Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2013/079460
Kind Code:
A1
Abstract:
This invention relates to compounds of formula (I) their use as positive allosteric modulators of mGlu5 receptor activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of neurological and psychiatric disorders associated with glutamate dysfunction such as schizophrenia or cognitive decline such as dementia or cognitive impairment. A, B, Ar, R1, R2, R3 have meanings given in the description.

Inventors:
GRAUERT MATTHIAS (DE)
BISCHOFF DANIEL (DE)
DAHMANN GEORG (DE)
KUELZER RAIMUND (DE)
RUDOLF KLAUS (DE)
Application Number:
PCT/EP2012/073668
Publication Date:
June 06, 2013
Filing Date:
November 27, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
International Classes:
C07D231/12; A61K31/415; A61K31/4439; A61K31/4709; A61K31/4725; A61K31/497; A61K31/501; A61K31/505; A61K31/52; A61K31/53; A61P25/00; C07D401/12; C07D401/14; C07D403/12; C07D473/34
Domestic Patent References:
WO2008145616A12008-12-04
WO2003105853A12003-12-24
WO2005056015A12005-06-23
Other References:
CRAIG W. LINDSLEY ET AL: "Discovery of Positive Allosteric Modulators for the Metabotropic Glutamate Receptor Subtype 5 from a Series of N -(1,3-Diphenyl-1 H - pyrazol-5-yl)benzamides That Potentiate Receptor Function in Vivo", JOURNAL OF MEDICINAL CHEMISTRY, vol. 47, no. 24, 1 November 2004 (2004-11-01), pages 5825 - 5828, XP055021138, ISSN: 0022-2623, DOI: 10.1021/jm049400d
RIEDEL ET AL., BEHAV. BRAIN RES., vol. 140, 2003, pages 1 - 47
FARBER ET AL., PROG. BRAIN RES., vol. 116, 1998, pages 421 - 437
COYLE ET AL., CELL. AND MOL NEUROBIOL., vol. 26, 2006, pages 365 - 384
JAVITT ET AL., AM J. PSYCHIATRY, vol. 148, 1991, pages 1301 - 1308
SCHOEPP ET AL., NEUROPHARMA, vol. 38, 1999, pages 1431 - 1476
LUJAN ET AL., EUR. J. NEUROSCI., vol. 8, 1996, pages 1488 - 1500
MANNAIONI ET AL., NEUROSCI., vol. 21, 2001, pages 5925 - 5924
ROSENBROCK ET AL., EUR. J. PHARMA., vol. 639, 2010, pages 40 - 46
CHAN ET AL., PSYCHOPHARMA, vol. 198, 2008, pages 141 - 148
O'BRIEN ET AL., MOL. PHARMA., vol. 64, 2003, pages 731 - 740
LINDSLEY ET AL., J. MED. CHEM, vol. 47, 2004, pages 5825 - 5828
BERGE, S.M. ET AL.: "Pharmaceutical salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
Attorney, Agent or Firm:
SIMON ET AL, Elke et al. (Binger Str. 173, Ingelheim Am Rhein, DE)
Download PDF:
Claims:
Claims

1. A compound of formula I

in which

A and B independently represent CH or N;

R1 represents C6-io-aryl, 5-14-membered heteroaryl, Ci_salkyl, C3_7cycloalkyl,

-0-Ci_salkyl which latter five groups are optionally substituted with one or more substituents selected from halogen, -CN, Ci_3alkyl, -0-Ci_3alkyl;

R2 and R3 independently represent -H, halogen,-CN, -COO-C i_4alkyl, Ci_5alkyl,

C3-5cycloalkyl, -0-Ci_5alkyl which latter four groups are optionally substituted with one or more fluorine atoms;

Ar represents

R4 and R5 independently represent -H, halogen, -OH, -CN, -NH2, Ci_5alkyl, phenyl,

-NH-Ci_5alkyl, -N(Ci_5alkyl)2, -0-Ci_5alkyl, -COO-Ci_5alkyl,

-CONH(Ci_5alkyl), -CON(Ci_5alkyl)2, -NHCONH-Ci_5alkyl,

-NHCON(Ci_5alkyl)2, -NHCO- Ci_5alkyl which latter eleven groups are optionally substituted with one or more substituents selected from halogen, -OH, morpholinyl-;

or a salt thereof, particularly a physiologically acceptable salt thereof.

2. A compound according to claim 1 , wherein

R1 represents phenyl, thienyl-, pyridinyl, Ci_4alkyl, C3-6cycloalkyl, -0-Ci_3alkyl which latter six groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, Ci_3alkyl, -0-Ci_3alkyl.

3. A compound according to any one of the preceding claims, wherein the group

represents

phenyl, 2-pyridyl which latter two groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, -CN, Ci_3alkyl, C3-5cycloalkyl, -0-Ci_3alkyl, -COO-Ci_4alkyl which latter four groups are optionally substituted with one or more fluorine atoms.

A compound according to any one of the preceding claims, wherein represents phenyl, 2-pyridinyl, 2-thienyl-, Ci_4alkyl, C3-6cycloalkyl, 0-Ci_3alkyl which latter six groups are optionally substituted with one or more substituents selected from chloro, fluoro, methyl, methoxy.

A compound according to any one of the preceding claims, wherein re resents phenyl,

which latter eleven groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, -CN, Ci_3alkyl, 0-Ci_3alkyl [which latter two groups are optionally substituted with one or more substituents selected from fluoro, -OH, 4-morpholinyl-].

6. A compound according to any one of the preceding claims, wherein

A represents N or CH;

B represents CH.

7. A compound according to any one of the preceding claims, wherein

R1 represents phenyl, methyl, ethyl, propyl, iso-propyl, cyclopropyl, cyclohexyl, methoxy,

A compound according to any one of the preceding claims, wherein

re resents

118

10. A compound according to any one of the preceding claims, namely a compound of formula I

represents N or CH;

represents CH; represents phenyl, methyl, ethyl, propyl, iso-propyl, cyclopropyl, cyclohexyl, methoxy,

Ar re resents

the group

or a salt thereof, particularly a physiologically acceptable salt thereof.



130

ı32









140

141

12. A compound according to any of the preceding claims for use as a medicament. 13. A pharmaceutical composition comprising at least one compound according to any of claims 1 to 11 or a pharmaceutically acceptable salt thereof in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.

14. A compound according to any one of claims 1 to 11 or a pharmaceutically acceptable salt thereof for use in the treatment of psychotic disorders including schizophrenia, schizoaffective disorder and substance induced psychotic disorder; cognitive disorders and dementias including age-associated learning and memory impairments or losses, post stroke dementia, deficits in concentration, mild cognitive impairment, the cognitive dysfunction in Alzheimers disease, and the cognitive dysfunction of schizophrenia.

Description:
Novel Compounds

Field of the invention

This invention relates to substituted pyrazoles and their use as positive allosteric modulators of mGlu5 receptor activity, pharmaceutical compositions containing the same, and methods of using the same as agents for treatment and/or prevention of neurological and psychiatric disorders associated with glutamate dysfunction such as schizophrenia or cognitive decline such as dementia or cognitive impairment.

Background of the invention

Glutamate is the primary excitatory amino acid in the mammalian central nervous system. Neurotransmisstion mediated by glutamate has been demonstrated to be critical in many physiological processes, such as synaptic plasticity, long term potentiation involved in both learning and memory as well as sensory perception (Riedel et al, Behav. Brain Res. 2003, 140: 1-47). Furthermore, it has been demonstrated that an imbalance of glutamate

neurotransmission plays a critical role in the pathophysiology of various neurological and psychiatric diseases.

The excitatory neurotransmission of glutamate is mediated through at least two different classes of receptors, the ionotropic glutamate receptors (NMD A, AMPA and kainate) and the metabo tropic glutamate receptors (mGluR). The ionotropic receptors are ligand gated ion channels and are thought to be responsible for the regulating rapid neuronal transmission between two neurons. The metabotropic glutamate receptors are G-protein coupled receptors (GPCRs) which appear to mediate not only synaptic transmission, but also to regulate the extent of neurotransmitter release as well as post synaptic receptor activation.

Disregulation in glutamatergic neurotransmission, for example through altered glutamate release or post-synaptic receptor activation, has been demonstrated in a variety of neurological ans well as psychiatric disorders. Hypo function of the NMDA receptor has not only been demonstrated in Alzheimer's patients, but is increasingly accepted as the putative cause of schizophrenia (Farber et al, Prog. Brain Res., 1998, 116: 421-437, Coyle et al, Cell, and Mol Neurobiol. 2006, 26: 365-384). This is supported by clinical studies showing that antagonists of the NMDA receptor induce symptoms indistinguishable to those suffered by schizophrenia patients (Javitt et al, Am J. Psychiatry, 1991, 148: 1301-1308). Therefore, approaches that could potentiate or normalize NMDA receptor signaling have the potential to treat neurological and psychiatric disorders. mGluR5 belongs to a superfamily of currently eight identified Type III GPCRs, which are unique in that the glutamate ligand binds to a large extraceluUar amino -terminal protein domain. This superfamily is further divided into three gropus (Group I, II and III) based on amino acid homology as well as the intracellular signalling cascades they regulate (Schoepp et al.,

Neuropharma, 1999, 38: 1431-1476). mGluR5 belongs to group I and is coupled to the phospho lipase C signalling cascade which regulates intracellular calcium mobilization .

In the CNS, mGluR5 has been demonstrared to be expressed mainly in the cortex, hippocampus, nucleus accumbens and the caudate-putamen. These brain regions are known to be involved in memory formation and cogntive function as well as emotional response. mGluR5 has been shown to be localized post-synaptically, adjacent to the post-synaptic density (Lujan et al., Eur. J. Neurosci. 1996, 8: 1488-1500). A functional interaction between mGluR5 and the NMDA receptor has also been demonstrated, where activation of mGluR5 potentiates the activation state of the NMDA receptor (Mannaioni et al, NeuroSci., 2001, 21 :5925-5924, Rosenbrock et al, Eur. J. Pharma., 2010, 639:40-46). Furthermore, activation of mGluR5 has been demonstrated in preclinical in vivo models to rescue cognitive impairment as well as psychotic disturbance induced by NMDA receptor antagonists (Chan et al., Psychopharma. 2008, 198: 141-148). Therefore, activation of mGluR5, and thereby potentiation or normalization of the NMDA receptor signaling, is a potential mechanism for the treatment of psychiatric and neurological disorders.

Most agonists of mGluR5 bind the orthosteric glutamate binding site. Since the glutamate binding site between the mGluR family members is highly conserved, it has been challenging to develop selective mGluR5 agonists which have acceptable CNS penetration and demonstrate in vivo activity. An alternative approach to achieve selectivity between the mGluR family members is to develop compounds which bind to an allosteric site, which is not as highly conserved between the family members. These allosteric binding compounds would not interfere with the natural glutamate binding and signaling, but modulate the receptor activation state.

Positive allosteric modulators of mGluR5 have recently been identified (O'Brien et al, Mol. Pharma. 2003, 64: 731-740, Lindsley et al, J. Med. Chem 2004, 47: 5825-5828). These compounds potentiate mGluR5 activity in the presence of bound glutamate. In the absence of bound glutamate, the mGluR5 positive modulators do not demonstrate intrinsic activity.

Therefore, these compounds potentiate the natural signaling of mGluR5 as opposed to agonists which activate the receptor in a permanent, unnatural manor. mGluR5 positive allosteric modulators therefore represent an approach to potentiate mGluR5 signaling which in turn potentiates and normalizes the NMDA receptor hypofunction detected in neurological and psychiatric disorders. WO 2003/105853 and WO 2005/056015 disclose substituted pyrazoles that are said to be CCRl receptor antagonists and to be useful for the treatment of inflammation and immune disorders. Quite surprisingly, according to the present invention, some selected pyrazole derivatives show positive modulatory activity on the mGluR5 receptor without having an inhibitory effect on the CCRl receptor. Such compounds are useful for the treatment of psychotic disorders, cognitive disorders and dementias.

DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to compounds of formula I:

independently represent CH or N;

represents aryl, heteroaryl, Ci_salkyl, C 3 _ 7 cycloalkyl, -0-Ci_salkyl which latter five groups are optionally substituted with one or more substituents selected from halogen, -CN, C 1-3 alkyl, -0-C 1-3 alkyl;

independently represent -H, halogen, -CN, -COO-Ci_ 4 alkyl, Ci_ 5 alkyl,

C 3 - 5 cycloalkyl, -0-Ci_ 5 alkyl which latter four groups are optionally substituted one or more fluorine atoms; Ar represents

R 4 and R 5 independently represent -H, halogen, -OH, -CN, -NH 2 , Ci_ 5 alkyl, phenyl,

-NH-Ci_ 5 alkyl, -N(Ci_ 5 alkyl) 2 , -0-Ci_ 5 alkyl, -COO-Ci_ 5 alkyl,

-CONH(Ci_ 5 alkyl), -CON(Ci_ 5 alkyl) 2 , -NHCONH-Ci_ 5 alkyl,

-NHCON(Ci_ 5 alkyl) 2 , -NHCO- Ci_ 5 alkyl which latter eleven groups are optionally substituted with one or more substituents selected from halogen,

-OH, morpholinyl-; or a salt thereof, particularly a physiologically acceptable salt thereof.

In second embodiment, in the general formula I, A, B, Ar, R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and represents phenyl, thienyl-, pyridinyl, Ci_ 4 alkyl, C3- 6 cycloalkyl, -0-Ci_ 3 alkyl which latter six groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, Ci_ 3 alkyl, -0-Ci_ 3 alkyl. In another embodiment, in the general formula I, A, B, R 1 , R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and

Ar represents phenyl, pyridinyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, isochinolinyl, purinyl which latter eight groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, -CN, Ci_ 3 alkyl, 0-Ci_ 3 alkyl [which latter two groups are optionally substituted with one or more substituents selected from fluoro, -OH, morpholinyl-]. In another embodiment, in the general formula I, Ar, R 1 have the same meaning as defined in any of the preceding embodiments, and the group

represents

phenyl, 2-pyridyl which latter two groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, -CN, Ci_ 3 alkyl, C3- 5 cycloalkyl, -0-Ci_ 3 alkyl, -COO-Ci_ 4 alkyl which latter four groups are optionally substituted with one or more fluorine atoms.

In another embodiment, in the general formula I, A, B, Ar, R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and

R 1 represents phenyl, 2-pyridinyl, 2-thienyl-, Ci_ 4 alkyl, C3- 6 cycloalkyl, 0-Ci_ 3 alkyl which latter six groups are optionally substituted with one or more substituents selected from chloro, fluoro, methyl, methoxy. In another embodiment, in the general formula I, A, B, R 1 , R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and

Ar represents phenyl,

which latter eleven groups are optionally substituted with one or more substituents selected from chloro, bromo, fluoro, -CN, Ci_3alkyl, 0-Ci_3alkyl [which latter two groups are optionally substituted with one or more substituents selected from fluoro, -OH,

4-morpholinyl-].

In another embodiment, in the general formula I, Ar, R 1 have the same meaning as defined in any of the preceding embodiments, and

A represents N or CH;

B represents CH.

In another embodiment, in the general formula I, A, B, Ar, R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and

R 1 represents phenyl, methyl, ethyl, propyl, iso-propyl, cyclopropyl, cyclohexyl, methoxy,

In another embodiment, in the general formula I, A, B, R 1 , R 2 , R 3 have the same meaning as defined in any of the preceding embodiments, and

Ar represents

In another embodiment, in the general formula I, Ar, R 1 have the same meaning as defined in any of the preceding embodiments, and the group

*

represents

A further embodiment of the present invention comprises compounds of formula I in which

A represents N or CH;

B represents CH;

R 1 represents phenyl, methyl, ethyl, propyl, iso-propyl, cyclopropyl, cyclohexyl, methoxy,

TERMS AND DEFINITIONS USED

General definitions: Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to.

In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example, Ci_6-alkyl means an alkyl group or radical having 1 to 6 carbon atoms. In general, for groups comprising two or more subgroups, the last named subgroup is the radical attachment point, for example, the substituent "aryl-Ci_3-alkyl-" means an aryl group which is bound to a Ci_3-alkyl-group, the latter of which is bound to the core or to the group to which the substituent is attached.

In case a compound of the present invention is depicted in form of a chemical name and as a formula in case of any discrepancy the formula shall prevail.

An asterisk is may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.

The numeration of the atoms of a substituent starts with the atom which is closest to the core or to the group to which the substituent is attached.

For example, the term "3-carboxypropyl-group" represents the following substituent:

wherein the carboxy group is attached to the third carbon atom of the propyl group. The terms "1-methylpropyl-", "2,2-dimethylpropyl-" or "cyclopropylmethyl-" group represent the following groups:

CH 3

The asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule as defined.

Stereochemistry/solvates/hydrates:

Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc...) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound. Salts:

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, and

commensurate with a reasonable benefit/risk ratio.

As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. For example, such salts include salts from ammonia, L-arginine, betaine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine (2,2'- iminobis(ethanol)), diethylamine, 2-(diethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-glucamine, hydrabamine, lH-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxy- ethyl)-morpholine, piperazine, potassium hydroxide, l-(2-hydroxyethyl)-pyrrolidine, sodium hydroxide, triethanolamine (2,2',2"-nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2.2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzene- sulfonic acid, benzoic acid, 2,5-dihydroxybenzoic acid, 4-acetamido-benzoic acid, (+)-camphoric acid, (+)-camphor-10-sulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, decanoic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy- ethanesulfonic acid, ethylenediaminetetraacetic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, D-glucoheptonic acid, D-gluconic acid, D-glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, hexanoic acid, hippuric acid, hydrobromic acid, hydrochloric acid, isobutyric acid, DL- lactic acid, lactobionic acid, lauric acid, lysine, maleic acid, (-)-L-malic acid, malonic acid, DL-mandelic acid, methane- sulfonic acid, galactaric acid, naphthalene- 1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1- hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, octanoic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid (embonic acid), phosphoric acid, propionic acid, (-)-L-pyroglu- tamic acid, salicylic acid, 4-amino-salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid. Further pharmaceutically acceptable salts can be formed with cations from metals like aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like, (also see Pharmaceutical salts, Berge, S.M. et al, J. Pharm. Sci., (1977), 66, 1-19). The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof.

Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention (e.g. trifluoro acetate salts,) also comprise a part of the invention.

Halogen:

The term halogen generally denotes fluorine, chlorine, bromine and iodine. Alkyl:

The term "Ci_ n -alkyl", wherein n is an integer from 2 to n, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n C atoms. For example the term Ci_5-alkyl embraces the radicals H 3 C-, H 3 C-CH 2 -, H 3 C-CH 2 - CH 2 -, H 3 C-CH(CH 3 )-, H 3 C-CH 2 -CH 2 -CH 2 -, H 3 C-CH 2 -CH(CH 3 )-, H 3 C-CH(CH 3 )-CH 2 -, H 3 C- C(CH 3 ) 2 -,

H 3 C-CH 2 -CH 2 -CH 2 -CH 2 -, H 3 C-CH 2 -CH 2 -CH(CH 3 )-, H 3 C-CH 2 -CH(CH 3 )-CH 2 -,

H 3 C-CH(CH 3 )-CH 2 -CH 2 -, H 3 C-CH 2 -C(CH 3 ) 2 -, H 3 C-C(CH 3 ) 2 -CH 2 -, H 3 C-CH(CH 3 )-CH(CH 3 )- and H 3 C-CH 2 -CH(CH 2 CH 3 )-. Alkylene:

The term "Ci_ n -alkylene" wherein n is an integer 2 to n, either alone or in combination with another radical, denotes an acyclic, straight or branched chain divalent alkyl radical containing from 1 to n carbon atoms. For example the term Ci_4-alkylene includes -CH 2 -,

-CH 2 -CH 2 -, -CH(CH 3 )-, -CH 2 -CH 2 -CH 2 -, -C(CH 3 ) 2 -, -CH(CH 2 CH 3 )-, -CH(CH 3 )-CH 2 -,

-CH 2 -CH(CH 3 )-, -CH 2 -CH 2 -CH 2 -CH 2 -, -CH 2 -CH 2 -CH(CH 3 )-, -CH(CH 3 )-CH 2 -CH 2 -,

-CH 2 -CH(CH 3 )-CH 2 -, -CH 2 -C(CH 3 ) 2 -, -C(CH 3 ) 2 -CH 2 -, -CH(CH 3 )-CH(CH 3 )-,

-CH 2 -CH(CH 2 CH 3 )-, -CH(CH 2 CH 3 )-CH 2 -, -CH(CH 2 CH 2 CH 3 )- , -CH(CH(CH 3 )) 2 - and

-C(CH 3 )(CH 2 CH 3 )-. Alkenyl:

The term "C 2 - n -alkenyl", is used for a group as defined in the definition for "Ci_ n -alkyl" with at least two carbon atoms, if at least two of those carbon atoms of said group are bonded to each other by a double bond.

Alkenylene:

The term "C 2 - n -alkenylene" is used for a group as defined in the definition for "Ci_ n -alkylene" with at least two carbon atoms, if at least two of those carbon atoms of said group are bonded to each other by a double bond.

Alkynyl:

The term "C 2 - n -alkynyl", is used for a group as defined in the definition for "Ci_ n -alkyl" with at least two carbon atoms, if at least two of those carbon atoms of said group are bonded to each other by a triple bond.

Alkynylene:

The term "C 2 - n -alkynylene" is used for a group as defined in the definition for "Ci_ n -alkylene" with at least two carbon atoms, if at least two of those carbon atoms of said group are bonded to each other by a triple bond.

Carbocyclyl:

The term "carbocyclyl" as used either alone or in combination with another radical, means a mono- bi- or tricyclic ring structure consisting of 3 to 14 carbon atoms. The term "carbocycle" refers to fully saturated and aromatic ring systems and partially saturated ring systems. The term "carbocycle" encompasses fused, bridged and spirocyclic systems.

Cycloalkyl:

The term "C3_ n -cycloalkyl", wherein n is an integer from 4 to n, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to n C atoms. For example the term C3_7-cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.

Cycloalkenyl:

The term "C3_ n -cycloalkenyl", wherein n is an integer 3 to n, either alone or in combination with another radical, denotes an cyclic, unsaturated but nonaromatic, unbranched hydrocarbon radical with 3 to n C atoms, at least two of which are bonded to each other by a double bond. For example the term C3-7-cycloalkenyl includes cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl cycloheptadienyl and cycloheptatrienyl.

Aryl:

The term "aryl" as used herein, either alone or in combination with another radical, denotes a carbocyclic aromatic monocyclic group containing 6 carbon atoms which may be further fused to a second 5- or 6-membered carbocyclic group which may be aromatic, saturated or unsaturated. Aryl includes, but is not limited to, phenyl, indanyl, indenyl, naphthyl, anthracenyl, phen- anthrenyl, tetrahydronaphthyl and dihydronaphthyl.

Heterocyclyl:

The term "heterocyclyl" means a saturated or unsaturated mono- or polycyclic-ring systems including aromatic ring system containing one or more heteroatoms selected from N, O or S(0) r , wherein r=0, 1 or 2, consisting of 3 to 14 ring atoms wherein none of the heteroatoms is part of the aromatic ring. The term "heterocycle" is intended to include all the possible isomeric forms. Heteroaryl:

The term "heteroaryl" means a mono- or polycyclic-ring systems containing one or more heteroatoms selected from N, O or S(0) r , wherein r=0, 1 or 2, consisting of 5 to 14 ring atoms wherein at least one of the heteroatoms is part of aromatic ring. The term "heteroaryl" is intended to include all the possible isomeric forms.

Thus, the term "heteroaryl" includes the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:

Many of the terms given above may be used repeatedly in the definition of a formula or group and in each case have one of the meanings given above, independently of one another.

GENERAL METHOD OF PREPARATION

Compounds of the present invention can be prepared in accordance with techniques that are well known to those skilled in the art.

Compounds of the present invention can be synthesized according to the following scheme: Schemel

separation of isomers and hydrolysis

hydrolysis and separation of isom Iers

Aryl ketones were deprotonated with with potassium tert butoxide and condensed with a methylesters to form a di-keton. Then, the di-keton was condensed with hydrazine to yield a pyrazole-system. The pyrazoles were coupled with 2-bromoacetic acid methyl ester under basic conditions to yield the desired pyrazol-l-yl-acetic acid methyl ester together with different quantities of the isomeric systhem. The pyrazol-l-yl-acetic acids methyl esters were hydro lyzed with LiOH to the coresponding acid. The isomeres were either seperated before ore after hydrolysis of the ester. Finally, the pyrazol-l-yl-acetic acides were coupled with aryl substituted piperazines to the desired products. Alternatively, the compounds of invention can be synthesized according to scheme2:

Scheme2

separation of isomers and hydrolysis

or

hydrolysis and separation of isomers

In this case the di-keton-systems were directly condensed with hydrazino-acetic acid ethyl ester to yield the two isomeric pyrazol-l-yl-acetic acid esters under acidic conditions. Then, the target compounds were obtained following the same synthesis strategy as for scheme 1.

As an additional altrenative, the compounds of invention can be synthesized according to scheme 3 :

Scheme3

The pyrazole-systems were formed following the same strategy as illustrated in scheme 1 or they are commerical available. However, in this case the aryl piperazines were first coupled with bromacetylbromide under basic conditions. Subsequently, the bromo-arylpiperazinyl-ethanon derivates were coupled with the pyrazoles to the desired products. Biological Assay

The positive modulation of mGluR5 is measured in a HEK 293 cell line expressing human recombinant mGluR5 and is detected with calcium based FLIPR assay. The cells are cultured with DMEM supplemented with 10% FCS, 2 μg/mL tetracycline, 100 μg/mL hygromycin and 500 μg/mL gneticin. The cell culture media is exchanged for tetracycline-free cell culture media 3-7 days before the assay. One day before the assay the cell culture medium is exchanged to DMEM without glutamine and phenol red and supplemented with 10% FCS, 100 μg/mL hygromycin and 500 μg/mL geneticin. On the assay day, the medium of the subconfluent cultures is removed and the cells are detached by addition of 2.5 ml EDTA(0.02%) per 175 cm2 culture flask for 1 minute. The cells are resuspend in Ringer solution (140 mM NaCl, 5 mM KC1, 2.5 mM CaC12, 1.5 mM MgC12, 5 mM Glucose, 10 mM Hepes; adjusted to pH 7.4 with NaOH), pooled and Ringer solution added to adjust the volume to 50 mL. The cell suspension is centrifuged for 5 min at 1500U/min (425 g). The supernatant is removed and the cells washed a second time with 50 ml fresh Ringer solution and centrifuged again as before. The supernatant is again removed and the pellet resuspended in Ringer solution to l,000,000cells/ml (1χ10 Λ 6 cells / mL). The cells are plated onto BD BioCoat Poly-D-Lysine 384 well plates (20.000 cells/well; 20 μΐ/well). The lid covered plates are then incubated until use at 37°C/10%> C02. For dye loading, 20 μΐ of Calcium-4 assay kit solution (prepared according to the manufacturer's description in Ringer solution) are added to the cells and the plates are incubated for 80 min 37°C and then 10 min at room temperature.

Controls, Compound dilution and assay execution:

Each assay plate contained wells with "high" and "low" controls:

Low controls l%DMSO/ringer solution + basal glutamate activation (defined as 100% CTL). High controls 10μΜ CDPPB + basal glutamate activation (defined as 200% CTL).

Test compounds are dissolved and diluted in DMSO to 100-fold the desired concentrations. In a second step, the compounds are diluted in Ringer solution such that the compounds are 4-fold more concentrated than the desired final assay concentration. The final DMSO concentration was 1%.

20μ1 of each compound solution are then transferred to the assay plate and the Ca2+ kinetic is measured to determine any intrinsic compound activity. After 5 min incubation in the FLIPR device, the second stimulation with 20 μΐ of glutamate in Ringer solution (glutamate concentration adjusted to approximately 5% basal stimulation of the maximal possible glutamate effect ) is added and the kinetic Ca2+ response of the wells was measured for the modulation effect.

Analysis:

The peak height of the Ca release related fluorescence signal (9-66) is used for the EC50.

The EC50 of the modulation is calculated over a nonlinear regression with GraphPad Prism (Table 1).

Table 1

EC50 EC50 EC50 EC50

Example Example Example Example

[nM] [nM] [nM] [nM]

7.02.07 853 7.01.54 205 7.01.99 129 7.01.143 20

7.02.08 934 7.01.55 501 7.01.100 126 7.01.144 57

7.02.09 298 7.01.56 393 7.01.101 17 7.01.145 216

7.02.10 948 7.01.57 81 7.01.102 20 7.01.146 25

7.02.11 91 7.01.58 211 7.01.103 24 7.01.147 19

7.02.12 1255 7.01.59 118 7.01.104 39 7.01.148 20

7.02.13 696 7.01.60 1228 7.02.21 77 7.01.149 12

7.02.14 129 7.01.61 246 7.02.22 85 7.01.150 5

7.03.01 31 7.01.62 48 7.02.23 60 7.01.151 19

7.01.13 14 7.01.63 94 7.02.24 340 7.01.152 26

7.01.14 22 7.01.64 677 7.02.25 92 7.01.153 276

7.01.15 30 7.01.65 253 7.01.105 4 7.01.154 11

7.01.16 32 7.01.66 957 7.01.106 35 7.01.155 24

7.04.01 696 7.04.03 392 7.01.107 64 7.01.156 33

7.04.02 81 7.01.67 61 7.01.108 1260 7.01.157 35

7.01.17 117 7.01.68 74 7.01.109 140 7.01.158 79

7.01.18 285 7.01.69 79 7.01.110 137 7.01.159 39

7.01.19 86 7.01.70 41 7.01.111 24 7.01.160 250

7.01.20 183 7.01.71 51 7.01.112 120 7.01.161 297

7.01.21 111 7.01.72 63 7.01.113 94 7.01.162 343

7.01.22 701 7.01.73 419 7.01.114 153 7.01.163 490

7.01.23 221 7.01.74 253 7.01.115 65 7.01.164 42

7.01.24 79 7.01.75 421 7.01.116 89 7.01.165 4

7.01.25 112 7.01.76 388 7.01.117 28 7.01.166 8

7.01.26 138 7.01.77 697 7.01.118 18 7.01.167 7 EC50 EC50 EC50 EC50

Example Example Example Example

[nM] [nM] [nM] [nM]

7.01.27 64 7.01.78 723 7.01.119 19 7.01.168 12

7.01.28 64 7.01.79 26 7.01.120 20 7.01.169 11

7.01.29 158 7.01.80 45 7.01.121 34 7.01.170 22

7.01.30 987 7.01.81 13 7.01.122 51 7.01.171 9

7.01.31 208 7.01.82 33 7.01.123 36 7.01.172 152

7.01.32 75 7.01.83 52 7.01.124 8 7.01.173 11

7.01.33 161 7.01.84 67 7.01.125 29 7.02.27 243

7.01.34 60 7.02.15 153 7.01.126 19 7.01.189 48

7.01.35 213 7.02.16 237 7.01.127 34 7.01.190 47

7.01.174 3 7.01.179 46 7.01.184 145 7.01.191 7

7.01.175 10 7.01.180 64 7.01.185 442 7.01.192 15

7.01.176 10 7.01.181 130 7.01.186 441

7.01.177 24 7.01.182 434 7.01.187 360

7.01.178 21 7.01.183 318 7.01.188 27

METHOD OF TREATMENT

The present invention is directed to compounds of general formula I which are useful in the treatment of a disease and/or condition wherein the activity of an mGluR5 positive modulator is of therapeutic benefit, including but not limited to the treatment of psychotic disorders, cognitive disorders and dementias.

The compounds of general formula I are useful for the treatment of psychotic disorders including schizophrenia, schizoaffective disorder and substance induced psychotic disorder; cognitive disorders and dementias including age-associated learning and memory impairments or losses, post stroke dementia, deficits in concentration, mild cognitive impairment, the cognitive dysfunction in Alzheimers disease, and the cognitive dysfunction of schizophrenia.

Therefore, the present invention also relates to a compound of general formula I as a

medicament. A further aspect of the present invention relates to the use of a compound of general formula I for the treatment of a disease and/or condition wherein the activity of mGluR5 positive modulator is of therapeutic benefit.

Furthermore, the present invention relates to the use of a compound of general formula I for the treatment of psychotic disorders, cognitive disorders and dementias.

Furthermore, the present invention relates to the use of a compound of general formula I for the treatment of psychotic disorders including schizophrenia, schizoaffective disorder and substance induced psychotic disorder; cognitive disorders and dementias including age-associated learning and memory impairments or losses, post stroke dementia, deficits in concentration, mild cognitive impairment, the cognitive dysfunction in Alzheimers disease, and the cognitive dysfunction of schizophrenia.

In a further aspect of the present invention the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of general formula I to a human being.

DOSAGE

The dose range of the compounds of general formula I applicable per day is usually from 0.1 to 5000 mg, preferably from 0.1 to 1000 mg, more preferably from 5 to 500 mg, most preferably, 10 or 100 mg. Each dosage unit may conveniently contain from 0.1 to 500 mg, preferably 10 to 100 mg.

The actual pharmaceutically effective amount or therapeutic dosage will of course depend on factors known by those skilled in the art such as age and weight of the patient, route of administration and severity of disease. In any case the combination will be administered at dosages and in a manner which allows a pharmaceutically effective amount to be delivered based upon patient's unique condition.

Pharmaceutical Compositions

Suitable preparations for administering the compounds of formula will be apparent to those with ordinary skill in the art and include for example tablets, pills, capsules, suppositories, lozenges, troches, solutions, syrups, elixirs, sachets, injectables, inhalatives and powders etc. The content of the pharmaceutically active compound(s) should be in the range from 1 to 99 wt.-%, preferably 10 to 90 wt.-%, more preferably 20 to 70 wt.-%, of the composition as a whole. Suitable tablets may be obtained, for example, by mixing one or more compounds according to formula I with known excipients, for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants . The tablets may also consist of several layers.

A further aspect of the invention is a pharmaceutical formulation including a compound of formula I in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier.

COMBINATION THERAPY In another aspect the present invention relates to a combination therapy in which an active compound according to the present invention is administered together with another active compound. Accordingly, the invention also refers to pharmaceutical formulations that provide such a combination of active ingredients, whereby one of which is an active compound of the present invention. Such combinations may be fixed dose combinations (the active ingredients that are to be combined are subject of the same pharmaceutical formulation) or free dose combinations (active ingredients are in separate pharmaceutical formulations).

Consequently, a further aspect of the present invention refers to a combination of each of the active compounds of the present invention, preferably at least one active compound according to the present invention, with another active compound for example selected from the group of antipsychotics such as haloperidol, clozapine, risperidone, quetiapine, aripripazole, and olanzapine; antidepressants such as selective serotonin re-uptake inhibitors and dual

serotonin/noradrenaline re-uptake inhibitors; mood stabilizers such as lithium valproate and lamotrigine; beta-secretase inhibitors; gamma-secretase inhibitors; gamma-secretase modulators; amyloid aggregation inhibitors such as e.g. scyllo-inositol; directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants, such as e.g. vitamin E , ginko biloba or ginkolide; anti-inflammatory substances, such as e.g. Cox inhibitors, NSAIDs additionally or exclusively having AB (Abeta) lowering properties; HMG-CoA reductase inhibitors, such as statins; acetylcholine esterase inhibitors, such as donepezil, rivastigmine, tacrine, galantamine; NMDA receptor antagonists such as e.g. memantine; AMPA receptor agonists; AMPA receptor positive modulators, AMPkines, glycine transporter 1 inhibitors;

monoamine receptor reuptake inhibitors; substances modulating the concentration or release of neurotransmitters; substances inducing the secretion of growth hormone such as ibutamoren mesylate and capromorelin; CB-1 receptor antagonists or inverse agonists; antibiotics such as minocyclin or rifampicin; PDE1, PDE2, PDE4, PDE5, PDE9 or PDE10 inhibitors, GABAA receptor inverse agonists; GABAA alpha5 receptor inverse agonists; GABAA receptor antagonists; nicotinic receptor agonists or partial agonists or positive modulators; alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators; alpha7 nicotinic receptor agonists or partial agonists; histamine receptor H3 antagonists; 5-HT4 receptor agonists or partial agonists; 5-HT6 receptor antagonists; alpha2-adrenoreceptor antagonists, calcium antagonists; muscarinic receptor Ml agonists or partial agonists or positive modulators;

muscarinic receptor M2 antagonists; muscarinic receptor M4 antagonists; muscarinic receptor M4 positive allosteric modulators; metabotropic glutamate receptor 5 positive allosteric modulators; metabotropic glutamate receptor 2 antagonists; metabotropic glutamate receptor 2/3 agonists; metabotropic glutamate receptor 2 positive allosteric modulators and other substances that modulate receptors or enzymes in a manner such that the efficacy and/or safety of the active compounds according to the invention is increased and/or unwanted side effects are reduced. The active compounds according to the invention may also be used in combination with immunotherapies such as e.g. active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies, nanobodies or antibody fragments for the treatment of the above mentioned diseases and conditions.

The active compounds according to the invention also may be combined with antipsychotics like haloperidol, flupentixol, fluspirilene, chlorprothixene, prothipendyl, levomepromazine, clozapine, olanzapine, quetiapine, risperidone, paliperidone, amisulpride, ziprasidone, aripiprazol, sulpiride, zotepine, sertindole, fluphenazine, perphenazine, perazine, promazine, chlorpromazine, levomepromazine, benperidol, bromperidol, pimozid, melperone, pipamperone, iloperidone, asenapine, perospirone, blonanserin, lurasidone.

The active compounds according to the invention also may be combined with antidepressants like amitriptyline imipramine hydrochloride, imipramine maleate, lofepramine, desipramine, doxepin, trimipramine.

Or the active compounds according to the invention also may be combined with serotonin (5- HT) reuptake inhibitors such as alaproclate, citalopram, escitalopram, clomipramine, duloxetine, femoxetine, fenfluramine, norfenfluramine, fluoxetine, fluvoxamine, indalpine, milnacipran, paroxetine, sertraline, trazodone, venlafaxine, zimelidine, bicifadine, desvenlafaxine, braso- fensme and tesofensine. The combinations according to the present invention may be provided simultaneously in one and the same dosage form, i.e. in form of a combination preparation, for example the two

components may be incorporated in one tablet, e. g. in different layers of said tablet. The combination may be also provided separately, in form of a free combination, i.e. the active compounds of the present invention are provided in one dosage form and one or more of the above mentioned combination partners is provided in another dosage form. These two dosage forms may be equal dosage forms, for example a co-administration of two tablets, one containing a therapeutically effective amount of the active compound of the present invention and one containing a therapeutically effective amount of the above mentioned combination partner. It is also possible to combine different administration forms, if desired. Any type of suitable administration forms may be provided.

The active compound according to the invention, or a physiologically acceptable salt thereof, in combination with another active substance may be used simultaneously or at staggered times, but particularly close together in time. If administered simultaneously, the two active substances are given to the patient together; if administered at staggered times the two active substances are given to the patient successively within a period of less than or equal to 12, particularly less than or equal to 6 hours.

The dosage or administration forms are not limited, in the frame of the present invention any suitable dosage form may be used. Exemplarily the dosage forms may be selected from solid preparations such as patches, tablets, capsules, pills, pellets, dragees, powders, troches, suppositories, liquid preparations such as solutions, suspensions, emulsions, drops, syrups, elixirs, or gaseous preparations such as aerosols, sprays and the like.

The dosage forms are advantageously formulated in dosage units, each dosage unit being adapted to supply a single dose of each active component being present. Depending from the administration route and dosage form the ingredients are selected accordingly.

The dosage for the above-mentioned combination partners may be expediently 1/5 of the normally recommended lowest dose up to 1/1 of the normally recommended dose.

The dosage forms are administered to the patient for example 1, 2, 3, or 4 times daily depending on the nature of the formulation. In case of retarding or extended release formulations or other pharmaceutical formulations, the same may be applied differently (e.g. once weekly or monthly etc.). It is preferred that the active compounds of the invention be administered either three or fewer times, more preferably once or twice daily. EXPERIMENTAL SECTION

Preparation of examples for compounds of the general formula I Unless otherwise stated, one or more tautomeric forms of compounds of the examples described hereinafter may be prepared in situ and/or isolated. All tautomeric forms of compounds of the examples described hereinafter should be considered to be disclosed.

The invention is illustrated by way of the following examples, in which the following abbreviations may be employed:

Abbreviations

RT room temperature

THF tetrahydrofuran

KotBu kalium tert butoxide

PFTU pentafluorphenol-tetramethyluronium hexafluorophosphat

ACN acetonitrile

MeOH methanol

DIPEA diisopropylamine

DEA diethylamine

EtOAC ethyl acetate

DMF dimethylformamide

TBTU [(Benzotriazol- 1 -yloxy)-dimethylamino-methylene]-dimethyl-ammonium

tetrafluoro borate

HATU (0-(7-Azobenzotriazol- 1 -yl)- 1 , 1 ,3,3-tetramethyluroniumhexafluorophosphate cone. concentrated

min. minutes

DCM dichlormethane Analytical methods

All compounds specified in the examples below gave the correct mass spectra matching the theoretical isotope pattern. For practical reasons, only one of the major isotope peaks is given as representative data for the mass spectrum.

List of HPLC purification methods: method 1:

Gilson HPLC

eluent:

A: water with 0.10% TFA

B: methanol

gradient

time in min %A %B flow in ml/min

00.00 90 10 50

02.00 90 10 50

11.00 0 100 50

14.00 0 100 50

column: Sunfire CI 8, 30 x 100 mm, 10 μιη (temperature: isocratic 60°C). method 2:

column: Daicel OJH, 250mmx4.6mm,

flow: 4ml/min,

run time: lOmin,

mobile phases: C0 2 , 20% Methanol with DEA

The HPLC/MS data, where specified, are obtained under the following conditions List of analytical HPLC-methods: method A:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient

time in min %A %B flow in ml/min

0.00 99 1 1.20

0.05 99 1 1.20

1.05 0 100 1.20

1.25 0 100 1.20

column: Sunfire C18, 2.1 x 30 mm, 2,5 μιη (temperature: isocratic 60°C).

diodenarray detektion: 210-400 nm. method B:

Waters ZQ MS, Alliance 2690/2695 HPLC, Waters 996/2996 diodenarraydetector eluent:

A: water with 0.10% TFA

D: methanol

gradient:

time in min %A %D flow in ml/min

0.00 95 5 4.00

0.20 95 5 4.00

1.60 0 100 4.00

2.10 0 100 4.00

column: Waters XBridge J M CI 8 3.5μιη, 4.6x20mm IS (temperature: isocratic 40°C). diodenarray detection: 210-400 nm. method C:

Waters ZMD, Alliance 2690/2695 HPLC, Waters 996/2996 diodenarraydetector

eluent:

A: water with 0.10% TFA

B: acetonitril with 0.10% TFA

gradient:

time in min %A %B flow in ml min

0.00 95 5 2.80

0.30 95 5 2.80

1.60 2 98 2.80

1.90 2 98 2.80

2.00 95 5 2.50

column: Merck Chromolith Flash RP-18e, 3 mm x 100 mm (temperature: isocratic 25°C) dioden array detection: 210-400 nm. method D:

Waters Acquity with diodenarraydetector

eluent:

A: water with 0.13% TFA

B: methanol with 0.05% TFA

gradient:

time in min %A %>B flow in ml min

0.00 99 1 1.20

0.05 99 1 1.20

1.05 0 100 1.20

1.25 0 100 1.20

column: Sunfire CI 8, 2.1 x 30 mm, 2,5 μηι (temperature: isocratic 60°C). method E:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.90

1.60 0 100 4.90

2.20 95 5 4.90

column: Sunfire CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method F:

Agilent 1200 mit DA- und MS-Detektor

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 1.80

0.20 95 5 1.80

1.70 0 100 1.80

1.75 0 100 2.50

2.20 0 100 2.50

column: Sunfire CI 8, 3 x 30 mm, 2.5 μιη (temperature: isocratic 60°C). method G:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol with 0.10% TFA

gradient:

time in min %A % 0 B flow in ml/min 0.00 95 5 4.00

0.20 95 5 4.00

1.50 0 100 4.00

1.75 0 100 4.00

1.85 95 5 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method H:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.90

1.50 0 100 4.90

2.20 95 5 4.90

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method I:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient

time in min %A %>B flow in ml/min

0.00 99 1 1.50

0.05 99 1 1.50

1.05 0 100 1.50

1.20 0 100 1.50

column: Xbridge CI 8, 2.1 x 30 mm, 1.7 μηι (temperature: isocratic 60°C). method J:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.13% TFA

B: methanol with 0.08% TFA

gradient:

time in min %A %>B flow in ml/min

0.00 99 1 1.30

0.05 99 1 1.30

0.35 0 100 1.30

0.50 0 100 1.30

column: Xbridge CI 8, 2.1 x 30 mm, 1.7 μιη (temperature: isocratic 60°C). method K:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% NH3

B: methanol with 0.10% NH3

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.20 95 5 4.00

1.50 0 100 4.00

1.75 0 100 4.00

column: XBridge C18, 4.6 x 30 mm, 3.5 μηι (temperature: isocratic 60°C). method L:

Waters ZQ 2000MS, Agilent HP 100, binare pumps

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min % 0 A %>B flow in ml/min

0.00 95 5 1.50 1.30 0 100 1.50

2.50 0 100 1.50

2.60 95 5 1.50

column: Sunfire CI 8, 4.6 x 50 mm, 3.5 μιη (temperature: isocratic 40°C). diodenarray detection: 210-400 nm. method M:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 99 1 1.30

0.15 99 1 1.30

1.10 0 100 1.30

1.25 0 100 1.30

column: Sunfire CI 8, 2.1 x 30 mm, 2,5 μιη (temperature: isocratic 60°C). diodenarray detektion: 210-400 nm. method N:

Agilent 1100 System with diodenarraydetector and massdetector

eluent:

A: water with 0.10% ammonia

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.15 95 5 4.00

1.70 0 100 4.00

2.10 0 100 4.00

column: Stable Bond CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method O:

Agilent 1100 System with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B : methano 1 with 0.10% TFA

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.15 95 5 4.00

1.70 0 100 4.00

2.25 0 100 4.00

column: Sunfire CI 8, 4.6 x 30 mm, 3.5 μηι (temperature: isocratic 60°C). method P:

Agilent 1200 mit DA- und MS-Detektor

eluent:

A: water with 0.20% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 2.20

0.05 95 5 2.20

1.40 0 100 2.20

1.80 0 100 2.20

column: Stable Bond C18, 3 x 30 mm, 1.8 μιη (temperature: isocratic 60°C). method Q:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient

time in min %A %B flow in ml/min

0.00 99 1 1.40 0.05 99 1 1.40

1.00 0 100 1.40

1.10 0 100 1.40

column: Xbridge CI 8, 2.1 x 20 mm, 2.5 μιη (temperature: isocratic 60°C). method R:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.80

1.60 0 100 4.80

1.85 0 100 4.80

1.90 95 5 4.80

column: XBridge C18, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method S.Agilent 1200 mit DA- und MS-Detektor

eluent:

A: water with 0.20% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 2.20

0.05 95 5 2.20

1.40 0 100 2.20

1.80 0 100 2.20

column: Sunfire C18, 3 x 30 mm, 2.5 μιη (temperature: isocratic 60°C). method T:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

1.60 0 100 4.00

1.85 0 100 4.00

1.90 95 5 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method U:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.13% TFA

B: methanol 0.05% TFA

gradient

time in min %A %B flow in ml/min

0.00 99 1 1.30

0.05 99 1 1.30

1.05 0 100 1.30

1.20 0 100 1.30

column: Xbridge BEH C 18, 2.1 x 30 mm, 1.7 μιη (temperature: isocratic 60°C method V:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00 1.60 0 100 4.00

1.85 0 100 4.00

1.90 95 5 4.00

column: SunFire CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method W:

Waters ZQ2000 with diodenarray detector and massdetector

eluent:

A: water with 0.15% formic acid

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 1.50

2.00 0 100 1.50

column: Xbridge C 18, 2.1 x 50 mm, 3.5 μιη (temperature: isocratic 40°C). method X:

Agilent 1200 mit DA- und MS-Detektor

eluent:

A: water with 0.10% ammonia

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 2.20

0.20 95 5 2.20

1.50 0 100 2.20

1.55 0 100 2.90

1.70 0 100 2.90

column: Xbridge C18, 3 x 30 mm, 2.5 μιη (temperature: isocratic 60°C). method Y:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% NH3

B : methano 1 with 0.10% NH3

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.20 95 5 4.00

1.50 0 100 4.00

1.75 0 100 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method Z:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.1% TFA

B: methanol

gradient

time in min %A %>B flow in ml/min

0.00 99 1 1.50

0.05 99 1 1.50

1.05 0 100 1.50

1.20 0 100 1.50

column: Xbridge BEH Phenyl, 2.1 x 30 mm, 1.7 μιη (temperature: isocratic 60°C) method AA:

Waters Acquity with diodenarraydetector and massdetector

eluent:

A: water with 0.1% TFA

B: methanol

gradient

time in min %A %B flow in ml/min

0.00 99 1 1.50 0.05 99 1 1.50

1.00 0 100 1.50

1.10 0 100 1.50

column: Xbridge Phenyl, 2.1 x 20 mm, 2.5 μιη (temperature: isocratic 60°C)

method AB:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

1.60 0 100 4.00

1.85 0 100 4.00

1.90 95 5 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method AC:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% TFA

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.20 95 5 4.00

1.50 0 100 4.00

1.90 0 100 4.00

2.20 95 5 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method AD:

Waters Alliance with diodenarraydetector and massdetector

eluent:

A: water with 0.10% NH3

B: methanol

gradient:

time in min %A %B flow in ml/min

0.00 95 5 4.00

0.20 95 5 4.00

1.50 0 100 4.00

1.75 0 100 4.00

column: XBridge CI 8, 4.6 x 30 mm, 3.5 μιη (temperature: isocratic 60°C). method AE:

Waters SQD MS, Acquity UPLC with diodenarraydetector

eluent:

A: water with 0.10% TFA

B: acetonitrile 0.08% TFA

gradient:

time in min %A %B flow in ml/min

0.00 95 5 1.50

0.70 0 100 1.50

0.80 0 100 1.50

0.81 95 5 1.50

column: Ascentis Express C 18, 2.1 x 50 mm, 2.7 μπι (temperature: isocratic 60°C).

Synthesis of intermediates

6.01. Synthesis of building blocks -methoxy-6-piperazin- 1 -yl-pyrimidine hydrochloride

6.01.01.1 4-chloro-6-methoxy-pyrimidine 5.8 g sodium methanolate was added to 10 g 4, 6-dichloropyrimidine in 150 mL MeOH at 0°C. The reaction was warmed up to RT and stirred over night at RT. Then 3.63 g sodium

methanolate was added and the reaction was stirred for 3 h. The mixture was treated with water and extracted with EtOAc. The organic layer was dried over magesiumsulfate and evaporated to give 9.7 g 4-chloro-6-methoxy-pyrimidine.

R t : 0.87 min (method B)

(M+H) + : 145

6.01.01.2 4-(6-methoxy-pyrimidin-4-yl)-piperazine- 1 -carboxylic acid tert-butyl ester 18.6 mL triethylamine and piperazine-1 -carboxylic acid tert-butyl ester were added to 9.7 g 4- chloro-6-methoxy-pyrimidine in 50 mL DMF. The reaction was sirred over night at 60°C. The reaction was cooled and the precipitate was filltered. The filtrate was evaporated. The residue was dissolved in DCM, extracted with water and washed with sodium chloride solution. The solvent was evaporated and the residue was crystallized with a mixture of petrolether and diethylether (8:2) to yield 8.7 g of the desired compound.

R t : 1.12min (method B)

(M+H) + : 195 6.01.01.3 4-methoxy-6-piperazin- 1 -yl-pyrimidine hydrochloride

8.61 g 4-(6-Methoxy-pyrimidin-4-yl)-piperazine-l-carboxylic acid tert-butyl ester were stirred in 4 mol/L HCl solution in dioxane for lh. The the precipitates was filtered, washed with dioxane and dried to yield 5.17 g of the desired compound.

R f : 0.75 (flash chromatography: silica gel, cyclohexane: EtOAc/NH3 (3: 1))

(M+H) + : 295

By using the same synthesis strategy as for 4-methoxy-6-piperazin-l -yl-pyrimidine

hydrochloride the following compounds were obtained:

6.01.04 1 -P ridin-3 -yl-piperazine

6.01.04.1 l-Benzyl-4-pyridin-3 -yl-piperazine

1.8 M phenyllithium solution in cyclohexane/diethylether (7/3) was dropped to cooled 25 g 3- fluorpyridine and 59 g n-benzylpiperazine in 600 mL diethylether. The reaction was warmed up to RT and stirred over night at RT. 150 mL water was dropped to the mixture and the organic layer was dried and evaporated. The residue was cleaned on silica gel

(dichlormethane/cyclohexane/methanol (5/5/1)) to yield 31.3 g of the desired compound. 6.01.04.2 l-Pyridin-3-yl-piperazine

31.3 g l-Benzyl-4-pyridin-3-yl-piperazin was hydrogenated at 5 bar on 4 g 10% palladium on charcoal in 300 mL methanol at 50°C for 25h. The reaction was filltered. The filtrate was evaporated and purified by chromatography on silica gel (dichlormethane/methanol/ammonia (9/1/0.1)). 12.5g of the desired compound was obtained. -bromo- 1 -(4-pyrimidin-2-yl-piperazin- 1 -yl)-ethanon

6.01.05.1 2-bromo- l-(4-pyrimidin-2-yl-piperazin-l-yl)-ethanon

20.2 g bromacetylbromide was dropped to 16.5 g l-pyrimidin-2-yl-piperazin and 10.2 g triethylamine in 250 mL THF. The reaction was stirred over night at RT and evaporated. The residue was extracted with DCM and water. The organic layer was evaporated and the residue was crystallized with petrolether and then purified by chromatography on silica gel

(DCM/MeOH: 95/5) to yield 36 mg of the desired compound.

R t : 1.09 min (method C)

(M+H) + : 286 -fluoro-pyridine-2-carboxylic acid methyl ester

2.7 mL Thionylchloride was dropped to 5 g 5-fluor-pyridine-2-carboxylic acid in 50 mL methanol. The reaction was stirred for 2 h at 65°C in a sealded micro wave vial. The solvents were removed and the residue was desolved in a mixture of DCM and methanol and filtered over silica gel. The filtrate was evaporated to give 5.9 g of the desired product.

R t : 0.77 (method K). (M+H) + : 156 By using the same synthesis strategy as for 5-fluoro-pyridine-2-carboxylic acid methyl ester the following compound was obtained:

-dimethyl-pyridine-2-carboxylic acid methyl ester

77.4 ml 2 mol/L trimethylsilyldiazomethan in hexane was added to 5, 6-dimethyl-pyridine-2- carboxylic acid in 150 mL methanol and 600 mL dichlormethane at -5°C. The reaction was stirred 0.5h at -5°C and then warm up to RT. The solvent was removed and the residue was purified by chromatorgaphie on Silica (cyclohexane 7: ethyl acetate 3) to give 12.8 g desired product.

R t : 0.49 (method M)

(M+H) + : 166 -chloro-6-piperazin- 1 -yl-pyrimidine

13. 3 g tert-butyl 1-piperazinecarboxylate and 11.2 mL DIPEA were added to 10 g 4, 6 dichloro pyrimidine in 1000 mL dichlormethane was stirred over the weekend at RT. The solvent was removed. The residue was dissolved in 200 mL dichlormethane and 200 mL trifluoro acetic acid was added. The reaction was stirred at RT. The solvent was removed.The residue was dissolved in methanol and passed through HC03 containing resin. The solvent was removed and the residue was purified by chromatographie on silica (dcyclohexane: ethylacetate 0-40%) to yield

13.07 g of the desired product. R t : 1.77 min (method W). (M+H) + : 199 5-fluoro-4-methyl-pyridine-2-carboxylic acid methyl ester

18 g 2-bromo-5-fluoro-4-methyl-pyridine, 1.5 g 1, Γ-bis (diphenylphosphino) ferrocene dichloropalladium (II) and 18 g sodium acetate was stirred 17 h at 80°C and 5 bar carbon monoxide. The reaction was filtered and the solvent was removed. Diethylether was added to the residue and the mixture was filtered. The filtrate was evaporated to give 12 g desired product. R t : 0.90 min. (method AB)

(M+H) + : 170

By using the same synthesis strategy as for 5-fluoro-4-methyl-pyridine-2-carboxylic acid methyl ester the following compound was obtained:

6.02. Synthesis of pyrazol-lyl-acids -(4-fluoro-3-mehyl-phenyl)-3-phenyl- 1 , 3-propanedione

17.3 g KOtBu was dissolved in 500 mL THF and 9 mL acetophenone was added. After 15 min at RT 25.9 g 4-fluoro-3-mehyl-benzoic acid methyl ester was added and stirred for 3 h at RT. The solvent was removed and the residue was purified by chromatography on silica gel

(cyclohexane/EE: 98:2) to yieled 19.3 g of the desired compound.

By using the same synthesis strategy as for l-(4-fluoro-3-mehyl-phenyl)-3-phenyl-l, 3- propanedione the following compounds were obtained:

6.02.01.16 223 method T 1.63

0 0

method

6.02.01.17 265 1.03

M

CI

1 0 0

method

6.02.01.18 273 1.00

M

6.02.01.19 250 method V 1.58

-Bis-(4-fluoro-phenyl)-l, 3-propanedione

9.27 g KOtBu was added to 109 mg 18-Krone-6 and 5 mL 4-fluoracetophenone in 150 mL THF. After 30 min at RT 10.7 g 4-fluoro-benzoic acid methyl ester was added and stirred for 3 h at RT. The reaction was decomposed with water and filtered, the filtrate was concentrated and the residue was purified by chromatography on silica gel (cyclohexane/EE: 98:2). The solvent was removed and 3.9 g of the desired compound was obtained.

(M+H) + : 261

By using the same synthesis strategy as for l-(4-fluoro-3-mehyl-phenyl)-3-phenyl-l, 3- propanedione the following compounds were obtained: MS

Product m/z HPLC Rt

Examples

[M+H] Method min

+

O 0

6.02.01.21 221 method B 1.48

6.02.01.22 223 method B 1.59

O 0

6.02.01.23 239

F

6.02.01.24 244 method D 1.08

6.02.01.25 262 method M 1.09

0 0

6.02.01.26 209 method A 1.05

0 0

6.02.01.27 195 method A 1.00

0 0

6.02.01.28 225 method P 1.20

0 0

6.02.01.29 205 method Q 0.86 0 0

6.02.01.39 292 method ΑΒ 0.97 -3-(4-fluoro-phenyl)-propane- 1 ,3-dione

0.88 g 60% sodiumhydrid was added 1-cyclohexyl-ethanone in 60 mL THF and stirred for 30 min. Then, 3.25 g 4-fluoro-benzoic acid methyl ester and 200 mg 18-Krone-6 was added to the reaction. The reaction was re fluxed 2h and stirred over night at RT. The reaction was added to 100 mL IN HC1 and extracted with diethylether. The organic layer was concentrated and the residue was purified by chromatography on silica gel and 3 g of the desired compound was obtained. R t : 2.01 min (method B). (M+H) + : 249

By using the same synthesis strategy as for l-cyclohexyl-3-(4-fluoro-phenyl)-propane-l,3-dione the following compounds were obtained:

3 -(4-fluoro-3-mehyl-phenyl)-5 -phenyl-pyrazo le

1.7 g l-(4-fluoro-3-mehyl-phenyl)-3-phenyl-l,3-propanedione was dissolved in 20 mL of a solution of 1 N hydrazine in THF and stirred for 3 h at 75 °C. The solvent was removed and the residue was crystallized to give 1.7 g of the desired compound.

R t : 1.07 min (method D)

(M+H) + : 253

By using the same synthesis strategy as for 3 -(4-fluoro-3-mehyl-phenyl)-5 -phenyl-pyrazo le the following compounds were obtained:

MS

Product HPLC Rt

Examples m/z

Method min

[M+H] +

F H F

6.02.02.02 method C 1.67

257

H

6.02.02.03 method C 1.61

245

H

6.02.02.04 method B 1.27

217

H

N -N

6.02.02.05 279 method C 1.62

O

6.02.02.06 257 method D 1.03

H

6.02.02.18 220 method P 1.10

H

N-N

6.02.02.19 205 method R 1.26

H

N-N

6.02.02.20 201 method Q 0.71

H

6.02.02.21 187 method R 1.20

H

6.02.02.22 249 method M 1.06

H

6.02.02.23 191 method M 0.78

H

6.02.02.24 249 method Q 0.76

H

6.02.02.25 236 method M 0.68

H

6.02.02.26 207 method Q 0.81

N-N \

6.02.02.27 233 method T 1.52

H

6.02.02.28 250 method M 0.64 -(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l-yl-acetic acid methyl ester

1.7 g 3-(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazole, 4 g K 2 CO 3 and 0.64 mL 2-bromoacetic acid methyl ester were dissolved in 100 mL acetone and stirred for 10 h under reflux. K 2 CO 3 was filtered and the solvent was removed to yield 1.9 g as a mixture of isomers. By using the same synthesis strategy as for l-(4-fluoro-3-mehyl-phenyl)-3-phenyl-l, 3- propanedione the following compounds were obtained:

6.02.03.21 259 method Q 0.81

6.02.03.22 321 method M 1.07 o /

6.02.03.23 263 method S 1.20

6.02.03.24 321 method Q 0.80

6.02.03.25 308 method M 0.66

6.02.03.26 279 method Q 1.44

6.02.03.27 305 method T 1.56 6.02.03.28 322 method M 0.95

/

V- o

6.02.03.29 322 method M 0.64

6.02.03.30 273 method T 1.50

\

0

6.02.03.31 322 method M 0.93

6.02.03.32 322 method M 0.71

6.02.03.33 291 method T 1.53

Isomere seperation of 6.02.03.41 and 6.02.03.42 column: Daicel ADH-Saule (250x20mm)

eluent: 25% EtOH+0.2% DEA : 75% C0 2 : flow: 65ml/min

dissolved in: MeOHxhloro form 9: 1 (concentration: 70mg/ml)

wave length: 210nm

6.02.04.01 3-(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l-yl acetic acid

1.9 g of 3-(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l-yl-acetic acid methyl ester was dissolved in 20 mL dioxane and a solution of 153 mg LiOH in 3 mL of water was added. The mixture was stirred for 6 h at 89 °C. The solvent was removed and the residue dissolved in a mixture of methanol and water, purified by HPLC (method 1) and subsequently separated into the two isomers by HPLC (method 2). Yield: 222 mg and 374 mg of the two isomers.

R t : 1.05 min (method D)

(M+H) + : 311

By using the same synthesis strategy as for 3-(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l-yl acetic acid the following compounds were obtained: 6.02.04.45 (3, 5-Bis-(4-methoxy-phenyl)-pyrazol-l-yl)-acetic acid

7 mL concentrated accetic acid was added to 500 mg l,3-bis-(4-methoxy-phenyl)-propane-l,3- dione and 299 mg hydrazino-acetic acid ethyl ester hydrochloride, stirred for 24 h at RT and additional 24h at 80°C. The reaction was cooled and the precipitate was filltered, washed with diethylether and dried to give 418 mg of the desired compound.

R f : 0.24 (flash chromatography: silica gel, DCM: MeOH: 19: 1+ two drops of cone, acetic acid) (M+H) + : 339. 6.02.05.01 (3, 5-Bis-(4-methoxy-phenyl)-pyrazol-l-yl)-acetic acid

700 mg (3-(4-fluoro-3-methyl-phenyl)-5-phenyl-pyrazol-l-yl)-acetic acid was dissolved in 10 mL DMF. 869 mg TBTU and 465 DIPEA were added to this solution and the mixture was stirred 1 min. at RT. 448 mg 4-chloro-6-piperazine-l-yl-pyrimidine was added and stirring was continued for 2h. Then 10 mL of a 1M NaOH solution and 50 mL CH 2 CI 2 were added, the organic phase was separated.The solvent was removed. The residue was dissolved in dichlormethane, water was added and the layers were seperated. The organic layer was evaporated and the residue was purified by flash chromatography (silica gel, DCM/MeOH 0-30 to yield 500 mg of the desired compound.

R t : 1.58 (method F)

(M+H) + : 491 7. Synthesis of target componds

7.01.001. 2-(3-(4-Fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol- 1 -yl)- 1 -(4-(2-pyrimidinyl)- 1 - piperazinyl)-ethanon

50 mg 3-(4-fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l-yl acetic acid was dissolved in 2 mL DMF. 83 mg PFTU and 33 μΐ ^ DIPEA were added to this solution and the mixture was stirred for 15 min at RT. Then, 25 2-(l-piperazinyl)-pyrimidine was added and stirring was continued for 8 h. Then, 1 mL of a K 2 CO 3 solution (5%) and 10 mL CH 2 CI 2 were added, the organic phase was separated and washed two times with water. The solvent was removed and the residue was crystallized from ACN to yield 46 mg of the desired compound.

R t : 1.04 min (method A)

(M+H) + : 457 By using the same synthesis strategy as for 2-(3-(4-Fluoro-3-mehyl-phenyl)-5-phenyl-pyrazol-l- yl)-l-(4-(2-pyrimidinyl)-l-piperazinyl)-ethanon the following compounds were obtained:

7.01.135 =N o 454 method M 0.73

7.01.178 492 method M 0,85

0

7.01.179 F 491 method M 0,70

7.01.180 522 method M 0,78

7.01.181 409 method M 0,90

7.01.182 408 method M 0,74 439 method M 0,82

499 method Q 0.78

424 method M 0,84

423 method M 0,67 454 method M 0,76

-(3, 5-di henyl-pyrazol- 1 -yl)- 1 -(4-(2-pyrimidinyl)- 1 -piperazinyl)-ethanon

278 mg (3,5-diphenyl-pyrazol-l-yl)-acetic acid was dissolved in 5 mL DMF. 321 mg TBTU and 172μί DIPEA were added to this solution and the mixture was stirred for a few minutes at RT. 164 mg 2-(l-piperazinyl)-pyrimidine was added and stirring was continued for 30 min. Then 10 mL of a 1M NaOH solution and 50 mL CH 2 CI 2 were added, the organic phase was separated.The solvent was removed and the residue was purified by flash chromatography (silica gel,

DCM/MeOH/NH3 0-5%). Diethylether was given to the residue and the precipitate was filltered to yield 310 mg of the desired compound.

R t : 1.48 (method F)

(M+H) + : 425

By using the same synthesis strategy as for 2-(3, 5-diphenyl-pyrazol- l-yl)-l -(4-(2-pyrimidinyl)- l-piperazinyl)-ethanon the following compounds were obtained:

7.03.01. 2-(3, 5-bis-(4-methoxy-phenyl))-pyrazol- 1 -yl)- 1 -(4-(2-pyrimidinyl)- 1 - piperazinyl)-ethanon

100 mg (3, 5 -bis-(4-methoxy-phenyl)-pyrazol-l-yl)-acetic acid acid was dissolved in 4 mL

DMF. 135 mg HATU and 200 DIPEA were added to this solution and the mixture was stirred for 15 min at RT. Then 77 mg 2-(l-piperazinyl)-pyrimidine hydrochloride was added. The mixture was stirred over night. The reaction was diluted with potassium carbonat solution and extracted wih dichlormethane. The organic layer was evaporated. The residue was purified by HPLC to give 36 mg of the desired compound.

R t : 1.52 (method C)

(M+H) + : 485 2-(3, 5-bis-(2-fluor-phenyl))-pyrazol- 1 -yl)- 1 -(4-(2-pyrimidinyl)- 1 -piperazinyl)-

200 mg 3,5-bis-(2-fiuoro-phenyl)-lH-pyrazole, 222 mg 2-bromo-l-(4-pyrimidin-2-yl-piperazin- l-yl)-ethanon and 500 mg potassiumcarbonat in 20 mL acetone was stirred 24h by RT. The reaction was filltered and the filtrate was evaporated. The residue was crystallized with acetonitrile to yield 254 mg of the desired compound. R t : 1.57 min (method C). (M+H) : 461.

By using the same synthesis strategy as for 2-(3, 5-bis-(2-fluor-phenyl))-pyrazol-l-yl)-l-(4-(2- pyrimidinyl)-l-piperazinyl)-ethanon the following compounds were obtained:

7.05.01. 2-[3-(4-nuoro-3-methyl-phenyl)-5-phenyl-pyrazol-l-yl]-l-{4-[ 6-(2-morpholin-4- -ethoxy)-pyrimidin-4-yl]-piperazin- 1 -yl} -ethanone

50 mg (3, 5-Bis-(4-methoxy-phenyl)-pyrazol-l-yl)-acetic acid was added to 68 mg N-(2- hydroxyethyl) morpholine and 14.1 mg potasiumcarbonate in 2 mL n-methyl-2-pyrrolidinone. The reaction was stirred 30 min. at 120 °C and 15 min. at 200 °C under microwave conditions. The solvent was removed and the residue was purified by HPLC.

R t : 1.34min (method X)

(M+H) + : 586

By using the same synthesis strategy as for 2-[3-(4-fluoro-3-methyl-phenyl)-5-phenyl-pyrazol-l- yl]- 1 - {4-[6-(2-morpholin-4-yl-ethoxy)-pyrimidin-4-yl]-piperazin- 1 -yl} -ethanone the following compounds were obtained: