NOVEL N-[(PYRIMIDINYLOXY)PROPANYL]BENZAMIDES

Information

  • Patent Application
  • 20190112295
  • Publication Number
    20190112295
  • Date Filed
    April 17, 2017
    7 years ago
  • Date Published
    April 18, 2019
    5 years ago
Abstract
This invention relates to compounds of formula (I), a process for their preparation, pharmaceutical compositions containing them and their use in the treatment of conditions having an association with the orexin sub-type 1 receptor. Ar, R1, R2, R3, R4, R5 have meanings given in the description.
Description
FIELD OF THE INVENTION

The present invention relates to novel N-[(pyrimidinyloxy)propanyl]benzamide derivatives, processes for their preparation, pharmaceutical compositions containing them and their use in therapy, particularly in the treatment or prevention of conditions having an association with the orexin sub-type 1 receptor.


BACKGROUND OF THE INVENTION

Orexins are hypothalamic neuropeptides that play an important role in the regulation of many physiological behaviours such as arousal, wakefulness, appetite, food intake, cognition, motivated behaviours, reward, mood and stress. Orexin A, also referred to as hypocretin 1, is a peptide composed of 33 amino acids and orexin B, also referred to as hypocretin 2, is a peptide composed of 28 amino acids. Both are derived from a common precursor peptide referred to as pre-pro-orexin [Sakurai et al., Cell, 1998 Feb. 20; 92(4):573-85, and De Lecea et al., Proc. Nat. Acad. Sci., 1998 Jan. 6; 95(1):322-7). Orexins bind to two orphan G-protein-coupled receptors, the orexin receptor type 1 (OX1R) and orexin receptor type 2 (OX2R), which are widely distributed in the central nervous system and peripheral organs such as adrenal glands, gonads, and gut. Whereas orexin A binds predominantly to OX1R, orexin B is able to bind to both OX1R and OX2R.


Orexins are involved in the regulation of a wide range of behaviours including for example the regulation of emotion and reward, cognition, impulse control, regulation of autonomic and neuroendocrine functions, arousal, vigilance and sleep-wakefulness states (Muschamp et al., Proc. Natl. Acad. Sci. USA 2014 Apr. 22; 111(16):E1648-55; for a recent review see Sakurai, Nat. Rev. Neurosci., 2014; Nov.; 15(11):719-31; Chen et al., Med. Res. Rev., 2015; January; 35(1):152-97; Gotter et al., Pharmacol. Rev., 2012, 64:389-420 and many more). Dual antagonism of OX1R and OX2R by small molecules is clinically efficacious in the treatment of insomnia, for which the drug suvorexant, [[(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl]methanone] has been granted marketing authorisation (Kishi et al., PLoS One, 2015; 10(8):e0136910). The sleep-inducing effects of dual orexin receptor antagonists are predominantly mediated via OX2R (Bonaventure et al., J. Pharmacol. Exp. Ther., March 2015, 352, 3, 590-601), whereas the other physiological states such as emotion and reward, cognition, impulse control, regulation of autonomic and neuroendocrine functions, arousal, and vigilance are rather mediated via OX1R.


Due to their sleep-inducing effects, dual OX1R and OX2R antagonists are not suitable for treating disorders related to impulse control deficits as seen in addictions such as substance use disorders, personality disorders such as borderline personality disorder, eating disorders such as binge eating disorder, or attention deficit hyperactivity disorder (ADHD). Therefore, it is desirable to provide an OX1R selective antagonist for the treatment of impulse control deficits.


Orexin receptor antagonists of various structural classes are reviewed in Roecker et al. (J. Med. Chem. 2015, 59, 504-530). WO2013/187466, WO2016/034882 and Bioorganic & Medicinal Chemistry 2015, 23, 1260-1275 describe orexin receptor antagonists.







DETAILED DESCRIPTION OF THE INVENTION

The present invention provides novel N-ethyl-N-[(2S)-1-(pyrimidin-2-yloxy)propan-2-yl]-benzamides of formula I




embedded image


in which


Ar represents




embedded image


R1 represents hydrogen, fluoro, methyl;


R2 and R3 independently represent hydrogen, fluoro, methyl, —OCH3;


R4 represents hydrogen or fluoro;


R5 represents bromo or —CF3;


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


In another embodiment, in the general formula I, Ar and R5 have the same meaning as defined in any of the preceding embodiments, and at least two of the substituents R1, R2, R3 and R4 represent hydrogen.


In another embodiment, in the general formula I, Ar, R1, R4 and R5 have the same meaning as defined in any of the preceding embodiments, and


R2 represents hydrogen, fluoro, methyl, —OCH3; and


R3 represents hydrogen, fluoro, methyl, —OCH3.


In another embodiment, in the general formula I, Ar, R4 and R5 have the same meaning as defined in any of the preceding embodiments, and R1, R2 and R3 independently represent hydrogen or fluoro.


In another embodiment, in the general formula I, Ar, R1, R2, R3 and R5 have the same meaning as defined in any of the preceding embodiments, and


R4 represents hydrogen.


In another embodiment, in the general formula I, Ar, R1, R2, R3 and R4 have the same meaning as defined in any of the preceding embodiments, and


R5 represents —CF3.


In another embodiment, in the general formula I, R1, R2, R3, R4 and R5 have the same meaning as defined in any of the preceding embodiments, and


Ar represents




embedded image


In another embodiment, in the general formula I,


Ar represents




embedded image


R1, R2 and R3 independently represent hydrogen or fluoro;


R4 represents hydrogen;


R5 represents —CF3.


Compounds of the present invention are potent OX1R antagonists. They are more selective over the OX2R than preferred examples disclosed in WO2013/187466. Compounds of the present invention differ structurally from those disclosed in WO2013/187466 in that they contain a substituted —O-pyrimidyl moiety in place of a Het1-Het2 moiety in which Het2 is phenyl or pyridyl. These structural differences unexpectedly result in an explicit enhancement in selectivity over the OX2R.


Compounds of the present invention differ structurally from Examples 14 and 91 in WO2016/034882 (closest prior art) in that they contain a central N-ethyl-(propan-2-yl)amino moiety in place of the N-methyl-[butan-2-yl]amino moiety or a N-methyl-(propan-2-yl)amino moiety and contain a —O-pyrimidyl instead of the —N-pyrimidyl or —N-pyridyl substituent. These structural differences unexpectedly result either in a marked increase in potency at the OX1R at comparable or higher selectivity over OX2R, or in an explicit enhancement in selectivity over the OX2R at comparable or increased potency at OX1R. Therefore, compounds of the present invention are expected either to be more efficacious in vivo or to have a larger therapeutic window or to demonstrate both characteristics. Either of these qualities results in a better drug safety. A drug compound that is more efficacious may be administered at lower doses thereby reducing the likelihood of OX1R independent side effects. A drug compound that is more selective over OX2R is expected to cause fewer side effects such as drowsiness or sleep. Consequently, compounds of the present invention are expected to be safer and must thus be more viable for human use.


General Definitions

Terms not specifically defined herein should be given the meanings that would be given to them by one skilled in the art in light of the disclosure and the context.


Stereochemistry:

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, diastereoisomers, E/Z isomers etc.) and racemates thereof, as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereoisomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof.


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.


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. trifluoroacetate salts) also comprise a part of the invention.


Biological Assays
Abbreviations:


















IP1
D-myo-Inositol-1-phosphate



IP3
D-myo-inositol-1,4,5-triphosphate



HEPES
4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid



HBSS
Hanks' Balanced Salt Solution



BSA
bovine serum albumin



DMSO
dimethyl sulfoxide



CHO
Chinese hamster ovary










Activation of the orexin receptors expressed in cell lines results in an increase in intracellular IP3 concentration. IP1, a downstream metabolite of IP3, accumulates in cells following receptor activation and is stable in the presence of LiCl. Using Homogeneous Time-Resolved Fluorescence technology with Lumi4-Tb cryptate (commercially available from Cisbio Bioassay.) and a suitable fluorescence plate reader. This functional response is detectable and quantifiable as described in Trinquet et al. Anal. Biochem. 2006, 358, 126-135, Degorce et al. Curr. Chem. Genomics 2009, 3, 22-32. This technique is used to characterize pharmacological modification of the orexin receptors.


The biological activity of compounds is determined by the following methods:


A. In Vitro Testing of OX1R Potency: OX1R IP1

IP1 measurements are performed in CHO-K1 cells stably expressing the full-length human Orexin 1 receptor and the aequorin photoprotein. Cells are cultivated in Ham's nutrient mixture F12 medium with 10% fetal calf serum, in a 37° C., 95% humidity and 5% CO2 incubator. The CHO-K1/hOx1 cell mass is expanded to larger cell numbers. The cells are obtained as frozen cells in cryo-vials and stored until use at −150° C. The viability of the cells after thawing is >90%. In preparation for the assay, 24 hours before the assay, the cells are thawed at 37° C. and immediately diluted with cell culture medium. After centrifugation, the cell pellet is re-suspended in medium and then distributed into the assay plates with a density of 10000 cells/25 μL per well. The plates are incubated for one hour at room temperature to reduce edge effects before they are incubated for 24 hours at 37° C./5% CO2. Compounds are prepared by an 8-point serial dilution in DMSO and a final dilution step into assay buffer (HBSS with 20 mM HEPES, 0.1% BSA and 50 mM LiCl, pH 7.4) to ensure a final DMSO concentration of 1% in the assay.


On the day of the assay, cells in the plate are washed twice with 60 μL assay buffer (20 μL buffer remained in the wells after washing), followed by adding 5 μL per well of compounds diluted in assay buffer. After 15 minutes of incubation at room temperature 5 μL per well of Orexin A peptide (final concentration: 0.5 nM and/or 50 nM) dissolved in assay buffer is added to the assay plate. The assay plate is incubated for 60 minutes at 37° C. Then 5 μl per well of Anti-IP1-Cryptate Tb solution and 5 μl per well of IP1-d2 dilution are added and the plate is incubated for a further 60 minutes light protected at room temperature. The emissions at 615 nm and 665 nm (Excitation wavelength: 320 nm) are measured using an EnVision reader (PerkinElmer). The ratio between the emission at 665 nm and 615 is calculated by the reader.


8-point four parametric non-linear curve fitting and determination of IC50 values and Hill slopes is performed using a regular analysis software e.g. AssayExplorer (Accelrys). In order to establish an agonist concentration independent parameter, Kb values are calculated using the following equation: IC50/((2+(A/EC50)n)1/n−1) (with A=concentration agonist, EC50=EC50 agonist, n=Hill slope agonist).


B. In Vitro Testing of OX2R Potency: OX2R IP1

IP1 measurements are performed in CHO-K1 cells stably expressing the full-length human orexin 2 receptor and the aequorin photoprotein. Cells are cultivated in Ham's nutrient mixture F12 medium with 10% fetal calf serum, in a 37° C., 95% humidity and 5% CO2 incubator. The CHO-K1/hOx2 cell mass is expanded to larger cell numbers. The cells are obtained as frozen cells in cryo-vials and stored until use at −150° C. The viability of the cells after thawing is >90%. In preparation for the assay, 24 hours before the assay, the cells are thawed at 37° C. and immediately diluted with cell culture medium. After centrifugation, the cell pellet is resuspended in medium and then distributed into the assay plates with a density of 5000 cells/25 μL per well. The plates are incubated for one hour at room temperature to reduce edge effects before they are incubated for 24 hours at 37° C./5% CO2. Compounds are prepared by a 8-point serial dilution in DMSO and a final dilution step into assay buffer (HBSS with 20 mM HEPES, 0.1% BSA and 50 mM LiCl, pH 7.4) to ensure a final DMSO concentration of 1% in the assay.


On the day of the assay, cells in the plate are washed twice with 60 μL assay buffer (20 μL buffer remained in the wells after washing), followed by adding 5 μL per well of compounds diluted in assay buffer. After 15 minutes of incubation at room temperature 5 μL per well of Orexin A peptide (final concentration: 0.5 nM) dissolved in assay buffer is added to the assay plate. The assay plate is incubated for 60 minutes at 37° C. Then 5 μl per well of Anti-IP1-Cryptate Tb solution and 5 μl per well of IP1-d2 dilution are added to all well of the plate and the plate is incubated for a further 60 minutes light protected at room temperature. The emission at 615 nm and 665 nm (Excitation wavelength: 320 nm) are measured using an EnVision reader (PerkinElmer). The ratio between the emission at 665 nm and 615 is calculated by the reader.


8-point four parametric non-linear curve fitting and determination of 1050 values and Hill slopes is performed using a regular analysis software e.g. AssayExplorer (Accelrys). In order to establish an agonist concentration independent parameter, Kb values are calculated using the following equation: IC50/((2+(A/EC50)n)1/n−1) (with A=concentration agonist, EC50=EC50 agonist, n=Hill slope agonist) (see P. Leff, I. G. Dougall, Trends Pharmacol. Sci. 1993, 14(4), 110-112).


Kb values from Assay A (OX1R) and Assay B (OX2R) can then provide a selectivity ratio which is independent of the agonist (Orexin A) concentration.


Biological Data

Comparison of Assays A and B with the Assays Described in WO2013/187466


Assays described in WO2013/187466 differ from assays A and B in:

    • The technology and readout: fluorescence measurement of intracellular Ca2+ changes (WO2013/187466) instead of luminescence measurement of IP1 (assays A and B)
    • OX1R and OX2R overexpressing cell lines used for the assays described in WO2013/187466 are of different origin as cell lines used for assays A and B
    • Use of modified orexin A (2 amino acids substituted) as agonist instead of orexin A
    • Agonist concentration of 300 pM used for the OX1R assay and 3 nM for the OX2R assay (EC75 vs. EC100; according to Okumura T. et al., Biochemical and Biophysical Research Communications, 2001) (WO2013/187466). IC50 values that have been reported are dependent on the agonist concentration. Selectivity ratios calculated from these IC50 values cannot be compared with the selectivity ratios calculated from the agonist concentration independent Kb values obtained from assay A and B.


Due to these differences between the assays, a direct comparison has to be established. Therefore, examples 69, 70 (the most selective ones) and 5 (one of the most potent ones) described in WO2013/187466 are tested in assays A and B so as to be directly compared with compounds of the present invention (see Table 1).









TABLE 1







In vitro potencies of compounds of WO2013/187466 as reported therein versus as


determined in the Assays A and B (described above)










As described in WO2013/187466
As determined in Assays A and B
















OX2R
OX1R Kb
OX2R Kb
OX2R



OX1R
OX2R
IC50/
[nM] (Orexin
[nM] (0.5 nM
Kb/


Structure
IC50
IC50
OX1R
A concentration
Orexin A
OX1R


Example # in WO2013/187466
[nM]
[nM]
IC50
used)
concentration)
Kb







embedded image


1.6
1896
1185
2.25 (0.5 nM)
98
43







embedded image


1.1
 452
411
0.72 (50 nM)
29
40







embedded image


0.5
 76
152
0.94 (50 nM)
28
30
















TABLE 2







In vitro potencies of the structurally closest prior art compounds (Example 14 and 91)


WO2016/034882 as reported therein:









As described in WO2016/034882 (Table 1, page 178)










Structure


OX2R IC50/


Example # in WO2016/034882
OX1R
OX2R
OX1R IC50







embedded image


Table 1: pIC50 = 8.3 corresponds to IC50 = 5.0 nM Table 2: pIC50 = 7.8 corresponds to IC50 = 16 nM Table 3: not reported
Table 1: pIC50 = 6.8 corresponds to IC50 = 158 nM Table 2: pIC50 = 7.2 corresponds to IC50 = 63 nM Table 3: not reported
Table 1: 32 Table 2: 4







embedded image


Table 1 and 2: not reported Table 3: pIC50 = 7.6 corresponds to IC50 = 25 nM
Table 1 and 2: not reported Table 3: pIC50 <5.1 corresponds to IC50 = >7950 nM
Table 3: >318









Table 3 shows a comparison of biological data on the OX1R and OX2R potencies of the compounds of the present invention with the closest prior art compounds in WO2016/034882.


Examples 1, 2, 5, 7, 10, 11, 13, 14, 20, 22, 24, 30, 34 and 35 of the present invention differ structurally from Example 91 in WO2016/034882, the closest prior art compound, in that they contain a) a central N-ethyl-(propan-2-yl)amino moiety in place of the N-methyl-[butan-2-yl]amino moiety, and b) a —O-pyrimidyl instead of the —N-pyrimidyl substituent. Further, the phenyl group may have a different substitution pattern.


In Examples 1, 7 and 20, the fluoro substituent of the phenyl group is in a different position than in Example 91 in WO2016/034882. In Examples 35 and 22, the phenyl group bears an additional fluoro substituent. These structural differences unexpectedly result in Examples 1, 7, 20, 35, 22 being more selective for OX1R over OX2R (selectivity ratio OX2R Kb/OX1R Kb) as compared to Example 91 in WO2016/034882.


In Example 2, the phenyl group carries two methyl groups. These structural differences lead to an increase in OX1R potency compared to Example 91 in WO2016/034882.


In Examples 5, 10, 11, 13, 14, 24, and 30, the phenyl group is unsubstituted or substituted with a methoxy, or one or two methyl groups instead of the fluoro substituent in Example 91 in WO2016/034882. These structural differences unexpectedly result in an increase in both OX1R potency and OX1R selectivity compared to Example 91 in WO2016/034882.


In Example 34, the phenyl group contains a methyl group in addition to the fluoro substituent in Example 91 in WO2016/034882. This structural difference unexpectedly leads to an increase in OX1R potency and an increase in OX1R selectivity compared to Example 91 in WO2016/034882.


Examples 8, 15, 18, 19, 21, 23, 31, and 36 of the present invention differ structurally from Example 91 in WO2016/034882, the closest prior art compound, in that they a) contain a central N-ethyl-(propan-2-yl)amino moiety in place of the N-methyl-[butan-2-yl]amino moiety and b) contain a —O-pyrimidyl instead of the —N-pyrimidyl substituent. They further have a differently substituted phenyl group: They contain a pyrazoyl, oxadiazoyl, or oxazoyl instead of the triazoyl, and further the phenyl group is unsubstituted or substituted with a methyl, a fluoro in a different position than in Example 91 in WO2016/034882. These structural differences lead to an increase in OX1R selectivity compared to Example 91 in WO2016/034882.


Example 29 of the present invention differs structurally from Example 91 in WO2016/034882, the closest prior art compound, in that it contains a) a central N-ethyl-(propan-2-yl)amino moiety in place of the N-methyl-[butan-2-yl]amino moiety and b) a —O-pyrimidyl instead of the —N-pyrimidyl substituent. It further contains the fluoro substituent on the phenyl group in a different position as compared to Example 91 in WO2016/034882, and contains a bromo substituted pyrimidine rather than the CF3-substituted pyrimidine. These structural differences unexpectedly lead to an increase in OX1R selectivity compared to Example 91 in WO2016/034882.


Examples 32, 28, and 25 of the present invention differ structurally from Example 14 in WO2016/034882, the closest prior art compound, in that they contain a) a central N-ethyl-(propan-2-yl)amino moiety in place of the N-methyl-[prop-2-yl]amino moiety and b) a —O-pyrimidyl instead of the —N-pyridyl substituent. They further contain a pyrimidine instead of the second phenyl group. Examples 32 and 25 further contain a fluoro substituent on the phenyl group. These structural differences unexpectedly result in an increase in OX1R selectivity compared to Example 14 in WO2016/034882.









TABLE 3







Comparison of biological data of the compounds of the present invention with the


closest prior art compounds in WO2016/034882













Assay A OX1R
Assay B OX2R





Kb [nM]
Kb [nM]





(Orexin A
(0.5 nM Orexin
OX2RKb/


Example
Structure
concentration)
A concentration)
OX1R Kb














Ex 91 in WO2016/ 034882


embedded image


3.63 (0.5 nM)
313
 86





 1


embedded image


1.7 (0.5 nM) 2.5 (50 nM)
425
250 170





 7


embedded image


4.5 (0.5 nM)
902
201





20


embedded image


2.6 (0.5 nM)
803
309





35


embedded image


2.5 (0.5 nM)
455
182





22


embedded image


3.6 (0.5 nM)
1461
406





 2


embedded image


0.39 (50 nM)
34
 87





 5


embedded image


1.0 (0.5 nM) 1.1 (50 nM)
156
156 142





10


embedded image


0.65 (50 nM)
565
869





11


embedded image


1.3 (0.5 nM) 1.4 (50 nM)
160
123 114





13


embedded image


0.5 (0.5 nM) 0.38 (50 nM)
142
284 374





14


embedded image


0.97 (0.5 nM) 1.21 (50 nM)
253
261 209





30


embedded image


1.8 (0.5 nM)
579
322





34


embedded image


1.8 (0.5 nM)
431
239





24


embedded image


1.8 (0.5 nM) 2.6 (50 nM)
237
132  91





 8


embedded image


2.2 (0.5 nM)
502
228





15


embedded image


7.8 (0.5 nM)
999
128





18


embedded image


16 (0.5 nM)
6637
415





19


embedded image


7.3 (0.5 nM)
6638
909





21


embedded image


2.0 (0.5 nM) 1.9 (50 nM)
540
270 284





23


embedded image


2.1 (0.5 nM)
435
207





31


embedded image


6.0 (0.5 nM)
746
124





36


embedded image


3.2 (0.5 nM)
511
160





29


embedded image


2.0 (0.5 nM)
350
178





Ex 14 in WO2016/ 034882


embedded image


0.171 (50 nM)
4.7
 27





32


embedded image


0.61 (0.5 nM) 0.46 (50 nM)
188
308 408





28


embedded image


0.62 (0.5 nM) 0.74 (50 nM)
176
284 238





25


embedded image


0.81 (0.5 nM) 1.4 (50 nM)
272
336 194









Use in Treatment/Method of Use

The present invention is directed to compounds which are useful in the treatment of a disease, disorder and condition wherein the antagonisms of OX1R is of therapeutic benefit, including but not limited to the treatment and/or prevention of psychiatric and neurological conditions associated with impulse control deficits. Such impulse control deficits are seen in addictions including substance use disorders; personality disorders such as borderline personality disorder; eating disorders such as binge eating disorder; or attention deficit hyperactivity disorder. According to a further aspect of the invention, compounds of the present invention are useful in the treatment of OX1R related pathophysiological disturbances in arousal/wakefulness, appetite/food intake, cognition, motivated behaviours/reward, mood and stress.


In view of their pharmacological effect, compounds of the present invention are suitable for use in the treatment of a disease or condition selected from the list consisting of


(1) treatment or prevention of substance abuse/dependence/seeking or addiction as well as relapse prevention (including but not limited to drugs, such as cocaine, opiates such as morphine, barbiturates, benzodiazepines, amphetamines, nicotine/tobacco and other psychostimulants), alcoholism and alcohol-related disorders, drug abuse or addiction or relapse, tolerance to narcotics or withdrawal from narcotics,


(2) eating disorders, such as binge eating, bulimia nervosa, anorexia nervosa, other specified feeding or eating disorders, obesity, overweight, cachexia, appetite/taste disorders, vomiting, nausea, Prader-Willi-Syndrome, hyperphagia, appetite/taste disorders,


(3) attention deficit hyperactivity disorder, conduct disorders, attention problems and related disorders, sleep disorders, anxiety disorders such as generalized anxiety disorder, panic disorder, phobias, post-traumatic stress disorder, schizophrenia, Alzheimer's disease, Parkinson's disease, Huntington's disease and Gilles de la Tourette's syndrome, restless legs syndrome, dementia, dyskinesia, severe mental retardation, neurodegenerative disorders including nosological entities such as disinhibition-dementia-parkinsonism-amyotrophy complex, pallido-ponto-nigral degeneration,


(4) cognitive dysfunction in psychiatric or neurological disorder, cognitive impairments associated with schizophrenia, Alzheimer's disease and other neurological and psychiatric disorders,


(5) mood disorders, bipolar disorder, mania, depression, manic depression, borderline personality disorder, antisocial personality disorder, aggression such as impulsive aggression, suicidality, frontotemporal dementia, obsessive compulsive disorder, delirium, affective neurosis/disorder, depressive neurosis/disorder, anxiety neurosis, dysthymic disorder,


(6) sexual disorder, sexual dysfunction, psychosexual disorder,


(7) impulse control disorders such as pathological gambling, trichotillomania, intermittent explosive disorder, kleptomania, pyromania, compulsive shopping, internet addiction, sexual compulsion,


(8) sleep disorders such as narcolepsy, jetlag, sleep apnea, insomnia, parasomnia, disturbed biological and circadian rhythms, sleep disturbances associated with psychiatric and neurological disorders,


(9) treatment, prevention and relapse control of impulsivity and/or impulse control deficits and/or behavioural disinhibition in any psychiatric and/or neurological condition,


(10) personality disorders such as borderline personality disorder, antisocial personality disorder, paranoid personality disorder, schizoid and schizotypal personality disorder, histrionic personality disorder, narcissistic personality disorder, avoidant personality disorder, dependent personality disorder, other specified and non-specified personality disorders


(11) neurological diseases, such as cerebral oedema and angioedema, cerebral dementia like e.g. Parkinson's and Alzheimer's disease, senile dementia; multiple sclerosis, epilepsy, temporal lobe epilepsy, drug resistant epilepsy, seizure disorders, stroke, myasthenia gravis, brain and meningeal infections like encephalomyelitis, meningitis, HIV as well as schizophrenia, delusional disorders, autism, affective disorders and tic disorders.


The applicable daily dose of compounds of the present invention may vary from 0.1 to 2000 mg. The actual pharmaceutically effective amount or therapeutic dose will 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 drug substance is to be administered at a dose and in a manner which allows a pharmaceutically effective amount to be delivered that is appropriate to the patient's condition.


Pharmaceutical Compositions

Suitable preparations for administering the compounds of the present invention 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, powders, etc. The content of the pharmaceutically active compound(s) may vary in the range from 0.1 to 95 wt.-%, preferably 5.0 to 90 wt.-% of the composition as a whole.


Suitable tablets may be obtained, for example, by mixing a compound of the present invention with known excipients, for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants and pressing the resulting mixture to form tablets.


Combination Therapy

Compounds according to the present invention can be combined with other treatment options known to be used in the art in connection with a treatment of any of the indications the treatment of which is in the focus of the present invention.


Among such treatment options that are considered suitable for combination with the treatment according to the present inventions are:

    • Antidepressants
    • Mood stabilizers
    • Antipsychotics
    • Anxiolytics
    • Antiepileptic drugs
    • Sleeping agents
    • Cognitive enhancer
    • Stimulants
    • Non-stimulant medication for attention deficit hyperactivity disorder
    • Additional psychoactive drugs.


General Synthetic Methods

The invention also provides a process for making compounds of Formula (I). Unless specified otherwise, R1, R2, R3, R4, R5 and Ar in the formulas below shall have the meaning as defined for formula I in the detailed description of the invention above.


Optimum reaction conditions and reaction times may vary depending on the particular reactants used. Unless otherwise specified, solvents, temperatures, pressures, and other reaction conditions may be readily selected by one of ordinary skill in the art. Specific procedures are provided in the Synthetic Examples section. Typically, reaction progress may be monitored by thin layer chromatography (TLC), liquid chromatography-mass spectrometry (LC-MS) if desired, and intermediates and products may be purified by chromatography and/or by recrystallization.


The examples which follow are illustrative and, as recognized by one skilled in the art, particular reagents or conditions could be modified as needed for individual compounds without undue experimentation. Starting materials and intermediates used, in the methods below, are either commercially available or easily prepared from commercially available materials by those skilled in the art.


Compounds of Formula (I) can be synthesized by the method illustrated in Scheme 1:




embedded image


As shown in scheme 1, reacting the alcohol of formula II with a halo pyrimidine III (X=halide) in a nucleophilic aromatic substitution reaction, in a suitable solvent such as dioxane, THF or DMF and in the presence of a suitable base such as potassium tert-butoxide or NaH, provides an ether of formula IV. Debenzylation reactions are described in ‘Protective Groups in Organic Synthesis’, 3′ edition, T. W. Greene and P. G. M. Wuts, Wiley-Interscience (1999). Debenzylation of compound IV using 1-chloroethylchloroformate as a hydrogenating reagent may be used in a suitable solvent such as 1,2-dichloroethane to provide an amine of formula V.


Peptide coupling reactions known to the person skilled in the art (see for example M. Bodanszky, 1984, The Practice of Peptide Synthesis, Springer-Verlag) can be applied to react the secondary amine of formula V with a carboxylic acid of formula VI to yield a compound of formula I. For example, amine V and carboxylic acid VI in a suitable solvent such as acetonitrile or DMF in the presence of a suitable base such as DIPEA yields upon treatment with the coupling agent 2-chloro-4,5-dihydro-1,3-dimethyl-1H-imidazolium hexafluorophosphate (CIP) or 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate (HATU) a compound of formula I.


Alternatively, compounds of Formula I can be synthesized as illustrated in Scheme 2:




embedded image


Debenzylation reactions are described in ‘Protective Groups in Organic Synthesis’, 3′ edition, T. W. Greene and P. G. M. Wuts, Wiley-Interscience (1999). Debenzylation of compound II in a suitable solvent such as MeOH, with under a pressure of hydrogen in the presence of a suitable catalyst such as Pd/C results in a secondary amine of formula VII.


Peptide coupling reactions known to the person skilled in the art (see for example M. Bodanszky, 1984, The Practice of Peptide Synthesis, Springer-Verlag) can be applied to react the secondary amine of formula VII with a carboxylic acid of formula VI to yield a compound of formula VIII. For example, carboxylic acid VI in a suitable solvent such as DCM, DMF and toluene upon treatment with thionyl chloride or oxalyl chloride yields an acid chloride which is then treated with an amine of formula VII, in a suitable solvent such as DCM and THF, in the presence of a suitable base such as TEA, to provide a compound of formula VIII. Other peptide coupling reagents such as HATU in a suitable solvent such as DMF in the presence of a suitable base such as DIPEA may be used.


Reacting the alcohol of formula VIII with a halo pyrimidine III (X=halide) in a nucleophilic aromatic substitution reaction, in a suitable solvent such as dioxane, DMSO or DMF and in the presence of a suitable base such as potassium tert-butoxide or NaH, provides a compound of formula I. Alternatively, the alcohol of formula VIII can be reacted with a hydroxy pyrimidine of formula III (X═OH) in a Mitsunobu reaction in the presence of diethylazodicarboxylate (DEAD) or diisopropylazodicarboxylate (DIAD) and in the presence of triphenylphosphine to provide a compound of formula I.


Alternatively, an alcohol of formula VIII can be synthesized as illustrated in Scheme 3:




embedded image


Peptide coupling reactions known to the person skilled in the art (see for example M. Bodanszky, 1984, The Practice of Peptide Synthesis, Springer-Verlag) can be applied to react a secondary amine of formula IX, in which the protecting group (PG) may be a tert-butyl-dimethylsilyl group, with a carboxylic acid of formula VI to yield a compound of formula X. For example, a peptide coupling reagents such as TBTU or HATU in a suitable solvent such as DMF in the presence of a suitable base such as DIPEA may be used. Alkylation of the amide X using a suitable alkylation agent such as ethyl iodide in a suitable solvent such as DMF and a suitable base such as potassium tert-butoxide yields amide XI.


Deprotection reactions described in ‘Protective Groups in Organic Synthesis’, 3′ edition, T. W. Greene and P. G. M. Wuts, Wiley-Interscience (1999) can be applied in providing alcohol VIII from compound XI. For example tetra-n-butylammonium fluoride in a suitable solvent such as THF may be used.


Intermediate carboxylic acids V are commercially available or they can be synthesized according or in analogy to methods described in the literature.


EXPERIMENTAL SECTION
List of Abbreviations



  • RT room temperature

  • CIP 2-chloro-4,5-dihydro-1,3-dimethyl-1H-imidazolium hexafluorophosphate

  • dppf 1,1′-bis(diphenylphosphanyl)ferrocene

  • ESI-MS electrospray ionisation mass spectrometry

  • aq. aqueous

  • MS mass spectrometry

  • MeOH methanol

  • EtOH ethanol

  • EA ethyl acetate

  • DMF N,N-dimethylformamide

  • DMSO dimethyl sulfoxide

  • DCM dichloromethane

  • TEA triethylamine

  • THF tetrahydrofuran

  • DIPEA N,N-diisopropylethylamine

  • HATU 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate

  • TBTU O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyl-uronium tetrafluoroborate

  • Rt retention time

  • h hour(s)

  • min minutes

  • sat. saturated

  • ACN acetonitrile

  • TFA trifluoroacetic acid

  • M molarity

  • N normality

  • HPLC high-performance liquid chromatography

  • HPLC-MS high-performance liquid chromatography-mass spectrometry

  • LC-MS liquid chromatography-mass spectrometry

  • TLC thin layer chromatography

  • DIAD diisopropyl azodicarboxylate

  • DEAD diethyl azodicarboxylate



HPLC-Methods:
Method Name: A
Column: Venusil XBP-C18, 2.1×50 mm, 5 μm
Column Supplier: Agela Technologies
















Gradient/
% Sol
% Sol




Solvent
[H2O,
[ACN,




Time
0.0375%
0.018%
Flow
Temp


[min]
TFA]
TFA]
[mL/min]
[° C.]



















0.00
90
10
0.8
50


0.40
90
10
0.8
50


3.40
0
100
0.8
50


3.85
0
100
0.8
50


3.86
90
10
0.8
50


4.50
90
10
0.8
50









Method Name: B
Column: Sunfire C18, 2.1×30 mm, 2.5 μm
Column Supplier: Waters
















Gradient/
% Sol





Solvent
[H2O,





Time
0.1%
% Sol
Flow
Temp


[min]
TFA]
[ACN]
[mL/min]
[° C.]



















0.00
99
1
1.5
60


0.02
99
1
1.5
60


1.00
0
100
1.5
60


1.10
0
100
1.5
60









Method Name: C
Column: Chromolith Flash RP-18e 25-2 mm
Column Supplier: Merck
















Gradient/
% Sol
% Sol




Solvent
[H2O,
[ACN,




Time
0.0375%
0.018%
Flow
Temp


[min]
TFA]
TFA]
[mL/min]
[° C.]



















0.00
95
5
1.5
40


0.70
5
95
1.5
40


1.15
5
95
1.5
40


1.16
95
5
1.5
40


1.60
5
95
1.5
40









Method Name: D
Column: XBridge BEH Phenyl, 2.1×30 mm, 1.7 μm
Column Supplier: Waters



















Gradient/
% Sol






Solvent
[H2O,






Time
0.1%
% Sol
Flow
Temp



[min]
NH3]
[Acetonitril]
[mL/min]
[° C.]






















0.00
95
5
1.3
60



0.02
95
5
1.3
60



1.00
0
100
1.3
60



1.10
0
100
1.3
60










Method Name: E
Column: XBridge C18, 4.6×30 mm, 3.5 μm
Column Supplier: Waters
















Gradient/
% Sol





Solvent
[H2O,





Time
0.1%
% Sol
Flow
Temp


[min]
NH3]
[ACN]
[mL/min]
[° C.]



















0.00
97
3
5
60


0.02
97
3
5
60


1.60
0
100
5
60


1.70
0
100
5
60









Method Name: F
Column: XBridge C18, 3×30 mm, 2.5 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol
% Sol
Flow
Temp


Time [min]
[H2O, 0.1% NH3]
[ACN]
[mL/min]
[° C.]



















0.00
97
3
2.2
60


0.02
97
3
2.2
60


1.20
0
100
2.2
60


1.25
0
100
3
60


1.40
0
100
3
60









Method Name: H
Column: Sunfire C18, 2.1×30 mm, 2.5 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol
% Sol
Flow
Temp


Time [min]
[H2O, 0.1% TFA]
[ACN]
[mL/min]
[° C.]



















0.00
99
1
1.3
60


0.02
99
1
1.3
60


1.00
0
100
1.3
60


1.10
0
100
1.3
60









Method Name: J
Column: Acquity BEH C18, 2.1×50 mm, 1.7 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol
% Sol [ACN,
Flow
Temp


Time [min]
[H2O, 0.1% TFA]
0.1% TFA]
[mL/min]
[° C.]



















0.0
3.0
97.0
0.6
25° C.


0.4
3.0
97.0
0.6
25° C.


3.2
98.0
2.0
0.6
25° C.


3.8
98.0
2.0
0.6
25° C.


4.2
3.0
97.0
0.6
25° C.


4.5
3.0
97.0
0.6
25° C.









Method Name: K
Column: Acquity BEH C18, 2.1×50 mm, 1.7 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol [H2O,
% Sol [ACN,
Flow
Temp


Time [min]
0.1% TFA]
0.1% TFA]
[mL/min]
[° C.]



















0.0
97.0
3.0
0.55
35


0.4
97.0
3.0
0.55
35


3.2
2.0
98.0
0.55
35


3.8
2.0
98.0
0.55
35


4.2
2.0
98.0
0.55
35


4.5
97.0
3.0
0.55
35









Method Name: L
Column: XBridge BEH C18, 2.1×30 mm, 1.7 μm
Column Sunnlier: Waters
















Gradient/Solvent
% Sol
% Sol
Flow
Temp


Time [min]
[H2O, 0.1% NH3]
[ACN]
[mL/min]
[° C.]



















0.00
95
5
1.3
60


0.02
95
5
1.3
60


1.00
0
100
1.3
60


1.10
0
100
1.3
60









Method Name: M
Column: BEH C18 1.7 μm 2.1×50 mm
Column Supplier: Waters

















% Sol
% Sol




Gradient/
[90% H2O + 10%
[90%




Solvent
ACN + NH4COOH
ACN +
Flow
Temp


Time [min]
5 mM]
10% H2O]
[mL/min]
[° C.]



















0.00
100
0
0.7
35


1.20
0
100
0.7
35


1.45
0
100
0.7
35


1.55
100
0
0.7
35


1.75
100
0
0.7
35









Method Name: N
Column: Xselect CSH, 2.5 μm, 4.6×50 mm
Column Supplier: Waters

















% Sol
% Sol




Gradient/
[90% H2O + 10%
[90% ACN +




Solvent
ACN + 0.1%
10% H2O +
Flow
Temp


Time [min]
HCOOH]
0.1% HCOOH]
[mL/min]
[° C.]



















0.00
100
0
1.4
RT


4.00
0
100
1.4
RT


5.30
0
100
1.4
RT


5.50
100
0
1.4
RT


6.00
100
0
1.4
RT









Method Name: O
Column: Synergi Hydro RP100A, 2.5 μm, 3×50 mm
Column Supplier: Phenomenex

















% Sol
% Sol




Gradient/
[90% H2O + 10%
[90%




Solvent
ACN + 5 mM
ACN +
Flow
Temp


Time [min]
NH4COOH]
10% H2O]
[mL/min]
[° C.]



















0.00
100
0
1.2
RT


4.00
0
100
1.2
RT


5.30
0
100
1.2
RT


5.50
100
0
1.2
RT


6.00
100
0
1.2
RT









Method Name: P
Column: Sunfire C18, 3.0×30 mm, 2.5 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol
% Sol
Flow
Temp


Time [min]
[H2O, 0.1% TFA]
[ACN]
[mL/min]
[° C.]



















0.0
98.0
2.0
2.0
60


1.2
0.0
100.0
2.0
60


1.4
0.0
100.0
2.0
60









Method Name: Q

Column: Atlantis dC18 5 μm 4.6×50 mm, Temp 35° C.


Column Supplier: Waters

















% Sol
% Sol




Gradient/
[90% H2O + 10%
[90%




Solvent
ACN + 0.05%
ACN +
Flow
Temp


Time [min]
TFA]
10% H2O]
[mL/min]
[° C.]



















0.00
100
0
1.3
35


0.70
100
0
1.3
35


4.5
0
100
1.3
35


5.80
0
100
1.3
35


6.00
100
0
1.3
35









Method Name: R
Column: Luna-C18 5 μm, 2.0*50 mm
Column Supplier: Phenomenex
















Gradient/
% Sol
% Sol




Solvent
[H2O, 0.0375%
[ACN, 0.018%
Flow
Temp


Time [min]
TFA]
TFA]
[mL/min]
[° C.]



















0.00
99
1
0.8
40


0.40
99
1
0.8
40


3.40
0
100
0.8
40


3.85
0
100
0.8
40


3.86
99
1
0.8
40


4.50
99
1
0.8
40









Method Name: S
Column: Luna-C18 3 μm, 2.0*30 mm
Column Supplier: Phenomenex
















Gradient/
% Sol
% Sol




Solvent
[H2O, 0.0375%
[ACN, 0.018%
Flow
Temp


Time [min]
TFA]
TFA]
[mL/min]
[° C.]



















0.00
90
10
1.0
40


1.15
20
80
1.0
40


1.55
20
80
1.0
40


1.56
90
10
1.0
40


2.00
90
10
1.0
40









Method Name: T
Column: Luna-C18 3 μm, 2.0*30 mm
Column Supplier: Phenomenex
















Gradient/
% Sol
% Sol




Solvent
[H2O, 0.0375%
[ACN, 0.018%
Flow
Temp


Time [min]
TFA]
TFA]
[mL/min]
[° C.]



















0.00
100
0
1.0
40


1.60
40
60
1.0
40


1.65
40
60
1.0
40


1.66
100
0
1.0
40


2.20
100
0
1.0
40









Method Name: U
Column: XBridge BEH C18, 2.1×30 mm, 1.7 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol
% Sol
Flow
Temp


Time [min]
[H2O, 0.1% TFA]
[ACN]
[mL/min]
[° C.]



















0.00
99
1
1.3
60


0.02
99
1
1.3
60


1.00
0
100
1.3
60


1.10
0
100
1.3
60









Method Name: V
Column: Sunfire C18, 3.0×30 mm, 2.5 μm
Column Supplier: Waters
















Gradient/Solvent
% Sol [H2O,
% Sol [ACN,
Flow
Temp


Time [min]
0.1% TFA]
0.08% TFA]
[mL/min]
[° C.]



















0.0
95
5
1.5
60


1.3
0
100
1.5
60


1.5
0
100
1.5
60


1.6
95
5
1.5
60









HPLC traces and NMR spectra of the examples and some advanced intermediates are of increased complexity due to the fact that these compounds exist in an equilibrium of multiple rotameric forms. In the case of multiple peaks in the HPLC spectrum, the retention time of the main peak is reported.


Preparation of Intermediates
Synthesis of Acid Intermediates















Acid
Name
Structure
Reference/Source







A-1 
2-[1,2,3]Triazol-2-yl- benzoic acid


embedded image


WO2008/143856, page 30, Compound B-1





A-2 
5-Methyl-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2008/8518 page 31, compound A-3





A-3 
4-Methyl-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2013/50938, Page 62, Intermediate B1.17





A-4 
3-Methyl-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2011/50200, Page 68-69, Intermediate 37





A-5 
4,5-Dimethyl-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2013/50938, Page 61, Intermediate B1.14





A-6 
3,4-Dimethyl-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2013/68935, Page 58; Intermediate E-20





A-7 
3-Fluoro-2-[1,2,3]triazol- 2-yl-benzoic acid


embedded image


WO2011/50198, Page 47, Intermediate 5





A-8 
4-Chloro-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2011/50198, Page 47, Intermediate 6





A-9 
4-Fluoro-2-[1,2,3]triazol- 2-yl-benzoic acid


embedded image


WO2011/50200, Page 54, Intermediate 16





A-10
5-Fluoro-2-[1,2,3]triazol- 2-yl-benzoic acid


embedded image


WO2011/50198, Page 45-46, Intermediate 1





A-11
4,5-Difluoro-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2013/68935, Page 58; Intermediate E-24





A-12
2-[1,2,3]Triazol-2-yl-5- trifluoromethyl-benzoic acid


embedded image


WO2012/85852, Page 50, Intermediate 37





A-13
5-Methoxy-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2011/50198, Page 49, Intermediate 10





A-14
5-Methyl-2-pyrazol-1-yl- benzoic acid


embedded image


WO2013/50938, Page 62, Intermediate B1.21





A-15
5-Fluoro-2-pyrazol-1-yl- benzoic acid


embedded image


commercially available from Matrix Scientific: catalog number 053712, MDL number MFCD09054728





A-16
2-(3-Methyl- [1,2,4]oxadiazol-5-yl)- benzoic acid


embedded image


commercially available from Apollo Scientific catalog number OR2076, MDL number: MFCD08741426





A-17
2-[1,2,3]Triazol-2-yl-4- trifluoromethyl-benzoic acid


embedded image


WO2013/68935, Page 58; Intermediate E-21





A-18
2-Pyrazol-1-yl-benzoic acid


embedded image


J. Am. Chem. Soc., 1958, 80, 6271





A-19
2-Pyrrol-1-yl-benzoic acid


embedded image


e.g. J. Chem. Soc., Perkin Trans. 1, 1980, 97; commercially available from Alfa Aesar reference L06069, registry number 10333-68-3 MDL number MFCD00051645





A-20
4-Methoxy-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2011/50198, Page 73-74, Intermediate 73





A-21
3-Fluoro-2-pyrimidin-2- yl-benzoic acid


embedded image


WO2011/50200, page 78, Intermediate 52





A-23
2-Oxazol-2-yl-benzoic acid


embedded image


WO2006/76644, Page 212-213, Example 184





A-24
5-Cyano-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2012/85852, Page 50, Intermediate 39





A-25
2-(5-Methyl- [1,3,4]oxadiazol-2-yl)- benzoic acid


embedded image


commercially available from Enamine-BB catalog ID EN300-90215, MDL number MFCD09880459





A-26
4-Fluoro-2-pyrimidin-2- yl-benzoic acid


embedded image


WO2011/50200, page 95, intermediate 85





A-27
2-Fluoro-6-[1,2,3]triazol- 2-yl-benzoic acid


embedded image


WO2012/145581, intermediate 12, page 49-50





A-34
3-Chloro-2- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2013/68935 E-23 in Table 6, p. 57





A-35
2-Fluoro-3-methyl-6- [1,2,3]triazol-2-yl- benzoic acid


embedded image


WO2012/85857 building block 34 p. 57-58









2-Pyrimidin-2-yl-benzoic acid A-22



embedded image


Step 1: A stirred mixture of A-22.1 (30.0 g, 140 mmol), A-22.2 (38.8 g, 154 mmol), Pd(dppf)Cl2.DCM (11.4 g, 14.0 mmol) and potassium acetate (54.7 g, 558 mmol) in dioxane (300 mL) is heated at 100° C. for 2 h. After cooling the reaction is quenched with water and EA and the mixture filtered through a pad of celite. The phases are separated and the organic layer is dried, filtered and concentrated. The residue is purified by flash column chromatography (1-2% EA in hexane) to obtain A-22.3. ESI-MS: 263 [M+H]+; HPLC (Rt): 2.54 min (method J)


Step 2: A stirred mixture of A-22.3 (25.0 g, 95.4 mmol), A-22.4 (12.0 g, 104.9 mmol), Pd(dppf)Cl2.DCM (7.78 g, 9.54 mmol) and sodium carbonate (40.4 g, 381.5 mmol) in 2-methyl tetrahydrofuran (300 mL) and water (150 mL) is heated at 100° C. for 15 h. The reaction is cooled to RT and quenched with water and EA and filtered through a pad of celite. The layers are separated, the organic layer is dried, filtered and concentrated. The residue is purified by flash column chromatography on silica gel (EA/hexane=1:4) to provide 15.0 g of A-22.5. ESI-MS: 215 [M+H]+; HPLC (Rt): 1.42 min (method K)


Step 3: A stirred mixture of A-22.5 (15.0 g, 70.0 mmol) and NaOH (4M aq. solution, 52.5 mL, 210 mmol) in 2-methyltetrahydrofuran (200 mL) is heated at 50° C. for 2 h. The organic solvent is distilled off, the aqueous layer acidified with conc. HCl and extracted with DCM. The combined organic phases are dried, filtered and concentrated. The residue is triturated with n-pentane yielding 5.8 g of A-22. ESI-MS: 201 [M+H]+; HPLC (Rt): 1.39 min (method J)


3,5-Difluoro-2-[1,2,3]triazol-2-yl-benzoic acid A-28



embedded image


Step 1: A mixture of A-28.1 (9.80 g, 53.6 mmol) in water (70 mL) and H2SO4 (25 mL, 475 mmol) are cooled to 0° C. NaNO2 (4.80 g, 69.0 mmol) in H2O (20 mL) is added dropwise and the reaction mixture is stirred for 1.5 h. To this mixture KI (44.5 g, 268 mmol) in H2O (40 mL) is added slowly. The reaction mixture is warmed to RT and then heated to 90° C. for 90 min. The mixture is allowed to cool to RT and Na2S2O3 (aq. solution) is added until the color disappears. The precipitate is filtered and then dissolved in NaOH (4M aq. solution). The mixture is filtered, the filtrate is acidified with HCl (4M aq. solution) and the precipitate is filtered off, washed with water and dried to give 9.0 g of A-28.2. ESI-MS: 285 [M+H]+; HPLC (Rt): 0.71 min (method C)


Step 2: A mixture of A-28.2, (3.50 g, 11.1 mmol), A-28.3 (1.56 g, 22.2 mmol), CuI (0.18 g, 0.89 mmol), A-28.4 (0.70 mL, 4.44 mmol) and K2CO3 (3.46 g, 23.9 mmol) in DMF (10 mL) is heated to 100° C. by microwave for 1.5 h. The mixture is poured into water and extracted with EA, the organic phase is washed with water. The combined aqueous phases are acidified with HCl (0.5M aq. solution) and extracted with EA. The organic phase is washed with brine, dried and concentrated to give the crude product which is purified by HPLC-MS (using a solvent gradient of H2O+0.075% TFA with 5-35% ACN) to provide 1.25 g of A-28. ESI-MS: 226 [M+H]+; HPLC (Rt): 1.88 min (method A)


4-Fluoro-2-(5-methyl-[1,3,4]oxadiazol-2-yl)-benzoic acid A-29



embedded image


Step 1: To a mixture of A-29.1 (2.00 g, 8.40 mmol) in dry DCM (50 mL) is added A-29.2 (0.83 g, 10 mmol) and the reaction mixture is stirred at RT for 1 h. Another portion of A-29.2 (0.83 g, 10 mmol) is added and the reaction is stirred overnight. MeOH (5 mL) is added and the solvent is reduced to half the volume. The precipitate is filtered to provide 0.50 g of A-29.3. The filtrate is concentrated and purified by flash column chromatography on silica gel (using a solvent gradient from 100% DCM to 95% DCM and 5% MeOH) to provide another 1.1 g of A-29.3. ESI-MS: 275 [M+H]+; HPLC (Rt): 0.47 min (method D)


Step 2: To a mixture of A-29.3 (1.60 g, 5.70 mmol) in DCM (50 mL) is added A-29.4 (2.70 g, 11.0 mmol) and the reaction is stirred overnight. Na2CO3 (2M aq. solution) is added, the organic phase is extracted with Na2CO3 (2M aq. solution), the combined organic phases are washed with brine, dried and concentrated to provide 0.80 g of A-29.5. ESI-MS: 257 [M+H]+; HPLC (Rt): 0.47 min (method D)


Step 3: To a mixture of A-29.5 (0.80 g, 3.1 mmol) in dry MeOH (10 mL) and TEA (1.1 mL, 7.5 mmol) is added Pd(dppf)Cl2.DCM (152 mg, 0.19 mmol). The reaction is stirred at 70° C. under a pressure of carbon monoxide (3 bar) for 4 h. The mixture is filtered, concentrated and purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4OH) to provide 0.55 g of compound A29.6. ESI-MS: 237 [M+H]+; HPLC (Rt): 0.88 min (method E)


Step 4: A mixture of A-29.6 (0.55 g, 2.3 mmol) in MeOH (4 mL) and NaOH (4M aq. solution, 2.9 mL, 12 mmol) is stirred at RT for 30 min. The mixture is concentrated and acidified with HCl (4M aq. solution) to pH2. The aqueous phase is extracted with EA, dried and concentrated to provide 0.42 g of compound A-29. ESI-MS: 223 [M+H]+; HPLC (Rt): 0.10 min (method D)


The following examples are prepared in analogy to the above described procedure using the corresponding starting material:





















HPLC






ESI-MS
(Rt)
HPLC


Intermediate
Name
Structure
[M + H]+
[min]
method







A-30
3-Fluoro-2-(5- methyl- [1,3,4]oxadiazol- 2-yl)-benzoic acid


embedded image


223
0.10
D





A-31
4-Methyl-2-(5- methyl- [1,3,4]oxadiazol- 2-yl)-benzoic acid


embedded image


271
0.22
D





A-32
3-Methyl-2-(5- methyl- [1,3,4]oxadiazol- 2-yl)-benzoic acid


embedded image


219
0.10
D









2-(2-Methyl-oxazol-5-yl)-benzoic acid A-33



embedded image


Step 1: To a mixture of A-33.2 (17.00 g, 538 mmol) in DCM (250 mL) is added A-33.3 (16.00 g, 106 mmol) and the mixture is stirred for 1 h. Then A-33.1 (6.5 g, 26 mmol) and A-33.4 (11 g, 264 mmol) are added and the reaction is stirred at 45° C. for 5 h. The pH of the reaction mixture is adjusted to pH8 with NaHCO3 (sat. aq. solution), and the mixture is extracted with DCM and the combined organic layers are dried and concentrated. The residue is purified by flash column chromatography on silica gel (using a solvent gradient of petroleum ether: EA from 40:1 to 20:1) to provide 2.20 g of A-33.5. ESI-MS: 286 [M+H]+; HPLC (Rt): 1.55 (method S)


Step 2: A mixture of A-33.5 (2.20 g, 7.89 mmol), TEA (4.00 g, 40.0 mmol) Pd(dppf)Cl2.DCM (0.58 g, 0.79 mmol) in MeOH (70 mL) is stirred at 50° C. and under an atmosphere of carbon monoxide (50 psi) for 16 h. The mixture is concentrated and purified by flash column chromatography on silica gel (using a solvent gradient of petroleum ether: EA from 80:1 to 40:1) to provide 2.0 g of A-33.6. ESI-MS: 218 [M+H]+; HPLC (Rt): 0.71 min (method C)


Step 3: A mixture of A-33.6 (2.00 g, 9.20 mmol), MeOH (10 mL) and LiOH monohydrate (0.46 g, 11 mmol) is stirred at 25° C. for 16 h. The organic solvent is evaporated, the residue is acidified with HCl (1M aq. solution) to pH3-4. The precipitate is filtered and dried to provide 1.4 g of A-33. ESI-MS: 204 [M+H]+; HPLC (Rt): 2.38 min (method R)


3,4-Difluoro-2-[1,2,3]triazol-2-yl-benzoic acid A-36



embedded image


A mixture of A-36.1 (9.0 g, 36 mmol), A-36.2 (5.3 g, 72 mmol), CuI (0.70 g, 3.6 mmol) and K2CO3 (11 g, 78 mmol) in DMF (100 mL) is heated to 120° C. for 16 h. The mixture is acidified with HCl (0.5M aq. solution) to pH2. The mixture is extracted with EA, the organic phase is washed with brine, dried and concentrated to give the crude product which is purified by HPLC-MS (using a solvent gradient H2O+0.075% TFA with 5-35% ACN) to provide 3.0 g of A-36. ESI-MS: 248 [M+Na]+; HPLC (Rt): 0.45 min (method B)


2-(5-Methyl-[1,2,4]oxadiazol-3-yl)-benzoic acid A-37



embedded image


Step 1: A mixture of NH2OH.HCl (29 g, 0.41 mol) and K2CO3 (57 g, 0.41 mol) in EtOH (500 mL) is stirred at 25° C. for 30 min. A-37.1 (30 g, 0.17 mol) is added and the reaction mixture is heated to 70° C. for 12 h. After filtration, the solvent is evaporated and the residue purified by flash column chromatography on silica gel (petroleum ether/EA=5:1 to 2:1) to obtain 25 g of A-37.2.


Step 2: To a mixture of A-37.2 (18.0 g, 0.084 mol) in ACN (200 mL) is added acetic anhydride (10.0 g; 0.10 mol) and TEA (17.0 g, 0.17 mol). The mixture is stirred at 120° C. for 48 h. The mixture is concentrated in vacuum and the residue purified by flash column chromatography on silica gel (petroleum ether/EA=1/0 to 10/1) to afford 9.0 g of A-37.3. ESI-MS: 239/241 [M+H]+; HPLC (Rt): 1.43 min (method S)


Step 3: To a mixture of A-37.3 (9 g, 0.038 mol) and TEA (11.5 g; 0.114 mol) in MeOH (200 mL) is added Pd(dppf)Cl2.DCM (1.0 g, 1.2 mmol). The mixture is stirred at 50° C. under an atmosphere of carbon monoxide (50 psi) for 16 h. The mixture is concentrated and the residue purified by flash column chromatography on silica gel (petroleum ether/EA=1/0 to 5/1) to afford 4.0 g of A-37.4. ESI-MS: 219 [M+H]+; HPLC (Rt): 1.28 min (method S)


Step 4: To a mixture of A-37.4 (4.0 g, 0.018 mol) in MeOH (40 mL) and H2O (4 mL) is added NaOH (1.5 g, 0.037 mol) at 25° C. under nitrogen. The mixture is stirred at 70° C. for 4 h. The mixture is concentrated. The residue is dissolved in H2O, acidified with HCl (4M aq. solution) to pH3 and filtered to obtain 2.2 g of A-37. ESI-MS: 205 [M+H]+; HPLC (Rt): 2.13 min (method T)


2-(1-Methyl-1H-pyrazol-4-yl)-benzoic acid A-38



embedded image


Step 1: A mixture of A-38.1 (2.0 g, 7.6 mmol), A-38.2 (1.8 g, 8.4 mmol), K2CO3 (1.6 g, 15 mmol), 1c) Pd(dppf)Cl2.DCM (0.28 g, 0.38 mmol) in dioxane (6 mL) and water (3 mL) is heated at 140° C. by microwave for 15 min and then for 25 h at 160° C. The mixture is filtered and concentrated. The crude mixture is purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4OH) to provide 1.32 g of compound 3. ESI-MS: 217 [M+H]+; HPLC (Rt): 0.49 min (method U)


Step 2: A mixture of 1.3 g (6.1 mmol) A-38.3, NaOH (4M aq. solution, 7.5 mL, 30 mmol) in MeOH (7.5 mL) is stirred at RT overnight. The mixture is concentrated, water is added and extracted with DCM/EA. The organic phases are dried and concentrated to provide 750 mg of A-38. ESI-MS: 203 [M+H]+; HPLC (Rt): 0.40 min (method U)


Synthesis of Amine Intermediates
Ethyl-[(S)-1-methyl-2-(5-trifluoromethyl-pyrimidin-2-yloxy)-ethyl]-amine hydrochloride B-1



embedded image


Step 1: A mixture of B-1.1 (5.0 g, 67 mmol) and B-1.2 (6.8 mL, 67 mmol) in 180 mL of THF is stirred at RT for 1 h. Sodium triacetoxyborohydride (45 g, 200 mmol) is added at 0° C. and the reaction mixture is stirred at RT for 30 min. B-1.3 (11.2 mL, 200 mmol) in 20 mL of THF is added dropwise within 10 min at 0° C. and the mixture is stirred at RT overnight. Another portion of B-1.3 (10 mL, 179 mmol) is added and the reaction mixture stirred at RT for 3 h. The precipitate is filtered and washed with THF and DCM. 200 mL of NaHCO3 (sat. aq. solution) and solid NaHCO3 is added to the filtrate until gas formation stopped. The aqueous phase is extracted with DCM, the organic phase is dried and concentrated to provide 11.8 g of B-1.4. ESI-MS: 194 [M+H]+; HPLC (Rt): 1.13 min (method E)


Step 2: To a mixture of B-1.4 (3.5 g, 18 mmol) in dry dioxane (60 mL) under nitrogen is added portion wise potassium tert-butoxide (4.5 g, 40 mmol) and the mixture is stirred for 15 min before B-1.5 (3.6 g, 20 mmol) is added. The mixture is heated to 60° C. for 3 h, stirred overnight at RT and concentrated. Water is added and the aq. phase extracted with EA. The organic phase is dried and concentrated. The crude product is purified by silica column (using a solvent gradient from 100% cyclohexane to 85% cyclohexane and 15% EA) to provide 1.30 g of B-1.6. ESI-MS: 340 [M+H]+; HPLC (Rt): 1.47 min (method M)


Step 3: To a mixture of B-1.6 (1.3 g, 3.5 mmol) in dry 1,2-dichloroethane (15 mL) at 0° C. under nitrogen is added B-1.7 (0.45 mL, 4.1 mmol). The mixture is heated to reflux and stirred for 4 h. MeOH (15 mL) is added and the reaction is heated to 60° C. and stirred for 1 h. The solvent is evaporated and the crude mixture is purified by flash column chromatography on silica gel (using a solvent gradient from 100% DCM to 94% DCM and 6% MeOH+0.6% NH3). The obtained compound is taken up into EA and 1.7 mL of HCl (2M aq. solution) is added under stirring. The solvent is removed and the residue obtained is triturated with Et2O and filtered to give 0.43 g of B-1. ESI-MS: 250 [M+H]+; HPLC (Rt): 3.17 min (method N), 1H NMR (400 MHz, DMSO-d6) δ ppm 1.23 (t, J=7.24 Hz, 3H) 1.36 (d, J=7.04 Hz, 3H) 3.05 (dd, J=7.04, 3.91 Hz, 2H) 3.62-3.72 (m, 1H) 4.52-4.71 (m, 2H) 9.03 (br. s., 2H) 9.12 (d, J=0.78 Hz, 2H)


(S)-2-(tert-Butyl-dimethyl-silanyloxy)-1-methyl-ethylamine B-2



embedded image


To a stirred mixture of B-1.1 (2.00 g, 26.6 mmol) and B-2.1 (4.01 g, 26.6 mmol) in dry DCM (15 mL) a solution of TEA (5.39 g, 53.3 mmol) in dry DCM (15 mL) is added dropwise. The mixture is stirred at RT overnight. NH4Cl (sat. aq. solution) is added and the aqueous phase is extracted with DCM, dried and concentrated to provide 4.7 g of B-2. ESI-MS: 190 [M+H]+; HPLC (Rt): 0.88 min (method M), 1H NMR (500 MHz, DMSO-d6) δ ppm 0.03 (s, 6H), 0.87 (s, 9H), 0.91 (d, J=6.36 Hz, 3H), 2.31 (s (broad), 2H), 2.79 (m, 1H), 3.27-3.35 (m, 2H)


Synthesis of Alcohol Intermediates
(S)-2-Ethylamin-propan-1-ol hydrochloride C-1



embedded image


To a mixture of B-1.4 (9.01 g, 46.6 mmol) in MeOH (200 mL) is added Pd/C (0.90 g). The reaction is stirred at RT and under a pressure of hydrogen (4 bar) for 4 h. The catalyst is filtered off, HCl (4M in dioxane, 14.0 mL, 56.0 mmol) is added and the mixture is concentrated to provide 6.00 g of C-1. ESI-MS: 104 [M+H]+; HPLC (Rt): 0.20 min (method L), 1H NMR (500 MHz, DMSO-d6) δ ppm 1.17-1.22 (m, 3H), 2.94 (dqd, 2H), 3.11-3.20 (m, 1H), 3.51 (dd, 1H), 3.62 (dd, 1H).


N-Ethyl-3-fluoro-N-((S)-2-hydroxy-1-methyl-ethyl)-2-[1,2,3]triazol-2-yl-benzamide C-2



embedded image


Step 1: A mixture of A-7 (1.24 g, 5.99 mmol), thionyl chloride (9.00 mL, 123 mmol) in DMF (0.25 mL) and DCM (7.0 mL) is stirred at RT for 1 h. The mixture is concentrated and concentrated to provide 1.68 g of C-2.1. ESI-MS: 222 [M+H]+; HPLC (Rt): 0.53 min (method H)


Step 2: To a mixture of C-2.1 (1.68 g, 5.96 mmol), TEA (2.09 mL, 14.9 mmol), THF (50 mL) and DCM (20 mL) is added C-1 (0.92 g, 6.55 mmol). The mixture is stirred at RT overnight. The precipitate is filtered off, washed with EA and the filtrate is concentrated. The crude mixture is purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4OH) to provide 1.33 g of C-2. ESI-MS: 291 [M+H]+; HPLC (Rt): 0.46 min (method H)


N-Ethyl-N-((S)-2-hydroxy-1-methyl-ethyl)-2-[1,2,3]triazol-2-yl-benzamide C-3



embedded image


A mixture of A-1 (0.72 g, 3.8 mmol) and thionyl chloride (0.36 mL, 5.0 mmol) in toluene (7 mL) and a drop of DMF is heated to 60° C. for 2 h, then cooled and concentrated. The residue is dissolved in 5 mL of dry DCM and added dropwise to a stirred mixture of C-1 (0.65 g, 4.64 mmol) and TEA (1.44 mL, 10.3 mmol) in dry DCM (10 mL) at 0° C. The reaction is stirred at RT for 2 h. Water is added and the organic layer is separated, washed with citric acid (10% aq. solution) and with NaHCO3 (sat. aq. solution). The organic layer is dried and concentrated to provide 0.94 g of C-3. ESI-MS: 275.1 [M+H]+, HPLC (Rt): 0.69 min (method M)


N-Ethyl-N-((S)-2-hydroxy-1-methyl-ethyl)-4-methoxy-2-[1,2,3]triazol-2-yl-benzamide C-4



embedded image


To a mixture of A-20 (314 mg, 1.43 mmol), C-1 (200 mg, 1.43 mmol) and HATU (0.60 g, 1.58 mmol) in dry DMF (5.0 mL) is added DIPEA (0.75 mL, 4.3 mmol) and the reaction is stirred at RT overnight. The reaction mixture is concentrated and the residue dissolved in EA, the mixture is washed with citric acid (10% aq. solution) and the organic phase is concentrated. The crude product is purified by silica column (using a solvent gradient from 100% DCM to 95% DCM and 5% MeOH) to provide 280 mg of C-4. ESI-MS: 305.2 [M+H]+; HPLC (Rt): 0.77 min (method M)


N-Ethyl-4-fluoro-N-((S)-2-hydroxy-1-methyl-ethyl)-2-pyrimidin-2-yl-benzamide C-5



embedded image


Step 1: A mixture of A-26 (0.20 g, 0.92 mmol), DIPEA (0.31 mL, 1.0 mmol) and C-5.1 (0.32 g, 1.0 mmol) in dry DMF (4.0 mL) is stirred under nitrogen for 30 min. B-2 (0.19 g, 1.0 mmol) is added and the reaction mixture is stirred for 2 h at RT. The crude mixture is poured into water and extracted with EA. The organic phase is dried and concentrated. The residue is purified by flash column chromatography on silica gel (using a solvent gradient from 100% cyclohexane to 40% cyclohexane and 60% EA) to provide 0.22 g of C-5.2. ESI-MS: 389, [M+H]+; HPLC (Rt): 1.38 min (method M)


Step 2: Under nitrogen, a mixture of C-5.2 (0.12 g, 0.31 mmol) and ethyl iodide (96 mg, 0.62 mmol) in dry DMF (2.5 mL) is stirred at 0° C. Potassium tert-butoxide (52 mg, 0.46 mmol) is added and the reaction is stirred at 0° C. for 1 h. Water is added and the crude mixture is extracted with EA. The organic phase is dried and concentrated. The residue is purified by flash column chromatography on silica gel (using a solvent gradient from 100% cyclohexane to 50% cyclohexane and 50% EA) to provide 140 mg (purity 90%) of C-5.3. ESI-MS: 418 [M+H]+; HPLC (Rt): 1.60 min (method M)


Step 3: To a stirred mixture of C-5.3 (140 mg, 0.30 mmol, purity 90%) in dry THF (3.0 mL) at 0° C. is added C-5.4 (595 mg, 0.67 mmol). The reaction is stirred at 0° C. for 1 h and is then concentrated. The residue is purified by flash column chromatography on silica gel (using a solvent gradient from 100% DCM to 90% DCM and 10% MeOH) to provide 80 mg of C-5. ESI-MS: 304 [M+H]+; HPLC (Rt): 0.75 min (method M)


Preparation of Compounds of the Present Invention
Example 1



embedded image


To a mixture of C-2 (40 mg, 0.14 mmol) in dry DMF (2 mL) is added NaH (60% in mineral oil, 7.0 mg, 0.17 mmol) and the mixture is stirred at RT for 1 h. 1.1 (60 mg, 0.33 mmol) is added and the mixture is stirred for 4 h, then diluted with MeOH, filtered and the crude product is purified by HPLC-MS (using a solvent gradient H2O/ACN with TFA) to provide 14 mg of Example 1. ESI-MS: 439 [M+H]+; HPLC (Rt): 1.14 min (method E)


The following examples are prepared in analogy to the above described procedure: For the synthesis of Examples 24 and 30 the work-up is done as follows: The reaction mixture is diluted with water and extracted with EA. The organic phase is dried and concentrated and then purified by HPLC-MS or flash column chromatography on silica gel.



















HPLC





ESI-MS
(Rt)
HPLC


Example
Structure
[M + H]+
[min]
method







24


embedded image


423 (M + Na)+
4.37
Q





30


embedded image


473 (M + Na)+
3.54
N





32


embedded image


472 (M + Na)+
3.29
N









Example 2



embedded image


To a mixture of A-5 (2.2 mg, 0.010 mmol) in ACN (0.10 mL) is added a mixture of B-1 (2.86 mg, 0.010 mmol) and DIPEA (5.19 μl, 0.030 mmol) in ACN (0.10 mL). Then CIP (3.62 mg, 0.010 mmol) in ACN (0.10 mL) is added and the reaction mixture is shaken at RT for 4 h. DMF (100 μL) and K2CO3 (3M aq. solution, 15 μL) is added and the mixture is shaken for 1 h. The mixture is then filtered through a pad of basic aluminium oxide, washed with DMF/MeOH (9/1) and concentrated to provide 4.8 mg of Example 2. ESI-MS: 449 [M+H]+; HPLC (Rt): 0.93 min (method P)


The following examples are prepared in analogy to the above described procedure using the corresponding acid (see Acid Intermediates) and amine (see Amine Intermediates) as described before.





















HPLC





ESI-MS
(Rt)
HPLC


Example
Structure
[M + H]+
[min]
method





 5


embedded image


435
0.88
P





 7


embedded image


439
0.86
P





 8


embedded image


434
0.87
P





10


embedded image


449
0.90
P





11


embedded image


435
0.87
P





15


embedded image


436
0.84
P





18


embedded image


454
0.76
P





19


embedded image


450
0.80
P





20


embedded image


439
0.85
P





21


embedded image


450
0.79
P





22


embedded image


457
0.86
P





34


embedded image


475.2 [M + Na]+
1.00
V





35


embedded image


479.2 [M + Na]+
0.93
V





36


embedded image


443.2 [M + Na]+
0.86
V







ESI
HPLC





pos.
(Rt)
HPLC


Example
Structure
(Loop-Inj.)
[min]
method





13


embedded image


435
1.08
F





14


embedded image


451
1.06
F









Example 31



embedded image


To a mixture of A-33 (43 mg, 0.21 mmol) in dry DMF (4.0 mL) 31.1 (120 mg, 0.32 mmol) and DIPEA (182 μL, 1.05 mmol) is added. After 10 min B-4 (60 mg, 0.21 mmol) is added and the mixture is stirred at RT for 4 h. The mixture is treated with water, washed with NH4Cl (aq. solution), dried and concentrated. The crude mixture is purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4COOH) to provide 35 mg of Example 31. ESI-MS: 435 [M+H]+; HPLC (Rt): 4.48 min (method O)


The following examples are prepared in analogy to the above described procedure using the corresponding acid (see Acid Intermediates) and amine (see Amine Intermediates) as described before.



















HPLC





ESI-MS
(Rt)
HPLC


Example
Structure
[M + H]+
[min]
method







23


embedded image


458 [M + Na]+
3.41
N









Example 25



embedded image


To a stirred mixture of polymer bound triphenylphosphine (from Aldrich: catalog number 366455, 3.0 mmol/g, 440 mg, 1.32 mmol) in dry THF (6.0 mL) is added 25.1 (162 mg, 0.99 mmol) and 25.2 (200 mg, 0.99 mmol). After 10 min C-5 (100 mg, 0.33 mmol) is added and the reaction mixture is stirred overnight. The reaction mixture is filtered and the filtrate is concentrated. The crude mixture is taken up in EA and washed with citric acid (10% aq. solution), Na2CO3 (aq. solution) and water. The mixture is dried and concentrated. The residue is purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4COOH). The obtained product is purified by flash column chromatography on silica gel (using a solvent gradient from 100% cyclohexane to 40% cyclohexane and 60% EA) to provide Example 25 (25 mg). ESI-MS: 472 [M+H]+; HPLC (Rt): 3.45 min (method N)


Example 28



embedded image


To a mixture of A-22 (25 mg, 0.13 mmol), B-1 (30 mg, 0.11 mmol) and DIPEA (54 μl, 0.32 mmol) in dry ACN (2.0 mL) is added CIP (38 mg, 0.14 mmol). The reaction mixture is stirred at 65° C. for 40 min. The reaction mixture is purified by HPLC-MS (using a solvent gradient H2O/ACN with NH4OH) to provide Example 28 (12 mg) ESI pos.+neg. (Loop-Inj.): 432 [M+H]+; HPLC (Rt): 1.03 min (method F)


Example 29



embedded image


To a mixture of C-2 (2.70 g, 9.24 mmol) and 29.1 (1.78 g, 10.2 mmol) in dry THF (30 mL) is added triphenylphosphine (3.15 g, 12.0 mmol) and 29.2 (2.39 g, 230 mmol) and the reaction mixture is stirred at 60° C. for 2 h. The reaction mixture is concentrated, dissolved in DMF and purified by HPLC-MS (using a solvent gradient H2O/ACN with TFA) to provide Example 29 (2.4 g). ESI pos.+neg. (Loop-Inj.): 449 [M+H]+; HPLC (Rt): 1.01 min (method F)

Claims
  • 1. A compound of formula I
  • 2. The compound according to claim 1, wherein R2 represents hydrogen, fluoro, methyl, or —OCH3; andR3 represents hydrogen, fluoro, methyl, or —OCH3.
  • 3. The compound according to claim 1, wherein R1, R2 and R3 independently represent hydrogen or fluoro.
  • 4. (canceled)
  • 5. The compound according to claim 1, wherein R5 represents —CF3.
  • 6. The compound according to claim 1, wherein Ar represents
  • 7. The compound according to claim 1, wherein Ar represents
  • 8. The compound according to claim 1, selected from the group consisting of
  • 9. (canceled)
  • 10. A pharmaceutical composition comprising the compound according to claim 1 or a pharmaceutically acceptable salt thereof in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.
  • 11. A method of treating a psychiatric or neurological condition associated with impulse control deficits, the method comprising administering a compound according to claim 1 or a pharmaceutically acceptable salt thereof, to a patient in need thereof.
  • 12. (canceled)
  • 13. A pharmaceutically acceptable salt of the compound according to claim 1.
Priority Claims (1)
Number Date Country Kind
16165520.4 Apr 2016 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2017/058312 4/17/2017 WO 00