composé selon l’une quelconque des revendications 1 à 6 ou agent pharmaceutique supplémentaire, sous forme de préparation combinée pour utilisation simultanée, séparée ou séquentielle dans le traitement prophylactique ou thérapeutique des états pathologiques ou des maladies du système nerveux central choisis parmi les suivants : troubles de l’humeur ; délire, démence, troubles amnésiques ou autres troubles cognitifs ; troubles généralement diagnostiqués pour la première fois dans la petite enfance, l’enfance ou l’adolescence ; troubles liés à des substances ; schizophrénie et autres troubles psychotiques ; troubles somatoformes ; et trouble du sommeil de type hypersomnie. 14. Produit comprenant un composé selon l’une quelconque des revendications 1 à 6 et agent pharmaceutique supplémentaire, sous forme d’une préparation combinée pour utilisation simultanée, séparée ou séquentielle, dans le traitement prophylactique ou thérapeutique des états pathologiques ou maladies du système nerveux central choisis parmi les suivants : troubles dépressifs ; troubles neurocognitifs ; troubles neurodéveloppementaux ; troubles liés aux substances et troubles d’addiction ; troubles du spectre schizophrénique et troubles apparentés ; symptôme somatique et troubles apparentés ; et trouble d’hypersomnolence. 15. Composé de Formule (V)
où R2a représente un groupement halogéno, et R3 et R4 sont tels que définis dans l’une quelconque des revendications 1 à 6. 16. Composé selon la revendication 15, répondant à la Formule (V’)
17. Composé selon la revendication 15 ou 16, répondant à la Formule (1-13) ou (1-13a)
ou sel de celui-ci. 18. Composé de Formule (II)
où R2, R3 et R4 sont tels que définis dans l’une quelconque des revendications 1 à 6. 19. Procédé de synthèse d’un composé de Formule (I) selon l’une quelconque des revendications 1 à 6, qui comprend les étapes a) et b) a) réaction d’un composé de Formule (V), où R2a représente un groupement halogéno, avec une espèce adaptée du bore, en présence d’un catalyseur au palladium et d’une base et d’un solvant adaptés, pour former un composé de Formule (II)
b) réaction du composé de Formule (II) obtenu dans l’étape a) avec un composé de Formule (III) où X représente un groupement halogéno, en présence d’un agent de couplage adapté ou d’un catalyseur et d’un ligand en présence d’une base dans un solvant adapté
20. Composé selon la revendication 1 ou 2, où le composé est
ou N-oxyde de celui-ci, ou sel pharmaceutiquement acceptable ou solvate de celui-ci. 21. Composé selon la revendication 20, où le composé est la base libre, le sel de chlorhydrate, le sel de sulfate, le sel de méthanesulfonate ou le sel de maléate. 22. Composé selon la revendication 20, où le composé est
ou l’un des sels pharmaceutiquement acceptables ou solvates de celui-ci. 23. Composé selon la revendication 1 ou 2, où le composé est
ou l’un des sels pharmaceutiquement acceptables ou solvates de celui-ci.
REFERENCES CITED IN THE DESCRIPTION
This list of references cited by the applicant is for the reader’s convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.
Patent documents cited in the description • RO 4995819 [0003] · WO 2014064028 A [0003] • WO 2013066736 A [0003] · WO 2009118292 A [0283] • WO 2013174822 A [0003] · WO 2010130424 A [0338]
Non-patent literature cited in the description • FERRAGUTI ; SHIGEMOTO. Cell &amp; Tissue Re- · SCHAFFHAUSER et al. Molecular Pharmacology, search, 2006, vol. 326, 483-504 [0002] 2003, vol. 4, 798-810 [0333] • NISWENDER ; CONN. Annual Review of Pharma- · CID et al. J. Med. Chem., 2012, vol. 55, 8770-8789 cology &amp; Toxicology, 2010, vol. 50, 295-322 [0002] [0338] • GOELDNER et al. Neuropharmacology, 2013, vol. · EM BRECHTS et al. Longitudinal characterization of 64, 337-346 [0002] the TauPS2APP mouse model of Alzheimer’s dis- • KELMENDI et al. Primary Psychiatry, 2006, vol. 13, ease in a two trial discrimination task of visuo-spatial 80-86 [0004] recognition memory. European Brain and Behaviour • KOIKE et al. Behavioural Brain Research, 2013, vol. Society Meeting, 2013 [0340] 238, 48-52 [0004] · CHEMICAL ABSTRACTS, 579473-69-1 [0343] • J. Fluorine Chem., 2008, 767-774 [0097] [0249] · SHIGEMOTO etal. The Journal of Neuroscience, 01 • Diagnostic &amp; Statistical Manual of Mental Disorders October 1997, vol. 17 (19), 7503-7522 [0346] (DSM-IV). American Psychiatric Association [0116] · GOELDNER. Neuropharmacology, 2013, vol. 64, • American Psychiatric Association: Diagnostic and 337-346 [0348]
Statistical Manual of Mental Disorders. American · DINKLO et al. J. Pharmacol. Exp. Ther., 2011, vol.
Psychiatric Association, 2013 [0117] 336 (2), 560-574 [0348] • GENNARO et al. Remington’s Pharmaceutical Sci- · HIGGINS et al. Neuropharmacology, 2004, vol. 46, ences. Mack Publishing Company, 1990 [0131] 907-917 [0349] • HARPER. Current Protocols in Pharmacology. John Wiley &amp; Sons, Inc, 1998, 2.6.1-10 [0333]

Claims (6)

1, ró késistá vsa^üi^t ||i
i| i|| |i ι
jm ji j ii J i Jü 11 |J
vagy ennék sztereo; zsanér tormája, ahol R' Jelébtésé lenti vagy 2-gtridmÍL amely egyenként adott esetben míbaatituili egy vagy több sznbsztitnenssek amely egynmstóí ligptlennl halogén, €v.<alkil menő· vagy poibbalogénCii^alkiL -Ö^Ci^alkil, '·€|-#ΙΜΐ~0'·€^ηΙΙΙΙ, Émm- vagy polbbalogétCMaikltoxh •-C^alkil-Oii mono- vagy poI1-hatogénCs,4atki kio, ciano, Cv.-mkloal.kik amely adott esetben szubsztitoáii sntloormeiil szubsztftuenssek és -SF< csoportjából.megválasztott; vagy·· jelentése
R:í jelentése
köztii mégyá 1 asztali., abor R5 és: R*: jélénlésé egy mlxíéi elattn, C iaaikll, CeaalktbClli, CeíétMoalkik mono- vagy pö|bfta!dgénCn4alkil, -€;, a I k,l 1-0-Cs,sa 1 kd.. -O-Cvidkik mono* vagy poIi-halogénC- I -acetilazettdjn*3-ik.#:iMR,Kf' csoportjából megválasztott; ahol R' jelentése hidrogén és C^alkll kiérni megválasztott!· R“ jelentése hidrogén és C;..:alkil köziül megválasztott; vagy R' és R" jelentése a kapcsolódó nitrogén atommal együtt heterociklusos: esoppriy amely i~azetidinii l-pirrolidinÜ, 1-piperidinik l-ptperazinil. és 4-moríbjmíl csoportjából megválasztott; áltól .mindegyik ihemrociklosos csoport adón esetben szuhsztimáit égj szuhszómenssel amely halogén, bilroxil, C...-.alkik mono* vagy poibhalogérrG„ndki 1, és --(CO>C j ^alktl közül megválasztott; R- jelentése; hidrogén ésCsaalktl: lözükmegvái ásított; R? jelentése nldtxtgén, Csoaiklls mono* vagyspU-haiogépCMá^ éf^palktb -OH csoportjából megválasztott; vagy ennek N-okid, vagy gyégyszerészetiieg stkatoa^satö só vagy szolvat: tormája,
2, Az 1. igénypont szerinti vegyidet vagy ennek sztcreolzomer tormája, ahol i> fenti vagy O-nsrkimih anteiv vgyenkén; adón őseiben stmhszuiuáft egy vágy » ^^ituen^t amely egymásul fóggeiieoíi! hído^sn C^lkil, moau- vagy poU-haiogébCMa&amp;ik #CX«íí&amp;I!, -C't aalk$l«0<\ ,alkit, ww- vagy poH^H^CMfttaiexi. G rtlkd ebi mono- vagy i50imfeiéHCí&amp;ía!kiitio. ciaoo.. MFv creprpMl rsegvaiavztoü vágj pteném
R;' jelentése
feMF megválasztott, ahol R' és ^jelentése egymástól függetlenül hidrogén, halogén, manó, Cs.gF;kH, e^alksFOB, Cajerkioalksh mono- vagy polFhologénC^alkil, "C>,,«lkü"0~CV»aikjl, -O-C.^&amp;iksh nmm~vm P&amp;i-halogéo€M&amp;lk«eM, «s NR'R - céoponiából megváteiptF ahol íF jelentése hidrogén és Cj^alM! közti! niegvsiavziety K” jelentése hidrogén έν CMaikii közli! megválasztott; vagy ÍR1 és R” jelentése s- kapasoié# sdtrogsa aiom?t>td «yöti hgferooíitoos csoportsaméig Iva^iiisil, i-pirtondinik és Ι*|φ^«ΐωΐ·^ορ^||# «agvíapíott;;: sh<i iwiödégyll hetetöeíynm csoport adón esetben szabsstkoák halegén sz*ífesztitoc»ss«I; Kijelentése hidrogén és C{.4alkihközttl megválasziott; iR* jelentése hidrogén, C^aHuk mono· vagy poikhaiogénCj^alkil,éáRAaalkiF -OH csoportjából megvslas/Κϊη; vagy «onek N-oakt, vagy gyégyszetószetiieg alkalmazható só vagy szóivá; formája.
3. Az. I. vagy 2. igénypont szerinti vegyidet, vágy- e^ék;s?3e^j^ö^'f#^|^ ^°i RJ jéientésé fomlgameiv adói; esetben s»IttttiáR;:«gy egymásáéi ÉtggMipnll halogén, CMa!kth «iife vagy -O-C^Érk mm®»· «agfc $ö#* «halogénCj^aikiioxi, dana és -SÍR csoportból megválasztott; vagy jelentése
R.': jeleniése
köm! megválasztod, ahol .RJ és jelentése egymástól fílggetlenlHi nidrogéty CwálKO,. νηοηο- vágy pofv^olQgenC^^aik!!,/C^íálklI-OC^^álkik 'O-Cí.^ilkii, és ME‘£o esoport]áöói mégválaáátott; ahol .R' jelentése hidrogén és C^alM! közét; megválasztod; R” jeleaíése hidrogén és-C^alk!·! közöl ntegválasztolt; R': jeieniése hidrogén és C· ^alkit közöl megválasztott;; le jelentése hidrogén, Üuálkik morm- vagy poikhalogé-isC^alkil, -Gg^alkilrö-CM^Mk és "Ci.mlkíl-ΌΗ esopotíj ábó! megvál aszioté; vagy ennek gyégyazeíészetheg alkalmazható s.6 vagy szölvát formája,
4. Az 1--3. igénypontok bármelyike szerinti ve gyű let, vagy ennek sztereo izomer tormája, ahol R: jelentése fenik amely adott esetben szobsztítoáh egy vagy töhhszubszéiínenssek amely egymástól: függetlenül halogén, Cjmdkik mono- vagy poli-'lmlogértGi.mlkik -•Cr-CMaikik menő- vagy polk -lnriogénC i,.ía!:kiloxk oiano és -SFj csoportjából megválasztott vagy jelentése
R* jelemére
közli! megválasztott, ahol Rs és R* jelentése egymástól1 függetlenül hidrogén, Cj»C$4álktK> -Ci.48tk.il·, -O-Ci^alkd, és NRIR" csoportlábol megválászíotg ahol Rt jelentése hidrogén; R" jelentése hidrogén· R·' jelentése hidrogén és Cj.gdkH közül megválasztott; R- jelentése hidrogén és (.áralku csoportjából megválasztott; vagy ennek gyógyszerészeiilég alkalmazható só vagy szóivá· tormája.
5, At 1-4· igénypontok bármelyiké szerinti vegyniek vagy ennek sztereöizrmter íormáiá, ahol: R: jeieniése fénll, amely adott esetben szabszthuáll egy vagy több szubsztnüenssel, amely egymástól függetlenül balogén, Ci^álkik.foli-halogóftCi^aikö és -SFs esoportjáböf megválasztottig ieientese
.- 4 - köaöl iköl Bf ék 1* jelentése egymástól ftlggetboiH y4r«|^. Q#! ** ssoportJáM! meipélasatett; B5 jeteöMse é§<Si^élk® kö^il:m^gváfss:xíótl| E^jeiaöíise hidrogén; vagy ennek gyögysK8?ásaetíkg aikalmsáia^ sd yap polkái: tpm?#15' € A&amp; k--4. if|»ypn&amp;&amp; iárnpylke énfrt#]te. ^tO*4h*> -ΜΛ §. jv lémévé
s^psr^Mixmig^ászisít,^ a:.t|l>l?i:^álí0i£ó jele8t#§' »14; % fíyagpMí'kásáítmány, amely mtpláma: Isatófe^ííy msaayiségbéB $&amp; 1-m ígeöypöíók hánoglylks szerinti vegyületet tartalmaiba, ^é'g^»mészstíl«g.. alkalmazható hosdOKoanyág Mgy ssgédanyííg mellett 8i Ar i ~6, %éöypsjÉ6k feteelytké pepiid vegyliet vagy a ?v. Íg4nyi>ös4 :SMf5ats mény gyópótókdnt: történd alkalmazásra,
0, Ab 1-6. igénypontok bármelyike szerinti vegyidet vagy 3 ?. Igénypont szerinti gyogysaerkésbi" meny központi idegrendszeri állapotok vagy betegségek kezelésében vagy tssgpiözésélpn történő alkalmazásra, amely kedélybetepégek; öetMuna, dementla, amnMa.éaímifö.ilkögftitíf Mv&amp;iöl; általában: afe^t kiagyemmkoktmk gyerekeknél, fiataloknál diagnosztizált feetepögek; miyággái összefüggő rendeitönesságek; sklzeÉénia és mm psxiákotite beíepégék, szomaloíőrm rendellenességeit; és y^j«2on9ni%-:k|vM^^i1«^§é'g ktgö'l megválasztott. 1:0. Ax T'C igénypontok bártnelyike szerinti vegyütet vagy a ?. igénypont érinti gyégyszerkésziri Oíény központi idegrendszeri gavarok vagy állapotok kezelésében vagy: megelőzésében történő álkari amely depressziós betegségek; oeurokognhiv zavarok; neumáüvslgpmeteMs-mnieE^tesM* gek; anyaggal összefüggő és addikeiös rendellenességek; skizofrénia spektrum ős «3ás pzklíOŐSüs betegségek, szomaÉkms s«impitm« rolón rendellenességek; is hipefszoroooleneiás rendellenesség közül 'megválasztott. Π. Λ ö. vagy 10. igiayponsszerkükvgpület ^gy gyégpmj'klgzfíménpazadoüalkatoazám s^öís központi idegrendszeri állapot vágy1 bétegsig dememla. vagy nsuröiDptiffy záygti ipöly iöpmsszlős rendellenesség, depresszió, kezelésre rspszpHá ieptesszié, figyelte; sig;ŐS1 sfétzoírésia kö?:üí;áipgválásztpi;k :||, lp*És a ^pi|in>ppü;5tszersstŐ .^ói^2efkékzftPá|P''#l^ll1lásáíai ijsxzal jdknlejp^· Migy M$W*' smrismibég jIkálptazkáíötbordttziapi agof alaposait elkeverünk az Ίν&amp; ige nypornók:btamlg ske sze-ripti vegyülei terápiásán htííékösiy meímy iségévs; p; lennék, amely tartalmazzaaz b&amp; igénypontok bármelyike szériái vegytikoet és további gyógyszer hatóanyagok kombinál? készítmény formájában központi idegrendszeri állapotok és betegségek kezáléséMt? vap j&amp;§gelSz#Ífem ebköl%:ls$. 1¾^ «gyónás mim alkátezasra, amely ksílálybetegséiek; déimmk dementia. amnézia és más kognitív zavarok; általában először kis-gyermekeknélg.gywskekaélylIá^tófeiVfW^nbS^dt beiegségtfe anyaggal összefüggő rendellenességek; skizofrénia, és más pzkhdtíkM reodöllenessépk; is bipemzömniás alvási rendellenesség közöl mepllasztötí. 1:4. Ipmlk, arnel>· rnrmlmazzam 1-®· Igéffponíok bármelyike szerinti vegyiintet is íovábbi győgyv szer háföanpgot. kombinált klpítpény íermájáte közgottíi idegrimdszsri állapotok és bitápipk fegzéiésébeft vagy tnegelőzésébtm történő egyidejű, eiküíöintet? vagy egymás mám aikaltnázásmí: amely depressziós betegségek; nénfökognitiv zavarok; neorodevsiopmenfaÜs rendéiiersességgk; anyáipl összefüggő és addlkciós rendellenességek; skizofrénia spektrum és mis pztéhotikus betep ségek, szomatikus szítnpíéma és tokon tendöllenességek; és hiperszomnolenciás rendellenesség közül megválasztott. 15, (V) képletü vegyibe?
almi RJ: jelentése halogén, és R* és lejelentése az 1-6. igénypontok bármely ikében megadott. lí A '..A„.............. .......:...4t . ..._____. .. .· /\ÍÍ\ í . J.< . t : i. / M.
fi. A 15, vagy sí#' igénypont vegyüiet, amely <M3> vagy {1-13=0 képlettel ábrázolható
vagy ennek sója.
18. tH) kepletü vegyüto
ahol R\ R? és R4 jelentése a?. }-6. igénypontok bármelyikében megadott. m Eljárás m M* Igénypontok bármelyike ^érinti tlt képletü vegyüld cldál1Öás^%#mé|f-.:iai^ mázzá a kivetkező a|és b) tépéseket; képleti: -y^jgyöletet, ahol lejelentése balogéit, megfelelő bor vegyöíettel rcngáltattmk.pliá^ m^áSzfer.&amp;t megfelelő bázis és oldószer jelenlétében, ül) képiéin vegyidet előállításához.
b) az a) lépésben kapott (li> képlett! vegyáleiei Ollj klplptü yé^ülgttéi reagállatjukyahol X jelentess halogén, megteiclö kapcsoló reagens vagy katalizátor és lípndpöi jblénlétében, bázis jelenlétében, megfelelő oldószer ben
:2il Az í. mm 2, ímmnoot s^ylbti veövhléf, ahot >*
vagy M«e'fc !N*o.xíd, vagy gyógysxereszeíiJeg alkalmazható se vagy szoívét .formája,. 2k Λ 20. igénypont szerinti vagyaiét áltól s vagyaiét szabad bázis,; Mdíokloridso, azofíáisrk meránszuHbníUsó vagy ssaieátso, 22, A 20. Igénypont szeristi vegyülek ahol is vágyaiét
vagy esnek gyogyszerészeriieg alkalmazható só vágy szóival fbfsiáia, 23:, Az 1, vagy 2, igénypont szeried vsgyűleL ahol a vegyidet
vagy ennek győgyszerészstüag alkalínazb&amp;tó só vagy szölvái fonoda.
HUE14727824A 2013-06-04 2014-06-03 6,7-dihidropirazolo[l,5-a]pirazin-4(5H)-on vegyületek és ezek alkalmazása mint az mGluR2 receptorok negatív alloszterikus modulátorai HUE033057T2 (hu)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP13170447 2013-06-04
EP13173939 2013-06-27
EP14166450 2014-04-29

Publications (1)

Publication Number Publication Date
HUE033057T2 true HUE033057T2 (hu) 2017-11-28

Family

ID=50884400

Family Applications (1)

Application Number Title Priority Date Filing Date
HUE14727824A HUE033057T2 (hu) 2013-06-04 2014-06-03 6,7-dihidropirazolo[l,5-a]pirazin-4(5H)-on vegyületek és ezek alkalmazása mint az mGluR2 receptorok negatív alloszterikus modulátorai

Country Status (31)

Country Link
US (2) US10106542B2 (hu)
EP (1) EP3004107B1 (hu)
JP (1) JP6357533B2 (hu)
KR (1) KR102223050B1 (hu)
CN (2) CN105263935B (hu)
AU (1) AU2014276963C1 (hu)
BR (1) BR112015029982A2 (hu)
CA (1) CA2910507C (hu)
CL (1) CL2015003499A1 (hu)
CY (1) CY1119499T1 (hu)
DK (1) DK3004107T3 (hu)
EA (1) EA030426B1 (hu)
ES (1) ES2637295T3 (hu)
HK (2) HK1216527A1 (hu)
HR (1) HRP20171051T1 (hu)
HU (1) HUE033057T2 (hu)
IL (1) IL242868B (hu)
JO (1) JO3368B1 (hu)
LT (1) LT3004107T (hu)
ME (1) ME02949B (hu)
MX (1) MX368861B (hu)
MY (1) MY192029A (hu)
PH (1) PH12015502685B1 (hu)
PL (1) PL3004107T3 (hu)
PT (1) PT3004107T (hu)
RS (1) RS56128B1 (hu)
SG (1) SG11201509938YA (hu)
SI (1) SI3004107T1 (hu)
TW (2) TWI674264B (hu)
UA (1) UA117016C2 (hu)
WO (1) WO2014195311A1 (hu)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JO3368B1 (ar) 2013-06-04 2019-03-13 Janssen Pharmaceutica Nv مركبات 6، 7- ثاني هيدرو بيرازولو [5،1-a] بيرازين- 4 (5 يد)- اون واستخدامها بصفة منظمات تفارغية سلبية لمستقبلات ميجلور 2
DK3096790T3 (da) 2014-01-21 2019-10-07 Janssen Pharmaceutica Nv Kombinationer omfattende positive allosteriske modulatorer eller orthosteriske agonister af metabotrop glutamaterg subtype 2-receptor og anvendelse af disse
UA127921C2 (uk) 2014-01-21 2024-02-14 Янссен Фармацевтика Нв Комбінації, які містять позитивні алостеричні модулятори або ортостеричні агоністи метаботропного глутаматергічного рецептора 2 підтипу, та їх застосування
EP3112369A4 (en) * 2014-02-27 2017-09-20 The University of Tokyo Fused pyrazole derivative having autotaxin inhibitory activity
JOP20150177B1 (ar) 2014-08-01 2021-08-17 Janssen Pharmaceutica Nv مركبات 6 ، 7 ثاني هيدرو بيرازولو [ 1، 5 الفا ] بيرازين – 4 (5 يد) – اون واستخدامها كمنظمات الوسترية سلبية لمستقبلات ملجور 2
JOP20150179B1 (ar) 2014-08-01 2021-08-17 Janssen Pharmaceutica Nv مركبات 6 ، 7 ثاني هيدرو بيرازولو [ 1، 5 الفا ] بيرازين – 4 (5 يد) – اون واستخدامها كمنظمات الوسترية سلبية لمستقبلات ملجور 2
US10967078B2 (en) 2014-12-03 2021-04-06 Janssen Pharmaceutica Nv Radiolabelled mGluR2 PET ligands
AU2015357167B2 (en) 2014-12-03 2020-06-25 Janssen Pharmaceutica Nv 6,7-dihydropyrazolo(1,5-alpha)pyrazin-4(5H)-one compounds and their use as negative allosteric modulators of mGluR2 receptors
MA41338B1 (fr) 2015-01-16 2019-07-31 Hoffmann La Roche Composés de pyrazine pour le traitement de maladies infectieuses
CA2984711C (en) 2015-07-15 2023-08-29 F. Hoffmann-La Roche Ag Ethynyl derivatives as metabotropic glutamate receptor modulators
JP2018154554A (ja) * 2015-07-29 2018-10-04 大日本住友製薬株式会社 新規リンカー部位を持つ縮合ピラゾール誘導体およびその医薬用途
US11033641B2 (en) 2015-12-18 2021-06-15 Janssen Pharmaceutica Nv Radiolabelled mGluR2/3 pet ligands
US11045562B2 (en) 2015-12-18 2021-06-29 Janssen Pharmaceutica Nv Radiolabelled mGluR2/3 PET ligands
SI3484889T1 (sl) 2016-07-18 2020-11-30 F. Hoffmann-La Roche Ag Derivati etinila
JP6681517B2 (ja) 2016-09-27 2020-04-15 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. mGluR2陰性アロステリック調節剤としてのクロマン、イソクロマン及びジヒドロイソベンゾフラン誘導体、組成物、及びそれらの使用
CN111601811B (zh) 2017-11-24 2023-05-05 住友制药株式会社 6,7-二氢吡唑并[1,5-a]吡嗪酮衍生物和其医药用途
JP7266010B2 (ja) * 2019-05-30 2023-04-27 住友ファーマ株式会社 6,7-ジヒドロピラゾロ[1,5-a]ピラジノン誘導体又はその塩を含有する医薬
CA3225285A1 (en) * 2021-07-13 2023-01-19 Alessandro Boezio Pi3k.alpha. inhibitors and methods of use thereof

Family Cites Families (488)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2976146A (en) 1958-11-28 1961-03-21 Eastman Kodak Co Novel cyan-forming couplers
BE790440A (hu) 1971-10-23 1973-04-24 Bayer Ag
JPS5752334B2 (hu) 1974-02-05 1982-11-06
JPS538707B2 (hu) 1974-02-05 1978-03-31
US3906953A (en) 1974-05-23 1975-09-23 American Optical Corp Endoscopic surgical laser system
SU509578A1 (ru) 1974-09-19 1976-04-05 Стерлитамакский Химический Завод Способ получени пропилендиаминов
IE43079B1 (en) 1975-03-20 1980-12-17 Ici Ltd Quinolone derivatives
GB1502312A (en) 1975-03-20 1978-03-01 Ici Ltd Quinolone derivatives
FR2311776A1 (fr) 1975-05-23 1976-12-17 Sogeras Diamino-2,4 bromo-5 chloro-6 pyrimidines et procede pour leur preparation
GB1570494A (en) 1975-11-28 1980-07-02 Ici Ltd Thienopyrimidine derivatives and their use as pesticides
JPS5382783A (en) 1976-12-29 1978-07-21 Yamanouchi Pharmaceut Co Ltd Novel pyridone derivs and process for their preparation
US4196207A (en) 1977-05-23 1980-04-01 Ici Australia Limited Process for controlling eradicating or preventing infestations of animals by Ixodid ticks
DE2750288A1 (de) 1977-11-10 1979-05-17 Thomae Gmbh Dr K Neue 9-(omega-heteroarylamino- alkylamino)-erythromycine, ihre salze, verfahren zu ihrer herstellung und diese enthaltende arzneimittel
US4432979A (en) 1981-10-26 1984-02-21 William H. Rorer, Inc. Pyridone compounds
EP0082023A3 (en) 1981-12-16 1983-07-20 Sankyo Company Limited Thienopyrimidine derivatives, their preparation and their medical use
US4358453A (en) 1982-01-08 1982-11-09 Schering Corporation 1,2,4-Triazolo[4,3-a]pyridines
US4520025A (en) 1982-07-21 1985-05-28 William H. Rorer, Inc. Bicyclic nitrogen heterocyclic ethers and thioethers, and their pharmaceutical uses
JPS60159184A (ja) 1984-01-27 1985-08-20 Agency Of Ind Science & Technol 水電解用陽極
DE3406329A1 (de) 1984-02-22 1985-08-22 Merck Patent Gmbh, 6100 Darmstadt Pyridone
US4550166A (en) 1984-05-21 1985-10-29 American Cyanamid Company (Pyridinyl)-1,2,4-triazolo[4,3-a]pyridines
US5175157A (en) 1985-11-27 1992-12-29 Boehringer Ingelheim Gmbh Cyclic amine derivatives, pharmaceutical compositions containing these compounds and methods for preparing them
DE3717561A1 (de) 1987-05-25 1988-12-08 Thomae Gmbh Dr K Indol-, isochinolin- und benzazepinderivate, diese verbindungen enthaltende arzneimittel und verfahren zu ihrer herstellung
US4866074A (en) 1987-05-08 1989-09-12 Rorer Pharmaceutical Corporation Naphtheridinone- and pyridooxazinone-pyridone compounds, cardiotonic compositions including same, and their uses
EP0308020A3 (en) 1987-09-18 1990-12-05 Merck & Co. Inc. 5-(aryl and heteroaryl)-6-(aryl and heteroaryl)-1,2-dihydro-2-oxo 3-pyridinecarboxylic acids and derivatives thereof
US5260293A (en) 1988-01-30 1993-11-09 Merck Sharp & Dohme Limited Pyrazines, pyrimidines and pyridazines useful in the treatment of senile dementia
GB8804448D0 (en) 1988-02-25 1988-03-23 Smithkline Beckman Intercredit Compounds
JP2614081B2 (ja) 1988-05-27 1997-05-28 大塚化学株式会社 光学活性β−ラクタム誘導体の製造法
JPH02124871A (ja) 1988-07-27 1990-05-14 Dainippon Pharmaceut Co Ltd 1位が置換された複素環式カルボン酸アミド誘導体
EP0365486B1 (en) 1988-10-20 1995-07-19 Sandoz Ag Fibre-reactive azo dyes
US5032602A (en) 1988-12-14 1991-07-16 Bayer Aktiengesellschaft Inhibiting HMG-CoA reductase with novel substituted 2-pyridones and pyrid-2-thiones
HU206337B (en) 1988-12-29 1992-10-28 Mitsui Petrochemical Ind Process for producing pyrimidine derivatives and pharmaceutical compositions
JPH02277044A (ja) 1989-04-19 1990-11-13 Fuji Photo Film Co Ltd ハロゲン化銀写真感光材料
US5236917A (en) 1989-05-04 1993-08-17 Sterling Winthrop Inc. Saccharin derivatives useful as proteolytic enzyme inhibitors and compositions and method of use thereof
US5280026A (en) 1989-05-30 1994-01-18 Smithkline Beecham Intercredit B.V. Thienopyrimidines
AU622330B2 (en) 1989-06-23 1992-04-02 Takeda Chemical Industries Ltd. Condensed heterocyclic compounds having a nitrogen atom in the bridgehead for use as fungicides
US4978663A (en) 1989-08-16 1990-12-18 Hoechst-Roussel Pharmaceuticals Inc. 5-(1-aminocyclohexyl)-2(1H)-pyridinone compounds which have pharmaceutical utility
GB8926560D0 (en) 1989-11-24 1990-01-17 Zambeletti Spa L Pharmaceuticals
IL96432A0 (en) 1989-11-30 1991-08-16 Schering Ag Pesticidal compositions containing pyridine derivatives and novel pyridine derivatives
DE3940480A1 (de) 1989-12-07 1991-06-13 Bayer Ag Chromogene enaminverbindungen, ihre herstellung und verwendung als farbbildner
WO1991009848A1 (en) 1989-12-22 1991-07-11 The Upjohn Company Pyridinones useful as antiatherosclerotic agents
FR2657610A1 (fr) 1990-01-29 1991-08-02 Rhone Poulenc Agrochimie Triazolopyridines herbicides.
GB9104238D0 (en) 1990-03-16 1991-04-17 Ici Pharma 3-tetrazolylthiomethyl cephalosporin antibiotics
DE4008726A1 (de) 1990-03-19 1991-09-26 Basf Ag Thieno(2,3-d)pyrimidinderivate
CA2039411A1 (en) 1990-03-30 1991-10-01 Ronnie Gerald Edie Thienopyrimidine derivatives
RU1796625C (ru) 1990-06-27 1993-02-23 Киевский Государственный Университет Им.Т.Г.Шевченко 3-Амино-7-нитро-4(2,3,4-триметоксифенил)-2-фенил-1(2Н)изохинолон, обладающий аналептическим действием
EP0478195B1 (en) 1990-09-21 1999-05-26 Rohm And Haas Company Dihydropyridazinones and pyridazinones as fungicides
KR920008026A (ko) 1990-10-24 1992-05-27 오노 화아마슈티칼 캄파니 리미팃드 이소퀴놀리논 유도체 또는 이의 무독성 산부가염 또는 이의 수화물, 이의 제조방법 및 이를 포함하는 약제 조성물
WO1992018115A1 (en) 1991-04-18 1992-10-29 Dr Lo Zambeletti S.P.A. Use of heterocyclic compounds for the treatment of inflammatory pain
DE4122240A1 (de) 1991-07-05 1993-01-07 Boehringer Mannheim Gmbh Dibenz(b,e)azepinderivate und diese enthaltende arzneimittel
US5332750A (en) 1991-09-04 1994-07-26 Merck Patent Gesellschaft Mit Beschrankter Haftung 1,2-dihydro-2-oxopyridines
DE4129340A1 (de) 1991-09-04 1993-03-11 Merck Patent Gmbh 1,2-dihydro-2-oxopyridine
DE4131924A1 (de) 1991-09-25 1993-07-08 Hoechst Ag Substituierte 4-alkoxypyrimidine, verfahren zu ihrer herstellung und ihre verwendung als schaedlingsbekaempfungsmittel
US5204198A (en) 1991-10-28 1993-04-20 Eastman Kodak Company Photoelectrographic elements utilizing nonionic sulfonic acid photogenerators
US5416099A (en) 1991-10-29 1995-05-16 Merck & Co., Inc. Fibrinogen receptor antagonists
DE4221583A1 (de) 1991-11-12 1993-05-13 Bayer Ag Substituierte biphenylpyridone
JP2878531B2 (ja) 1991-12-16 1999-04-05 富士写真フイルム株式会社 ハロゲン化銀写真感光材料
US5378720A (en) 1991-12-19 1995-01-03 Sterling Winthrop Inc. Saccharin derivative proteolytic enzyme inhibitors
AU668694B2 (en) 1991-12-19 1996-05-16 Sanofi-Synthelabo Saccharin derivative proteolytic enzyme inhibitors
TW219935B (hu) 1991-12-25 1994-02-01 Mitsubishi Chemicals Co Ltd
GB9200293D0 (en) 1992-01-08 1992-02-26 Wyeth John & Brother Ltd Piperazine derivatives
GB9201694D0 (en) 1992-01-27 1992-03-11 Smithkline Beecham Intercredit Compounds
JPH05204071A (ja) 1992-01-29 1993-08-13 Konica Corp ハロゲン化銀写真感光材料
JP3042156B2 (ja) 1992-02-20 2000-05-15 田辺製薬株式会社 ナフタレン誘導体、その製法及びその合成中間体
DE4206045A1 (de) 1992-02-27 1993-09-02 Bayer Ag Sulfonylbenzyl substituierte pyridone
US5922773A (en) 1992-12-04 1999-07-13 The Children's Medical Center Corp. Glaucoma treatment
AU660132B2 (en) 1992-12-21 1995-06-08 Bayer Aktiengesellschaft Substituted 4-phenyl-pyridones and 4-phenyl-2-alkoxypyridine
JPH06211798A (ja) 1993-01-19 1994-08-02 Tosoh Corp テトラヒドロイソキノリノン誘導体及びその製造方法
JPH06211797A (ja) 1993-01-19 1994-08-02 Tosoh Corp テトラヒドロイソキノリン誘導体、その製法及び抗真菌剤
SE9300657D0 (sv) 1993-02-26 1993-02-26 Astra Ab New compounds
US5814645A (en) 1993-03-24 1998-09-29 Bayer Aktiengesellschaft Arylor hetaryl substituted nitrogen heterocycles and their use as pesticides
DE4316077A1 (de) 1993-05-13 1994-11-17 Bayer Ag Substituierte Mono- und Bihydridylmethylpyridone
WO1994029273A1 (en) 1993-06-09 1994-12-22 Smithkline Beecham Corporation Bicyclic fibrinogen antagonists
JP3333271B2 (ja) 1993-06-17 2002-10-15 大日本印刷株式会社 自動丁合装置
JPH07101861A (ja) 1993-08-10 1995-04-18 Tanabe Seiyaku Co Ltd 抗喘息薬
DE4326758A1 (de) 1993-08-10 1995-02-16 Basf Ag [1,3,4]Triazolo[1,5-a]pyridine
NZ271994A (en) 1993-08-19 1997-06-24 Janssen Pharmaceutica Nv Aryloxyalkylaminoalkyl-guanidine derivatives, cyclised analogues thereof and medicaments
CZ287771B6 (en) 1993-08-19 2001-01-17 Janssen Pharmaceutica Nv Dihydrobenzopyran derivatives, process and intermediates for their preparation, their use and pharmaceutical preparations based thereon
AU7467294A (en) 1993-08-20 1995-03-21 Banyu Pharmaceutical Co., Ltd. Tyrosine kinase inhibitor
JP3701984B2 (ja) 1993-08-31 2005-10-05 サントリー株式会社 ラベル化シクロプロパン誘導体およびその製造方法
US5424435A (en) 1993-10-18 1995-06-13 Olin Corporation 1-hydroxy-6-substituted-2-pyridones
US5500420A (en) 1993-12-20 1996-03-19 Cornell Research Foundation, Inc. Metabotropic glutamate receptor agonists in the treatment of cerebral ischemia
US5654307A (en) 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
KR100383305B1 (ko) 1994-03-10 2003-11-01 후지사와 야꾸힝 고교 가부시키가이샤 프로스타글란딘i₂작용물질로서의나프탈렌유도체
ES2079323B1 (es) 1994-06-21 1996-10-16 Vita Invest Sa Derivados de indol utiles para el tratamiento de la migraña, composicion y uso correspondientes.
GB9416554D0 (en) 1994-08-19 1994-10-12 Ciba Geigy Ag Glutamate receptor
AU3325295A (en) 1994-08-24 1996-03-14 Eli Lilly And Company Pyrrolidinyl di-carboxylic acid derivatives as metabotropic glutamate receptor antagonists
US6017697A (en) 1994-11-14 2000-01-25 Eli Lilly And Company Excitatory amino acid receptor protein and related nucleic acid compounds
US5473077A (en) 1994-11-14 1995-12-05 Eli Lilly And Company Pyrrolidinyl di-carboxylic acid derivatives as metabotropic glutamate receptor agonists
US5512576A (en) 1994-12-02 1996-04-30 Sterling Winthrop Inc. 2-substituted 1,2,5,-thiadiazolidin-3-one 1,1-dioxides and compositions and method of use thereof
US5789426A (en) 1995-01-20 1998-08-04 Cornell Research Foundation, Inc. Method for the treatment of fibroproliferative disorders by application of inhibitors of protein hydroxylation
US5869486A (en) 1995-02-24 1999-02-09 Ono Pharmaceutical Co., Ltd. Fused pyrimidines and pyriazines as pharmaceutical compounds
DE19507522C2 (de) 1995-03-03 2003-05-28 Basf Ag Verfahren zur Herstellung von 3,4-Dihydroisochinolinverbindungen und 3,4-Dihydroisochinolinium-Salzen
US5869428A (en) 1995-03-13 1999-02-09 Ishihara Sangyo Kaisha Ltd. Pyridonesulfonylurea compounds, process for their production and herbicides containing them
DE19510965A1 (de) 1995-03-24 1996-09-26 Asta Medica Ag Neue Pyrido/3,2-e/pyrazinone mit antiasthmatischer Wirksamkeit und Verfahren zu deren Herstellung
CN1188401C (zh) 1995-04-27 2005-02-09 三菱制药株式会社 杂环类酰胺化合物及其医药用途
JPH08325248A (ja) 1995-05-26 1996-12-10 Chugoku Kayaku Kk テトラゾール類の新規な合成試薬及びそれを用いたテトラゾール類の製造方法
US5849587A (en) 1995-06-09 1998-12-15 Cornell Research Foundation, Inc. Method of inhibiting viral replication in eukaryotic cells and of inducing apoptosis of virally-infected cells
EP0756200B1 (en) 1995-07-26 1999-11-10 Konica Corporation Silver halide color photographic light-sensitive material
US5659033A (en) 1995-09-13 1997-08-19 Neurogen Corporation N-aminoalkylfluorenecarboxamides; a new class of dopamine receptor subtype specific ligands
TR199800464T1 (xx) 1995-09-15 1998-05-21 Synthelabo Serotonin antagonistleri olarak kinolein-2(1H)-on t�revleri
JPH1045750A (ja) 1995-09-20 1998-02-17 Takeda Chem Ind Ltd アゾール化合物、その製造方法及び用途
US6130217A (en) 1995-09-20 2000-10-10 Pfizer Inc Compounds enhancing antitumor activity of other cytotoxic agents
AR004010A1 (es) 1995-10-11 1998-09-30 Glaxo Group Ltd Compuestos heterociclicos
CA2231105C (en) 1995-12-08 2005-09-13 Janssen Pharmaceutica N.V. Farnesyl protein transferase inhibiting (imidazol-5-yl)methyl-2-quinolinone derivatives
GB9602166D0 (en) 1996-02-02 1996-04-03 Zeneca Ltd Aminoheterocyclic derivatives
WO1997028128A1 (en) 1996-02-02 1997-08-07 Zeneca Limited Heterocyclic compounds useful as pharmaceutical agents
GB9602294D0 (en) 1996-02-05 1996-04-03 Zeneca Ltd Heterocyclic compounds
US6084084A (en) 1996-02-21 2000-07-04 Nps Pharmaceuticals, Inc. Human metabotropic glutamate receptor
US5710274A (en) 1996-02-28 1998-01-20 Neurogen Corporation N-aminoalkyldibenzofurancarboxamides; new dopamine receptor subtype specific ligands
US5756518A (en) 1996-04-02 1998-05-26 Kowa Co., Ltd. Phenylene derivatives
JPH1029979A (ja) 1996-04-12 1998-02-03 Ajinomoto Co Inc 新規ピリジン誘導体
US5741798A (en) 1996-06-03 1998-04-21 Boehringer Ingelheim Pharmaceuticals, Inc. 2-benzyl-4-sulfonyl-4H-isoquinolin-1,3-diones and their use as antiinflammatory agents
AU3783497A (en) 1996-08-09 1998-03-06 Yamanouchi Pharmaceutical Co., Ltd. Metabotropic glutamate receptor agonists
DE19632423A1 (de) 1996-08-12 1998-02-19 Merck Patent Gmbh Thienopyrimidine
ATE230399T1 (de) 1996-08-14 2003-01-15 Astrazeneca Ab Substituierte pyrimidinderivate und ihre pharmazeutische anwendung
JP2001502300A (ja) 1996-09-16 2001-02-20 デュポン ファーマシューティカルズ カンパニー ピラジノン類およびトリアジノン類およびその誘導体類
DE19638486A1 (de) 1996-09-20 1998-03-26 Basf Ag Hetaroylderivate
DE19638484A1 (de) 1996-09-20 1998-03-26 Basf Ag Hetaroylderivate
DE19644228A1 (de) 1996-10-24 1998-04-30 Merck Patent Gmbh Thienopyrimidine
US6284794B1 (en) 1996-11-05 2001-09-04 Head Explorer Aps Method for treating tension-type headache with inhibitors of nitric oxide and nitric oxide synthase
HUP0001140A3 (en) 1996-12-05 2002-05-28 Amgen Inc Thousand Oaks Substituted pyrimidinone and pyridone compounds and methods of use
DE19653647A1 (de) 1996-12-20 1998-06-25 Hoechst Ag Vitronectin - Rezeptorantagonisten, deren Herstellung sowie deren Verwendung
US6482821B2 (en) 1996-12-20 2002-11-19 Hoechst Aktiengellschaft Vitronectin receptor antagonists, their preparation and their use
JP4352115B2 (ja) 1997-01-24 2009-10-28 クラヴィス・ファルマ・アーエスアー ゲムシタビン誘導体
US5855654A (en) 1997-01-30 1999-01-05 Rohm And Haas Company Pyridazinones as marine antifouling agents
FR2759366B1 (fr) 1997-02-11 1999-04-16 Centre Nat Rech Scient Composes constituant notamment des effecteurs de recepteurs du systeme nerveux central sensibles aux amino acides neuroexcitateurs, leur preparation et leurs applications biologiques
US6262068B1 (en) 1997-02-21 2001-07-17 Bristol-Myers Squibb Company Lactam derivatives as antiarrhythmic agents
WO1998038168A1 (en) 1997-02-27 1998-09-03 Tanabe Seiyaku Co., Ltd. Isoquinolinone derivatives, process for preparing the same, and their use as phosphodiesterase inhibitors
ES2131463B1 (es) 1997-04-08 2000-03-01 Lilly Sa Derivados de ciclopropilglicina con propiedades farmaceuticas.
GB9708945D0 (en) 1997-05-01 1997-06-25 Merck Sharp & Dohme Therapeutic agents
DE19728996A1 (de) 1997-07-07 1999-01-14 Basf Ag Triazolverbindungen und deren Verwendung
SI0891978T1 (en) 1997-07-18 2002-06-30 F. Hoffmann-La Roche Ag 5H-Thiazolo (3,2-a) pyrimidine derivatives
AU743899B2 (en) 1997-07-18 2002-02-07 Georgetown University Bicyclic metabotropic glutamate receptor ligands
CN1265590A (zh) 1997-07-31 2000-09-06 赛尔金有限公司 取代的链烷异羟肟酸及降低肿瘤坏死因子α水平的方法
CN1154489C (zh) 1997-08-14 2004-06-23 弗·哈夫曼-拉罗切有限公司 抗神经学疾病的杂环乙烯基醚
US6358975B1 (en) 1997-08-15 2002-03-19 Johns Hopkins University Method of using selective parp inhibitors to prevent or treat neurotoxicity
AU9298098A (en) 1997-09-03 1999-03-22 Guilford Pharmaceuticals Inc. Amino-substituted compounds, methods, and compositions for inhibiting parp activity
US20020022636A1 (en) 1997-09-03 2002-02-21 Jia-He Li Oxo-substituted compounds, process of making, and compositions and methods for inhibiting parp activity
US6121278A (en) 1997-09-03 2000-09-19 Guilford Pharmaceuticals, Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
US6197785B1 (en) 1997-09-03 2001-03-06 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US20020028813A1 (en) 1997-09-03 2002-03-07 Paul F. Jackson Thioalkyl compounds, methods, and compositions for inhibiting parp activity
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
WO1999012532A2 (en) 1997-09-08 1999-03-18 F.Hoffmann-La Roche Ag Piperidine derivatives against malaria
DE69815008T2 (de) 1997-09-19 2004-04-01 Ssp Co., Ltd. Alfa-substituierte Phenylpropionsäurederivate und diese enthaltende Arzneimittel
EP1017682A4 (en) 1997-09-26 2000-11-08 Merck & Co Inc NEW ANGIOGENESIS INHIBITORS
US6465484B1 (en) 1997-09-26 2002-10-15 Merck & Co., Inc. Angiogenesis inhibitors
US6162804A (en) 1997-09-26 2000-12-19 Merck & Co., Inc. Tyrosine kinase inhibitors
WO1999018096A1 (en) 1997-10-02 1999-04-15 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
IL135588A0 (en) 1997-10-14 2001-05-20 Welfide Corp Piperazine derivatives and pharmaceutical compositions containing the same
WO1999021992A2 (en) 1997-10-23 1999-05-06 Ganimed Pharmaceuticals Gmbh Nucleic acid molecules encoding a glutamate receptor
JP4321737B2 (ja) 1997-12-17 2009-08-26 塩野義製薬株式会社 新規ピリジン化合物
US6013672A (en) 1997-12-18 2000-01-11 Uab Research Foundation Agonists of metabotropic glutamate receptors and uses thereof
EP1045836B1 (en) 1997-12-18 2008-03-05 Boehringer Ingelheim Pharmaceuticals Inc. Pyridones as src family sh2 domain inhibitors
ATE389636T1 (de) 1997-12-19 2008-04-15 Amgen Inc Substituierte pyridin- und pyridazinderivate und ihre pharmazeutische verwendung
FR2772763B1 (fr) 1997-12-24 2004-01-23 Sod Conseils Rech Applic Nouveaux analogues tetracycliques de camptothecines, leurs procedes de preparation, leur application comme medicaments et les compositions pharmaceutiques les contenant
IT1298155B1 (it) 1998-01-19 1999-12-20 Moreno Paolini Composti pirimidin 3-ossido per il trattamento delle patologie muscolo-scheletriche, in particolare per il trattamento della
US6664250B2 (en) 1998-01-20 2003-12-16 Bristol-Myers Squibb Co. Lactam derivatives as antiarrhythmic agents
CA2318800C (en) 1998-01-28 2005-12-20 Taisho Pharmaceutical Co., Ltd. Fluorine-containing amino acid derivatives
CA2321153A1 (en) 1998-02-17 1999-08-19 Timothy D. Cushing Anti-viral pyrimidine derivatives
IL138342A0 (en) 1998-03-17 2001-10-31 Pfizer Prod Inc Bicyclo[2.2.1] heptanes and related compounds
CN1295570A (zh) 1998-04-08 2001-05-16 诺瓦提斯公司 N-吡啶酮基除草剂
WO1999053956A1 (fr) 1998-04-16 1999-10-28 Yamanouchi Pharmaceutical Co., Ltd. Remedes contre l'obesite
EP0955301A3 (en) 1998-04-27 2001-04-18 Pfizer Products Inc. 7-aza-bicyclo[2.2.1]-heptane derivatives, their preparation and use according to their affinity for neuronal nicotinic acetylcholine receptors
DE19822198C2 (de) 1998-05-16 2003-02-13 Wella Ag Oxonolfarbstoffe enthaltende Mittel und Verfahren zur Erzeugung von semipermanenten Färbungen auf Haaren
US6288054B1 (en) 1998-06-02 2001-09-11 Merck & Co., Inc. Carbapenem antibacterial compounds, compositions containing such compounds and methods of treatment
SK18542000A3 (sk) 1998-06-04 2001-12-03 Abbott Laboratories Protizápalové zlúčeniny inhibujúce bunkovú adhéziu
DE19826671A1 (de) 1998-06-16 1999-12-23 Hoechst Schering Agrevo Gmbh 1,3-Oxazolin- und 1,3-Thiazolin-Derivate, Verfahren zu ihrer Herstellung und ihre Verwendung als Schädlingsbekämpfungsmittel
FR2781218B1 (fr) 1998-07-15 2001-09-07 Lafon Labor Compositions pharmaceutiques comprenant des 2-quinolones
JP2000072751A (ja) 1998-08-26 2000-03-07 Tanabe Seiyaku Co Ltd イソキノリノン誘導体
CA2341865C (en) 1998-08-31 2006-01-17 Taisho Pharmaceutical Co., Ltd. 6-fluorobicyclo[3.1.0]hexane derivatives
JP2000072731A (ja) 1998-08-31 2000-03-07 Taisho Pharmaceut Co Ltd 4−置換−2−アミノビシクロ[3.1.0]ヘキサン−2,6−ジカルボン酸誘導体及び製薬用組成物
AU760020B2 (en) 1998-08-31 2003-05-08 Merck & Co., Inc. Novel angiogenesis inhibitors
CH694053A5 (de) 1998-09-03 2004-06-30 Hoffmann La Roche Verfahren zur Herstellung von 2-Amino-bicyclo[3.1.0]hexan-2,6-dicarbonsäure-Derivaten.
US6284759B1 (en) 1998-09-30 2001-09-04 Neurogen Corporation 2-piperazinoalkylaminobenzo-azole derivatives: dopamine receptor subtype specific ligands
SE9803518D0 (sv) 1998-10-15 1998-10-15 Astra Pharma Prod Novel compounds
PE20001236A1 (es) 1998-11-13 2000-11-10 Lilly Co Eli Moduladores del receptor de aminoacidos excitadores
US6133271A (en) 1998-11-19 2000-10-17 Cell Pathways, Inc. Method for inhibiting neoplastic cells and related conditions by exposure thienopyrimidine derivatives
US5948911A (en) 1998-11-20 1999-09-07 Cell Pathways, Inc. Method for inhibiting neoplastic cells and related conditions by exposure to thienopyrimidine derivatives
EP1006112A1 (en) 1998-12-01 2000-06-07 Cerebrus Pharmaceuticals Limited 3-Hydroxy-2(1H)-pyridinone or 3-hydroxy-4(1H)-pyridinone derivatives useful as reactive oxygen species (ROS) scavengers
WO2000034244A1 (en) 1998-12-04 2000-06-15 Bristol-Myers Squibb Company 3-substituted-4-arylquinolin-2-one derivatives as potassium channel modulators
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
TW564247B (en) 1999-04-08 2003-12-01 Akzo Nobel Nv Bicyclic heteraromatic compound
GB9908175D0 (en) 1999-04-09 1999-06-02 Lilly Co Eli Method of treating neurological disorders
US6972296B2 (en) 1999-05-07 2005-12-06 Encysive Pharmaceuticals Inc. Carboxylic acid derivatives that inhibit the binding of integrins to their receptors
US6723711B2 (en) 1999-05-07 2004-04-20 Texas Biotechnology Corporation Propanoic acid derivatives that inhibit the binding of integrins to their receptors
WO2000069816A1 (en) 1999-05-17 2000-11-23 Eli Lilly And Company Metabotropic glutamate receptor antagonists
DK1196397T3 (da) 1999-06-02 2006-01-02 Nps Pharma Inc Metabotrope glutamatreceptorantagonister og anvendelse deraf til behandling af sygdomme i centralnervesystemet
CA2390948A1 (en) 1999-06-03 2000-12-14 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
AU5073600A (en) 1999-06-03 2000-12-28 Lilly, S.A. Excitatory amino acid receptor modulators
JP4783967B2 (ja) 1999-07-21 2011-09-28 大正製薬株式会社 含フッ素アミノ酸誘導体を有効成分とする医薬
EP1210338A2 (en) 1999-08-05 2002-06-05 IGT Pharma Inc. 1,4-diazepine derivatives for the treatment of diseases related to the central nervous system
US6660753B2 (en) 1999-08-19 2003-12-09 Nps Pharmaceuticals, Inc. Heteropolycyclic compounds and their use as metabotropic glutamate receptor antagonists
US7040838B2 (en) 1999-08-27 2006-05-09 Kristar Enterprises, Inc. High capacity catch basin filtration system with adjustable deflector ring
RU2259360C2 (ru) 1999-10-15 2005-08-27 Ф.Хоффманн-Ля Рош Аг Производные бензодиазепина и лекарственное средство, их содержащее
AU779874B2 (en) 1999-10-15 2005-02-17 F. Hoffmann-La Roche Ag Benzodiazepine derivatives
IL148891A0 (en) 1999-10-19 2002-09-12 Merck & Co Inc Tyrosine kinase inhibitors
SV2002000205A (es) 1999-11-01 2002-06-07 Lilly Co Eli Compuestos farmaceuticos ref. x-01095
GB2355982A (en) 1999-11-03 2001-05-09 Lilly Co Eli Heterocyclic amino acids
WO2001046190A1 (fr) 1999-12-22 2001-06-28 Kyorin Pharmaceutical Co., Ltd. Composes tricycliques et sels d'addition de ceux-ci
AU779442B2 (en) 2000-01-20 2005-01-27 Eisai Co. Ltd. Novel piperidine compounds and drugs containing the same
GB0002100D0 (en) 2000-01-28 2000-03-22 Novartis Ag Organic compounds
US6800651B2 (en) 2000-02-03 2004-10-05 Eli Lilly And Company Potentiators of glutamate receptors
CZ20023199A3 (cs) 2000-02-25 2003-05-14 F. Hoffmann-La Roche Ag Modulátory receptoru adenosinu
DE10012373A1 (de) 2000-03-14 2001-09-20 Dresden Arzneimittel Verwendung von Pyrido[3,2-e]-pyrazinonen als Inhibitoren der Phosphodiesterase 5 zur Therapie von erektiler Dysfunktion
GB0007108D0 (en) 2000-03-23 2000-05-17 Novartis Ag Organic compounds
JP2001270871A (ja) 2000-03-24 2001-10-02 Sumitomo Chem Co Ltd オキシラン化合物の製造方法
US6469026B2 (en) 2000-03-24 2002-10-22 Millennium Pharmaceuticals, Inc. Isoquinolone inhibitors of factor Xa
GB0007193D0 (en) 2000-03-25 2000-05-17 Univ Manchester Treatment of movrmrnt disorders
US6403588B1 (en) 2000-04-27 2002-06-11 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
CN1173975C (zh) 2000-04-27 2004-11-03 山之内制药株式会社 咪唑并吡啶衍生物
CZ301809B6 (cs) 2000-04-28 2010-06-30 Nihon Nohyaku Co., Ltd. Zpusob prípravy 2-halogenbenzoových kyselin a jejich derivátu
EP1288199A4 (en) 2000-04-28 2005-10-12 Shionogi & Co INHIBITORS OF MMP-12
MXPA02011053A (es) 2000-05-11 2004-08-19 Kenneth Curry Compuestos novedosos de carboxi amino espiro (2,4)heptano y derivados de los mismos.
US20020009713A1 (en) 2000-05-11 2002-01-24 Miller Freda D. Methods for identifying modulators of neuronal growth
WO2001085716A1 (en) 2000-05-11 2001-11-15 Kyowa Hakko Kogyo Co., Ltd 2-piperidone compounds for the treatment of cancer
US7081481B2 (en) 2000-05-31 2006-07-25 Eli Lilly And Company Excitatory amino acid receptor modulators
US6949571B2 (en) 2000-06-12 2005-09-27 Eisai Co., Ltd. 1,2-dihydropyridine compounds, process for preparation of the same and use thereof
JP2002012533A (ja) 2000-06-27 2002-01-15 Kao Corp 染毛剤組成物
JP2002003401A (ja) 2000-06-27 2002-01-09 Japan Science & Technology Corp 脳由来神経栄養因子誘導剤
WO2002000715A2 (en) 2000-06-27 2002-01-03 Centre National De La Recherche Scientifique-Cnrs Mammal 2p domain mechano-sensitive k+ channel, cloning and applications thereof
WO2002000605A1 (fr) 2000-06-28 2002-01-03 Taisho Pharmaceutical Co., Ltd. Nouveaux derives d'acide dicarboxylique
DE10031390A1 (de) 2000-07-03 2002-01-17 Knoll Ag Pyrimidinderivate und ihre Verwendung zur Prophylaxe und Therapie der zerebralen Ischämie
US20020041880A1 (en) 2000-07-05 2002-04-11 Defeo-Jones Deborah Method of treating cancer
US6861530B2 (en) 2000-07-07 2005-03-01 Kyowa Hakko Kogyo Co., Ltd. Piperidine derivatives
JP2002040252A (ja) 2000-07-27 2002-02-06 Shiseido Co Ltd コレステリック液晶層を含む光学シート、それを用いた情報記録体、情報記録方法並びに情報判別方法
DE10038019A1 (de) 2000-08-04 2002-02-14 Bayer Ag Substituierte Triazolopyrid(az)ine
CN1446202A (zh) 2000-08-11 2003-10-01 卫材株式会社 2-氨基吡啶化合物及其作为药物的用途
KR100732206B1 (ko) 2000-09-11 2007-06-27 노바티스 백신즈 앤드 다이아그노스틱스 인코포레이티드 티로신 키나제 억제제로서의 퀴놀리논 유도체
US6599940B2 (en) 2000-09-13 2003-07-29 Georgetown University Synthesis of 2-hydroxymethylglutamic acid and congeners thereof
JP2002105085A (ja) 2000-09-28 2002-04-10 Yamanouchi Pharmaceut Co Ltd 新規イミダゾチアゾール誘導体
BR0114253A (pt) 2000-10-02 2003-07-01 Janssen Pharmaceutica Nv Antagonistas de receptor de glutamato metabotrópico
DE10058663A1 (de) 2000-11-25 2002-05-29 Merck Patent Gmbh Verwendung von Thienopyrimidinen
WO2002051849A1 (fr) 2000-12-26 2002-07-04 Daiichi Pharmaceutical Co., Ltd. Inhibiteurs cdk4
ATE438624T1 (de) 2000-12-28 2009-08-15 Shionogi & Co 2-pyridonderivate mit affinität für den cannabinoid-typ-2-rezeptor
JP2002308882A (ja) 2001-02-08 2002-10-23 Yamanouchi Pharmaceut Co Ltd チエノピリミジン誘導体
AU2002258400A1 (en) 2001-02-16 2002-08-28 Tularik Inc. Methods of using pyrimidine-based antiviral agents
ES2360205T3 (es) 2001-03-02 2011-06-01 Agennix Ag Sistema de ensayo de tres híbridos.
AU2002308354A1 (en) 2001-03-08 2002-09-24 Ilfa Industrieelektronik Und Leiterplattenfertigung Aller Art Gmbh Multilayer printed circuit board
US6831074B2 (en) 2001-03-16 2004-12-14 Pfizer Inc Pharmaceutically active compounds
US6596731B2 (en) 2001-03-27 2003-07-22 Hoffmann-La Roche Inc. Substituted imidazo[1,2-A] pyridine derivatives
JP2004527244A (ja) 2001-04-02 2004-09-09 メルク エンド カムパニー インコーポレーテッド 受容体型キナーゼ阻害剤の活性のインビボ測定方法
ATE299868T1 (de) 2001-04-12 2005-08-15 Hoffmann La Roche Dihydro-benzo (b) (1,4) diazepin-2-on-derivate als mglur2 antagonisten
KR100566178B1 (ko) 2001-04-12 2006-03-29 에프. 호프만-라 로슈 아게 mGluR2 길항물질 I로서의디하이드로-벤조[b][1,4]디아제핀-2-온 유도체
SE0101579D0 (sv) 2001-05-04 2001-05-04 Astrazeneca Ab New compounds
CZ20033053A3 (en) 2001-05-14 2004-05-12 Bristol@Myersásquibbápharmaácompany Substituted pyrazinones, pyridines and pyrimidines as corticotropin releasing factor ligands
US7144903B2 (en) 2001-05-23 2006-12-05 Amgen Inc. CCR4 antagonists
CA2448306A1 (en) 2001-05-30 2002-12-05 Alteon Inc. Method for treating fibrotic diseases or other indications
EP1414464A4 (en) 2001-05-30 2005-06-22 Alteon Inc METHOD OF TREATING GLAUCOMA
US20030114448A1 (en) 2001-05-31 2003-06-19 Millennium Pharmaceuticals, Inc. Inhibitors of factor Xa
AU2002310156A1 (en) 2001-06-05 2002-12-16 Boehringer Ingelheim Pharmaceuticals, Inc. 1,4-disubstituted benzo-fused cycloalkyl urea compounds
CA2451057A1 (en) 2001-06-14 2002-12-27 Banyu Pharmaceutical Co., Ltd. Novel isoxazolopyridone derivatives and use thereof
JP2005500294A (ja) 2001-06-19 2005-01-06 ブリストル−マイヤーズ スクイブ カンパニー ホスホジエステラーゼ7に対するピリミジン阻害剤
JP2003012653A (ja) 2001-06-28 2003-01-15 Yamanouchi Pharmaceut Co Ltd キナゾリン誘導体
JP2005504034A (ja) 2001-08-02 2005-02-10 ニューロクライン バイオサイエンシーズ, インコーポレイテッド ゴナドトロピン放出性ホルモンレセプターアンタゴニストとしてのピリジノン誘導体およびピリダジノン誘導体
JP2005508904A (ja) 2001-09-11 2005-04-07 スミスクライン ビーチャム コーポレーション 血管新生阻害剤としてのフロ−及びチエノピリミジン誘導体
JP4459619B2 (ja) 2001-10-02 2010-04-28 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー 化合物
TWI330183B (hu) 2001-10-22 2010-09-11 Eisai R&D Man Co Ltd
TW200406466A (en) 2001-11-13 2004-05-01 Ciba Sc Holding Ag Compositions comprising at least one oxonol dye and at least one metal complex
US6921762B2 (en) 2001-11-16 2005-07-26 Amgen Inc. Substituted indolizine-like compounds and methods of use
GB0129260D0 (en) 2001-12-06 2002-01-23 Eisai London Res Lab Ltd Pharmaceutical compositions and their uses
WO2003051841A2 (en) 2001-12-14 2003-06-26 Novo Nordisk A/S Compounds and uses thereof for decreasing activity of hormone-sensitive lipase
EP1465869B1 (en) 2001-12-21 2013-05-15 Exelixis Patent Company LLC Modulators of lxr
DE10164139A1 (de) 2001-12-27 2003-07-10 Bayer Ag 2-Heteroarylcarbonsäureamide
AU2002359923B2 (en) 2001-12-27 2007-12-20 Taisho Pharmaceutical Co., Ltd. 6-fluorobicyclo[3.1.0]hexane derivatives
JP2005170790A (ja) 2002-01-09 2005-06-30 Ajinomoto Co Inc N−アルキルスルフォニル置換アミド誘導体
EP1465862A1 (en) 2002-01-17 2004-10-13 SmithKline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin k inhibitors
EP1513522A2 (en) 2002-01-18 2005-03-16 Sri International Methods of treating conditions associated with an edg receptor
US20050113283A1 (en) 2002-01-18 2005-05-26 David Solow-Cordero Methods of treating conditions associated with an EDG-4 receptor
WO2003064428A1 (en) 2002-01-29 2003-08-07 H. Lundbeck A/S Furano- and thienopyrimidines as neurokinase inhibitors
US6949542B2 (en) 2002-02-06 2005-09-27 Hoffman-La Roche Inc. Dihydro-benzo[b][1,4]diazepin-2-one derivatives
EP1482916A4 (en) 2002-02-07 2007-12-12 Univ Miami THERAPY BASED ON SCHWANN CELLS AND PHOSPHODIESTERASE INHIBITORS
US20040116489A1 (en) 2002-02-12 2004-06-17 Massey Steven Marc Synthetic excitatory amino acids
US7402595B2 (en) 2002-02-13 2008-07-22 Takeda Pharmaceutical Company Limited JNK inhibitor
GEP20063937B (en) 2002-02-14 2006-10-10 Pharmacia Corp Substituted pyridinones as modulators of p38 map kinase
AU2003205558A1 (en) 2002-02-19 2003-09-09 H. Lundbeck A/S Thioibotenic acid and derivatives thereof
US6833380B2 (en) 2002-03-07 2004-12-21 Warner-Lambert Company, Llc Compounds that modulate PPAR activity and methods of preparation
US20060046999A1 (en) 2002-03-14 2006-03-02 Cristina Alonso-Alija Monocyclic aroylpyridinones as antiinflammatory agents
WO2003082191A2 (en) 2002-03-28 2003-10-09 Merck & Co., Inc. Substituted 2,3-diphenyl pyridines
NZ535438A (en) 2002-03-29 2006-08-31 Janssen Pharmaceutica Nv Radiolabelled quinoline and quinolinone derivatives and their use as metabotropic glutamate receptor ligands
JP2005528378A (ja) 2002-04-03 2005-09-22 イーライ・リリー・アンド・カンパニー 非定型抗精神病薬およびmGLU2/3受容体アゴニストを組み合わせた精神病治療
US6864261B2 (en) 2002-05-02 2005-03-08 Euro-Celtique S.A. Therapeutic agents useful for treating pain
CA2483084A1 (en) 2002-05-02 2003-11-13 Merck & Co., Inc. Tyrosine kinase inhibitors
US7026326B2 (en) 2002-05-21 2006-04-11 Amgen Inc. Substituted heterocyclic compounds and methods of use
EA011231B1 (ru) 2002-06-11 2009-02-27 Эли Лилли Энд Компани Пролекарство возбуждающей аминокислоты и его применение
WO2003105846A1 (en) 2002-06-13 2003-12-24 Vertex Pharmaceuticals Incorporated 2-ureido-6-heteroaryl-3h-benzoimidazole-4-carboxylic acid derivatives and related compounds as gyrase and/or topoisomerase iv inhibitors for the treatment of bacterial infections
GB0214268D0 (en) 2002-06-20 2002-07-31 Celltech R&D Ltd Chemical compounds
MY141867A (en) 2002-06-20 2010-07-16 Vertex Pharma Substituted pyrimidines useful as protein kinase inhibitors
US20060063782A1 (en) 2002-07-03 2006-03-23 Murray Christopher W 3-Hetero arylmethoxy ! pyridines and their analogues as p38 map kinase inhibitors
US7262194B2 (en) 2002-07-26 2007-08-28 Euro-Celtique S.A. Therapeutic agents useful for treating pain
US20040138238A1 (en) 2002-08-08 2004-07-15 Dhanoa Dale S. Substituted aminopyrimidine compounds as neurokinin antagonists
GB0218630D0 (en) 2002-08-10 2002-09-18 Tanabe Seiyaku Co Novel compounds
GB0218800D0 (en) 2002-08-13 2002-09-18 Celltech R&D Ltd Chemical compounds
WO2004017950A2 (en) 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents
US20050288346A1 (en) 2002-08-26 2005-12-29 Cube Rowena V Acetophenone potentiators of metabotropic glutamate receptors
AU2003258305A1 (en) 2002-08-28 2004-03-19 Intermune, Inc. Combination therapy for treatment of fibrotic disorders
MY139563A (en) 2002-09-04 2009-10-30 Bristol Myers Squibb Co Heterocyclic aromatic compounds useful as growth hormone secretagogues
US8354447B2 (en) 2002-09-10 2013-01-15 The Scripps Research Institute mGLU receptors antagonists for treating disorders associated with mGLU receptors including addiction and depression
EP1546201A4 (en) 2002-09-11 2006-08-16 Merck & Co Inc NUCLEIC ACID SEQUENCES CODING TO MGLUR2 AND MGLUR3 FOR NEW POINT MUTATIONS
ES2268499T3 (es) 2002-09-19 2007-03-16 Boehringer Ingelheim (Canada) Ltd. Inhibidores no nucleosidos de transcriptasa inveresa.
AU2003269628A1 (en) 2002-09-19 2004-04-08 Boehringer Ingelheim (Canada) Ltd. Non-nucleoside reverse transcriptase inhibitors
TW200410975A (en) 2002-09-26 2004-07-01 Nihon Nohyaku Co Ltd New pesticide and method for using it, new substituted thienopyrimidine derivative, its intermediate, and method for producing it
US7067658B2 (en) 2002-09-30 2006-06-27 Bristol-Myers Squibb Company Pyridino and pyrimidino pyrazinones
US7998163B2 (en) 2002-10-03 2011-08-16 Boston Scientific Scimed, Inc. Expandable retrieval device
DE60330376D1 (de) 2002-10-23 2010-01-14 Daiichi Pure Chemicals Co Ltd Defructosylierungsverfahren
US20040138204A1 (en) 2002-10-30 2004-07-15 Harrington James Frederick Compositions and methods for pain reduction
US7902203B2 (en) 2002-11-01 2011-03-08 Abbott Laboratories, Inc. Anti-infective agents
MXPA05004670A (es) 2002-11-01 2005-08-18 Abbott Lab Agentes anti-infecciones.
US6930117B2 (en) 2002-11-09 2005-08-16 The Procter & Gamble Company N-alkyl-4-methyleneamino-3-hydroxy-2-pyridones
AU2003295776B2 (en) 2002-11-21 2011-05-12 Novartis Vaccines And Diagnostics, Inc. 2,4,6-trisubstituted pyrimidines as phosphotidylinositol (PI) 3-kinase inhibitors and their use in the treatment of cancer
WO2004054979A1 (ja) 2002-12-18 2004-07-01 Takeda Pharmaceutical Company Limited Jnk阻害剤
CN1802353A (zh) 2002-12-30 2006-07-12 细胞基因公司 氟烷氧基取代的1,3-二氢-异吲哚化合物及其药物用途
EP1599468B1 (en) 2003-01-14 2007-10-03 Arena Pharmaceuticals, Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
ITMI20030151A1 (it) 2003-01-30 2004-07-31 Recordati Ind Chimica E Farma Ceutica S P A Uso di antagonisti selettivi del recettore mglu5 per il trattamento di disfunzioni neuromuscolari del tratto urinario inferiore.
PL378405A1 (pl) 2003-02-04 2006-04-03 F. Hoffmann-La Roche Ag Pochodne malonoamidu jako inhibitory <$Egamma> - sekretazy
DE10306250A1 (de) 2003-02-14 2004-09-09 Aventis Pharma Deutschland Gmbh Substituierte N-Arylheterozyklen, Verfahren zu ihrer Herstellung und ihre Verwendung als Arzneimittel
US20070066590A1 (en) 2003-02-24 2007-03-22 Jones Robert M Phenyl-and pyridylpiperidine-derivatives as modulators of glucose metabolism
WO2004078176A1 (en) 2003-03-03 2004-09-16 F. Hoffmann-La Roche Ag 2,5- and 2,6-substituted tetrahydroisoquinolines for use as 5-ht6 modulators
ITFI20030058A1 (it) 2003-03-06 2004-09-07 Univ Firenze Formulazioni farmaceutiche contenenti tiazolidinedioni
DE10311065A1 (de) 2003-03-13 2004-09-23 Abbott Gmbh & Co. Kg Pyrimidin-2-on-Verbindungen und ihre therapeutische Verwendung
WO2004092123A2 (en) 2003-04-10 2004-10-28 Microbia, Inc. Inhibitors of fungal invasion
CA2527170A1 (en) 2003-04-15 2004-10-28 Astrazeneca Ab Therapeutic compounds
JP2004339080A (ja) 2003-05-13 2004-12-02 Tokyo Institute Of Technology ピラゾ−ル誘導体を含有する高血圧治療剤
BRPI0410348A (pt) 2003-05-14 2006-05-30 Torreypines Therapeutics Inc compostos e usos dos mesmos na modulação de amilóide-beta
JP2007504283A (ja) 2003-05-20 2007-03-01 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア β−アミロイド斑に作用物質を結合させる方法
GB0315950D0 (en) 2003-06-11 2003-08-13 Xention Discovery Ltd Compounds
EP1680125A1 (en) 2003-07-02 2006-07-19 Warner-Lambert Company LLC Combination of an allosteric inhibitor of matrix metalloproteinase-13 and a ligand to an alpha-2-delta receptor
TWI372050B (en) 2003-07-03 2012-09-11 Astex Therapeutics Ltd (morpholin-4-ylmethyl-1h-benzimidazol-2-yl)-1h-pyrazoles
WO2005007144A2 (en) 2003-07-14 2005-01-27 Decode Genetics Ehf Method of diagnosis and treatment for asthma based on haplotype association
BRPI0413861A (pt) 2003-08-29 2006-10-24 Vernalis Cambridge Ltd compostos de pirimidotiofeno
GB0320300D0 (en) 2003-08-29 2003-10-01 Cancer Rec Tech Ltd Pyrimidothiophene compounds
GB0322016D0 (en) 2003-09-19 2003-10-22 Merck Sharp & Dohme New compounds
WO2005061507A1 (en) 2003-12-16 2005-07-07 Pfizer Products Inc. Bicyclic pyrazol-4-one cannabinoid receptor ligands and uses thereof
AU2005214379B2 (en) 2004-02-18 2012-03-22 Astrazeneca Ab Tetrazole compounds and their use as metabotropic glutamate receptor antagonits
DE102004009039A1 (de) 2004-02-23 2005-09-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-Amino-piperidin-1-yl]-xanthine, deren Herstellung und Verwendung als Arzneimittel
US7306631B2 (en) 2004-03-30 2007-12-11 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
US20070219234A1 (en) 2004-04-12 2007-09-20 Kiyoshi Oizumi Thienopyridine Derivatives
US7459562B2 (en) 2004-04-23 2008-12-02 Bristol-Myers Squibb Company Monocyclic heterocycles as kinase inhibitors
GB0413605D0 (en) 2004-06-17 2004-07-21 Addex Pharmaceuticals Sa Novel compounds
US8063004B2 (en) 2004-07-22 2011-11-22 Malcera, L.L.C. Chemical composition of matter for the liquefaction and dissolution of asphaltene and paraffin sludges into petroleum crude oils and refined products at ambient temperatures and method of use
CA2574971A1 (en) 2004-07-30 2006-02-09 Merck & Co., Inc. Indanone potentiators of metabotropic glutamate receptors
CA2574956A1 (en) 2004-07-30 2006-02-09 Merck & Co., Inc. Heterocyclic acetophenone potentiators of metabotropic glutamate receptors
CA2568850A1 (en) 2004-08-02 2006-02-16 Schwarz Pharma Ag Indolizine carboxamides and the aza and diaza derivatives thereof
JP5276322B2 (ja) 2004-08-11 2013-08-28 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ 虚血性心疾患の超音波診断方法及び装置
TW200613272A (en) 2004-08-13 2006-05-01 Astrazeneca Ab Isoindolone compounds and their use as metabotropic glutamate receptor potentiators
EP1781655A2 (en) 2004-08-18 2007-05-09 Pharmacia & Upjohn Company LLC Triazolopyridine compounds useful for the treatment of inflammation
DE102004044884A1 (de) 2004-09-14 2006-05-24 Grünenthal GmbH Substituierte bizyklische Imidazo-3-yl-amin-Verbindungen
WO2006030847A1 (ja) 2004-09-17 2006-03-23 Dainippon Sumitomo Pharma Co., Ltd. 新規二環性ピラゾール誘導体
GB0420719D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
GB0420722D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
JP2008517920A (ja) 2004-10-25 2008-05-29 メルク エンド カムパニー インコーポレーテッド 代謝調節型グルタミン酸受容体の複素環式インダノン増強因子
DE102004054665A1 (de) 2004-11-12 2006-05-18 Bayer Cropscience Gmbh Substituierte bi- und tricyclische Pyrazol-Derivate Verfahren zur Herstellung und Verwendung als Herbizide und Pflanzenwachstumsregulatoren
US7718921B2 (en) 2004-11-17 2010-05-18 Metal Improvement Company Llc Active beam delivery system with variable optical path segment through air
PL1817301T3 (pl) 2004-11-22 2012-08-31 Lilly Co Eli Związki wzmacniające receptory glutaminianu
US7434262B2 (en) 2004-12-08 2008-10-07 At&T Intellectual Property I, L.P. Methods and systems that selectively resurrect blocked communications between devices
DE102004061288A1 (de) 2004-12-14 2006-06-29 Schering Ag 3-Amino-Pyrazolo[3,4b]pyridine als Inhibitoren von Proteintyrosinkinasen, deren Herstellung und Verwendung als Arzneimittel
CN101128435A (zh) 2004-12-27 2008-02-20 阿斯利康(瑞典)有限公司 作为代谢型谷氨酸受体激动剂用于治疗神经病学及精神病学病症的吡唑酮化合物
US7456289B2 (en) 2004-12-31 2008-11-25 National Health Research Institutes Anti-tumor compounds
JP2008536802A (ja) 2005-02-24 2008-09-11 メルク エンド カムパニー インコーポレーテッド 代謝調節型グルタミン酸受容体のベンズアゾール増強剤
KR100968989B1 (ko) 2005-03-23 2010-07-09 에프. 호프만-라 로슈 아게 mGluR2 길항제로서 아세틸렌일-피라졸로-피리미딘 유도체
AU2006234627C1 (en) 2005-04-08 2009-11-26 Eisai R & D Management Co., Ltd. Therapeutic agent for dyskinesia
US7572807B2 (en) 2005-06-09 2009-08-11 Bristol-Myers Squibb Company Heteroaryl 11-beta-hydroxysteroid dehydrogenase type I inhibitors
US7579360B2 (en) 2005-06-09 2009-08-25 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
JPWO2006137350A1 (ja) 2005-06-22 2009-01-15 キッセイ薬品工業株式会社 新規なフロピリジン誘導体、それを含有する医薬組成物およびそれらの用途
US20080318999A1 (en) 2005-08-05 2008-12-25 Methvin Isaac Tricyclic Benzimidazoles and Their Use as Metabotropic Glutamate Receptor Modulators
CN101309905A (zh) 2005-08-12 2008-11-19 阿斯利康(瑞典)有限公司 取代的异吲哚酮及其作为代谢型谷氨酸受体增效剂的用途
WO2007027669A1 (en) 2005-08-29 2007-03-08 Cps Biofuels, Inc. Improved biodiesel fuel, additives, and lubbricants
EP1948654B1 (en) 2005-09-17 2009-06-24 Novartis Ag 5-amino-4-hydroxy-7- (imidazo [1,2-a] pyridin-6- ylmethyl)-8-methyl-nonamide derivatives and related compounds as renin inhibitors for the treatment of hypertension
EP1764099A3 (en) 2005-09-17 2007-05-09 Speedel Experimenta AG Diaminoalcohol derivatives for the treatment of Alzheimer, malaria, HIV
WO2008051197A2 (en) 2005-09-20 2008-05-02 Mayo Foundation For Medical Education And Research Small-molecule botulinum toxin inhibitors
CN101273040B (zh) 2005-09-27 2011-11-09 弗·哈夫曼-拉罗切有限公司 作为mglur2拮抗剂的*二唑基吡唑并嘧啶类化合物
CA2755268C (en) 2005-11-15 2013-12-31 Array Biopharma, Inc. Erbb inhibitors
AR057218A1 (es) 2005-12-15 2007-11-21 Astra Ab Compuestos de oxazolidinona y su uso como pontenciadores del receptor metabotropico de glutamato
WO2007084314A2 (en) 2006-01-12 2007-07-26 Incyte Corporation MODULATORS OF 11-ß HYDROXYL STEROID DEHYDROGENASE TYPE 1, PHARMACEUTICAL COMPOSITIONS THEREOF, AND METHODS OF USING THE SAME
US7553836B2 (en) * 2006-02-06 2009-06-30 Bristol-Myers Squibb Company Melanin concentrating hormone receptor-1 antagonists
TW200804281A (en) 2006-02-16 2008-01-16 Astrazeneca Ab New metabotropic glutamate receptor-potentiating isoindolones
WO2007103760A2 (en) 2006-03-02 2007-09-13 Smithkline Beecham Corporation Thiazolones for use as pi3 kinase inhibitors
TWI417095B (zh) 2006-03-15 2013-12-01 Janssen Pharmaceuticals Inc 1,4-二取代之3-氰基-吡啶酮衍生物及其作為mGluR2-受體之正向異位性調節劑之用途
GB0606774D0 (en) 2006-04-03 2006-05-10 Novartis Ag Organic compounds
GB0608263D0 (en) 2006-04-26 2006-06-07 Glaxo Group Ltd Compounds
WO2007135527A2 (en) 2006-05-23 2007-11-29 Pfizer Products Inc. Benzimidazolyl compounds
WO2007135529A2 (en) 2006-05-23 2007-11-29 Pfizer Products Inc. Azabenzimidazolyl compounds as mglur2 potentiators
JP5228911B2 (ja) 2006-06-19 2013-07-03 東レ株式会社 多発性硬化症の治療又は予防剤
EP2049119A2 (en) 2006-06-29 2009-04-22 Astex Therapeutics Limited Pharmaceutical combinations of 1-cyclopropyl-3-[3-(5-morphoolin-4-ylmethyl-1h-benzoimidazol-2-yl)-1h-1-pyrazol-4-yl]-urea
WO2008006540A1 (en) 2006-07-12 2008-01-17 Syngenta Participations Ag Triazolopyridine derivatives as herbicides
US8198448B2 (en) 2006-07-14 2012-06-12 Amgen Inc. Fused heterocyclic derivatives and methods of use
PE20121506A1 (es) 2006-07-14 2012-11-26 Amgen Inc Compuestos triazolopiridinas como inhibidores de c-met
US8217177B2 (en) 2006-07-14 2012-07-10 Amgen Inc. Fused heterocyclic derivatives and methods of use
WO2008012622A2 (en) 2006-07-25 2008-01-31 Pfizer Products Inc. Azabenzimidazolyl compounds as potentiators of mglur2 subtype of glutamate receptor
WO2008012623A1 (en) 2006-07-25 2008-01-31 Pfizer Products Inc. Benzimidazolyl compounds as potentiators of mglur2 subtype of glutamate receptor
EP2061775A2 (en) 2006-09-13 2009-05-27 Astra Zeneca AB Spiro-oxazolidinone compounds and their use as metabotropic glutamate receptor potentiators
ATE542818T1 (de) 2006-10-11 2012-02-15 Amgen Inc Imidazo- und triazolopyridinverbindungen und verfahren zu deren anwendung
WO2008057855A2 (en) 2006-11-01 2008-05-15 Bristol-Myers Squibb Company Heterocyclic compounds as modulators of glucocorticoid receptor, ap-i, and/or np-kappa-b activity
TW200831085A (en) 2006-12-13 2008-08-01 Merck & Co Inc Non-nucleoside reverse transcriptase inhibitors
JP5442449B2 (ja) 2006-12-22 2014-03-12 アステックス、セラピューティックス、リミテッド 新規化合物
US8895745B2 (en) 2006-12-22 2014-11-25 Astex Therapeutics Limited Bicyclic heterocyclic compounds as FGFR inhibitors
EP2114940A1 (en) 2007-02-09 2009-11-11 AstraZeneca AB Aza-isoindolones and their use as metabotropic glutamate receptor potentiators - 613
ES2320955B1 (es) 2007-03-02 2010-03-16 Laboratorios Almirall S.A. Nuevos derivados de 3-((1,2,4)triazolo(4,3-a)piridin-7-il)benzamida.
TW200845978A (en) 2007-03-07 2008-12-01 Janssen Pharmaceutica Nv 3-cyano-4-(4-tetrahydropyran-phenyl)-pyridin-2-one derivatives
MX2009009520A (es) 2007-03-07 2009-11-18 Janssen Pharmaceutica Nv Fenoxiaminotiazolonas sustituidas como moduladores del receptor alfa relacionados con estrogenos.
MX2009009519A (es) 2007-03-07 2009-11-18 Janssen Pharmaceutica Nv Fenoxitiazolidinodionas n-alquiladas sustituidas como moduladores de receptor- alfa relacionados con estrogeno.
TW200900065A (en) 2007-03-07 2009-01-01 Janssen Pharmaceutica Nv 3-cyano-4-(4-pyridinyloxy-phenyl)-pyridin-2-one derivatives
TW200900391A (en) 2007-03-07 2009-01-01 Janssen Pharmaceutica Nv 3-cyano-4-(4-phenyl-piperidin-1-yl)-pyridin-2-one derivatives
CN101679322A (zh) 2007-03-07 2010-03-24 詹森药业有限公司 用作雌激素相关性受体-α调节剂的取代苯氧基噻唑烷二酮类化合物
GB0704407D0 (en) 2007-03-07 2007-04-18 Glaxo Group Ltd Compounds
KR101547573B1 (ko) 2007-03-09 2015-08-26 사노피 치환된 디하이드로 및 테트라하이드로 옥사졸로피리미디논, 이의 제조방법 및 용도
WO2008124085A2 (en) 2007-04-03 2008-10-16 Exelixis, Inc. Methods of using combinations of mek and jak-2 inhibitors
WO2008130853A1 (en) 2007-04-17 2008-10-30 Astrazeneca Ab Hydrazides and their use as metabotropic glutamate receptor potentiators - 681
PT2155747E (pt) 2007-05-10 2012-12-19 Ge Healthcare Ltd Imidazol(1,2-a)piridinas e compostos relacionados com actividade nos receptores de canabinóides cb2
EP2167464B1 (en) 2007-05-25 2014-12-03 AbbVie Deutschland GmbH & Co KG Heterocyclic compounds as positive modulators of metabotropic glutamate receptor 2 (mglu2 receptor)
TWI417100B (zh) 2007-06-07 2013-12-01 Astrazeneca Ab 二唑衍生物及其作為代謝型麩胺酸受體增效劑-842之用途
TW200911255A (en) 2007-06-07 2009-03-16 Astrazeneca Ab Metabotropic glutamate receptor oxadiazole ligands and their use as potentiators-841
WO2009004430A1 (en) 2007-06-29 2009-01-08 Pfizer Inc. N-benzyl oxazolidinones and related heterocycleic compounds as potentiators of glutamate receptors
US20090017943A1 (en) 2007-07-13 2009-01-15 Ken Landry Golf green reader
TWI445532B (zh) 2007-09-14 2014-07-21 Janssen Pharmaceuticals Inc 1’,3’-二取代-4-苯基-3,4,5,6-四氫-2h,1’h-〔1,4’〕聯吡啶基-2’-酮化物
JP5433579B2 (ja) 2007-09-14 2014-03-05 ジャンセン ファーマシューティカルズ, インコーポレイテッド. 1,3−二置換−4−フェニル−1h−ピリジン−2−オン
TW200922566A (en) 2007-09-14 2009-06-01 Ortho Mcneil Janssen Pharm 1,3 disubstituted 4-(aryl-X-phenyl)-1H-pyridin-2-ones
WO2009041567A1 (ja) 2007-09-27 2009-04-02 Banyu Pharmaceutical Co., Ltd. メラニン凝集ホルモン受容体拮抗作用を有するジアリールケチミン誘導体
US8119658B2 (en) 2007-10-01 2012-02-21 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
ES2637794T3 (es) 2007-11-14 2017-10-17 Janssen Pharmaceuticals, Inc. Derivados de imidazo[1,2-A]piridina y su uso como moduladores alostéricos positivos de receptores MGLUR2
WO2009094265A1 (en) 2008-01-24 2009-07-30 Merck & Co., Inc. 3,5-substituted-1,3-oxazolidin-2-one derivatives
EP2085390A1 (en) 2008-01-31 2009-08-05 Institut National De La Sante Et De La Recherche Medicale (Inserm) Labelled analogues of halobenzamides as multimodal radiopharmaceuticals and their precursors
US9446995B2 (en) 2012-05-21 2016-09-20 Illinois Institute Of Technology Synthesis of therapeutic and diagnostic drugs centered on regioselective and stereoselective ring opening of aziridinium ions
JP5309162B2 (ja) 2008-03-06 2013-10-09 サノフイ 置換されたジヒドロ、トリヒドロ及びテトラヒドロシクロアルキルオキサゾロピリミジノン、その製造及び使用
MX2010010525A (es) 2008-03-24 2010-10-25 Novartis Ag Inhibidores de metaloproteasa de matriz basados en aril-sulfonamida.
DE102008001056A1 (de) 2008-04-08 2009-10-15 Robert Bosch Gmbh Umlenkeinrichtung für einen Strahl einer elektromagnetischen Welle
WO2009130232A1 (en) 2008-04-24 2009-10-29 Glaxo Group Limited Pyrazolo [1, 5 -a] pyrazine derivatives as antagonists of v1b receptors
CA2723727A1 (en) 2008-05-15 2009-11-19 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
AU2009246629A1 (en) 2008-05-15 2009-11-19 Merck Sharp & Dohme Corp. Oxazolobenzimidazole derivatives
US7790760B2 (en) 2008-06-06 2010-09-07 Astrazeneca Ab Metabotropic glutamate receptor isoxazole ligands and their use as potentiators 286
TW201006801A (en) 2008-07-18 2010-02-16 Lilly Co Eli Imidazole carboxamides
UY32049A (es) 2008-08-14 2010-03-26 Takeda Pharmaceutical Inhibidores de cmet
WO2010022081A1 (en) 2008-08-19 2010-02-25 Array Biopharma Inc. Triazolopyridine compounds as pim kinase inhibitors
TWI496779B (zh) 2008-08-19 2015-08-21 Array Biopharma Inc 作為pim激酶抑制劑之三唑吡啶化合物
JPWO2010024258A1 (ja) 2008-08-29 2012-01-26 塩野義製薬株式会社 Pi3k阻害活性を有する縮環アゾール誘導体
CA2735764C (en) 2008-09-02 2016-06-14 Ortho-Mcneil-Janssen Pharmaceuticals, Inc. 3-azabicyclo[3.1.0]hexyl derivatives as modulators of metabotropic glutamate receptors
ES2466341T3 (es) 2008-10-16 2014-06-10 Janssen Pharmaceuticals, Inc. Derivados de indol y benzomorfolina como moduladores de receptores de glutamato metabotrópicos
WO2010060589A1 (en) 2008-11-28 2010-06-03 Ortho-Mcneil-Janssen Pharmaceuticals, Inc. Indole and benzoxazine derivatives as modulators of metabotropic glutamate receptors
AT507619B1 (de) 2008-12-05 2011-11-15 Oesterreichisches Forschungs Und Pruefzentrum Arsenal Ges M B H Verfahren zur approximation des zeitlichen verlaufs von verkehrsdaten
CA2744946A1 (en) 2009-02-04 2010-08-12 Boehringer Ingelheim International Gmbh Cyclic inhibitors of 11.beta.-hydroxysteroid dehydrogenase 1
WO2010114726A1 (en) 2009-03-31 2010-10-07 Merck Sharp & Dohme Corp. Aminobenzotriazole derivatives
EP2417135A1 (en) 2009-04-07 2012-02-15 Schering Corporation Substituted triazolopyridines and analogs thereof
MX2011011964A (es) 2009-05-12 2012-02-23 Janssen Pharmaceuticals Inc Derivados de 1,2,3-triazolo[4,3-a]piridina y su uso para el tratamiento o prevencion de trastornos neurologicos y psiquiatricos.
MY153913A (en) 2009-05-12 2015-04-15 Janssen Pharmaceuticals Inc 7-aryl-1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mglur2 receptors
SG176018A1 (en) 2009-05-12 2011-12-29 Janssen Pharmaceuticals Inc 1,2,4-triazolo[4,3-a]pyridine derivatives and their use as positive allosteric modulators of mglur2 receptors
WO2010141360A1 (en) 2009-06-05 2010-12-09 Merck Sharp & Dohme Corp. Biaryl benzotriazole derivatives
EP2470527A4 (en) 2009-08-20 2013-02-27 Merck Sharp & Dohme BENZOTRIAZOLE D ETHER DERIVATIVES
CN102002040A (zh) 2009-09-01 2011-04-06 上海药明康德新药开发有限公司 一种三唑并吡啶环化合物的合成方法
AR078173A1 (es) 2009-09-15 2011-10-19 Sanofi Aventis Bifeniloximetil dihidro oxazolopirimidinonas sustituidas, su preparacion y su uso
AR078171A1 (es) 2009-09-15 2011-10-19 Sanofi Aventis Dihidrobenzocicloalquiloximetil-oxazolopirimidinonas sustituidas, preparacion y uso de las mismas
AR078172A1 (es) 2009-09-15 2011-10-19 Sanofi Aventis Fenoximetil dihidro oxazolopirimidinonas sustituidas y uso de las mismas como moduladores de receptores metabotropicos de mglur
WO2011034741A1 (en) 2009-09-15 2011-03-24 Merck Sharp & Dohme Corp. Imidazopyridin-2-one derivatives
JP5204071B2 (ja) 2009-09-25 2013-06-05 パナソニック株式会社 電気かみそり
EP2496569A2 (en) 2009-11-02 2012-09-12 MSD Oss B.V. Heterocyclic derivatives
EP2542083B1 (en) 2010-03-04 2015-05-06 Merck Sharp & Dohme Corp. Positive allosteric modulators of mglur2
WO2011116356A2 (en) 2010-03-19 2011-09-22 Sanford-Burnham Medical Research Institute Positive allosteric modulators of group ii mglurs
US8664214B2 (en) 2010-03-30 2014-03-04 AbbVie Deutschland GmbH & Co. KG Small molecule potentiators of metabotropic glutamate receptors I
US8314120B2 (en) 2010-03-30 2012-11-20 Abbott Gmbh & Co. Kg Small molecule potentiators of metabotropic glutamate receptors
WO2011137046A1 (en) 2010-04-29 2011-11-03 Merck Sharp & Dohme Corp. Substituted 1,3-benzothiazol-2(3h)-ones and [1,3]thiazolo[5,4-b]pyridin-2(1h)-ones as positive allosteric modulators of mglur2
CN102869334A (zh) 2010-04-30 2013-01-09 荷兰联合利华有限公司 组合物
WO2011136723A1 (en) 2010-04-30 2011-11-03 Astrazeneca Ab Polymorphs of a metabotropic glutamate receptor positive allosteric modulator
US8765784B2 (en) 2010-06-09 2014-07-01 Merck Sharp & Dohme Corp. Positive allosteric modulators of MGLUR2
CN101893589B (zh) 2010-06-29 2012-10-17 中国人民解放军第三0二医院 一种无菌检查方法及其使用的全封闭集菌安瓿培养器
WO2012021382A1 (en) 2010-08-12 2012-02-16 Merck Sharp & Dohme Corp. Positive allosteric modulators of mglur2
WO2012035078A1 (en) 2010-09-16 2012-03-22 Novartis Ag 17α-HYDROXYLASE/C17,20-LYASE INHIBITORS
US8785481B2 (en) 2010-09-29 2014-07-22 Merck Sharp & Dohme Corp. Ether benzotriazole derivatives
EP2638040A1 (en) 2010-11-08 2013-09-18 Janssen Pharmaceuticals, Inc. RADIOLABELLED mGLuR2 PET LIGANDS
PL2649069T3 (pl) 2010-11-08 2016-01-29 Janssen Pharmaceuticals Inc Pochodne 1,2,4-triazolo[4,3-a]pirydyny i ich zastosowanie jako dodatnich allosterycznych modulatorów receptorów mGluR2
ES2536433T3 (es) 2010-11-08 2015-05-25 Janssen Pharmaceuticals, Inc. Derivados de 1,2,4-triazolo[4,3-a]piridina y su uso como moduladores alostéricos positivos de receptores mGluR2
JP5852666B2 (ja) 2010-11-08 2016-02-03 ジヤンセン・フアーマシユーチカルズ・インコーポレーテツド 1,2,4−トリアゾロ[4,3−a]ピリジン誘導体およびmGluR2受容体のポジティブアロステリックモジュレーターとしてのそれらの使用
JP2013545822A (ja) * 2010-12-17 2013-12-26 ヴァンダービルト ユニバーシティー mGluR5受容体のアロステリック調節剤としての、二環式トリアゾールラクタムおよびピラゾールラクタム
GB201106817D0 (en) 2011-04-21 2011-06-01 Astex Therapeutics Ltd New compound
EP2704573A4 (en) 2011-05-03 2014-10-15 Merck Sharp & Dohme AMINOMETHYL biaryl BENZOTRIAZOL DERIVATIVES
EP2705025A4 (en) 2011-05-03 2015-01-21 Merck Sharp & Dohme Hydroxymethyl-biaryl BENZOTRIAZOL DERIVATIVES
US8772276B2 (en) 2011-05-03 2014-07-08 Merck Sharp & Dohme Corp. Alkyne benzotriazole derivatives
AU2012284088B2 (en) 2011-07-19 2015-10-08 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
AR088218A1 (es) 2011-07-19 2014-05-21 Infinity Pharmaceuticals Inc Compuestos heterociclicos utiles como inhibidores de pi3k
KR20140088887A (ko) 2011-11-03 2014-07-11 머크 샤프 앤드 돔 코포레이션 mGluR2-음성 알로스테릭 조절제로서의 퀴놀린 카르복스아미드 및 퀴놀린 카르보니트릴 유도체, 조성물, 및 그의 용도
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US20130281397A1 (en) 2012-04-19 2013-10-24 Rvx Therapeutics Inc. Treatment of diseases by epigenetic regulation
JP6211797B2 (ja) 2012-05-14 2017-10-11 パナソニック株式会社 除臭剤を備えた除臭装置、および除臭装置を備えた冷蔵庫
EP2666775A1 (en) 2012-05-21 2013-11-27 Domain Therapeutics Substituted pyrazoloquinazolinones and pyrroloquinazolinones as allosteric modulators of group II metabotropic glutamate receptors
WO2013192350A1 (en) 2012-06-20 2013-12-27 Vanderbilt University Substituted bicyclic aralkyl pyrazole lactam analogs as allosteric modulators of mglur5 receptors
WO2013192347A1 (en) 2012-06-20 2013-12-27 Vanderbilt University Substituted bicyclic cycloalkyl pyrazole lactam analogs as allosteric modulators of mglur5 receptors
BR112014031896A2 (pt) 2012-06-20 2017-06-27 Univ Vanderbilt composto, composição farmacêutica, e, método para o tratamento de um distúrbio
AR093077A1 (es) 2012-10-23 2015-05-20 Hoffmann La Roche ANTAGONISTAS DE mGlu2/3 PARA EL TRATAMIENTO DE LOS TRASTORNOS AUTISTAS
US9191739B2 (en) 2013-03-25 2015-11-17 Bose Corporation Active reduction of harmonic noise from multiple rotating devices
JP6211798B2 (ja) 2013-05-17 2017-10-11 富士機械製造株式会社 基板上の異物検査方法および異物検査装置
JO3368B1 (ar) 2013-06-04 2019-03-13 Janssen Pharmaceutica Nv مركبات 6، 7- ثاني هيدرو بيرازولو [5،1-a] بيرازين- 4 (5 يد)- اون واستخدامها بصفة منظمات تفارغية سلبية لمستقبلات ميجلور 2

Also Published As

Publication number Publication date
US20190055257A1 (en) 2019-02-21
JO3368B1 (ar) 2019-03-13
CA2910507C (en) 2021-07-27
HK1216527A1 (zh) 2016-11-18
RS56128B1 (sr) 2017-10-31
KR102223050B1 (ko) 2021-03-04
TW201524981A (zh) 2015-07-01
BR112015029982A2 (pt) 2017-07-25
EP3004107B1 (en) 2017-05-10
PH12015502685A1 (en) 2016-03-07
NZ713458A (en) 2021-06-25
KR20160015247A (ko) 2016-02-12
CN107474050B (zh) 2019-09-10
US10106542B2 (en) 2018-10-23
ME02949B (me) 2018-07-20
CY1119499T1 (el) 2018-03-07
EA030426B1 (ru) 2018-08-31
CN105263935B (zh) 2018-02-27
ES2637295T3 (es) 2017-10-11
CL2015003499A1 (es) 2016-05-27
HRP20171051T1 (hr) 2017-10-06
EA201592252A1 (ru) 2016-04-29
TWI674264B (zh) 2019-10-11
JP2016524620A (ja) 2016-08-18
AU2014276963C1 (en) 2018-11-08
IL242868B (en) 2019-06-30
CN107474050A (zh) 2017-12-15
UA117016C2 (uk) 2018-06-11
US10584129B2 (en) 2020-03-10
TWI632144B (zh) 2018-08-11
PT3004107T (pt) 2017-08-16
PH12015502685B1 (en) 2016-03-07
SI3004107T1 (sl) 2017-09-29
WO2014195311A1 (en) 2014-12-11
DK3004107T3 (en) 2017-08-28
PL3004107T3 (pl) 2017-10-31
HK1246792A1 (zh) 2018-09-14
JP6357533B2 (ja) 2018-07-11
MY192029A (en) 2022-07-23
CN105263935A (zh) 2016-01-20
CA2910507A1 (en) 2014-12-11
IL242868A0 (en) 2016-02-01
MX2015016692A (es) 2016-04-04
MX368861B (es) 2019-10-18
US20160115169A1 (en) 2016-04-28
SG11201509938YA (en) 2016-01-28
TW201835084A (zh) 2018-10-01
AU2014276963A1 (en) 2015-11-12
LT3004107T (lt) 2017-08-25
EP3004107A1 (en) 2016-04-13
AU2014276963B2 (en) 2018-05-10

Similar Documents

Publication Publication Date Title
US10584129B2 (en) Substituted 6,7-dihydropyrazolo[1,5-a]pyrazines as negative allosteric modulators of mGluR2 receptors
EP3177623B1 (en) 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5h)-one compounds and their use as negative allosteric modulators of mglu2 receptors
EP3174882B1 (en) 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5h)-one compounds and their use as negative allosteric modulators of mglu2 receptors
AU2015295301B2 (en) 6,7-dihydropyrazolo[1,5-alpha]pyrazin-4(5H)-one compounds and their use as negative allosteric modulators of mGluR2 receptors
AU2015295299B2 (en) 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one compounds and their use as negative allosteric modulators of mGluR2 receptors
EP3174883B1 (en) 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5h)-one compounds and their use as negative allosteric modulators of mglu2 receptors
NZ713458B2 (en) 6,7-DIHYDROPYRAZOLO[1,5-a]PYRAZIN-4(5H)-ONE COMPOUNDS AND THEIR USE AS NEGATIVE ALLOSTERIC MODULATORS OF MGLUR2 RECEPTORS