CYCLIC ETHER DERIVATIVES OF PYRAZOLO[1,5-A]PYRIMIDINE-3-CARBOXYAMIDE

Information

  • Patent Application
  • 20200039991
  • Publication Number
    20200039991
  • Date Filed
    October 14, 2019
    4 years ago
  • Date Published
    February 06, 2020
    4 years ago
Abstract
The invention relates to Spirocyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide of general formula (I) which are inhibitors of phosphodiesterase 2, useful in treating central nervous system diseases and other diseases.
Description
FIELD OF THE INVENTION

The invention relates to cyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide of general formula (I) which are inhibitors of phosphodiesterase 2, useful in treating central nervous system diseases and other diseases.


In addition, the invention relates to processes for preparing pharmaceutical compositions as well as processes for manufacture the compounds according to the invention.


BACKGROUND OF THE INVENTION

Phosphodiesterase 2 (PDE2) inhibitors are promising therapeutic targets for treatment of cognitive impairment in diseases such as Schizophrenia, Alzheimer's disease and depression. Inhibitors of PDE2 have emerged as potential candidates to improve synaptic plasticity and memory function.


Phosphodiesterases (PDE) are expressed in nearly all mammalian cells. To date eleven families of phosphodiesterases have been identified in mammals. It is well established that PDEs are critically involved in cell signalling. Specifically, PDEs are known to inactivate the cyclic nucleotides cAMP and/or cGMP.


PDE2 hydrolyses both, cGMP and cAMP. It is both abundantly expressed in the brain indicating their relevance in CNS function.


The expression of PDE2 in the hippocampus, the cortex and in the striatum indicate an involvement in the mechanism of learning and memory/cognition. This is further supported by the fact that increased levels of both cGMP and cAMP are involved in the process of short and long term potentiation (LTP) forming. Further data support the procognitive effect of PDE2 and a synergistic effect of PDE2 on cognition. Furthermore, the expression of PDE2 in the nucleus accumbens (part of the striatum), the olfactory bulb, the olfactory tubercle and the amygdala supports additional involvement of PDE2 in the pathophysiology of anxiety and depression. This is supported by in vivo studies.


It is commonly accepted (free drug hypothesis) that unbound or free drug concentration at the site of action is responsible for pharmacological activity in vivo at steady state and, in the absence of active transport, the free drug concentration is the same in any biomembrane.


For drugs with an intended action in the central nervous system (CNS), it is assumed that unbound drug in interstitial spaces (ISF) in the brain is in direct contact or in equilibrium with the site of action. Because cerebrospinal fluid (CSF) is in direct contact with the brain tissue, it is assumed to readily equilibrate with brain interstitial fluid concentration so that CSF concentration is used as a common surrogate measure for drug unbound concentration in pre-clinical pharmacology studies. Accordingly, for compounds with an intended action in the central nervous system it is important that they reach a high CSF concentration and a high CSF to plasma ratio in order to have high pharmacological activity in the CNS.


At steady state and in the absence of active transport, the unbound brain concentration can also be estimated with the experimentally more accessible unbound plasma concentration by measuring the plasma protein binding (PPB) across species.


High membrane permeability and absence of active transport process at the BBB (blood brain barrier) together with plasma/brain tissue binding are recognised as the primary determinant of drug disposition within CNS.


High metabolic stability is desirable in order to achieve significant exposure of a drug within the body.


Several families of PDE2 inhibitors are known. Imidazotriazinones are claimed in WO 2002/068423 for the treatment of e.g. memory deficiency, cognitive disorders, dementia and Alzheimer's disease. Oxindoles are described in WO 2005/041957 for the treatment of dementia. Further inhibitors of PDE2 are known from WO 2007/121319 for the treatment of anxiety and depression, from WO 2013/034761, WO 2012/104293 and WO2013/000924 for the treatment of neurological and psychiatric disorders, from WO 2006/072615, WO 2006/072612, WO 2006/024640 and WO 2005/113517 for the treatment of arthritis, cancer, edema and septic shock, from WO 2005/063723 for the treatment of renal and liver failure, liver dysfunction, restless leg syndrome, rheumatic disorders, arthritis, rhinitis, asthma and obesity, from WO 2005/041957 for the treatment of cancer and thrombotic disorders, from WO 2006/102728 for the treatment of angina pectoris and hypertension from WO 2008/043461 for the treatment of cardiovascular disorders, erectile dysfunction, inflammation and renal failure and from WO 2005/061497 for the treatment of e.g. dementia, memory disorders, cancer and osteoporosis.


Benzodiazepine like PDE2 inhibitors are described in WO 2005/063723 for the general treatment of CNS diseases including anxiety, depression, ADHD, neurodegeneration, Alzheimer's disease and psychosis.


Newer PDE2 inhibitor families are described in WO 2015/096651, WO 2015/060368 and WO 2015/012328.


Aim of the Invention

It has now been found that compounds of the present invention according to general formula (I) are effective inhibitors of phosphodiesterase 2.


Besides the inhibition property toward phosphodiesterase 2 enzymes, the compounds of the present invention provide further advantageous properties such as high selectivity with regard to PDE 10, low plasma protein binding across species, high CSF to plasma ratio, adequate tissue permeability and high metabolic stability.


For example the compounds of the present invention show low plasma protein binding across species and as a consequence high fraction unbound in plasma, high concentration in cerebrospinal fluid (CSF) and have a high CSF to plasma ratio, which translates in lower efficacious doses of the compounds for disease treatment and as a consequence in further potential advantages such as minimization of side effects. Furthermore, compounds of the present inventions show good metabolic stability both in rodents and non rodents species, good membrane permeability with no active transport at the BBB. In addition the compounds of the present invention have very high IC50 values for PDE 10.


Accordingly, one aspect of the invention refers to compounds according to formula (I), or salts thereof as inhibitors of phosphodiesterase 2.


Another aspect of the invention refers to compounds according to formula (I), or pharmaceutically acceptable salts thereof as inhibitors of phosphodiesterase 2 and reaching high concentrations in cerebrospinal fluid (CSF) and/or having high CSF to plasma ratio.


Another aspect of the invention refers to compounds according to formula (I), or pharmaceutically acceptable salts thereof as inhibitors of phosphodiesterase 2 with low plasma protein binding and thus high fraction unbound across species.


Another aspect of the invention refers to compounds according to formula (I), or pharmaceutically acceptable salts thereof as inhibitors of phosphodiesterase 2 and showing good membrane permeability and low to moderate in vitro efflux.


Another aspect of the invention refers to according to formula (I), or pharmaceutically acceptable salts thereof as inhibitors of phosphodiesterase 2 and showing good metabolic stability.


In a further aspect this invention relates to pharmaceutical compositions, containing at least one compound according to formula (I), or pharmaceutically acceptable salts thereof, optionally together with one or more inert carriers and/or diluents.


A further aspect of the present invention relates to compounds according to formula (I), or pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula (I), or pharmaceutically acceptable salts thereof for the use in the prevention and/or treatment of disorders associated with PDE2 hyperactivity and/or cAMP and/or cGMP hypofunction.


Another aspect of the invention relates to processes of manufacture of the compounds of the present invention.


A further aspect of the present invention relates to compounds according to formula (I), or pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula (I), or pharmaceutically acceptable salts thereof for the use in the prevention and/or treatment of diseases or conditions which can be influenced by inhibition of PDE2 hyperactivity and/or cAMP and/or cGMP hypofunction, such as (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood affective disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) including Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders; (17) obsessive-compulsive disorders; (18) depression; (19) neuropsychiatric symptoms (e.g. depressive symptoms in Alzheimer's disease); (20) mixed dementia; (21) cognitive impairment in schizoaffective disorder; (22) cognitive impairment in bipolar disorder and (23) cognitive impairment in major depressive disorder.


In addition, the compounds of the present invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning, attention or memory.


In addition, the compounds of the present invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis.


In addition, the compounds of the present invention can be used for the treatment of Alzheimer's disease.


In addition compounds of the present invention can be used for the treatment of pain disorders, including but not limited to inflammatory, neuropathic and osteoarthritic pain.


In addition, the compounds of the present invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.


Other aims of the present invention will become apparent to the skilled man directly from the foregoing and following remarks.







DETAILED DESCRIPTION

In a first aspect the present invention relates to compounds of general formula (I)




embedded image


wherein


A is selected from the group Aa consisting of




embedded image




    • wherein above mentioned groups are substituted with one R5 and one R6;





R1 is selected from the group R1a consisting of

    • halogen, C1-3-alkyl- and C3-6-cycloalkyl-
      • wherein the above mentioned C1-3-alkyl-, and C3-6-cycloalkyl- groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, NC— and HO—;


R2 is selected from the group R2a consisting of

    • aryl and heteroaryl,
      • wherein the above mentioned aryl and heteroaryl-groups may optionally be substituted with 1 to 5 substituents R4;


R3 is selected from the group R3a consisting of

    • H— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-groups may optionally be substituted with 1 to 7 substituents independently from each other selected from the group consisting of halogen;


R4 is independently from each other selected from the group R4a consisting of

    • halogen, NC—, HO—, C1-4-alkyl- and C1-3-alkyl-O—
      • wherein the above mentioned C1-4-alkyl- and C1-3-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO— and F—;


R5 is selected from the group R5a consisting of

    • H—, halogen, NC—, HO— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-group may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO— and F—
      • or R5 and R6 together form an group O═;


R6 is selected from the group R6a consisting of

    • H—, halogen, NC—, HO— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-group may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO— and F—
      • or R5 and R6 together form a group O═;


or a salt thereof.


Unless otherwise stated, the groups, residues, and substituents, particularly R1, R2, R3, R4 and R5 are defined as above and hereinafter. If residues, substituents, or groups occur several times in a compound they may have the same or different meanings. Some preferred meanings of groups and substituents of the compounds according to the invention will be given hereinafter.


In a further embodiment of the present invention


A is selected from the group Ab consisting of




embedded image




    • wherein above mentioned groups are substituted with with one R5 and one R6.





In a further embodiment of the present invention


A is selected from the group Ac consisting of




embedded image


In a further embodiment of the present invention


A is selected from the group Ad consisting of




embedded image


In a further embodiment of the present invention


A is selected from the group Ae consisting of




embedded image


In a further embodiment of the present invention


R1 is selected from the group R1b consisting of

    • F—, Cl—, C1-3-alkyl- and C3-6-cycloalkyl-,
      • wherein the above mentioned C1-3-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of F—.


In a further embodiment of the present invention


R1 is selected from the group R1c consisting of

    • F—, H3C— and cyclopropyl-.


In a further embodiment of the present invention


R1 is selected from the group R1d consisting of

    • H3C— and cyclopropyl-.


In a further embodiment of the present invention


R2 is selected from the group R2b consisting of

    • quinolinyl, phenyl and pyridynyl,
      • wherein the above mentioned quinoline, phenyl and pyridyl-groups may optionally be substituted with 1 to 5 substituents R4.


In a further embodiment of the present invention


R2 is selected from the group R2C consisting of

    • phenyl and pyridyl,
      • wherein the above mentioned phenyl and pyridyl-groups may optionally be substituted with 1 to 2 substituents R4.


In a further embodiment of the present invention


R2 is selected from the group R2d being




embedded image


In a further embodiment of the present invention


R2 is selected from the group R2e being




embedded image


In a further embodiment of the present invention


R2 is selected from the group R2f being




embedded image


In a further embodiment of the present invention


R3 is selected from the group R3b consisting of

    • H—, H3C—, F3C—, F2HC—, FH2C— and F3C—.


In a further embodiment of the present invention


R3 is selected from the group R3c consisting of

    • H— and H3C—.


In a further embodiment of the present invention


R3 is selected from the group R3d being H—.


In a further embodiment of the present invention


R4 is independently from each other selected from the group R4b consisting of

    • halogen, C1-4-alkyl- and C1-3-alkyl-O—
      • wherein the above mentioned C1-4-alkyl- and C1-3-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and F—.


In a further embodiment of the present invention


R4 is independently from each other selected from the group R4C consisting of

    • halogen, C1-3-alkyl-, F3C—O—, F2HC—O—, FH2C—O— and H3C—O—,
      • wherein the above mentioned C1-3-alkyl-groups may optionally be substituted with 1 to 5 F—.


In a further embodiment of the present invention


R4 is independently from each other selected from the group R4d consisting of

    • F, Cl, Br, F3C—, F2HC—, FH2C—, H3C—, F3C—O—, F2HC—O—, FH2C—O— and H3C—O—.


In a further embodiment of the present invention


R4 is independently from each other selected from the group R4e consisting of

    • F, Cl, F3C—, F3C—O— and H3C—O—.


In a further embodiment of the present invention


R4 is independently from each other selected from the group R4f consisting of

    • F and F3C—.


In a further embodiment of the present invention


R5 is selected from the group R5b consisting of

    • H—, HO— and C1-2-alkyl-,
      • wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,
      • or R5 and R6 together form an group O═.


In a further embodiment of the present invention


R5 is selected from the group R5C consisting of

    • H— and HO—.


In a further embodiment of the present invention


R5 is selected from the group R5d being

    • HO—.


In a further embodiment of the present invention


R6 is selected from the group R6b consisting of

    • H— and C1-2-alkyl-,
      • wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,
    • or R5/R6 together form a group O═.


In a further embodiment of the present invention


R6 is selected from the group R6c consisting of

    • H and H3C—,
      • wherein the above mentioned methyl-group may optionally be substituted with 1 to 3 F—.


In a further embodiment of the present invention


R6 is selected from the group R6d consisting of

    • H— and H3C—.


Each Ax, R1x, R2x, R3x, R4x, R5x and R6x represents a characterized, individual embodiment for the corresponding substituent as described above. Thus given the above definitions, individual embodiments of the first aspect of the invention are fully characterized by the term (Ax, R1x, R2x, R3x, R4x, R5x and R6x), wherein for each index x an individual figure is given that ranges from “a” to the highest letter given above. All individual embodiments described by the term in parentheses with full permutation of the indices x, referring to the definitions above, shall be comprised by the present invention.


The following Table 1 shows such embodiments E-1 to E-39 of the invention that are considered preferred. Embodiment E-39, represented by the entries in the last row of Table 1, is the most preferred embodiment.









TABLE 1







Embodiments E-1 to E-39 of the invention















Ax
R1x
R2x
R3x
R4x
R5x
R6x





E-1
Aa
R1a
R2a
R3a
R4b
R5a
R6a


E-2
Aa
R1a
R2a
R3b
R4b
R5a
R6a


E-3
Aa
R1b
R2b
R3b
R4c
R5a
R6a


E-4
Aa
R1c
R2b
R3b
R4d
R5a
R6a


E-5
Aa
R1c
R2b
R3c
R4e
R5b
R6b


E-6
Ab
R1b
R2b
R3b
R4b
R5a
R6a


E-7
Ab
R1c
R2b
R3c
R4e
R5b
R6b


E-8
Ac
R1c
R2b
R3b
R4b
R5b
R6b


E-9
Ac
R1c
R2b
R3c
R4e
R5b
R6b


E-10
Ac
R1c
R2c
R3b
R4c
R5b
R6b


E-11
Ac
R1d
R2c
R3c
R4d
R5b
R6b


E-12
Ac
R1d
R2c
R3d
R4e
R5b
R6b


E-13
Ac
R1d
R2c
R3d
R4f
R5c
R6c


E-14
Ac
R1d
R2d
R3b
R4c
R5b
R6b


E-15
Ac
R1d
R2d
R3c
R4d
R5b
R6b


E-16
Ac
R1d
R2d
R3c
R4f
R5d
R6d


E-17
Ac
R1d
R2d
R3d
R4e
R5b
R6b


E-18
Ac
R1d
R2d
R3d
R4f
R5c
R6c


E-19
Ac
R1d
R2e
R3b
R4b
R5b
R6b


E-20
Ac
R1d
R2e
R3b
R4b
R5c
R6c


E-21
Ac
R1d
R2e
R3c
R4d
R5b
R6b


E-22
Ac
R1d
R2e
R3c
R4d
R5c
R6c


E-23
Ac
R1d
R2e
R3c
R4e
R5b
R6b


E-24
Ac
R1d
R2e
R3c
R4e
R5c
R6c


E-25
Ac
R1d
R2e
R3d
R4e
R5b
R6b


E-26
Ac
R1d
R2e
R3d
R4e
R5c
R6c


E-27
Ac
R1d
R2f
R3c

R5d
R6d


E-28
Ac
R1d
R2f
R3d

R5d
R6d


E-29
Ad
R1c
R2c
R3b
R4d
R5b
R6b


E-30
Ad
R1c
R2d
R3b
R4e
R5c
R6c


E-31
Ad
R1c
R2e
R3c
R4e
R5d
R6d


E-32
Ad
R1d
R2f
R3c

R5d
R6d


E-33
Ae
R1c
R2c
R3b
R4d
R5b
R6b


E-34
Ae
R1c
R2d
R3b
R4e
R5c
R6c


E-35
Ae
R1c
R2e
R3c
R4e
R5d
R6d


E-36
Ae
R1c
R2f
R3b

R5b
R6b


E-37
Ae
R1d
R2f
R3c

R5c
R6c


E-38
Ae
R1d
R2f
R3c

R5d
R6d


E-39
Ae
R1d
R2f
R3d

R5d
R6d









Accordingly, for example E-1 covers compounds of formula (I),


wherein


A is selected from the group Aa consisting of




embedded image




    • wherein above mentioned groups are substituted with one R5 and one R6;





R1 is selected from the group R1a consisting of

    • halogen, C1-3-alkyl- and C3-6-cycloalkyl-
      • wherein the above mentioned C1-3-alkyl-, and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, NC— and HO—;


R2 is selected from the group R2a consisting of

    • aryl and heteroaryl,
      • wherein the above mentioned aryl and heteroaryl-groups may optionally be substituted with 1 to 5 substituents R4;


R3 is selected from the group R3a consisting of

    • H— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-groups may optionally be substituted with 1 to 7 substituents independently from each other selected from the group consisting of halogen;


R4 is independently from each other selected from the group R4b consisting of

    • halogen, C1-4-alkyl- and C1-3-alkyl-O—
    • wherein the above mentioned C1-4-alkyl- and C1-3-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and F—;


R5 is selected from the group R5a consisting of

    • H—, halogen, NC—, HO— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-group may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO— and F—
      • or R5 and R6 together form an group O═;


R6 is selected from the group R6a consisting of

    • H—, halogen, NC—, HO— and C1-3-alkyl-,
      • wherein the above mentioned C1-3-alkyl-group may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO— and F—
      • or R5 and R6 together form a group O═;


or a salt thereof.


Accordingly, for example E-5 covers compounds of formula (I),


wherein


A is selected from the group Aa consisting of




embedded image




    • wherein above mentioned groups are substituted with one R5 and one R6;





R2 is selected from the group R2b consisting of

    • quinolinyl, phenyl and pyridynyl,
      • wherein the above mentioned quinoline, phenyl and pyridyl-groups may optionally be substituted with 1 to 5 substituents R4;


R3 is selected from the group R3c consisting of

    • H— and H3C—;


R4 is independently from each other selected from the group R4e consisting of

    • F, Cl, F3C—, F3C—O— and H3C—O—;


R5 is selected from the group R5b consisting of

    • H—, HO— and C1-2-alkyl-,
      • wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,
    • or R5 and R6 together form an group O═:


R6 is selected from the group R6b consisting of

    • H— and C1-2-alkyl-,
      • wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,
    • or R5/R6 together form a group O═;


or a salt thereof.


Accordingly, for example E-39 covers compounds of formula (I), wherein


A is selected from the group Ae consisting of




embedded image


R1 is selected from the group R1d consisting of

    • H3C— and cyclopropyl-;


R2 is selected from the group R2f being




embedded image


R3 is selected from the group R3d being H—;


R5 is selected from the group R5d being

    • HO—;


R6 is selected from the group R6d consisting of

    • H— and methyl-;


or a salt thereof.


Further preferred are the following compounds listed in Table 2:













No.
Structure







I


embedded image







II


embedded image







III


embedded image







IV


embedded image







V


embedded image







VI


embedded image







VII


embedded image







VIII


embedded image







IX


embedded image







X


embedded image







XI


embedded image







XII


embedded image







XIII


embedded image







XIV


embedded image







XV


embedded image







XVI


embedded image







XVII


embedded image







XVIII


embedded image







XIX


embedded image







XX


embedded image







XXI


embedded image







XXII


embedded image







XXIII


embedded image







XXIV


embedded image







XXV


embedded image







XXVI


embedded image







XXVII


embedded image







XXVIII


embedded image







XXIX


embedded image







XXX


embedded image







XXXI


embedded image







XXXII


embedded image







XXXIII


embedded image







XXXIV


embedded image







XXXV


embedded image







XXXVI


embedded image







XXXVII


embedded image







XXXVIII


embedded image







XXXIX


embedded image







XL


embedded image







XLI


embedded image







XLII


embedded image







XLIII


embedded image







XLIV


embedded image







XLV


embedded image







XLVI


embedded image







XLVII


embedded image







XLVIII


embedded image







XLIX


embedded image







L


embedded image







LI


embedded image







LII


embedded image







LIII


embedded image







LIV


embedded image







LV


embedded image







LVI


embedded image







LVII


embedded image







LVIII


embedded image







LIX


embedded image







LX


embedded image







LXI


embedded image







LXII


embedded image







LXIII


embedded image







LXIV


embedded image







LXV


embedded image







LXVI


embedded image







LXVII


embedded image







LXVIII


embedded image







LXIX


embedded image







LXX


embedded image







LXXI


embedded image







LXXII


embedded image







LXXIII


embedded image







LXXIV


embedded image







LXXV


embedded image







LXXVI


embedded image







LXXVII


embedded image











or the salts thereof.


Some terms used above and hereinafter to describe the compounds according to the invention will now be defined more closely.


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 C1-6-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-C1-3-alkyl-” means an aryl group which is bound to a C1-3-alkyl-group, the latter of which is bound to the core molecule or to the group to which the substituent is attached.


Within the present invention, the term “core molecule” is defined by the following structure:




embedded image


In general, the attachment site of a given residue to another group shall be variable, i.e. any capable atom, bearing hydrogens to be replaced, within this residue may be the linking spot to the group being attached, unless otherwise indicated.


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 may be used in sub-formulas to indicate the bond or attachment point which is connected to the core molecule, rest of the molecule or to the substituent to which it is bound as defined.


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.


The phrase “pharmaceutically acceptable” or “physiologically 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” or “physiologically 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 or physiologically 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, 1H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, 1-(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, benzenesulfonic 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, methanesulfonic 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-pyroglutamic 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.


The term “substituted” as used herein means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's viable valence number is not exceeded, and that the substitution results in a stable compound.


The term “partially unsaturated” as used herein means that in the designated group or moiety 1, 2, or more, preferably 1 or 2, double bonds are present. Preferably, as used herein, the term “partially unsaturated” does not cover fully unsaturated groups or moieties.


The term “halogen” generally denotes fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).


The term “C1-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 C1-5-alkyl embraces the radicals H3C—, H3C—CH2—, H3C—CH2—CH2—, H3C—CH(CH3)—, H3C—CH2—CH2—CH2—, H3C—CH2—CH(CH3)—, H3C—CH(CH3)—CH2—, H3C—C(CH3)2—, H3C—CH2—CH2—CH2—CH2—, H3C—CH2—CH2—CH(CH3)—, H3C—CH2—CH(CH3)—CH2—, H3C—CH(CH3)—CH2—CH2—, H3C—CH2—C(CH3)2—, H3C—C(CH3)2—CH2—, H3C—CH(CH3)—CH(CH3)— and H3C—CH2—CH(CH2CH3)—.


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.


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, phenanthrenyl, tetrahydronaphthyl and dihydronaphthyl.


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


In one embodiment the term “heteroaryl” means a mono- or bicyclic-ring system containing one to three heteroatoms selected from N, O or S(O)r, wherein r=0, 1 or 2, consisting of 5 to 10 ring atoms wherein at least one of the heteroatoms is part of an aromatic ring.


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:




embedded image


embedded image


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.


The compounds according to the invention may be obtained using methods of synthesis known in principle. Preferably, the compounds are obtained by the following methods according to the invention which are described in more detail hereinafter.


Preparation


The following Schemes shall illustrate generally how to manufacture the compounds of the present invention by way of example. The abbreviated substituents may be as defined above if not defined otherwise within the context of the schemes.


The preparation process might comprises:

    • a) Reacting a compound of formula (II)




embedded image


or a derivatives thereof, with a compound of formula (III)




embedded image


Wherein R1, R2, R3, R5, R6 and A are as defined above and L is a suitable leaving group such as halogen atom (e.g. chlorine or bromine) or hydroxyl group.


In case of L=halogen, process a) typically comprises the reaction of a compound of formula (II) with a compound of formula (III) in an appropriate solvent such as acetonitrile or N,N-dimethylformamide in the presence of a base such as TEA or DIPEA at room temperature.


In case of L=OH, process a) typically comprises the reaction of a compound of formula (II) with a compound of formula (III) in an appropriate solvent such as N,N-dimethylformamide and in the presence of a suitable coupling agent (e.g. HATU or TBTU)


Compounds of formula (III) are either commercially available or can be prepared as described in the following Schemes, following known reported procedures.




embedded image


In Scheme 1, Step 1 typically involves reaction of commercially available amino pyrazole derivatives with 2-Bromo-malonaldehyde in the presence of acetic acid in a suitable solvent such as EtOH under heating. In Step 2, the cyclopropyl group is introduced by a cross coupling palladium catalyzed reaction using for example potassium cyclopropyltrifluoroborate, a suitable palladium catalyst such as Palladium(II) acetate and 2-dicyclohexylphosphino-2′,6′-diisopropoxy 1,1′-biphenyl as ligand in an appropriate solvent such as toluene under heating. In Step 3 the ethyl ester is then hydrolyzed under basic conditions using sodium hydroxide or lithium hydroxide monohydrate in an appropriate solvent such as EtOH or a mixture of THF/water.




embedded image


In Scheme 2, Step 1 typically involves reaction of commercially available amino pyrazole derivative with 1,1,3,3,-tetraethoxy-propane in the presence of hydrochloric acid in a suitable solvent such as EtOH under heating. Bromination using bromine in acetic acid as solvent at room temperature provides the bromo derivative and the cyclopropyl group is then introduced as described in Scheme 1.




embedded image


In Scheme 3, Step 1 typically involves reaction of commercially available amino pyrazole derivative with 1,1,3,3,-tetraethoxy-2-methyl-propane in the presence of hydrochloric acid in a suitable solvent such as EtOH under heating. Basic hydrolysis provides the desired carboxylic acid derivative


Compounds of formula (II) are either commercially available or can be prepared as described in the following Schemes.




embedded image


In Scheme 4, R2 is aryl or heteroaryl


In Scheme 4, Step 1, commercially available ketone is converted in the corresponding 2-methyl-propane-2-sulfinyl-imine using titanium (IV) ethoxide and 2-Methyl-propane-2-sulfinic acid amide, as described in WO 2005087751.


The obtained intermediate is then added dropwise to a previously prepared solution of organo lithium derivatives of the appropriate halogen compounds (R2X, where X is of organo lithium derivatives of the appropriate halogen compounds (R2X, where X is bromine or iodine) prepared using for example commercially available solution of tert-butyllithium or n-buthyllitium in hexane or pentane at low temperature (−75° C.) in a suitable solvent such as toluene or THF. Cleavage of the resulting sulphinic amide by treatment with acid such as a 4N solution of HCl in a suitable solvent such as dioxane provides the desired intermediates amines.


The above described synthesis applies also for the analogues with 5 and 4 membered ring, starting from commercially available cyclopentanone and oxetane-3-one.




embedded image


In Scheme 5, R2 is aryl derivatives.


Step 1 involves a cross coupling Suzuky reaction with commercially available boronic acid or pinacol ester derivatives and the appropriate halogen derivatives (X=Br or I) using for example 1,1′-bis(diphenylphosphino)ferrocenedichloro palladium(II) as catalyst, potassium carbonate as base in an appropriate solvent such as a mixture of toluene/water under heating. The epoxidation step is performed using MCPBA as oxidating agent in DCM at room temperature. The desired amino alcohol intermediate is then obtained by opening the epoxide with a modified Ritter procedure using trifluoromethane sulfonic acid and acetonitrile followed by basic hydrolysis of the formed intermediate, in analogy to the procedure described in Tetrahedron Asymmetry, 1996, 5, 1501-1506.


The relative stereochemistry of the above described aminoalcohols is reported in the Experimental description.




embedded image


In Scheme 6, R2 is aryl or heteroaryl.


In Scheme 6, the opening of the epoxide is performed using sodium azide in the presence of ammonium chloride under heating in a suitable solvent, such as dimethyl formamide. After separation of the two regioisomers, (see experimental), the azide group is then converted into amino group by reduction following well known reported procedure such as for example using zinc and ammonium formate in a suitable solvent such as methanol at room temperature.


The relative stereochemistry of the above described amino alcohols is reported in the Experimental description.




embedded image


In Scheme 7, R2 is aryl or heteroaryl


In Scheme 7, the regioisomeric mixture of azide intermediates, obtained following the approach described in Scheme 6, is reduced under catalytic hydrogenation conditions, using for example Pd/C in a suitable solvent such as ethanol and in the presence of di.tert-butyldicarbonate to obtain the protected amino alcohols derivatives. Oxidation to ketone is performed using Dess Martin periodinane in a suitable solvent such as DCM at room temperature or using Swern's procedure Formation of the tertiary alcohols is accomplished by addition of methyl magnesium chloride to the carbonyl group at low temperature (−20° C.) in a suitable solvent such as THF. The cleavage of the Boc protecting group is performed under acid conditions using for example trifluoroacetic acid in a suitable solvent such as DCM at room temperature.


The regioisomeric ratio of epoxide opening and the relative stereochemistry of the above described amino alcohols are reported in the Experimental description.




embedded image


In Scheme 8 R2 is aryl.


In Scheme 8, the desired tetrahydrofuran-3-ol-intermediates are obtained by addition of the appropriate lithium derivatives, prepared reacting a suitable halogen compounds (R2X, X=halogen) with commercially available solution of tert-butyllithium or n-buthyllitium in hexane or pentane at low temperature (−75° C.) in a suitable solvent such as toluene or THF, to the carbonyl group. Treatment with pTsOH in toluene under reflux provides the double bond derivatives which are transformed into the desired aminoalcohols following the approaches described in Scheme 5.


The relative stereochemistry of the aminoalcohols compounds are reported in the Experimental description.




embedded image


In Scheme 9, R2 is aryl or heteroaryl.


The desired amino alcohols are obtained following the approaches described above in Scheme 6 and 7.


The regiochemistry ratio of the epoxide opening and the relative stereochemistry of the aminoalcohols compounds are reported in the Experimental description.


BIOLOGICAL EXAMPLES

In-Vitro Effect:


The in-vitro effect of the active compounds of the invention can be shown with the following biological assays.


a) Phosphodiesterase (PDE) 2A and 10 Assay with Fluorescent Substrate


Assay Principle:


The PDE reaction cleaves cAMP to AMP. The IMAP system (Molecular Device) using fluorescence polarization (FP) as detection principle was used to measure enzyme activity. A fluorescent labeled cAMP was used as substrate for the reaction, generating a labeled AMP. The fluorescent AMP binds specifically to the large M(III)-based nano-particles which reduces the rotational speed of the substrate and thus increases its polarization.


Detailed Method:


The inhibition of PDE 2A or 10 enzyme activity was assessed using IMAP-Phosphodiesterase-cAMP fluorescence labeled substrate (Molecular Devices, Order No. R7506), IMAP TR-FRET screening express (Molecular Devices, Order No. R8160, the TR-FRET component will not be used) and PDE 2A or PDE10 protein expressed upon baculovirus infection in SF9 cells. The cells were incubated after infection for 3 days and protein production was confirmed by Western Blot. The cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min). Since SF 9 cells do not express cAMP hydrolyzing enzymes to a high extent, no further purification of the protein was needed.


All reactions were performed in 384 well plates, Perkin Elmer black optiplates and IMAP reaction buffer with 0.1% Tween20 (kit component)


Compounds were serial diluted in DMSO. With an intermediate dilution step with reaction buffer DMSO concentration was reduced to achieve 1% DMSO in the assay reaction. Setup of the assay started with 10 μl enzyme (˜10 ng/well, depending on prep. batch), 5 μl compound, reaction was started by addition of 5 μl labeled cAMP (30 nM, final concentration), immediately mixed for 15 seconds on a Eppendorf mixmate (2000 rpm) followed by an incubation at room temperature for 90 minutes in the dark. Reaction is stopped by adding of 60 μl binding buffer for FP/cAMP (kit component). After at least 90 min of further incubation (room temperature, dark) the assay was measured at 485 nm excitation/525 nm emission in an Envision multilabel reader (PerkinElmer).


Each assay plate contained wells with vehicle controls (1% DMSO) for the measurement of non-inhibited reaction (=100% control) and wells without enzyme as 0% controls.


The analysis of the data was performed by calculation of the percentage of inhibition in the presence of test compound compared to the vehicle control samples (100% control, no inhibition) and a low control (0% control, no enzyme).


IC50 values are calculated with Assay Explorer or other suited software based on curve fitting of results of at least 8 different compound concentrations. The compound concentrations may vary according to the needed range, but typically cover the range between 10 μM and 0.1 μM.









TABLE 3a







PDE2A Activity of the examples (Ex) compiled in the experimental part,


based on above described assay (IMAP fluorescent).















PDE2

PDE2

PDE2

PDE2


Ex.
IC50 [nM]
Ex.
IC50 [nM]
Ex.
IC50 [nM]
Ex.
IC50 [nM]

















 1
14
27
534
53
100
81b
149


 2
22
28
834
54
12
82a
1.2


 3
266
29
661
55
21
82b
40


 4
48
30
12
56
229
83a
5.9


 5
258
31
30
57
103
83b
1820


 6
199
32
67
58
14
84a
120


 7
77
33
83
59
60
84b
2790


 8
69
34
27
60
22
85a
130


 9
117
35
31
61
19
86a
3.9


10
80
36
180
62
127
86b
2550


11
297
37
192
63
124
87a
1000


12
1650
38
331
64
496
87b
184


13
359
39
568
65
30
88a
32


14
456
40
84
66
25
88b
3030


15
746
41
291
67
50
89a
1000


16
537
42
130
68
24
90a
200


17
39
43
359
69
1740
90b
14


18
129
44
840
70
1250
91a
63


19
519
45
239
71
13
91b
1590


20
172
46
5.7
72
122
92a
93


21
74
47
240
73
1142
92b
712


22
119
48
1.35
74
2530
93a
11


23
232
49
70
75
42
93b
1520


24
754
50
596
80b
75




25
88
51
59
80a
313




26
174
52
231
81a
3.4
















TABLE 3b







PDE10 Activity of the examples (Ex) compiled in the experimental part,


based on above described assay (IMAP fluorescent).















PDE10

PDE10

PDE10

PDE10



IC50

IC50

IC50

IC50


Ex.
[nM]
Ex.
[nM]
Ex.
[nM]
Ex.
[nM]





 1
  10100
27
>10000
53
>10000
81b
>10000


 2
>10000
28
>10000
54
   9670
82a
>10000


 3
>10000
29
   5930
55
>10000
82b
   8461


 4
   550
30
>10000
56
>10000
83a
>10000


 5
  12200
31
>10000
57
>10000
83b
>10000


 6
>10000
32
>10000
58
>10000
84a
>10000


 7
   9110
33
>10000
59
>10000
84b
>10000


 8
>10000
34
  10800
60
>10000
85a
>10000


 9
>10000
35
>10000
61
   6650
86a
>10000


10
   9820
36
>10000
62
   7160
86b
   9940


11
   1470
37
>10000
63
>10000
87a
>10000


12
>10000
38
   6710
64
>10000
87b
>10000


13
   9910
39
>10000
65
   9760
88a
   7560


14
   8430
40
   5730
66
>10000
88b
>10000


15
>10000
41
   7950
67
>10000
89a
>10000


16
>10000
42
   5590
68
>10000
90a
   8590


17
   6940
43
   6860
69
>10000
90b
   7350


18
   8630
44
   9680
70
>10000
91a
   7700


19
>10000
45
   7850
71
>10000
91b
   5670


20
>10000
46
>10000
72
>10000
92a
>10000


21
>10000
47
>10000
73
>10000
92b
>10000


22
   9920
48
   6620
74
>10000
93a
>10000


23
>10000
49
>10000
75
>10000
93b
>10000


24
>10000
50
>10000
80b
   5280




25
   9070
51
   9040
80a
   7760




26
>10000
52
>10000
81a
>10000









In-Vivo Effect:


Animal Experiments and Sample Analysis (CSF):


Test compounds were administered to animals (rat) different routes at doses of 10.0 or 5 μmol/kg, (both oral and intravenous). CSF samples were carefully collected by puncture of the cisterna magna under anesthaesia. Immediately after CSF sampling, blood was taken by heart puncture and brains were dissected out. Blood was collected in EDTA-coated microvettes and plasma was prepared by centrifugation. Concentration of the test compounds in plasma, CSF or brain homogenate was determined using HPLC-MS-MS.









TABLE 4







Plasma, brain and CSF concentration















conc
conc

conc




Time(*)
plasma
brain
c(brain)/
CSF
c(CSF)/


Ex.
(h)
(nmol/L)
(nmol/L)
c(plasma)
(nmol/L)
c(plasma)





 1
0.5
 243
 471
1.96
 11
0.04


21
0.5
1210
1320
1.17
106
0.09


25
0.5
1040
 957
0.92
111
0.12


81a
0.5
2460
1070
0.42
261
0.10


82a
0.5
3320
1180
0.36
157
0.05


83a
0.5
 794
 449
0.6
 61
0.08





(*)Time between administration and CSF sampling






For the skilled in the art it is evident from the experimental results shown above that compounds of the present invention are not only potent phosphodiesterase 2 inhibitors but also reach high CSF concentrations and adequate CSF to plasma ratios.


Plasma Protein Binding (Determination of Human and Rat Plasma Protein Binding with Equilibrium Dialysis)


This equilibrium dialysis (ED) technique is used to determine the approximate in vitro fractional binding of test compounds to human and rat plasma proteins.


Dianorm Teflon dialysis cells (micro 0.2) are used. Each cell consists of a donor and an acceptor chamber, separated by an ultrathin semipermeable membrane with a 5 kDa molecular weight cutoff.


Stock solutions for each test compound are prepared in DMSO at 1 mM and diluted to a final concentration of 1.0 μM. The subsequent dialysis solutions are prepared in pooled human and rat plasma (with NaEDTA)


Aliquots of 200 μL dialysis buffer (100 mM potassium phosphate, pH 7.4) are dispensed into the buffer chamber. Aliquots of 200 μL test compound dialysis solution are dispensed into the plasma chambers. Incubation is carried out for 2 hours under rotation at 37° C.


At the end of the dialysis period, the dialysate is transferred into reaction tubes. The tubes for the buffer fraction contain 0.2 ml Acetonitril/water (80/20). Aliquots of 25 μL of the plasma dialysate are transferred into deep well plates and mixed with 25 μl Acetonitril/water (80/20), 25 μl buffer, 25 μL calibration solution and 25 μl Internal Standard solution. Protein precipitation is done by adding 200 μl Acetonitrile.


Aliquots of 50 μl of the buffer dialysate are transferred into deep well plates and mixed with 25 μl blank plasma, 25 μl Internal Standard solution and 200 μl Acetonitril.


Samples are measured on HPLC-MS/MS-Systems and evaluated with Analyst-Software.


Percent bound is calculated with the formula: % bound=(plasma concentration−buffer concentration/plasma concentration)×100 and % free is calculated as difference.









TABLE 4







PPB (Plasma Protein Binding) of compounds of the present


invention in human and rat plasma.













PPB HUM
PPB RAT

PPB HUM
PPB RAT


EX
% BINDING
% BINDING
EX
% BINDING
% BINDING















2
91.5
94.7
70
79.4



1
96
96.8
71
81.1



31
95.3
97.6
51
46.9



35
92.4
93.7
53
81.7



37
94.4
93.50
88b
44.2



17
91.4
90.7
82a
78.7
86.5


25
75.4
83.1
81a
63.0
74.3


21
83.8
87.2
46
67.1
68.7


3
84.3

81b
65.3
66.2


68
84.4

83a
84.4
82.2


69
90.7

86a
89.5
93.5









For the skilled in the art it is evident from the experimental results shown above that compounds of the present invention are not only potent phosphodiesterase 2 inhibitors but also have low plasma protein binding.


Assessment of Efflux in Madin-Darby Canine Kidney Cells Transfected with the Human MDR1 Gene (MDCK Assay)


Apparent permeability coefficients (PE) of the compounds across the MDCK-MDR1 cell monolayers are measured (pH 7.4, 37° C.) in apical-to-basal (AB) and basal-to-apical (BA) transport direction. AB permeability (PEAB) represents drug absorption from the blood into the brain and BA permeability (PEBA) drug efflux from the brain back into the blood via both passive permeability as well as active transport mechanisms mediated by efflux and uptake transporters that are expressed on the MDCK-MDR1 cells, predominantly by the overexpressed human MDR1 P-gp. The compounds are assigned to permeability/absorption classes by comparison of the AB permeabilities with the AB permeabilities of reference compounds with known in vitro permeability and oral absorption in the human. Identical or similar permeabilities in both transport directions indicate passive permeation, vectorial permeability points to additional active transport mechanisms. Higher PEBA than PEAB indicates the involvement of active efflux mediated by MDR1 P-gp. Active transport is concentration-dependently saturable.


MDCK-MDR1 cells (1-2×10e5 cells/1 cm2 area) are seeded on filter inserts (Costar transwell polycarbonate or PET filters, 0.4 μm pore size) and cultured (DMEM) for 7 days. Subsequently, the MDR1 expression is boosted by culturing the cells with 5 mM sodium butyrate in full medium for 2 days. Compounds are dissolved in appropriate solvent (like DMSO, 1-20 mM stock solutions). Stock solutions are diluted with HTP-4 buffer (128.13 mM NaCl, 5.36 mM KCl, 1 mM MgSO4, 1.8 mM CaCl2, 4.17 mM NaHCO3, 1.19 mM Na2HPO4×7H2O, 0.41 mM NaH2PO4xH2O, 15 mM HEPES, 20 mM glucose, 0.25% BSA, pH 7.4) to prepare the transport solutions (0.1-300 μM compound, final DMSO<=0.5%). The transport solution (TL) is applied to the apical or basolateral donor side for measuring A-B or B-A permeability (3 filter replicates), respectively. The receiver side contains the same buffer as the donor side. Samples are collected at the start and end of experiment from the donor and at various time intervals for up to 2 hours also from the receiver side for concentration measurement by HPLC-MS/MS or scintillation counting. Sampled receiver volumes are replaced with fresh receiver solution. Efflux ratio is calculated dividing the Papp (b-a) values by the Papp (a-b) values.









TABLE 5







Papp (PEBA) and efflux of compounds of the present invention













Papp (a-b)


Papp (a-b)
efflux


Ex.
mean [10−6 cm/s]
efflux ratio
Ex.
mean [10−6 cm/s]
ratio















1
78
0.6
53
15
1.8


2
84
0.6
82a
59
1.0


37
86
0.5
51
11
5.0


17
85
0.6
81a
60
1.4


25
100
0.8
46
60
1.4


21
94
0.6
81b
64
1.2


3
97
0.7
83a
34
1.4


69
25
1.3
84a
31
2.2


70
20
2.1
84b
23
2.2


71
23
1.9









For the skilled in the art it is evident from the experimental results shown above that compounds of the present invention are not only potent phosphodiesterase 2 inhibitors but also have good membrane permeability and low to moderate in vitro efflux.


Metabolic Stability


The metabolic stability of the compounds according to the invention has been investigated as follows:


The metabolic degradation of the test compound was assayed at 37° C. with pooled liver microsomes from various species. The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at room temperature (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/mL for human and dog, 0.5 mg/mL for other species) and the test compound at a final concentration of 1 μM. Following a short preincubation period at 37° C., the reactions were initiated by addition of betanicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM), and terminated by transferring an aliquot into solvent after different time points. After centrifugation (10000 g, 5 min), an aliquot of the supernatant was assayed by LC10 MS/MS for the amount of parent compound. The half-life was determined by the slope of the semi-logarithmic plot of the concentration-time profile.









TABLE 4







Stability of compounds of the present invention in


human liver microsomes.













Half-life -

Half-life -




t½ [min]

t½ [min]



Ex.
human
Ex.
human
















1
>130
71
>130



2
120
51
>130



37
>130
53
120



17
>130
88b
>130



25
>130
82a
>130



21
>130
81a
>130



3
72
46
>130



68
>130
81b
>130



69
53
83a
>130



70
63
86a
>130










For the skilled in the art it is evident from the experimental results shown above that compounds of the present invention are not only potent phosphodiesterase 2 inhibitors but also have good metabolic stability.


In view of their ability to inhibit the activity of phosphodiesterase 2 activity and their advantageous pharmacokinetics properties the compounds of general formula (I) according to the invention, or the physiologically acceptable salts thereof, are suitable for the treatment and/or preventative treatment of all those diseases or conditions which can be influenced by inhibition of PDE2 hyperactivity and/or cAMP and/or cGMP hypofunction. Therefore, compounds according to the invention, including the physiologically acceptable salts thereof, are particularly suitable for the prevention or treatment of diseases, particularly (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood affective disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) including Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders; (17) obsessive-compulsive disorders; (18) depression; (19) neuropsychiatric symptoms (e.g. depressive symptoms in Alzheimer's disease); (20) mixed dementia; (21) cognitive impairment in schizoaffective disorder; (22) cognitive impairment in bipolar disorder and (23) cognitive impairment in major depressive disorder.


In addition, the compounds of the present invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning, attention or memory.


In addition, the compounds of the present invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis. In addition, the compounds of the present invention can be used for the treatment of Alzheimer's disease.


In addition compounds of the present invention can be used for the treatment of pain disorders, including but not limited to inflammatory, neuropathic and osteoarthritic pain.


In addition, the compounds of the present invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.


Preferably the compounds according to the invention are suitable for the treatment of Alzheimer's Disease and for the treatment schizophrenia.


More preferably the compounds according to the invention are suitable for symptomatic treatment of Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia.


In particular the compounds according to the invention are suitable for symptomatic treatment of prodromal and mild-to-moderate Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia and symptomatic treatment of cognitive impairment associated with 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), or the pharmaceutically acceptable salts thereof, to a human being.


The dose range of the compounds of general formula (I) applicable per day is usually from 0.1 to 1000 mg, preferably from 1 to 500 mg by oral route, in each case administered 1 to 4 times a day.


Each dosage unit may conveniently contain from 0.1 to 500 mg, preferably 1 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.


Suitable preparations for administering the compounds of formula I, including the pharmaceutically acceptable salts thereof, 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) should be 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 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.


For this purpose, the compounds of formula I prepared according to the invention may be formulated, optionally together with other active substances, together with one or more inert conventional carriers and/or diluents, e.g. with corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, citric acid, tartaric acid, water, polyvinylpyrrolidone, water/ethanol, water/glycerol, water/sorbitol, water/polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof.


The compounds according to the invention may also be used in conjunction with other active substances, particularly for the treatment and/or prevention of the diseases and conditions mentioned above. Other active substances which are suitable for such combinations include, for example, BACE inhibitors; amyloid aggregation inhibitors (e.g. ELND-005); directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants (e.g. vitamin E or ginkolide); anti-inflammatory substances (e.g. Cox inhibitors, NSAIDs additionally or exclusively having Abeta lowering properties); HMG-CoA reductase inhibitors (statins); acetylcholinesterase inhibitors (e.g., donepezil, rivastigmine, tacrine, galantamine); NMDA receptor antagonists (e.g. memantine); AMPA receptor agonists; AMPA receptor positive modulators, AMPAkines, monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmitters; substances inducing the secretion of growth hormone (e.g., ibutamoren mesylate and capromorelin); CB-1 receptor antagonists or inverse agonists; antibiotics (e.g., minocyclin or rifampicin); PDE2, PDE4, PDE5, PDE9, PDE10 inhibitors, GABAA 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 or positive modulators; histamine H3 antagonists, 5 HT-4 agonists or partial agonists, 5HT-6 antagonists, alpha2-adrenoreceptor antagonists, calcium antagonists, muscarinic receptor M1 agonists or partial agonists or positive modulators, muscarinic receptor M2 antagonists, muscarinic receptor M4 antagonists, metabotropic glutamate-receptor 5 positive modulators, glycine transporter 1 inhibitors, antidepressants, such as citalopram, fluoxetine, paroxetine, sertraline and trazodone; anxiolytics, such as lorazepam and oxazepam; antiphychotics, such as aripiprazole, clozapine, haloperidol, olanzapine, quetiapine, risperidone and ziprasidone, and other substances that modulate receptors or enzymes in a manner such that the efficacy and/or safety of the compounds according to the invention is increased and/or unwanted side effects are reduced. The compounds according to the invention may also be used in combination with immunotherapies (e.g., active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or nanobodies) for the treatment of the above-mentioned diseases and conditions.


The dosage for the combination partners mentioned above is usefully ⅕ of the lowest dose normally recommended up to 1/1 of the normally recommended dose.


Therefore, in another aspect, this invention relates to the use of a compound according to the invention or a pharmaceutically acceptable salt thereof combined with at least one of the active substances described above as a combination partner, for preparing a pharmaceutical composition which is suitable for the treatment or prevention of diseases or conditions which can be affected by inhibitors of phosphodiesterase 2. These are preferably pathologies related to PDE2 hyperactivity and/or cAMP and/or cGMP hypofunction, particularly one of the diseases or conditions listed above, most particularly prodromal and mild-to-moderate Alzheimer's Disease and cognitive impairment associated with schizophrenia. The use of the compound according to the invention in combination with another active substance may take place simultaneously or at staggered times, but particularly within a short space of time. If they are administered simultaneously, the two active substances are given to the patient together; while if they are used at staggered times the two active substances are given to the patient within a period of less than or equal to 12 hours, but particularly less than or equal to 6 hours.


Consequently, in another aspect, this invention relates to a pharmaceutical composition which comprises a compound according to the invention or a pharmaceutically acceptable salt thereof and at least one of the active substances described above as combination partners, optionally together with one or more inert carriers and/or diluents.


The compound according to the invention may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts.


EXAMPLES

The following examples are intended to illustrate the invention, without restricting its scope.


Chemical Manufacture
Abbreviations



  • ACN acetonitrile

  • APCI Atmospheric pressure chemical ionization

  • d day

  • Cy cyclohexane

  • DCM dichloromethane

  • DIPEA diisopropylethylamine

  • DMF dimethylformamide

  • ESI electrospray ionization (in MS)

  • EtOAc ethylacetate

  • EtOH ethanol

  • Exp. Example

  • GC gas chromatography

  • GC-MS coupled gas chromatography-mass spectrometry

  • h hour(s)

  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium-Hexafluorophosphate

  • HCl hydrochloric acid

  • HPLC high performance liquid chromatography

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

  • LC liquid chromatography

  • LC-MS liquid chromatography-mass spectrometry

  • M molar (mol/L)

  • MeOH methanol

  • min minute(s)

  • MS mass spectrometry

  • NaOH sodiumhydroxide

  • NMP 1-methyl-2-pyrrolidinone

  • NOE Nuclear Overhauser effect

  • PE petroleum ether

  • rt room temperature

  • Rt retention time (in HPLC)

  • 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′-tetramethyluronium tetrafluoroborate

  • TEA triethylamine

  • TFA trifluoroacetic acid

  • THF tetrahydrofuran

  • TLC thin-layer chromatography

  • UPLC-MS ultra performance liquid chromatography-mass spectrometry



Analytical Methods:


UPLC-MS, HPLC-MS, LC-MS:


Method 1:


Instrument: LC/MS ThermoFinnigan HPLC Surveyor DAD, MSQ single quadrupole


Column: Synergi Hydro RP100A, 2.5 μm, 3×50 mm


Mobile phase: A=H2O 90%+10% CH3CN+NH4COOH 10 mM

    • B=CH3CN 90%+H2O 10%+NH4COOH 10 mM


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
1.2



0.50
100
0
1.2



6.50
0
100
1.2



7.50
0
100
1.2



8.00
100
0
1.2



9.00
100
0
1.2










Detection: UV 254 nm


Detection: Finnigan MSQ, single quadrupole


Ion source: APCI+/APCI−


Scan range: 100-900 amu


Method 2:


Instrument: LC/MS Waters Acquity UPLC System DAD, SQD single quadrupole


Column: BEH C18 1.7 μm 2.1×50 mm, Temp 35° C.


Mobile phase: A=H2O 90%+CH3CN 10%+NH4COOH 5 mM

    • B=CH3CN 90%+H2O 10%


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
0.7



1.20
0
100
0.7



1.45
0
100
0.7



1.55
100
0
0.7



1.75
100
0
0.7










Detection: UV 254 nm


Detection: SQD, single quadrupole


Ion source: ES+/ES−


Scan range: 90-900 amu


Method 3:


Instrument: LC/MS Waters Alliance 2695 HPLC System DAD, Quattro Micro Triple quadrupole


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


Mobile phase: A=H2O 90%+10% CH3CN+CF3COOH 0.05%

    • B=CH3CN 90%+10% H2O


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
1.3



0.70
100
0
1.3



4.50
0
100
1.3



5.80
0
100
1.3



6.00
100
0
1.3










Detection: UV 254 nm


Detection: Quattro Micro, triple quadrupole


Ion source: ES+


Scan range: 90-1000 amu


Method 4:


Instrument: LC/MS Waters Alliance 2695 HPLC System DAD, Quattro Micro Triple quadrupole


Column: XBridge Phenyl 3.5 μm 3×30 mm, Temp 35° C.


Mobile phase: A=H2O 90%+10% CH3CN+NH4HCO3 5 mM

    • B=CH3CN 90%+10% H2O


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
1.3



4.50
0
100
1.3



5.80
0
100
1.3



6.00
100
0
1.3










Detection: UV 254 nm


Detection: Quattro Micro, triple quadrupole


Ion source: ES+


Scan range: 90-1000 amu


Method 5:


Instrument: LC/MS Waters Acquity UPLC System DAD, SQD single quadrupole


Column: BEH C18 1.7 μm 2.1×50 mm, Temp 35° C.


Mobile phase: A=H2O 90%+CH3CN 10%+NH4HCO3 5 mM

    • B=CH3CN 90%+H2O 10%


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
0.70



1.20
100
0
0.70



1.45
0
100
0.70



1.55
0
100
0.70



1.75
100
0
0.70










Detection: UV 254 nm


Detection: SQD, single quadrupole


Ion source: ES+/ES−


Scan range: 90-900 amu


Method 6:


Instrument: LC/MS Waters Acquity System DAD, SQD single quadrupole


Column: XBridge C18 2.5 μm 3.0×30 mm, Temp 60° C.


Mobile phase: A=H2O+TFA 0.1%

    • B=CH3CN


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
98
2
1.5



1.3
1
99
1.5



1.5
1
99
1.5



1.6
98
2
1.5










Method 7:


Instrument: LC/MS Waters Acquity System DAD, SQD single quadrupole


Column: XBridge C18 2.5 μm 3.0×30 mm, Temp 60° C.


Mobile phase: A=H2O+NH4OH 0.1%

    • B=CH3CN


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
95
5
1.5



1.3
1
99
1.5



1.5
1
99
1.5



1.6
95
5
1.5










Method 8-:


Instrument: LC/MS Agilent 1100 System DAD


Column: Sunfire C18 2.5 μm 3.0×30 mm, Temp 60° C.


Mobile phase: A=H2O+TFA 0.1%

    • B=CH3CN


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
98
2.0
2.0



1.2
0.0
100
2.0



1.4
0.0
100
2.0










Method 10:


Instrument: LC/MS ThermoFinnigan HPLC Surveyor DAD, LCQFleet Ion Trap


Column: Xselect CSH, 2.5 μm, 4.6×50 mm


Mobile phase: A=H2O 90%+10% CH3CN+HCOOH 0.1%

    • B=CH3CN 90%+H2O 10%+HCOOH 0.1%


















Time in min:
% A
% B
Flow rate in mL/min





















0.00
100
0
1.4



4.00
0
100
1.4



5.30
0
100
1.4



5.50
100
0
1.4



6.00
100
0
1.4










Detection: UV 254 nm


Detection: Finnigan Fleet, Ion Trap


Ion source: ES+


Scan range: 100-900 amu


GC/MS Method


Method 9:


Instrument: GC/MS Thermo Scientific TRACE GC ULTRA, DSQ II MS single quadrupole


Column: Agilent DB-5MS, 25 m×0.25 mm×0.25 um


Carrier gas: Helium, 1 mL/min constant flow


Oven Program: 50° C., to 100° C. in 10° C./min, to 200° C. in 20° C./min, to 320° C. in 30° C./min (hold 10 min).


Detection: DSQ II MS single quadrupole

    • Ion source: EI
    • Scan range: 50-450 amu


Chiral HPLC Methods:


Instrument: HPLC Agilent 1100 (DAD equipped; UV Detection: 230 nm); flow rate: 1 mL/min; column temperature: 25° C.


Method C1


column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=70:30


Method C2


column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=60:40


Method C3


column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=80:20


Method C4


column: Daicel Chiralcel OJ-H; eluent: Hexane:EtOH=80:20


Method C5


column: Daicel Chiralcel OJ-H; eluent: Hexane:EtOH=85:15


Method C6


column: Daicel Chiralcel OJ-H; eluent: Hexane:EtOH=70:30


Method C7


column: Daicel Chiralcel AS-H; eluent: Hexane:EtOH=75:25


NMR Equipment:


The 1H NMR spectra were recorded on a Bruker Avance III (500 MHz) or a Varian 400 (400 MHz) or Varian Mercury (300 MHz) instrument using deuterated dimethylsulfoxide (DMSO-d6) as the solvent with tetramethylsilane (TMS) and residual solvent peak as an internal standard. Chemical shifts are reported in δ values (ppm) relative to TMS.


Purification:


The most suitable purification techniques applied for the purification of compounds of the present invention are direct phase silica gel flash chromatography and reverse phase chromatography, unless otherwise specifically stated.


General Comment Concerning the Presentation of the Structures


Compounds with stereogenic centre(s): The structures depicted in the experimental section will not necessarily show all the stereochemical possibilities of the compounds but only one.


The structural presentation of the compounds in the experimental section will show a stereochemical bond only in case where the absolute stereochemistry is known.


The structural presentation of the compounds in the experimental section with unknown absolute stereochemistry will show a planar bond plus an additional comment that indicates if the described compound is a racemic mixture, a single stereoisomer and where applicable the relative stereochemistry.


Two examples are given below.


Example 1: the presented chemical structure is depicted as:




embedded image


The added term racemic mixture points to the two stereochemical options and thus the manufactured compounds is a mixture of:




embedded image


When racemic mixtures of above depicted structures are separated, the single stereoisomers are depicted as:




embedded image


The added term ‘single stereoisomer’ and the planar bond indicates that the absolute configuration is unknown.


Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC, single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.


Example 2: the presented chemical structure is depicted as:




embedded image


The added term ‘TRANS-racemic mixture’ points to the two stereochemical options and thus the manufactured compounds is a mixture of:




embedded image


The same principles applies to ‘CIS-racemic mixture’.


When racemic mixtures of above depicted structures are separated, the single stereoisomers are depicted as:




embedded image


The added term ‘TRANS-single stereoisomer’ indicates a relative configuration known (trans) and the planar bond indicates the unknown absolute configuration.


The same principles applies to ‘CIS-single stereoisomer’.


Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC, single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.


EXPERIMENTAL

The following intermediates and examples are intended to illustrate the invention, without restricting its scope.


Intermediates


Intermediate 1:




embedded image


To a solution of 3-amino-4-carbethoxypyrazole (4 g, 25.27 mmol) in absolute EtOH (40 mL), 1,1,3,3-Tetraethoxy-2-methyl-propane (6.34 g, 26.53 mmol) was added followed by 13.90 mL of a 1M solution of HCl in dioxane. The mixture was heated at 80 C overnight. Solvents were evaporated, then DCM and water were added. Phases were separated, organics washed with a saturated solution of NaCl, dried over sodium sulphate and evaporated to obtain 5.17 g of the title compound


LC-MS (Method 2): Rt=0.73 min


MS (ESI pos): m/z=206 (M+H)+


Intermediate 2




embedded image


Intermediate 1 (5 g) was dissolved in a mixture of THF/water (1:1, 100 mL) and stirred at room temperature for 48 hrs. The resulting suspension was diluted with water and 70 mL of EtOAc were added. Phases were separated, aqueous phases were treated with a 4N solution of HCl (ca 20 mL). A white solid formed. The mixture was cooled at 0° C., then the white solid formed collected by filtration and dried under vacuum at 65° C. to obtain 3.50 g of the title compound.


LC-MS (Method 3): Rt=1.62 min


MS (ESI pos): m/z=178 (M+H)+


Intermediate 3:




embedded image


To a solution of of 2-Bromo-malonaldehyde (9.73 g; 64 mmol) in EtOH (100 mL) at 70° C., 3-amino-4-carbethoxypyrazole (10 g, 64 mmol)_and AcOH (100 mL) were added and the mixture stirred at 70° C. for 1 h. Solvents were evaporated, the residue treated with DCM (100 mL) and a 1N solution of NaOH (100 mL). Phases were separated, organics washed with a saturated solution of NaCl, dried over sodium sulphate and evaporated. The crude was purified flash chromatography (eluent 10:1 PE/EtOAc) to obtain 15 g of the title compound as white solid.


LC-MS (Method 2): Rt=0.98 min


MS (ESI pos): m/z=271 (M+H)+


Intermediate 4:




embedded image


Intermediate 3 (5 g, 18.5 mmol) was suspended in dry toluene (50 mL) and 5 mL of water were added. To this mixture, potassium cyclopropyltrifluoroborate (4 g, 28 mmol) was added followed by 2-dicyclohexylphosphino-2′,6′-di isopropoxy 1,1′-biphenyl (0.864 g, 1.85 mmol), palladium acetate (0.208 g, 0.93 mmol) and potassium carbonate (7.7 g, 55 mmol). Mixture was refluxed at 130° C. for 3 hrs, then cooled to room temperature, filtered over celite and washed with AcOEt and then EtOH.


Solvent was evaporated under vacuum and the crude used in the next step without further purification.


LC-MS (Method 2): Rt=0.9 min


MS (ESI pos): m/z=232 (M+H)+


Intermediate 5:




embedded image


Intermediate 4 (4 g, 17.5 mmol) was suspended in 50 ml of EtOH, 8 ml of 4N NaOH and 30 ml of water and stirred overnight. EtOH was evaporated and a 4 N solution of HCl added. The solid formed was filtered, washed with water and dried under vacuum at 70° C. overnight to obtain 3.6 g of the title compound.


LC-MS (Method 3): Rt=2.75 min


MS (ESI pos): m/z=204 (M+H)+


Intermediate 6:




embedded image


Intermediate 6 was prepared as described in WO 2010/007074 starting from commercially available (Z) 3-(diethylamino)-2-fluoroprop-2-enal (1.34 mL, 9.0 mmol) and 3-amino-4-carbethoxypyrazole (2.1 g, 13.6 mmol) to obtain 0.53 g of the title compound.



1H NMR (300 MHz, CDCl3): δ ppm 1.44-1.39 (t, 3H), 4.47-4.40 (q 2H) 8.57 (s, 1H) 8.7 (m, 1H), 8.8 (d, 1H)


Intermediate 7:




embedded image


To a solution of 5-amino-3-methyl-1H-pirazole-4-carboxylic acid ethyl ester (1 g, 5.91 mmol) in absolute EtOH (25 mL), 1,1,3,3-Tetraethoxy-2-methyl-propane (1.4 g, 6.2 mmol) was added followed by 1.63 mL of a 4N solution of HCl in dioxane. Mixture was heated at 80° C. for 5 hrs, left at room temperature overnight and then solvents were evaporated to dryness. The violet solid obtained was dissolved in DCM, water was added and the phases separated.


The organic phases were dried over sodium sulfate and concentrated under vacuum to obtain 1.26 g of title compound used for next step without further purification.


LC-MS (Method 2): Rt=0.79 min


MS (ESI pos): m/z=224 (M+H)+


Intermediate 8:




embedded image


To a solution of intermediate 7 (1.26 g, 5.75 mmol) in THF (25 mL) and water (25 mL) 1.5 mL of a 1N solution of sodium hydroxide were added and the mixture heated at heated at 60° C. for 2 h. Solvent was evaporated, water was added and 30 ml of a 12N solution of HCl added until pH 2. The solid formed was filtered, washed with water and dried at 70° C. under vacuum to obtain 0.9 g of title compound as white solid.


LC-MS (Method 1): Rt=0.27 min


MS (APCI): m/z=192 (M+H)+


Intermediate 9:




embedded image


To a solution of 5-amino-3-methyl-1H-pyrazole-4-carboxilic acid ethyl ester (4 g, 23.64 mmol) in absolute EtOH (80 mL), 1,1,3,3-tetraethoxypropane (5.96 mL, 23.64 mmol) and 5.9 mL of a 4N solution of HCl in dioxane were added. The resulting mixture was heated at 80° C. 3 hrs. Solvents were evaporated, the residue was diluted with DCM and water. Organic layer was separated, dried over sodium sulphate and evaporated to obtain the title compound as white solid (3.6 g)


LC-MS (Method1): Rt=263 min


MS (APCI): m/z=206 (M+H)+


Intermediate 10:




embedded image


To a solution of intermediate 9 (3.6 g, 17.54 mmol) in AcOH (70 mL) bromine (2.26 mL) was added drowpwise. The mixture was stirred at room temperature overnight then carefully poured into 500 mL of water and extracted with EtOAc (3×100 mL). Organic phases were collected and washed with 100 mL of a 5% solution of Na2S2O3 and then with 100 mL of a saturated solution of NaCl, dried over sodiumsulphate and concentrated under vacuum.


Crude was purified by flash chromatography (eluent: DCM/EtOAc; gradient from 100% to 70%) to obtain the title compound as white solid (2.1 g)


LC-MS (Method 1): Rt=3.52 min


MS (APCI): m/z=284 (M+H)+


Intermediate 11:




embedded image


To a solution of intermediate 10 (2.05 g, 7.22 mmol) in toluene (40 mL) water (4 mL) was added followed by potassium cyclopropyltrifluroborate (1.6 g, 10.82 mmol), palladium(II) acetate (0.08 g, 0.36 mmol), dicyclohexylphosphino-2′,6′-di-i-propoxy dl-1,1′-biphenyl (RUPHOS, 0.34 g, 0.72 mmol) and potassium carbonate (3 g, 21.65 mmol). Mixture was heated at 130° C. for 3 hrs then cooled to room temperature, filtered over celite and washed with AcOEt. Organic layer was dried and evaporated to obtain the title compound (1.5 g) used for the next step without further purification.


LC-MS (Method 2): Rt=0.92 min


MS (ESI pos): m/z=246 (M+H)+


Intermediate 12:




embedded image


To a solution of intermediate 11 (1.5 g, 6.2 mmol) in absolute EtOH (30 mL) water (10 mL) was added followed by 7.7 mL of a 8N solution of NaOH. Mixture was stirred at room temperature overnight, then solvent evaporated and a 4N solution of HCl added until pH=1. The solid formed was filtered, washed with water and dried under vacuum at 70° C. overnight (1.5 g).


LC-MS (Method 1): Rt=0.6 min


MS (APCI): m/z=218 (M+H)+


Intermediate 13:




embedded image


To a solution of 4-bromo-3-fluoro-benzotrifluoride (585 mg, 2.36 mmol) in 15 mL of THF, stirred at −75° C. under nitrogen atmosphere, 1.53 mL (2.6 mmol) of a 1.7M solution of tert-butyllithium in pentane were added dropwise. The reaction mixture was stirred at −60° C. for 15 minutes then a solution of 2-methyl-propane-2-sulfinic acid-(tetrahydro-pyran-4-ylidene)-amide (400 mg, 1.97 mmol; prepared as described in literature: WO2005/87751 A2) in 10 mL of THF was added dropwise. The reaction mixture was allowed to reach room temperature and stirred for 1 hr. A saturated ammonium chloride solution was added and the reaction mixtures was extracted with ethyl acetate. The organic phases were collected, dried over sodium sulfate and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 12% to 100% of AcOEt).


The oil obtained was diluted in 2 mL of 1,4-dioxane, 0.4 mL of a 4 M solution of hydrochloric acid in 1,4-dioxane were added, the reaction mixture was stirred at room temperature for 1 hr and then concentrated under vacuum to obtain 100 mg of the title compound as white solid.


LC-MS (Method 2): Rt=0.90 min


MS (ESI pos): m/z=264 (M+H)+


The following Amine Intermediates were prepared in analogy to Intermediate 13 starting from the corresponding commercially available bromo-aryl/heteroaryl or iodo-aryl/heteroaryl derivative:



















Rt



Starting
Amine intermediate
MS m/z
(min)
Method




















4-Chloro- 2-fluoro- iodobenzene
14


embedded image


230, 232 (M + H)+
0.79
Method 2





4-Iodo- benzo- trifluoride
15


embedded image


246 (M + H)+
0.66
Method 2






16 Commercially available from ENAMINE-BB (Cat. Number EN300- 185595)


embedded image










2-Bromo- quinoline
17


embedded image


229 (M + H)+
0.69
Method 2






18 Commercially available from ENAMINE-BB (Cat. Number EN300- 50665)


embedded image










4-Bromo- 1- (difluoro- methoxy)-2- fluoro- benzene
19


embedded image


262 (M + H)+
0.68
Method 2





4-Chloro- 3-fluoro- iodobenzene
20


embedded image


230, 232 (M + H)+
0.77
Method 2





2-Bromo- 3-fluoro-5- (trifluoro- methyl) pyridine
21


embedded image


265 (M + H)+
0.83
Method 2





2-Iodo-5- (trifluoro- methyl) pyridine
22


embedded image


247 (M + H)+
0.75
Method 2





5-Iodo-2- (trifluoro- methyl) pyridine
23


embedded image


247 (M + H)+
0.71
Method 2





5-Bromo- 3-fluoro-2- (trifluoro- methyl) pyridine
24


embedded image


265 (M + H)+
0.81
Method 2





2-Chloro- 4-fluoro- iodobenzene
25


embedded image


230, 232 (M + H)+
0.59
Method 2









Intermediate 26:




embedded image


Intermediate 26 was prepared as described for Intermediate 13 starting from commercially available 4-bromo-3-fluoro-benzotrifluoride (560 mg, 2.30 mmol) and 2-methyl-propane-2-sulfinic acid [dihydro-pyran-(3Z)-ylidene]-amide (390 mg, 1.92 mmol; prepared in analogy to 2-methyl-propane-2-sulfinic acid-(tetrahydro-pyran-4-ylidene)-amide, described in WO2005/87751 A2) to obtain 120 mg of the title compound, as racemic mixture.


LC-MS (Method 2): Rt=1.00 min


MS (ESI pos): m/z=264 (M+H)+


The following Amine Intermediates were prepared in analogy to Intermediate 26 starting from the corresponding commercially available bromo-aryl derivative:



















Rt



Starting
Amine intermediate
MS m/z
(min)
Method




















4-Bromo- benzo- trifluoride
27 Racemic mixture


embedded image


246 (M + H)+
0.92
Method 2





1-Bromo- 4- (trifluoro- methoxy) benzene
28 Racemic mixture


embedded image


262 (M + H)+
0.95
Method 2









Intermediate 29:




embedded image


Intermediate 29 was prepared as described for Intermediate 13 starting from commercially available 4-bromo-3-fluoro-benzotrifluoride (462 g, 1.90 mmol) and 2-methyl-propane-2-sulfinic acid [dihydro-furan-(3Z)-ylidene]-amide (300 mg, 1.58 mmol; prepared in analogy to 2-methyl-propane-2-sulfinic acid-(tetrahydro-pyran-4-ylidene)-amide, described in WO2005/87751 A2) to obtain 50 mg of the title compound, as racemic mixture.


LC-MS (Method 2): Rt=0.90 min


MS (ESI pos): m/z=250 (M+H)+


The following Amine Intermediates were prepared in analogy to Intermediate 29 starting from the corresponding commercially available bromo-aryl/heteroaryl or iodo-aryl/heteroaryl derivative:



















Rt



Starting
Amine intermediate
MS m/z
(min)
Method




















4- Iodobenzo- trifluoride
30 Racemic mixture


embedded image


232 (M + H)+
0.83
Method 2





4-Chloro- 2- fluoroiodo- benzene
31 Racemic mixture


embedded image


216, 218 (M + H)+
0.75
Method 2





2-Bromo- 3-fluoro-5- (trifluoro- methyl) pyridine
32 Racemic mixture


embedded image


251 (M + H)+
0.76
Method 2





5-Iodo-2- (trifluoro- methyl) pyridine
33 Racemic mixture


embedded image


233 (M + H)+
0.68
Method 2





2-Iodo-5- (trifluoro- methyl) pyridine
34 Racemic mixture


embedded image


233 (M + H)+
0.70
Method 2









Intermediate 35:




embedded image


Step 1:


3.6-Dihydro-2H-pyran-4-boronic acid pinacol ester (5.62 g, 26.75 mmol), 4-bromo-3-fluorobenzotrifluoride (5.00 g, 20.58 mmol), potassium carbonate (8.53 g, 61.73 mmol) and 1,1′-bis(diphenylphosphino)ferrocenedichloro palladium(II) (753 mg, 1.03 mmol) were suspended in 50 mL of 1,4-dioxane and 10 mL of water. The reaction mixture was refluxed for 3 hrs, solvents were evaporated and the crude was extracted with ethyl acetate (50 mL) and water (50 mL). Organic layer was separated, dried over sodium sulfate and evaporated. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 40% to 100% of AcOEt) to obtain 4.0 g of the title compound as clear oil.


GC-MS (Method 9): Rt=7.76 min


MS: m/z=246 (M)+


Step 2:


To a solution of 4-(2-Fluoro-4-trifluoromethyl-phenyl)-3,6-dihydro-2H-pyran (obtained as described in Step 1; 7.0 g, 25.18 mmol) in 150 mL of dichloromethane, stirred at 0° C., 3-chloroperoxybenzoic acid (11.3 g, 50.37 mmol) was added portionwise. The reaction mixture was allowed to reach room temperature and stirred overnight. The reaction mixture was cooled to 0° C. and the precipitate formed was filtered off. The organic solution was washed twice with an aqueous saturated solution of potassium carbonate, dried over sodium sulfate and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 50% to 100% of AcOEt) to obtain 4.2 g of the title compound.


GC-MS (Method 9): Rt=7.68 min


MS: m/z=262 (M)+


Step 3:


To a solution of 6-(2-Fluoro-4-trifluoromethyl-phenyl)-3,7-dioxa-bicyclo[4.1.0]heptane (obtained as described in Step 2; 1.64 g, 6.25 mmol) in 10 mL of acetonitrile, stirred under nitrogen atmosphere at −45° C., trifluoromethane sulfonic acid (1.88 g, 12.5 mmol) was added dropwise. The reaction mixture was allowed to reach room temperature and stirred for 2.5 hrs. 10 mL of water were added, the reaction mixture was warmed to 100° C. and acetonitrile was distilled out. The reaction mixture was stirred at 100° C. for 5 hrs, then cooled to room temperature and stirred overnight. The reaction mixture was diluted with dichloromethane and phases were separated. The aqueous phase was treated with a 4M solution of NaOH until basic pH and extracted with dichloromethane. The organic phase was dried over sodium sulfate and concentrated under vacuum to give 290 mg of the final compound (crude colorless oil), 4-Amino-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-pyran-3-ol, as racemic mixture (TRANS/CIS diastereoisomeric ratio 85:15, determined by NMR).


The crude was used in the next step without any further purification.


LC-MS (METHOD 2): Rt=0.77 min


MS (ESI pos): m/z=280 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 7.59-7.47 (m, 3H), 4.74 (d, J=5.8 Hz, 1H), 4.04 (dd, J=11.7, 1.5 Hz, 1H), 3.90 (ddd, J=12.5, 11.0, 2.0 Hz, 1H), 3.74 (d, J=5.8 Hz, 1H), 3.67 (dd, J=11.1, 4.1 Hz, 1H), 3.56-3.51 (m, 1H), 2.51-2.44 (m, 1H), 2.09 (br s, 1H), 1.56 (m, 1H).


NOE: 2.09 (NH2): 3.74; 4.04. 4.74 (OH): 3.55; 2.45


The following Amino-alcohol Intermediate were prepared in analogy to Intermediate 34 starting from the corresponding commercially available bromo-heteroaryl:



















Rt



Starting
Amino-alcohol intermediate
MS m/z
(min)
Method




















5-Bromo- 2- (trifluoro- methyl) pyridine
36 Trans racemate


embedded image


263 (M + H)+
0.90
Method 1









Relative stereochemistry of intermediate 36 assigned by NMR and NOE:


1H NMR (500 MHz, DMSO-d6) δ 8.87 (d, J=2.3 Hz, 1H, 13), 8.10 (ddd, J=8.3, 2.4, 0.8 Hz, 1H), 7.81 (dd, J=8.3, 0.8 Hz, 1H), 4.78 (d, J=5.9 Hz, 1H), 4.05 (dd, J=11.6, 1.5 Hz, 1H), 3.91 (td, J=11.7, 2.3 Hz, 1H), 3.68 (ddd, J=11.1, 4.9, 2.2 Hz, 1H), 3.56 (dd, J=11.7, 2.4 Hz, 1H), 3.47 (d, J=5.9 Hz, 1H), 2.49-2.44 (m, 1H), 2.12 (s, 2H), 1.49 (dd, J=13.1, 1.9 Hz, 1H).


NOE: 2.12 (NH2): 3.47; 3.91; 4.05. 4.78 (OH): 3.56; 2.48


Intermediate 37




embedded image


Step 1:


tert-Butyllithium (21.8 mL, 1.7M in pentane, 37.0 mmol) was added dropwise to 4-bromo-3-fluorobenzotrifluoride (5.00 g, 20.58 mmol) in THF (50 mL) at −70° C. After 1 h, 3-oxotetrahydrofuran (1.78 g, 20.58 mmol) in THF was added dropwise. The reaction mixture was warmed to −10° C. mixture and quenched with NH4Cl saturated solution. Ethyl acetate was added, the organic layer was separated, dried over sodium sulfate and evaporated. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 0% to 80% of AcOEt) to obtain 1.6 g of the 3-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-furan-ol.


GC-MS (Method 9): Rt=7.58 min


MS: m/z=250 (M)+


Step 2:


p-Toluenesulfonic acid monohydrate (1.75 g, 9.19 mmol) was added to 3-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-furan-ol (obtained as described in Step 1; 2.3 g, 9.19 mmol) in toluene (20 mL). After refluxing for 1 h, volatiles were evaporated, DCM and water were added, the organic layer was separated, dried over sodium sulfate and evaporated to obtain 2.0 g (77% content) of crude 3-(2-fluoro-4-trifluoromethyl-phenyl)-2,5-dihydro-furan, that was used without further purification.


GC-MS (Method 9): Rt=7.12-7.21 min


MS: m/z=232 (M)+


Step 3:


To a solution of 3-(2-fluoro-4-trifluoromethyl-phenyl)-2,5-dihydro-furan (obtained as described in Step 2; 2.0 g 77% content, 6.63 mmol) in 50 mL of dichloromethane, stirred at 0° C., 3-chloroperoxybenzoic acid (2.63 g, 15.26 mmol) was added portionwise. The reaction mixture was allowed to reach room temperature and stirred overnight. The reaction mixture was cooled to 0° C. and the precipitate formed was filtered off. The organic solution was washed twice with an aqueous saturated solution of potassium carbonate, dried over sodium sulfate and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 50% to 100% of AcOEt) to obtain 1.2 g (98% content) of 1-(2-fluoro-4-trifluoromethyl-phenyl)-3,6-dioxa-bicyclo[3.1.0]hexane. 1H NMR (300 MHz, DMSO-d6): δ ppm 3.91-3.95 (m, 2H), 4.06-4.19 (m, 3H) 7.37 (dd, J=10.2, 1.3 Hz, 1H), δ 7.47 (dd, J=8.4, 1.1 Hz, 1H), 7.59 (m, 1H)


Step 4:


To a solution of 1-(2-fluoro-4-trifluoromethyl-phenyl)-3,6-dioxa-bicyclo[3.1.0]hexane (obtained as described in Step 3; 1.20 g, 98% content, 4.74 mmol) in 20 mL of acetonitrile, stirred under nitrogen atmosphere at −40° C., trifluoromethane sulfonic acid (0.84 mL, 9.48 mmol) was added dropwise. The reaction mixture was allowed to reach room temperature and stirred for 2.5 hrs. 20 mL of water were added, the reaction mixture was warmed to 100° C. and acetonitrile was distilled out. The reaction mixture was stirred at 100° C. for 20 hrs, then cooled to room temperature and stirred overnight. The reaction mixture was diluted with dichloromethane and phases were separated. The aqueous phase was treated with a 4M solution of NaOH until basic pH and extracted with dichloromethane. The organic phase was dried over sodium sulfate and concentrated under vacuum to give 200 mg of 4-amino-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-furan-3- -ol, as racemic mixture (TRANS/CIS diastereoisomeric ratio 88/12, determined by NMR).


The crude was used in the next step without any further purification.


LC-MS (METHOD 1): Rt=2.52-3.04 min


MS (ESI pos): m/z=266 (M+H)+


1H NMR (400 MHz, DMSO-d6) δ 7.59-7.54 (m, 1H), 7.52-7.45 (m, 2H), 5.08 (d, J=4.8 Hz, 1H), 4.29 (q, J=3.8 Hz, 1H), 4.25 (dd, J=8.8, 3.8 Hz, 1H), 4.14 (d, J=7.6 Hz, 1H), 3.95 (dd, J=8.0, 2.2 Hz, 1H), 3.65 (d, J=8.8 Hz, 1H), 2.06 (s, 2H).


NOE: 2.06 (NH2): 3.95; 4.29; 4.25. 5.08 (OH): 4.14; 3.65


Intermediate 38:




embedded image


Step 1:


6-(2-Fluoro-4-trifluoromethyl-phenyl)-3,7-dioxa-bicyclo[4.1.0]heptane (obtained as described in Step 2 in the preparation of Intermediate 34; 4.20 g, 16.02 mmol), sodium azide (2.08 g, 32.04 mmol) and ammonium chloride (1.72 g, 32.04 mmol) were suspended in 50 mL of methanol and 10 mL of water. The reaction mixture was stirred at reflux for 18 hrs. Solvents were removed, the crude was suspended in water and extracted twice with ethyl acetate. The organic layer was dried over sodium sulfate and concentrated under vacuum to give 4.70 g of the final compound as a mixture of the desired regioisomer 4-Azido-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-pyran-3-ol and the undesired regioisomer 3-azido-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-pyran-4-ol in a regioisomeric ratio of 76/24 determined by NMR. The regioisomeric mixture was used in the next step without separation.


GC-MS (METHOD 9): Rt=9.57 min


MS: m/z=248 (M)+


1H NMR (500 MHz, DMSO-d6) δ 7.77-7.74 (m, 1H), 7.72 (d, J=7.9 Hz, 1H), 7.67-7.64 (m, 1H), 5.37 (d, J=6.0 Hz, 1H), 3.93-3.89 (m, 1H), 3.85-3.80 (m, 1H), 3.78 (dd, J=12.3, 1.5 Hz, 1H), 3.74-3.66 (m, 2H), 2.65 (ddd, J=13.8, 11.9, 4.8 Hz, 1H), 2.02-1.95 (m, 1H).


Step 2:


A mixture of 4-Azido-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-pyran-3-ol and 3-azido-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-pyran-4-ol (obtained as described in Step 1, 2.0 g, 6.55 mmol), Pd/C (300 mg, 2.82 mmol) and di-tert-butyldicarbonate (1.86 g, 8.52 mmol) were suspended in 150 mL of ethanol. The reaction mixture was stirred at room temperature under hydrogen atmosphere (2.5 bar) for 1 hr. The reaction mixture was filtered on a celite pad and the organic solution was concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: cyclohexane/AcOEt; gradient from 10% to 100% of AcOEt) to obtain 1.75 g of the title compound (yellow solid) as a mixture of the desired regioisomer [4-(2-fluoro-4-trifluoromethyl-phenyl)-3-hydroxy-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester and the undesired regioisomer [4-(2-Fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-pyran-3-yl]-carbamic acid tert-butyl ester in a regioisomeric ratio of 85/15 determined by NMR. The regioisomeric mixture was used in the next step without separation.


GC-MS (METHOD 9): Rt=10.92-10.99 min


MS: m/z=323 (M)+


Step 3:


A mixture of [4-(2-Fluoro-4-trifluoromethyl-phenyl)-3-hydroxy-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester and 4-(2-Fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-pyran-3-yl]-carbamic acid tert-butyl ester (obtained as described in Step 2, 3.7 g, 7.30 mmol) was dissolved in 20 mL of dichlorometane, Dess-Martin periodinane (2.18 g, 9.5 mmol) was added portionwise and the reaction mixture was stirred at room temperature for 2 hrs. The reaction mixture was diluted with dichloromethane, washed with aqueous bicarbonate saturated solution, washed with aqueous sodium bisulfite saturated solution, the organic layer was separated, dried over sodium sulfate and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: dichloromethane/AcOEt; gradient from 0% to 70% of AcOEt) to give 2.4 g of the desired compound.


LC-MS (METHOD 1): Rt=4.23-4.83 min


MS (ESI pos): m/z=278 (fragment) (M+H)+


Step 4:


[4-(2-Fluoro-4-trifluoromethyl-phenyl)-3-oxo-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester (obtained as described in Step 3; 340 mg, 0.9 mmol) was suspended in 10 mL of dry THF. The reaction mixture was stirred at −20° C. and 0.29 mL of a 3.4M solution of methylmagnesium bromide in methyl-tetrahydrofurane was added dropwise. The reaction mixture was stirred at −20° C. for 1 hr, then quenched with aqueous saturated ammonium chloride solution. Organic layer was separated, dried over sodium sulfate and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: dichloromethane/AcOEt; gradient from 0% to 30% of AcOEt) to give 200 mg of the title compound, 4-(2-fluoro-4-trifluoromethyl-phenyl)-3-hydroxy-3-methyl-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester, as racemic mixture (TRANS/CIS diastereoisomeric ratio 82/12, determined by NMR).


GC-MS (METHOD 9): Rt=11.01 min


MS: m/z=292 (fragment) (M)+


1H NMR (500 MHz, DMSO-d6) δ 7.58-7.53 (m, 1H), 7.52-7.48 (m, 1H), 7.45 (d, J=12.8 Hz, 1H), 6.92 (s, 1H), 4.78 (s, 1H), 3.88 (d, J=12.2 Hz, 1H), 3.70 (t, J=6.7 Hz, 1H), 3.60 (q, J=12.8, 12.2 Hz, 1H), 3.28-3.26 (d, J=12.2 Hz, 1H), 2.88 (t, J=11.4 Hz, 1H), 1.33 (s, 9H), 0.9 (s, 3H).


NOE: 6.92 (NH): 0.90; 3.88. 4.78 (OH): 2.88; 3.27


Step 5:


4-(2-Fluoro-4-trifluoromethyl-phenyl)-3-hydroxy-3-methyl-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester (obtained as described in Step 4, as preferred diastereoisomer; 200 mg, 0.51 mmol) was dissolved in 5 mL of dichloromethane. Trifluoroacetic acid (0.39 mL, 5.1 mmol) was added, the reaction mixture was stirred at room temperature for 1 hr and then concentrated under vacuum. The crude obtained was stripped twice with ethyl ether to give 198 mg of the title compound, 4-amino-4-(2-fluoro-4-trifluoromethyl-phenyl)-3-methyl-tetrahydro-pyran-3-ol trifluoroacetate salt as racemic mixture (TRANS/CIS diastereoisomeric ratio 85/15, determined by NMR).


LC-MS (METHOD 1): Rt=3.35 min


MS (ESI pos): m/z=294 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 8.61 (s, 3H), 7.96 (t, J=8.2 Hz, 1H), 7.81 (dd, J=13.3, 2.0 Hz, 1H), 7.70-7.66 (m, 1H), 5.40 (s, 1H), 3.98 (ddd, J=13.3, 10.7, 2.8 Hz, 1H), 3.92-3.85 (m, 1H), 3.62 (d, J=12.6 Hz, 1H), 3.40-3.38 (d, J=12.6 Hz, 1H), 2.99 (ddd, J=14.4, 10.7, 5.2 Hz, 1H), 1.79 (dt, J=14.4, 3.0 Hz, 1H), 1.11 (d, J=1.8 Hz, 3H).


Intermediate 39:




embedded image


Step 1:


Step 1 was performed in analogy to Step 1 in the preparation of Intermediate 35, starting from 2-bromo-3-fluoro-5-(trifluoromethyl)pyridine (5 g, 20.49 mmol) to obtain 2-(3,6-dihydro-2H-pyran-4-yl)-3-fluoromethyl-pyridine (5.7 g).


LC-MS (METHOD 2): Rt=1.17 min


MS (ESI pos): m/z=248 (M+H)+


Step 2:


Step 2 was performed in analogy to Step 2 in the preparation of Intermediate 35, starting from 2-(3,6-Dihydro-2H-pyran-4-yl)-3-fluoromethyl-pyridine (5.7 g, 23.06 mmol) to obtain 2-(3,7-dioxa-bicyclo[4.1.0]hept-6-yl)-3-fluoro-5-trifluoromethyl-pyridine (3.25 g).


LC-MS (METHOD 2): Rt=0.95 min


MS (ESI pos): m/z=264 (M+H)+


Step 3:


Step 3 was performed in analogy to Step 1 in the preparation of Intermediate 38, starting from 2-(3,7-Dioxa-bicyclo[4.1.0]hept-6-yl)-3-fluoro-5-trifluoromethyl-pyridine (250 mg, 0.95 mmol) to obtain after purification by flash chromatography (eluent: cyclohexane/EtOAc; gradient from 0% to 30% of EtOAc), 4-azido-4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-tetrahydro-pyran-3-ol (160 mg) as major regioisomer


LC-MS (METHOD 2): Rt=1.05 min


MS (ESI pos): m/z=307 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 8.90 (dq, J=2.0, 0.9 Hz, 1H), 8.39 (ddd, J=11.5, 1.9, 0.7 Hz, 1H), 5.42 (s, 1H), 4.01 (s, 1H), 3.93-3.86 (m, 1H), 3.78-3.64 (m, 3H), 2.73 (ddd, J=14.6, 12.6, 4.9 Hz, 1H), 2.02 (dq, J=14.7, 2.0 Hz, 1H).


The minor regioisomer, 3-Azido-4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-tetrahydro-pyran-4-ol, was also isolated (40 mg).


LC-MS (METHOD 2): Rt=1.04 min


MS (ESI pos): m/z=307 (M+H)+


Step 4:


To a solution of 4-azido-4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-tetrahydro-pyran-3-ol (160 mg, 0.52 mmol) in 5 mL of methanol stirred under nitrogen atmosphere, ammonium formate (165 mg, 2.61 mmol) and zinc (51.2 mg, 0.78 mmol) were added. The reaction mixture was stirred at room temperature overnight and concentrated. A saturated ammonium chloride water solution was added and the reaction mixture was extracted with dichloromethane. The organic phase was separated, washed with brine, dried over sodium sulfate and concentrated under vacuum to give 115 mg of 4-Amino-4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-tetrahydro-pyran-3-ol, as TRANS-racemic mixture.


LC-MS (METHOD 5): Rt=0.71 min


MS (ESI pos): m/z=281 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 8.75 (dq, J=2.0, 1.0 Hz, 1H), 8.18-8.12 (m, 1H), 4.80 (d, J=5.6 Hz, 1H), 4.01 (dd, J=11.7, 1.5 Hz, 1H), 3.89-3.83 (m, 1H), 3.78 (dt, J=5.5, 1.9 Hz, 1H), 3.74-3.66 (m, 1H), 3.55 (dd, J=11.7, 1.7 Hz, 1H), 2.71-2.61 (m, 1H), 2.09 (s, 2H), 1.65-1.58 (m, 1H).


NOE: 2.09 (NH2): 3.55; 3.70; 3.78 4.80 (OH): 1.61; 3.78


The following intermediate was prepared in analogy to Intermediate 39, starting from 2-Bromo-5-trifluoromethyl-pyridine



















Rt



Starting
Amino-alcohol intermediate
MS m/z
(min)
Method




















2-Bromo-5- trifluoromethyl pyridine
40


embedded image


263
0.68
METHOD 2









1H NMR (500 MHz, DMSO-d6) δ 8.89 (dq, J=2.6, 0.9 Hz, 1H), 8.13 (ddd, J=8.4, 2.5, 0.8 Hz, 1H), 7.69 (dt, J=8.4, 0.8 Hz, 1H), 4.69 (d, J=5.6 Hz, 1H), 4.05 (dd, J=11.4, 1.7 Hz, 1H), 3.84 (td, J=11.3, 2.4 Hz, 1H), 3.72 (ddd, J=10.9, 4.7, 2.7 Hz, 1H), 3.60 (ddd, J=5.7, 2.8, 1.4 Hz, 1H), 3.54 (dd, J=11.5, 2.8 Hz, 1H), 2.56-2.51 (m, 1H), 2.02 (s, 2H), 1.62-1.52 (m, 1H).


NOE: 2.09 (NH2): 3.55; 3.70; 3.78 4.80 (OH): 1.61; 3.78


Intermediate 41:




embedded image


Step 1:


Step 1 was performed in analogy to Step 1 in the preparation of Intermediate 38, starting from 1-(2-fluoro-4-trifluoromethyl-phenyl)-3,6-dioxa-bicyclo[3.1.0]hexane (750, 3.02 mmol, prepared as described in Step 3 in the preparation of Intermediate 37) to obtain 4-azido-4-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-furan-3-ol as major regioisomer and 4-Azido-3-(2-fluoro-4-trifluoromethyl-phenyl)-tetrahydro-furan-3-ol as minor regioisomer. (900 mg, regioisomer ratio 82/18 determined by NMR)


GC-MS (METHOD 9): Rt=9.22 min


MS: m/z=190 (fragment) (M)+


1H NMR (500 MHz, DMSO-d6) δ 7.82-7.77 (m, 1H), 7.66-7.60 (m, 2H), 5.72 (d, J=5.4 Hz, 1H), 4.59-4.55 (m, 1H), 4.39 (dd, J=9.7, 1.8 Hz, 1H), 4.23 (d, J=9.7 Hz, 1H), 4.18 (dd, J=9.6, 4.1 Hz, 1H), 3.79-3.75 (d, 1H).


Step 2:


Step 2 was performed in analogy to Step 1 in the preparation of Intermediate 38, starting from the regioisomeric mixture, to obtain [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester as preferred regioisomer and 4-(2-Fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester (670 mg) (regioisomer ratio 80/20 determined by NMR).


GC-MS (METHOD 9): Rt=10.67 min


MS: m/z=265 (fragment) (M)+


Step 3:


Step 3 was performed in analogy to Step 3 in the preparation of Intermediate 38, starting from the regioisomeric mixture of [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester and 4-(2-Fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester (670 mg, 1.47 mmol), to obtain [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-oxo-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester (455 mg).


GC-MS (METHOD 9): Rt=10.15 min


MS: m/z=249 (fragment) (M)+


Step 4:


Step 4 was performed in analogy to Step 4 in the preparation of Intermediate 38, starting from [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-oxo-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester (455 mg, 1.23) to obtain [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-4-methyl-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester as -racemic mixture (TRANS/CIS diastereoisomeric ratio 91/9, determined by NMR). (365 mg).


LC-MS (METHOD 10): Rt=3.46-3.62 min


MS (ESI pos): m/z=280 (fragment) (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 7.64 (t, J=7.9 Hz, 1H), 7.51 (d, J=9.5 Hz, 2H), 7.04 (s, 1H), 4.98 (s, 1H), 4.72-4.65 (m, 1H), 4.13 (d, J=8.4 Hz, 1H), 3.94 (d, J=8.8 Hz, 1H), 3.60 (d, J=8.8 Hz, 1H), 1.31 (s, 12H).


NOE: 7.04 (NH): 3.94; 4.13; 1.31 4.98 (OH): 1.31; 4.68 1.31 (Me): 7.04; 4.13; 4.98


Step 5:


Step 5 was performed in analogy to Step 5 in the preparation of Intermediate 38, starting from [3-(2-fluoro-4-trifluoromethyl-phenyl)-4-hydroxy-4-methyl-tetrahydro-furan-3-yl]-carbamic acid tert-butyl ester (365 mg, 1.0 mmol) to obtain 4-amino-4-(2-fluoro-4-trifluoromethyl-phenyl)-3-methyl-tetrahydro-furan-3-ol trifluoroacetate, as racemic mixture (TRANS/CIS diastereoisomeric ratio 90/10, determined by NMR). (378 mg)


LC-MS (METHOD 1): Rt=2.91-3.19 min


MS (ESI pos): m/z=280 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 8.67 (s, 3H), 7.88-7.79 (m, 2H), 7.70 (dd, J=8.5, 1.9 Hz, 1H), 5.62 (s, 1H), 4.62 (dd, J=10.0, 1.1 Hz, 1H), 4.19 (dd, J=10.0, 1.5 Hz, 1H), 3.98 (d, J=9.6 Hz, 1H), 3.80 (d, J=9.6 Hz, 1H), 1.48 (d, J=1.3 Hz, 3H).


Intermediate 42:




embedded image


Intermediate 42 was prepared in analogy to Intermediate 35, starting from 4-iodobenzotrifluoride (3 g, 10.7 mmol) to obtain, after chromatographic purification in the third step (eluent: cyclohexane/EtOAc; gradient from 0% to 100% of EtOAc), 105 mg the title compound, as racemic mixture (TRANS/CIS diastereoisomeric ratio 93/7, determined by NMR).


LC-MS (METHOD 5): Rt=0.77 min


MS (ESI pos): m/z=262 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ 7.71-7.67 (m, 2H), 7.65-7.61 (m, 2H), 4.58 (d, J=5.9 Hz, 1H), 4.05 (dd, J=11.5, 1.5 Hz, 1H), 3.92-3.86 (m, 1H), 3.67 (ddd, J=11.1, 4.9, 2.3 Hz, 1H), 3.57-3.52 (m, 1H), 3.48 (dq, J=5.7, 1.5 Hz, 1H), 2.49-2.41 (m, 1H), 1.97 (s, 2H), 1.49-1.42 (m, 1H).


NOE 1.97 (NH2): 3.48; 4.05; 1.47 4.58 (OH): 2.46; 3.55 3.48 (CH): 1.97; 3.89


Intermediate 43:




embedded image


Step 1:


Step 1 was performed in analogy to Step 2 in the preparation of Intermediate 38, starting from 4-Azido-4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-tetrahydro-pyran-3-ol (1.7 g, 4.77 mmol) to obtain after filtration on silica, 1.3 g of the title compound as TRANS-racemic mixture


LC-MS (METHOD 1): Rt=3.93 min


MS (APCI): m/z=381 (M+H)+


Step 2:


To a solution of oxalylchloride (0.25 mL, 2.6 mmol) in dry DCM (20 mL) at −55° C., DMSO (0.37 mL, 0.51 mmol) was added dropwise. After 20 min, a solution of (3-fluoro-5-trifluoromethyl-pyridin-2-yl)-3-hydroxy-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester (1.0 g, 2.37 mmol) dissolved in 3 mL of dry DCM was added dropwise. Mixture is stirred for 1 h at −70° C., then TEA was added dropwise, stirred for 1.30 hrs at −40° C. and then allowed to reach room temperature. Mixture was stirred at room temperature for 48 hrs. Solvent was evaporated, residue diluted with 50 ml of ethyl acetate and washed with 3×10 ml of water. Organic phase was separated, dried over sodium sulfate to obtain 0.92 g of the desired compound used in the next step without further purification.


LC-MS (METHOD 1): Rt=3.79 min


MS (APCI): m/z=475 (M+H)+


Step 3:


Step 3 was performed in analogy to Step 4 in the preparation of Intermediate 38, starting from 4-(3-fluoro-5-trifluoromethyl-pyridin-2-yl)-3-oxo-tetrahydro-pyra n-4-yl]-carbamic acid tert-butyl ester (0.92 g, 2.41 mmol) to obtain after chromatographic purification (eluent: cyclohexane/EtOAc; gradient from 100% to 40% of EtOAc), 0.190 g of CIS stereoisomer and 0.35 g of TRANS stereoisomer, both as racemic mixture.


LC-MS (METHOD 9): Rt=10.49 min (CIS stereoisomer)


MS (ESI): m/z=394 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.88 (s, 3H) 1.22-1.38 (m, 9H) 2.26-2.42 (m, 1H) 2.80 (td, J=13.54, 4.65 Hz, 1H) 3.25-3.29 (m, 1H) 3.54-3.63 (m, 2H) 3.78 (br dd, J=11.37, 3.42 Hz, 1H) 5.31 (s, 1H) 6.40 (br s, 1H) 8.18 (br d, J=11.61 Hz, 1H) 8.81-8.86 (m, 1H)


NOE: 6.40 (NH): 5.31 5.31 (OH): 6.40


LC-MS (METHOD 9): Rt=10.76 min (TRANS stereoisomer)


MS (ESI): m/z=394 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 1.03 (d, J=2.93 Hz, 3H) 1.10-1.44 (m, 9H) 1.96-2.03 (m, 1H) 3.06-3.18 (m, 1H) 3.24 (d, J=11.98 Hz, 1H) 3.54-3.69 (m, 1H) 3.72-3.86 (m, 2H) 4.71 (s, 1H) 7.01 (br s, 1H) 8.09 (br d, J=11.74 Hz, 1H) 8.79 (s, 1H)


NOE: 7.01 (NH): 1.02; 3.62; 3.77 4.71 (OH): 3.12; 3.24


Step 4:


Step 4 was performed in analogy to Step 5 in the preparation of Intermediate 38, starting from TRANS [(R)-4-(3-Fluoro-5-trifluoromethyl-pyridin-2-yl)-3-hydroxy-3-methyl-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester (0.350 g, 0.89 mmol) to obtain 0.25 g of desired intermediate 42, 4-Amino-4-(3-fluoro-5-trifluoro methyl-pyridin-2-yl)-3-methyl-tetrahydro-pyran-3-ol, as trifloroacetate salt.


LC-MS (METHOD 1): Rt=2.90 min


MS (APCI): m/z=295 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 1.00 (s, 3H) 1.87-1.93 (m, 1H) 3.14 (ddd, J=14.49, 9.72, 4.65 Hz, 1H) 3.39-3.43 (m, 1H) 3.66-3.69 (m, 1H) 3.81-3.88 (m, 1H) 4.04 (dt, J=11.86, 4.34 Hz, 1H) 5.48 (br s, 1H) 8.43-8.48 (m, 1H) 8.72 (br s, 3H) 8.94 (s, 1H)


NOE: 8.72 (NH3+): 1.00; 1.90; 3.68; 3.84 1.0 (Me): 8.72; 1.89; 3.66


Intermediate 44:




embedded image


[4-(2-Fluoro-4-trifluoromethyl-phenyl)-3-oxo-tetrahydro-pyran-4-yl]-carbamic acid tert-butyl ester (obtained from Step 3 in the preparation of Intermediate 38; 90 mg, 0.23 mmol) was dissolved in 1 mL of 1.4-dioxane. 1.67 mL of a 1.4M solution of hydrogen chloride in 1.4-dioxane was added, the reaction mixture was stirred at room temperature for 1 hr and then concentrated under vacuum. The crude obtained was stripped twice with ethyl ether to give 70 mg of the title compound, used in the next step without further purification.


LC-MS (METHOD 1): Rt=3.70 min


MS (ESI pos): m/z=278 (M+H)+


EXAMPLE COMPOUNDS
Example 1



embedded image


To a suspension of Intermediate 5 (40.68 mg, 0.20 mmol) in 1.0 mL of dry DMF, HATU (82.47 mg, 1.3 mmol) and DIPEA (0.11 mL 0.67 mmol) were added and the reaction mixture was stirred at room temperature. A solution of Intermediate 13 (50 mg, 0.17 mmol) in 1.0 mL of dry DMF was added, and the reaction mixture was stirred at room temperature overnight. The reaction mixture was treated with basic alumina and concentrated under vacuum. The crude obtained was purified by flash chromatography (eluent: water/acetonitrile; gradient from 10% to 100% of acetonitrile) to give 62 mg of the desired compound.


LC-MS (METHOD 1): Rt=4.78 min


MS (ESI pos): m/z=463 (M+H)+


The following Examples were prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from the corresponding Acid and the corresponding Amine Intermediates:


















Starting
Starting


MS m/z
Rt



Acid
Amine
Ex.
Structure
[M + H]+
(min)
Method







 2
13
 2


embedded image


423
4.53
METHOD 1





 6
13
 3


embedded image


427
3.50
METHOD 10





 8
14
 4


embedded image


403
0.90
METHOD 6





 2
14
 5


embedded image


389
4.28
METHOD 1





 5
25
 6


embedded image


415
4.67
METHOD 1





 5
14
 7


embedded image


415
4.78
METHOD 1





 5
15
 8


embedded image


431
4.98
METHOD 1





 2
15
 9


embedded image


405
4.40
METHOD 1





 5
16
10


embedded image


411
0.80
METHOD 7





12
17
11


embedded image


428
3.21
METHOD 10





 2
17
12


embedded image


388
2.86
METHOD 10





 5
18
13


embedded image


441
0.87
METHOD 8





 5
19
14


embedded image


447
3.54
METHOD 10





 2
19
15


embedded image


421
3.26
METHOD 10





 5
20
16


embedded image


415
4.70
METHOD 1





12
21
17


embedded image


464
4.93
METHOD 1





 8
21
18


embedded image


438
3.38
METHOD 10





 2
21
19


embedded image


424
3.27
METHOD 10





 5
21
20


embedded image


450
3.59
METHOD 10





12
22
21


embedded image


446
4.52
METHOD 1





 8
22
22


embedded image


420
3.23
METHOD 10





 5
22
23


embedded image


432
3.45
METHOD 10





 2
22
24


embedded image


406
3.12
METHOD 10





12
23
25


embedded image


446
4.75
METHOD 1





 8
23
26


embedded image


420
3.13
METHOD 10





 5
23
27


embedded image


432
3.32
METHOD 10





 2
23
28


embedded image


406
3.01
METHOD 10





12
24
29


embedded image


464
3.70
METHOD 10





 5
26
30 Racemic mixture


embedded image


449
3.87
METHOD 10





 2
26
31 Racemic mixture


embedded image


423
3.60
METHOD 10





 5
27
32 Racemic mixture


embedded image


[M + Na]+ 453
4.78
METHOD 3





 5
28
33 Racemic mixture


embedded image


447
4.83
METHOD 3





 5
29
34 Racemic mixture


embedded image


435
4.92
METHOD 1





 2
29
35 Racemic mixture


embedded image


409
3.44
METHOD 10





 2
30
36 Racemic mixture


embedded image


391
3.36
METHOD 10





 5
30
37 Racemic mixture


embedded image


417
4.77
METHOD 1





 5
31
38 Racemic mixture


embedded image


401
4.71
METHOD 1





 2
31
39 Racemic mixture


embedded image


[M + Na]+ 397
2.81
METHOD 4





12
32
40 Racemic mixture


embedded image


450
3.63
METHOD 10





 8
32
41 Racemic mixture


embedded image


424
3.32
METHOD 10





12
33
42 Racemic mixture


embedded image


432
3.47
METHOD 10





 8
33
43 Racemic mixture


embedded image


406
3.14
METHOD 10





 8
34
44 Racemic mixture


embedded image


406
3.25
METHOD 10





12
34
45 Racemic mixture


embedded image


432
3.53
METHOD 10









Example 46



embedded image


Example 46 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (34.6 mg, 0.2 mmol) and amino alcohol Intermediate 35 (65 mg, 0.19 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 100% of acetonitrile), 27 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=3.70 min


MS (ESI pos): m/z=439 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 2.40 (d, J=1.00 Hz, 3H) 2.59-2.73 (m, 2H) 3.62-3.71 (m, 1H) 3.79-3.85 (m, 2H) 3.82 (br d, J=11.62 Hz, 2H) 4.02 (br d, 1H) 4.10 (m, 1H) 5.33 (br s, 1H) 7.46-7.50 (m, 1H) 7.55 (d, J=7.99 Hz, 1H) 7.73 (t, J=8.19 Hz, 1H) 8.34 (s, 1H) 8.42 (s, 1H) 8.81 (d, J=2.08 Hz, 1H) 9.19 (dd, J=1.96, 1.10 Hz, 1H)


NOE: 8.42 (NH): 4.10; 4.03; 3.65; 3.82 5.33 (OH): 4.10; 3.70


Example 47



embedded image


Further elution from the column in the preparation of Example 46 gave 5 mg of the title compound, as CIS-racemic mixture.


LC-MS (METHOD 1): Rt=3.97 min


MS (ESI pos): m/z=439 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 2.35-2.48 (m, 4H) 3.12 (br d, J=14.18 Hz, 1H) 3.47 (br t, J=11.86 Hz, 1H) 3.58 (t, J=10.76 Hz, 1H) 3.72 (br dd, J=11.62, 2.57 Hz, 1H) 3.74-3.84 (m, 1H) 4.10 (dd, J=10.03, 5.14 Hz, 1H) 5.66 (s, 1H) 7.47-7.59 (m, 2H) 7.61-7.67 (m, 1H) 8.49 (s, 1H) 8.62 (s, 1H) 8.76 (d, J=1.96 Hz, 1H) 9.21 (dd, J=2.08, 1.10 Hz, 1H)


NOE: 8.62 (NH): 4.10; 5.66; 2.49; 3.78; 3.12 5.66 (OH): 8.62; 4.10; 3.78; 3.12


The following Examples were prepared in analogy to Example 46 and Example 47 starting from the corresponding acid and amino alcohol Intermediates:



















Starting







Starting
Amino-


MS m/z
Rt



Acid
alcohol
Ex.
Structure
[M + H]+
(min)
Method







5
35
48 TRANS- Racemic mixture


embedded image


465
4.12
METHOD 1





5
35
49 CIS- Racemic mixture


embedded image


465
4.33
METHOD 1









Relative stereochemistry assigned by NMR:
















Relative




Example
stereochemistry
1H-NMR
NOE



















48


embedded image


TRANS Racemic mixture
1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.11 (m, 4H) 2.11 (tt, J = 8.47, 5.10 Hz, 1 H) 2.56-2.76 (m, 2 H) 3.66 (td, J = 11.68, 2.08 Hz, 1 H) 3.81 (br d, J = 11.25 Hz, 2 H) 4.03 (d, J = 11.74 Hz, 1 H) 4.09 (m, 1 H) 5.33 (br s, 1 H) 7.46-7.50 (m, 1 H) 7.54 (d, J = 8.03 Hz, 1 H) 7.73 (t, J = 7.98 Hz, 1 H)
8.40 (NH): 4.09 5.33 (OH): 4.09





8.33 (s, 1 H) 8.40






(s, 1 H) 8.77 (d,






J = 2.20 Hz, 1 H)






9.13 (d, J = 2.20 Hz,






1 H)






49


embedded image


CIS- Racemic mixture
1H NMR (500 MHz, DMSO-d6) δ ppm 0.86-1.07 (m, 4 H) 2.06- 2.16 (m, 1 H) 2.42- 2.48 (m, 1 H) 3.12 (br d, J = 14.18 Hz, 1 H) 3.26- 3.29 (m, 1 H) 3.47 (br t, J = 11.98 Hz, 1 H) 3 58 (t, J = 10.76 Hz, 1 H) 3.65-3.75 (m, 1 H) 3.78 (dd, J = 11.49, 5.14 Hz, 1 H) 4.09 (dd, J = 10.27, 5.14 Hz, 1 H) 5.66 (br s, 1
8.58 (NH): 5.66; 3.58; 3.47; 3.12 5.66 (OH): 8.58; 3.77 4.09 (CH): 2.46





H) 7.48-7.60 (m,






2 H) 7.60-7.65






(m, 1 H) 8.48 (s, 1






H) 8.58 (s, 1 H)






8.71 (d, J = 2.20 Hz,






1 H) 9.14 (d,






J = 1.71 Hz, 1 H)









Example 50



embedded image


Example 50 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (100 mg, 0.56 mmol) and amino alcohol Intermediate 36 (171.4 mg, 0.62 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 0% to 60% of acetonitrile), 209 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=3.04 min


MS (ESI pos): m/z=422 (M+H)+


1H NMR (400 MHz, DMSO-d6) δ ppm 2.41 (s, 3H) 2.45-2.48 (m, 1H) 2.64-2.74 (m, 1H) 3.63-3.75 (m, 1H) 3.76-3.87 (m, 2H) 3.94 (br d, J=4.89 Hz, 1H) 4.08 (d, J=11.74 Hz, 1H) 5.30 (d, J=5.67 Hz, 1H) 7.83 (d, J=8.22 Hz, 1H) 8.10 (dd, J=8.31, 1.66 Hz, 1H) 8.38 (s, 1H) 8.44 (s, 1H) 8.82 (d, J=1.96 Hz, 1H) 8.85 (d, J=1.76 Hz, 1H) 9.20 (dd, J=1.96, 0.98 Hz, 1H)


NOE: 8.44 (NH): 4.08; 3.94; 3.68; 2.48 5.32 (OH): 8.58; 3.78, 2.68 3.94 (CH): 8.44; 2.48


The following Examples were prepared in analogy to Example 50 starting from the corresponding acid and amino alcohol Intermediates:



















Starting







Starting
Amino-


MS m/z
Rt



Acid
alcohol
Ex.
Structure
[M + H]+
(min)
Method







12
36
51 TRANS- Racemic mixture


embedded image


462
3.87
METHOD 1





 5
36
52 TRANS- Racemic mixture


embedded image


448
3.58
METHOD 1









Relative stereochemistry assigned by NMR:















Relative



Example
stereochemistry
1H-NMR


















51


embedded image


TRANS Racemic mixture
1H NMR (400 MHz, DMSO-d6) δ ppm 0.86-1.07 (m, 4 H) 2.03- 2.17 (m, 1 H) 2.47 (s, 3 H) 2.49- 2.5 (m, 1H) 2.68 (td, J = 12.57, 4.40 Hz, 1 H) 3.64-3.72 (m, 1 H) 3.77-3.88 (m, 3 H) 4.10 (d, J = 12.13 Hz, 1 H) 5.28 (d, J = 5.67 Hz, 1 H) 7.83 (d, J = 8.22 Hz, 1 H) 8.08 (d, J = 7.73 Hz, 1 H) 8.65 (s, 1 H) 8.71 (d, J = 1.96 Hz, 1 H) 8.83 (s, 1 H) 9.03 (d, J = 1.96 Hz, 1 H)





52


embedded image


TRANS Racemic mixture
1H NMR (400 MHz, DMSO-d6) δ ppm 0.91-1.09 (m, 4 H) 2.08- 2.15 (m, 1 H) 2.46 (s, 1 H) 2.59- 2.78 (m, 1 H) 3.64-3.75 (m, 1 H) 3.76-3.86 (m, 2 H) 3.92 (br d, J = 4.21 Hz, 1 H) 4.08 (d, J = 11.93 Hz, 1 H) 5.29 (d, J = 5.77 Hz, 1 H) 7.83 (d, J = 8.31 Hz, 1 H) 8.10 (dd, J = 8.12, 2.15 Hz, 1 H) 8.37 (s, 1 H) 8.43 (s, 1 H) 8.77 (d, J = 2.15 Hz, 1 H) 8.84 (d, J = 1.96 Hz, 1 H) 9.14 (d, J = 2.15 Hz, 1 H)









Example 53



embedded image


Example 53 was synthesized in analogy to Example 1, starting from acid


Intermediate 5 (98 mg, 0.48 mmol) and amino alcohol Intermediate 38 (198 mg, 0.48 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 80% of acetonitrile), 7 mg of the title compound, as CIS-racemic mixture.


LC-MS (METHOD 1): Rt=4.45 min


MS (ESI pos): m/z=479 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.07 (m, 4H) 1.12 (s, 3H) 2.12 (tt, J=8.38, 5.20 Hz, 1H) 2.72 (br t, J=11.98 Hz, 1H) 2.98 (br d, J=13.69 Hz, 1H) 3.47 (d, J=11.25 Hz, 1H) 3.56 (br t, J=11.74 Hz, 1H) 3.72-3.76 (m, 1H) 3.76-3.87 (m, 1H) 5.38 (s, 1H) 7.46-7.56 (m, 2H) 7.70 (t, J=8.07 Hz, 1H) 8.35 (s, 1H) 8.72 (d, J=2.20 Hz, 1H) 8.88-8.95 (m, 1H) 9.12 (d, J=2.20 Hz, 1H)


NOE: 8.92 (NH): 5.38; 3.80 5.38 (OH): 8.92; 3.80


Example 54



embedded image


Further elution from the column in the preparation of Example 53 gave 29 mg of the title compound, as racemic mixture (TRANS/CIS diastereoisomeric ratio 92/8 determined by NMR).


LC-MS (METHOD 1): Rt=4.45 min


MS (ESI pos): m/z=479 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.95-1.07 (m, 4H) 1.10 (d, 3H) 2.12 (tt, J=8.44, 5.14 Hz, 1H) 2.51-2.56 (m, 1H) 3.03 (td, J=12.65, 4.28 Hz, 1H) 3.53-3.65 (m, 2H) 3.74-3.76 (m, 1H) 3.88 (d, J=12.23 Hz, 1H) 5.10 (s, 1H) 7.44-7.48 (dd, 1H) 7.53 (d, J=8.27 Hz, 1H) 7.66 (t, J=8.05 Hz, 1H) 8.38 (s, 1H) 8.48 (s, 1H) 8.79 (d, J=2.20 Hz, 1H) 9.16 (d, J=2.20 Hz, 1H)


NOE: 8.48 (NH): 1.09; 3.88; 2.53 5.10 (OH): 3.04, 3.57 1.09 (Me): 8.48; 3.57; 2.53; 3.88


Example 55



embedded image


Example 55 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (304 mg, 1.68 mmol) and amino alcohol Intermediate 38 (700 mg, 1.68 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 100% of acetonitrile), 300 mg of the title compound, as racemic mixture (TRANS/CIS diastereoisomeric ratio 96/4 determined by NMR).


LC-MS (METHOD 1): Rt=4.02 min


MS (ESI pos): m/z=453 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 1.10 (d, J=1.00 Hz, 3H) 2.41 (d, J=0.86 Hz, 3H) 2.52-2.58 (m, 1H) 3.03 (br d, J=4.16 Hz, 1H) 3.46-3.66 (m, 2H) 3.72-3.82 (m, 1H) 3.88 (d, J=12.23 Hz, 1H) 5.10 (s, 1H) 7.46 (br d, J=12.72 Hz, 1H) 7.50-7.57 (m, 1H) 7.67 (t, J=8.23 Hz, 1H) 8.35-8.39 (m, 1H) 8.50 (s, 1H) 8.83 (d, J=1.96 Hz, 1H) 9.18-9.24 (m, 1H)


NOE: 8.50 (NH): 1.10; 3.89; 3.64 5.10 (OH): 3.03, 3.58 1.10 (Me): 8.50; 3.56; 3.89


Example 56



embedded image


Example 56 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (36 mg, 0.21 mmol) and amino alcohol Intermediate 39 (55 mg, 0.2 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 65% of acetonitrile), 52 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=3.57 min


MS (ESI pos): m/z=440 (M+H)+


1H NMR (400 MHz, DMSO-d6) δ ppm 2.40 (s, 3H) 2.42-2.48 (m, 1H) 2.89 (td, J=13.35, 4.40 Hz, 1H) 3.65 (br t, J=11.30 Hz, 1H) 3.79-3.89 (m, 2H) 4.01 (d, J=12.42 Hz, 1H) 4.09 (s, 1H) 5.35 (br s, 1H) 8.11 (d, J=11.84 Hz, 1H) 8.35 (s, 1H) 8.46 (s, 1H) 8.81 (d, J=1.76 Hz, 2H) 9.20 (s, 1H)


Example 57



embedded image


Example 57 was synthesized in analogy to Example 1, starting from acid


Intermediate 5 (42 mg, 0.21 mmol) and amino alcohol Intermediate 39 (55 mg, 0.2 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 48 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=4.00 min


MS (ESI pos): m/z=466 (M+H)+


1H NMR (400 MHz, DMSO-d6) δ ppm 0.87-1.11 (m, 4H) 2.07-2.15 (m, 1H) 2.47 (m, 1H) 2.89 (td, J=13.11, 4.11 Hz, 1H) 3.64 (br t, J=11.54 Hz, 1H) 3.78-3.89 (m, 2H) 4.01 (d, J=12.52 Hz, 1H) 4.09 (br s, 1H) 5.35 (br s, 1H) 8.11 (d, J=11.93 Hz, 1H) 8.34 (s, 1H) 8.44 (s, 1H) 8.77 (s, 1H) 8.81 (s, 1H) 9.14 (d, J=1.96 Hz, 1H)


Example 58



embedded image


Example 58 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (187 mg, 1.06 mmol) and amino alcohol Intermediate 41 (378 mg, 0.96 mmol) to give, after flash chromatographic purification (eluent: dichloromethane/MeOH; gradient from 0% to 60% of MeOH), 22 mg of the title compound, as racemic mixture (TRANS/CIS diastereoisomeric ratio 91/9)


LC-MS (METHOD 10): Rt=3.16 min


MS (ESI pos): m/z=439 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 1.56 (s, 3H) 2.40 (d, J=0.73 Hz, 4H) 3.78 (d, J=9.05 Hz, 1H) 3.90 (d, J=9.05 Hz, 1H) 4.39 (d, J=8.80 Hz, 1H) 4.90 (d, J=8.80 Hz, 1H) 5.31 (s, 1H) 7.48-7.56 (m, 2H) 7.71 (t, J=8.07 Hz, 1H) 8.37 (s, 1H) 8.67 (s, 1H) 8.82 (d, J=1.96 Hz, 1H) 9.09-9.26 (m, 1H) 9.21 (dd, J=2.08, 1.10 Hz, 1H) NOE: 8.67 (NH): 1.56; 3.90; 4.38 5.31 (OH): 3.78, 4.90 1.56 (Me): 8.67


Example 59



embedded image


Example 59 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (35.6 mg, 0.20 mmol) and amino alcohol Intermediate 42 (50 mg, 0.19 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 65% of acetonitrile), 42 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=3.77 min


MS (ESI pos): m/z=421 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 2.40 (d, J=0.98 Hz, 3H) 2.51-2.54 (m, 1H) 2.62-2.70 (m, 1H) 3.67 (td, J=11.68, 1.83 Hz, 1H) 3.75-3.82 (m, 2H) 3.82-3.89 (m, 1H) 4.06 (d, J=11.49 Hz, 1H) 5.14 (br d, J=4.16 Hz, 1H) 7.62-7.70 (m, 4H) 8.35 (s, 1H) 8.37 (s, 1H) 8.80 (d, J=1.96 Hz, 1H) 9.19 (dd, J=2.08, 1.10 Hz, 1H)


Example 60



embedded image


Example 60 was synthesized in analogy to Example 1, starting from acid


Intermediate 5 (40.8 mg, 0.2 mmol) and amino alcohol Intermediate 42 (50 mg, 0.19 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 39 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=4.07 min


MS (ESI pos): m/z=424 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.86-1.06 (m, 4H) 2.07-2.14 (m, 1H) 2.51-2.53 (m, 1H) 2.62-2.70 (m, 1H) 3.66 (td, J=11.74, 1.71 Hz, 1H) 3.75-3.87 (m, 3H) 4.07 (d, J=11.25 Hz, 1H) 5.14 (d, J=5.62 Hz, 1H) 7.62-7.70 (m, 4H) 8.35 (s, 1H) 8.36 (s, 1H) 8.76 (d, J=2.20 Hz, 1H) 9.13 (d, J=2.20 Hz, 1H)


Example 61



embedded image


Example 61 was synthesized in analogy to Example 1, starting from acid


Intermediate 12 (82 mg, 0.29 mmol) and amino alcohol Intermediate 39 (66.7 mg, 0.31 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 110 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=4.22 min


MS (ESI pos): m/z=480 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.90-1.06 (m, 4H) 2.09 (tt, J=8.44, 5.14 Hz, 1H) 2.45 (s, 3H) 2.47 (m, 1H) 2.88 (td, J=13.27, 4.28 Hz, 1H) 3.59-3.67 (m, 1H) 3.77-3.88 (m, 2H) 4.01-4.05 (m, 1H) 4.08 (br d, J=5.14 Hz, 1H) 5.32 (d, J=5.62 Hz, 1H) 8.11 (dd, J=11.86, 1.59 Hz, 1H) 8.69 (s, 1H) 8.71 (d, J=2.36 Hz, 1H) 8.81 (s, 1H) 9.03 (d, J=2.20 Hz, 1H)


NOE: 8.69 (NH): 4.08; 2.47 3.64 5.32 (OH): 3.82, 2.88 4.08 (CH): 8.69; 2.47


Example 62



embedded image


Example 62 was synthesized in analogy to Example 1, starting from acid


Intermediate 12 (103 mg, 0.6 mmol) and amino alcohol Intermediate 43 (250 mg, 0.6 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 115 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=4.40 min


MS (ESI pos): m/z=494 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.92-1.06 (m, 4H) 1.21 (d, J=3.18 Hz, 3H) 2.10 (tt, J=8.47, 5.23 Hz, 1H) 2.30-2.36 (m, 1H) 2.47-2.49 (m, 3H) 3.22-3.29 (m, 1H) 3.49-3.54 (m, 1H) 3.62 (br t, J=10.88 Hz, 1H) 3.76-3.88 (m, 2H) 4.96 (s, 1H) 8.08 (dd, J=11.98, 1.47 Hz, 1H) 8.72 (d, J=1.96 Hz, 1H) 8.78 (s, 1H) 8.84 (s, 1H) 9.05 (d, J=2.20 Hz, 1H)


NOE: 8.78 (NH): 1.21; 2.34; 3.62 4.96 (OH): 3.51, 3.35 1.21 (Me): 8.78; 2.34


Example 63



embedded image


Example 63 was synthesized in analogy to Example 1, starting from acid


Intermediate 5 (45 mg, 0.22 mmol) and amino alcohol Intermediate 40 (55 mg, 0.21 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 55 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=3.82 min


MS (ESI pos): m/z=448 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.91-1.08 (m, 4H) 2.06-2.17 (m, 1H) 2.41-2.48 (m, 1H) 2.76-2.84 (m, 1H) 3.65-3.76 (m, 2H) 3.92 (dt, J=11.23, 3.58 Hz, 1H) 3.95-4.00 (m, 1H) 4.04 (br s, 1H) 5.24 (d, J=4.03 Hz, 1H) 7.75 (d, J=8.44 Hz, 1H) 8.11 (dd, J=8.50, 2.02 Hz, 1H) 8.33 (s, 1H) 8.43 (s, 1H) 8.76 (d, J=2.08 Hz, 1H) 8.88 (dd, J=1.53, 0.79 Hz, 1H) 9.13 (d, J=2.20 Hz, 1H)


NOE 8.43 (NH): 4.04; 2.44; 3.98 5.24 (OH): 3.66, 2.80 4.04 (CH): 8.43; 2.44


Example 64



embedded image


Example 64 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (39 mg, 0.22 mmol) and amino alcohol Intermediate 40 (55 mg, 0.21 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 65% of acetonitrile), 31 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 10): Rt=2.66 min


MS (ESI pos): m/z=422 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 2.40 (s, 3H) 2.42 (br s, 1H) 2.76-2.85 (m, 1H) 3.64-3.77 (m, 2H) 3.90-3.95 (m, 1H) 3.97 (br d, J=12.23 Hz, 1H) 4.05 (br s, 1H) 5.24 (d, J=5.56 Hz, 1H) 7.75 (d, J=8.44 Hz, 1H) 8.11 (dd, J=8.47, 2.11 Hz, 1H) 8.33 (s, 1H) 8.45 (s, 1H) 8.80 (d, J=1.90 Hz, 1H) 8.88 (s, 1H) 9.19 (s, 1H)


NOE: 8.45 (NH): 4.05; 2.44; 3.98; 3.68 5.24 (OH): 3.72; 3.97; 2.80 4.05 (CH): 8.45; 2.44


Example 65



embedded image


Example 65 was synthesized in analogy to Example 1, starting from acid


Intermediate 12 (78 mg, 0.36 mmol) and amino alcohol Intermediate 40 (90 mg, 0.34 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 10% to 70% of acetonitrile), 110 mg of the title compound, as TRANS-racemic mixture.


LC-MS (METHOD 1): Rt=4.07 min


MS (ESI pos): m/z=462 (M+H)+


1H NMR (500 MHz, DMSO-d6) δ ppm 0.90-1.07 (m, 4H) 2.09 (tt, J=8.59, 4.98 Hz, 1H) 2.42-2.47 (m, 4H) 2.79 (ddd, J=13.66, 11.34, 4.34 Hz, 1H) 3.64-3.76 (m, 2H) 3.91 (dt, J=11.28, 3.59 Hz, 1H) 3.99 (d, J=12.35 Hz, 1H) 3.98-3.99 (m, 1H) 4.02 (br s, 1H) 5.21 (d, J=4.52 Hz, 1H) 7.73 (d, J=8.44 Hz, 1H) 8.12 (dd, J=8.50, 2.14 Hz, 1H) 8.69 (d, J=10.11 Hz, 2H) 8.88 (dd, J=1.47, 0.73 Hz, 1H) 9.02 (d, J=1.83 Hz, 1H)


Example 66



embedded image


Example 66 was synthesized in analogy to Example 1, starting from acid


Intermediate 5 (16.8 mg, 0.08 mmol) and amino alcohol Intermediate 44 (20 mg, 0.08 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 0% to 100% of acetonitrile), 11 mg of the title compound.


LC-MS (METHOD 1): Rt=4.78 min


MS (ESI pos): m/z=463 (M+H)+


Example 67



embedded image


Example 67 was synthesized in analogy to Example 1, starting from acid


Intermediate 2 (19.7 mg, 0.11 mmol) and amino alcohol Intermediate 44 (35 mg, 0.11 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 0% to 100% of acetonitrile), 24 mg of the title compound.


LC-MS (METHOD 1): Rt=4.40 min


MS (ESI pos): m/z=437 (M+H)+


Examples 68, 69, 70, 71

Examples 68, 69, 70 and 71 were synthesized in analogy to Example 1, starting from acid Intermediate 5 (150 mg, 0.74 mmol) and amino alcohol Intermediate 37 (230 mg, 0.71 mmol) to give, after flash chromatographic purification (eluent: water/acetonitrile; gradient from 0% to 80% of acetonitrile), 143 mg of mixture of the title compounds, which were obtained as single stereoisomers by chiral HPLC separation.


















MS
Rt (min)
Rt (min)




m/z
[LC-MS
[LC-MS


Ex. #
Structure
[M + H]+
Method]
Method]







68 CIS single stereo- isomer a


embedded image


451
4.28 METHOD 1
 9.66 [C3]





69 CIS single stereo- isomer b


embedded image


451
4.28 METHOD 1
11.30 [C3]





70 TRANS single stereo- isomer a


embedded image


451
4.21 METHOD 1
16.32, [C3]





71 TRANS single stereo- isomer b


embedded image


451
4.23 METHOD 1
22.71 [C3]









Relative stereochemistry assigned by NMR:
















Relative




Example
stereochemistry
1H-NMR
NOE



















68


embedded image


CIS single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.06 (m, 4 H) 2.07- 2.14 (m, 1 H) 3.58 (dd, J = 9.29, 5.14 Hz, 1 H) 4.02 (dd, J = 9.29, 5.87 Hz, 1 H) 4.14 (d, J = 9.78 Hz, 1 H) 4.40 (br s, 1 H) 4.80 (d, J = 10.03 Hz, 1 H) 6.34 (br s, 1 H) 7.54-7.63 (m, 2 H) 7.63-7.68 (m, 1 H) 8.39 (s, 1 H) 8.72 (d, J = 2.20 Hz,
9.11 (NH): 6.34; 4.14; 3.58 6.34 (OH): 9.11, 4.14; 3.58 4.40 (CH): 4.80





1 H) 9.09-9.13






(m, 2 H)






69


embedded image


CIS single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.06 (m, 4 H) 2.07- 2.14 (m, 1 H) 3.58 (dd, J = 9.29, 5.14 Hz, 1 H) 4.02 (dd, J = 9.29, 5.87 Hz, 1 H) 4.14 (d, J = 9.78 Hz, 1 H) 4.40 (br s, 1 H) 4.80 (d, J = 10.03 Hz, 1 H) 6.34 (br s, 1 H) 7.54-7.63 (m, 2 H) 7.63-7.68 (m, 1 H) 8.39 (s, 1 H) 8.72 (d, J = 2.20 Hz, 1 H) 9.09-9.13
9.11 (NH): 6.34; 4.14; 3.58 6.34 (OH): 9.11, 4.14; 3.58 4.40 (CH): 4.80





(m, 2 H)






70


embedded image


TRANS single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.92-1.07 (m, 4 H) 3.71- 3.80 (m, 1 H) 4.25 (dd, J = 9.78, 4.40 Hz, 1 H) 4.35 (d, J = 8.80 Hz, 1 H) 4.69 (d, J = 8.80 Hz, 1 H) 4.76 (br s, 1 H) 5.64 (br s, 1 H) 7.51-7.56 (m, 2 H) 7.73 (t, J = 7.82 Hz, 1 H) 8.28 (s, 1 H) 8.36 (s, 1 H) 8.74 (d, J = 1.96 Hz, 1 H) 9.09-9.11 (m, 1 H)
8.28 (NH): 4.69; 4.76; 4.25 5.64 (OH): 4.35; 3.76 4.76 (CH): 8.28





71


embedded image


TRANS single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 0.92-1.07 (m, 4 H) 3.71- 3.80 (m, 1 H) 4.25 (dd, J = 9.78, 4.40 Hz, 1 H) 4.35 (d, J = 8.80 Hz, 1 H) 4.69 (d, J = 8.80 Hz, 1 H) 4.76 (br s, 1 H) 5.64 (br s, 1 H) 7.51-7.56 (m, 2 H) 7.73 (t, J = 7.82 Hz, 1 H) 8.28 (s, 1 H) 8.36 (s, 1 H) 8.74 (d, J = 1.96 Hz, 1 H) 9.09-9.11 (m, 1 H)
8.28 (NH): 4.69; 4.76; 4.25 5.64 (OH): 4.35; 3.76 4.76 (CH): 8.28









Examples 72, 73, 74, 75

Examples 72, 73, 74 and 75 were synthesized in analogy to Example 1, starting from acid Intermediate 2 (92 mg, 0.52 mmol) and amino alcohol Intermediate 37 (160 mg, 43% content, 0.26 mmol) to give, after two subsequent flash chromatographic purifications (eluent: water/acetonitrile; gradient from 0% to 80% of acetonitrile; eluent: DCM/isopropyl alcohol; gradient from 0% to 30% of isopropyl alcohol), 110 mg of mixture of the title compounds, which were obtained as single stereoisomers by chiral HPLC separation.


















MS
Rt (min)
Rt (min)




m/z
[LC-MS
[Chiral HPLC


Ex. #
Structure
[M + H]+
Method]
Method]







72 CIS single stereo- isomer a


embedded image


425
3.84 METHOD 1
12.08 [C3]





73 CIS single stereo- isomer b


embedded image


425
3.85 METHOD 1
13.41 [C3]





74 TRANS single stereo- isomer a


embedded image


425
3.75 METHOD 1
22.54 [C3]





75 TRANS single stereo- isomer b


embedded image


425
3.78 METHOD 1
26.59 [C3]









The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding racemic mixture:

















Starting







Racemic


MS
Rt (min)
Rt (min)


Mixture


m/z
[LC/MS
[Chiral HPLC


Ex. #
Ex. #
Structure
[M + H]+
Method]
Method]







40
80a Single stereo- isomer a


embedded image


450
3.89 METHOD 10
 5.98 [C1]





40
80b single stereo- isomer b


embedded image


450
3.89 METHOD 10
 6.76 [C1]





46 TRANS- Racemic mixture
81a TRANS- single stereo- isomer a


embedded image


439
3.67 METHOD  1
11.19 [C4]





46 TRANS- Racemic mixture
81b TRANS- single stereo- isomer b


embedded image


439
3.67 METHOD  1
13.99 [C4]





48 TRANS- Racemic mixture
82a TRANS- single stereo- isomer a


embedded image


465
3.41 METHOD 10
11.78 [C4]





48 TRANS- Racemic mixture
82b TRANS- single stereo- isomer b


embedded image


465
3.41 METHOD 10
14.22 [C4]





55 TRANS/ CIS 96/4 Racemic mixture
83a TRANS- single stereo- isomer a


embedded image


453
METHOD  1
11.71 [C3]





55 TRANS/ CIS 96/4 Racemic mixture
83b TRANS- single stereo- isomer b


embedded image


453
4.07 METHOD  1
26.90 [C3]





55 TRANS/ CIS 96/4 Racemic mixture
84a CIS- single stereo- isomer a


embedded image


453
4.07 METHOD  1
13.67 [C3]





55 TRANS/ CIS 96/4 Racemic mixture
84b CIS- single stereo- isomer b


embedded image


453
4.07 METHOD  1
18.13 [C3]





54 TRANS/ CIS 92/8 Racemic mixture
85a CIS- single stereo- isomer a


embedded image


479
4.48 METHOD  1
 5.46 [C1]





54 TRANS/ CIS 92/8 Racemic mixture
85b CIS- single stereo- isomer b


embedded image


479
4.48 METHOD  1
 6.73 [C1]





54 TRANS/ CIS 92/8 Racemic mixture
86a TRANS- single stereo- isomer a


embedded image


479
4.48 METHOD  1
 6.08 [C1]





54 TRANS/ CIS 92/8 Racemic mixture
86b TRANS- single stereo- isomer b


embedded image


479
4.48 METHOD  1
10.75 [C1]





50 TRANS- Racemic mixture
87a TRANS- single stereo- isomer a


embedded image


422
3.08 METHOD  1
 9.97 [C6]





50 TRANS- Racemic mixture
87b TRANS- single stereo- isomer b


embedded image


422
3.08 METHOD  1
12.98 [C6]





51 TRANS- Racemic mixture
88a TRANS- single stereo- isomer a


embedded image


462
3.87 METHOD  1
 7.71 [C1]





51 TRANS- Racemic mixture
88b TRANS- single stereo- isomer b


embedded image


462
3.87 METHOD  1
 9.49 [C1]





52 TRANS- Racemic mixture
89a TRANS- single stereo- isomer a


embedded image


448
3.60 METHOD  1
11.82 [C6]





52 TRANS- Racemic mixture
89b TRANS- single stereo- isomer b


embedded image


448
3.60 METHOD  1
16   [C6]





61 TRANS- Racemic mixture
90a TRANS single stereo- isomer a


embedded image


480
4.22 METHOD  1
 4.9  [C7]





61 TRANS- Racemic mixture
90b TRANS single stereo- isomer b


embedded image


480
4.22 METHOD  1
 5.54 [C7]





62 TRANS- Racemic mixture
91a TRANS single stereo- isomer a


embedded image


494
4.4 METHOD  1
 6.68 [C1]





62 TRANS- Racemic mixture
91b TRANS single stereo- isomer b


embedded image


494
4.4 METHOD  1
12.94 [C1]





58 TRANS/ CIS 91/9 Racemic mixture
92a CIS Single stereo- isomer a


embedded image


439
3.16 METHOD 10
 4.76 [C1]





58 TRANS/ CIS 91/9 Racemic mixture
92b CIS Single stereo- isomer b


embedded image


439
3.16 METHOD 10
 8.24 [C1]





58 TRANS/ CIS 91/9 Racemic mixture
93a TRANS Single stereo- isomer a


embedded image


439
3.16 METHOD 10
 7.20 [C1]





58 TRANS/ CIS 91/9 Racemic mixture
93b TRANS Single stereo- isomer b


embedded image


439
3.16 METHOD 10
16.60 [C1]









Relative stereochemistry assigned by NMR:
















Relative




Example
stereochemistry
1H-NMR
NOE



















81a


embedded image


TRANS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 2.40 (s, 3 H) 2.60-2.71 (m, 2 H) 3.63-3.69 (m, 1 H) 3.82 (br d, J = 11.98 Hz, 2 H) 4.03 (d, J = 11.74 Hz, 1 H) 4.08- 4.11 (m, 1 H) 5.32 (d, J = 5.38 Hz, 1 H) 7.46-7.56 (m, 2 H) 7.73 (t, J = 7.51 Hz, 1 H) 8.34 (s, 1 H) 8.42 (s, 1 H) 8.81 (d, J = 2.20 Hz,
8.42 (NH): 4.08; 4.03; 3.65; 3.82 5.32 (OH): 4.10; 3.70.





1 H) 9.17-9.19






(m, 1






82a


embedded image


TRANS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.93-1.06 (m, 4 H) 2.10 (tt, J = 8.38, 5.20 Hz, 1 H) 2.59-2.71 (m, 2 H) 3.66 (td, J = 11.68, 2.08 Hz, 1 H) 3.81 (br d, J = 11.25 Hz, 2 H) 4.03 (d, J = 11.86 Hz, 1 H) 4.09 (br d, J = 4.65 Hz, 1 H) 5.32 (d, J = 5.38 Hz, 1 H) 7.48 (d, J = 12.47 Hz, 1 H) 7.54 (dd, J = 8.31, 1.22 Hz, 1 H) 7.73
8.40 (NH): 4.09 5.32 (OH): 4.09





(t, J = 8.19 Hz, 1 H)






8.33 (s, 1 H) 8.40






(s, 1 H) 8.77 (d,






J = 2.20 Hz, 1 H)






9.13 (s, 1 H)






83a


embedded image


TRANS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 1.10 (d, J = 2.57 Hz, 3 H) 2.41 (d, J = 0.73 Hz, 3 H) 2.52-2.57 (m, 1 H) 3.03 (br d, J = 4.40 Hz, 1 H) 3.56-3.58 (m, 1 H) 3.60-3.65 (m, 1 H) 3.75 (br dd, J = 11.49, 2.57 Hz, 1 H) 3.88 (d, J = 12.23 Hz, 1 H) 5.10 (s, 1 H) 7.46 (dd, J = 12.78,
8.50 (NH): 1.10; 3.88; 2.53 5.10 (OH): 3,57, 3.03 1.10 (Me): 8.50; 3.88; 2.53





1.41 Hz, 1 H) 7.53 (dd,






J = 8.31, 1.34 Hz, 1






H) 7.67 (t, J =






8.25 Hz, 1 H) 8.39 (s, 1






H) 8.50 (s, 1 H)






8.83 (d, J = 2.08 Hz,






1 H) 9.23 (dd,






J = 1.96, 1.10 Hz, 1






H)






84a


embedded image


CIS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 1.12 (s, 2 H) 2.36-2.47 (m, 3 H) 2.67-2.84 (m, 1 H) 2.98 (br d, J = 14.18 Hz, 1 H) 3.47 (d, J = 11.00 Hz, 1 H) 3.56 (br t, J = 11.98 Hz, 1 H) 3.75 (br dd, J = 11.37, 3.55 Hz, 1 H) 3.81 (d, J = 11.25 Hz, 1 H) 5.39 (s, 1 H) 7.46-
8.96 (NH): 5.39; 2.73 5.39 (OH): 8.96; 2.73





7.55 (m, 2 H) 7.70






(t, J = 8.07 Hz, 1 H)






8.35 (s, 1 H) 8.77






(d, J = 1.96 Hz, 1 H)






8.96 (s, 1 H) 9.18






(dd, J = 2.08,






1.10 Hz, 1 H)






84b


embedded image


CIS-single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 1.12 (s, 2 H) 2.36-2.47 (m, 3 H) 2.67-2.84 (m, 1 H) 2.98 (br d, J = 14.18 Hz, 1 H) 3.47 (d, J = 11.00 Hz, 1 H) 3.56 (br t, J = 11.98 Hz, 1 H) 3.75 (br dd, J = 11.37, 3.55 Hz, 1 H) 3.81 (d, J = 11.25 Hz, 1 H) 5.39 (s, 1 H) 7.46-
8.96 (NH): 5.39; 2.73 5.39 (OH): 8.96; 2.73





7.55 (m, 2 H) 7.70






(t, J = 8.07 Hz, 1 H)






8.35 (s, 1 H) 8.77






(d, J = 1.96 Hz, 1 H)






8.96 (s, 1 H) 9.18






(dd, J = 2.08,






1.10 Hz, 1 H)






83b


embedded image


TRANS-single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 1.10 (d, J = 2.57 Hz, 3 H) 2.41 (d, J = 0.73 Hz, 3 H) 2.52-2.57 (m, 1 H) 3.03 (br d, J = 4.40 Hz, 1 H) 3.56-3.58 (m, 1 H) 3.60-3.65 (m, 1 H) 3.75 (br dd, J = 11.49, 2.57 Hz, 1 H) 3.88 (d, J = 12.23 Hz, 1 H) 5.10 (s, 1 H) 7.46
8.50 (NH): 1.10; 3.88; 2.53 5.10 (OH): 3.57, 3.03 1.10 (Me): 8.50; 3.88; 2.53





(dd, J = 12.78,






1.41 Hz, 1 H) 7.53 (dd,






J = 8.31, 1.34 Hz, 1






H) 7.67 (t, J =






8.25 Hz, 1 H) 8.39 (s, 1






H) 8.50 (s, 1 H)






8.83 (d, J = 2.08 Hz,






1 H) 9.23 (dd,






J = 1.96, 1.10 Hz, 1






H)






85a


embedded image


CIS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.07 (m, 4 H) 1.12 (s, 3 H) 2.12 (tt, J = 8.38, 5.20 Hz, 1 H) 2.72 (br t, J = 11.98 Hz, 1 H) 2.98 (br d, J = 13.69 Hz, 1 H) 3.47 (d, J = 11.25 Hz, 1 H) 3.56 (br t, J = 11.74 Hz, 1 H) 3.75 (br dd, J = 11.37, 3.30 Hz, 1 H) 3.81 (d, J = 11.25 Hz, 1 H) 5.38 (s, 1 H) 7.46-
8.92 (NH): 5.38; 3.81; 3.56 5.38 (OH): 8.92; 1.12 (Me): 3.47; 2.72





7.56 (m, 2 H) 7.70






(t, J = 8.07 Hz, 1 H)






8.35 (s, 1 H) 8.72






(d, J = 2.20 Hz, 1 H)






8.92 (s, 1 H) 9.12






(d, J = 2.20 Hz, 1 H)






86a


embedded image


TRANS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.95-1.07 (m, 5 H) 1.10 (d, J = 2.57 Hz, 2 H) 2.12 (tt, J = 8.44, 5.14 Hz, 1 H) 2.51- 2.56 (m, 1 H) 3.03 (td, J = 12.65, 4.28 Hz, 1 H) 3.54- 3.59 (m, 1 H) 3.62 (t, J = 11.25 Hz, 1 H) 3.75 (br dd, J = 11.25, 2.45 Hz, 1 H) 3.88 (d, J = 12.23 Hz, 1 H)
8.48 (NH): 1.10; 3.88; 2.53 5.10 (OH): 3.57; 3.03 1.10 (Me): 8.48; 3.88; 2.53





5.10 (s, 1 H) 7.46






(br d, J = 12.96 Hz,






1 H) 7.47-7.56






(m, 1 H) 7.59-






7.71 (m, 1 H) 8.35-






8.40 (m, 1 H)






8.48 (s, 1 H) 8.79






(d, J = 2.20 Hz, 1 H)






9.16 (d, J = 2.20 Hz,






1 H)






85b


embedded image


CIS-single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 0.94-1.07 (m, 4 H) 1.12 (s, 3 H) 2.12 (tt, J = 8.38, 5.20 Hz, 1 H) 2.72 (br t, J = 11.98 Hz, 1 H) 2.98 (br d, J = 13.69 Hz, 1 H) 3.47 (d, J = 11.25 Hz, 1 H) 3.56 (br t, J = 11.74 Hz, 1 H) 3.75 (br dd, J = 11.37, 3.30 Hz, 1 H) 3.81 (d, J = 11.25 Hz, 1 H)
8.92 (NH): 5.38; 3.81; 3.56 5.38 (OH): 8.92; 1.12 (Me): 3.47; 2.72





5.38 (s, 1 H) 7.46-






7.56 (m, 2 H) 7.70






(t, J = 8.07 Hz, 1 H)






8.35 (s, 1 H) 8.72






(d, J = 2.20 Hz, 1 H)






8.92 (s, 1 H) 9.12






(d, J = 2.20 Hz, 1 H)






86b


embedded image


TRANS-single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 0.95-1.07 (m, 5 H) 1.10 (d, J = 2.57 Hz, 2 H) 2.12 (tt, J = 8.44, 5.14 Hz, 1 H) 2.51- 2.56 (m, 1 H) 3.03 (td, J = 12.65, 4.28 Hz, 1 H) 3.54- 3.59 (m, 1 H) 3.62 (t, J = 11.25 Hz, 1 H) 3.75 (br dd, J = 11.25, 2.45 Hz, 1 H) 3.88 (d, J = 12.23 Hz, 1 H)
8.48 (NH): 1.10; 3.88; 2.53 5.10 (OH): 3.57; 3.03 1.10 (Me): 8.48; 3.88; 2.53





5.10 (s, 1 H) 7.46






(br d, J = 12.96 Hz,






1 H) 7.47-7.56






(m, 1 H) 7.59-






7.71 (m, 1 H) 8.35-






8.40 (m, 1 H)






8.48 (s, 1 H) 8.79






(d, J = 2.20 Hz, 1 H)






9.16 (d, J = 2.20 Hz,






1 H)






87a


embedded image


TRANS-single stereoisomer a
1H NMR (400 MHz, DMSO-d6) δ ppm 2.41 (s, 3 H) 2.47 (m, 1 H) 2.63- 2.74 (m, 1 H) 3.63-3.75 (m, 1 H) 3.76-3.87 (m, 2 H) 3.94 (br d, J = 4.89 Hz, 1 H) 4.08 (d, J = 11.74 Hz, 1 H) 5.30 (d, J = 5.67 Hz, 1 H) 7.83 (d, J = 8.22 Hz, 1 H) 8.10 (dd, J = 8.31, 1.66 Hz, 1
8.44 (NH): 4.08; 3.94; 2.47 5.30 (OH): 3.81, 2.68 3.94 (CH): 8.44; 2.47





H) 8.38 (s, 1 H)






8.44 (s, 1 H) 8.82






(d, J = 1.96 Hz, 1 H)






8.85 (d, J = 1.76 Hz,






1 H) 9.20 (dd,






J = 1.96, 0.98 Hz, 1






H)






88a


embedded image


TRANS-single stereoisomer a
1H NMR (500 MHz, DMSO-d6) δ ppm 0.87-0.99 (m, 2 H) 1.00- 1.06 (m, 2 H) 2.10 (tt, J = 8.47, 5.10 Hz, 1 H) 2.46 (s, 3 H) 2.51-2.53 (m, 1 H) 2.64-2.71 (m, 1 H) 3.64- 3.72 (m, 1 H) 3.77- 3.88 (m, 3 H) 4.10 (d, J = 11.49 Hz, 1 H) 5.29 (br s, 1 H) 7.84 (d, J = 8.07 Hz, 1 H) 8.08 (dd, J = 8.19,






2.08 Hz, 1 H) 8.65






(s, 1 H) 8.71 (d,






J = 2.20 Hz, 1 H)






8.83 (d, J = 2.20 Hz,






1 H) 9.04 (d,






J = 1.71 Hz, 1 H)






89b


embedded image


TRANS-single stereoisomer b
1H NMR (500 MHz, DMSO-d6) δ ppm 0.92-1.07 (m, 4 H) 2.12 (tt, J = 8.47, 5.10 Hz, 1 H) 2.44-2.48 (m, 1 H) 2.64-2.72 (m, 1 H) 3.65- 3.73 (m, 1 H) 3.76- 3.86 (m, 2 H) 3.92 (br d, J = 5.87 Hz, 1 H) 4.09 (d, J = 11.49 Hz, 1 H) 5.29 (d, J = 6.11 Hz, 1 H) 7.83 (d, J = 8.07 Hz, 1 H) 8.10 (dd, J = 8.19, 2.08 Hz, 1 H) 8.37






(s, 1 H) 8.43 (s, 1






H) 8.77 (d, J =






2.20 Hz, 1 H) 8.84 (d,






J = 2.20 Hz, 1 H)






9.14 (d, J = 2.20 Hz,






1 H)








Claims
  • 1-14. (canceled)
  • 15. A method of treating, ameliorating or preventing one or more conditions comprising the symptom of cognitive deficiency; organic, including symptomatic, mental disorders, dementia; mental retardation; mood affective disorders; neurotic, stress-related and somatoform disorders including anxiety disorders; behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) and Autism spectrum disorders; disorders of psychological development, developmental disorders of scholastic skills; schizophrenia and other psychotic disorders; disorders of adult personality and behaviour; mental and behavioural disorders due to psychoactive substance use; extrapyramidal and movement disorders; episodic and paroxysmal disorders, epilepsy; systemic atrophies primarily affecting the central nervous system, ataxia; behavioural syndromes associated with physiological disturbances and physical factors; sexual dysfunction comprising excessive sexual drive; factitious disorders; cognitive impairment being related to perception, concentration, cognition, learning or memory; cognitive impairment being related to age-associated learning and memory impairments; age-associated memory losses; vascular dementia; craniocerebral trauma; stroke; dementia occurring after strokes (post stroke dementia); post-traumatic dementia; general concentration impairments; concentration impairments in children with learning and memory problems; Alzheimer's disease; lewy body dementia; dementia with degeneration of the frontal lobes including Pick's syndrome; parkinson's disease; progressive nuclear palsy; dementia with corticobasal degeneration; amyotropic lateral sclerosis (ALS); Huntington's disease; Multiple Sclerosis; thalamic degeneration; Creutzfeld-Jacob dementia; HIV dementia; schizophrenia with dementia or Korsakoff's psychosis; sleep disorders; bipolar disorder; metabolic syndrome; obesity; diabetis mellitus; hyperglycemia; dyslipidemia; impaired glucose tolerance; disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen; pain disorders; neuropsychiatric symptoms (e.g. depressive symptoms in Alzheimer's disease); mixed dementia; cognitive impairment in schizoaffective disorder; cognitive impairment in bipolar disorder or cognitive impairment in major depressive disorder; in a subject comprising administering to the subject an effective amount of a compound of formula (I)
  • 16. The method of claim 15, wherein A is selected from the group Ab consisting of
  • 17. The method of claim 15, wherein A is selected from the group Ac consisting of
  • 18. The method of claim 15, wherein A is selected from the group Ad consisting of
  • 19. The method of claim 15, wherein R1 is selected from the group R1b consisting of F—, Cl—, C1-3-alkyl- and C3-6-cycloalkyl-, wherein the above mentioned C1-3-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of F—,or a pharmaceutically acceptable salt thereof.
  • 20. The method of claim 15, wherein R1 is selected from the group Rio consisting of F—, H3C— and cyclopropyl-,or a pharmaceutically acceptable salt thereof.
  • 21. The method of claim 15, wherein R2 is selected from the group R2b consisting of quinolinyl, phenyl and pyridynyl, wherein the above mentioned quinoline, phenyl and pyridyl-groups may optionally be substituted with 1 to 5 substituents R4,or a pharmaceutically acceptable salt thereof.
  • 22. The method of claim 15, wherein R2 is selected from the group R2C consisting of phenyl and pyridyl, wherein the above mentioned phenyl and pyridyl-groups may optionally be substituted with 1 to 2 substituents R4,or a pharmaceutically acceptable salt thereof.
  • 23. The method of claim 15, wherein R3 is selected from the group R3b consisting of H—, H3C—, F3C—, F2HC—, FH2C— and F3C—,or a pharmaceutically acceptable salt thereof.
  • 24. The method of claim 15, wherein R4 is independently from each other selected from the group R4b consisting of halogen, C1-4-alkyl- and C1-3-alkyl-O— wherein the above mentioned C1-4-alkyl- and C1-3-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and F—,or a pharmaceutically acceptable salt thereof.
  • 25. The method of claim 15, wherein R5 is selected from the group R5b consisting of H—, HO— and C1-2-alkyl-, wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,or R5 and R6 together form an group O═,or a pharmaceutically acceptable salt thereof.
  • 26. The method of claim 15, wherein R6 is selected from the group R6b consisting of H— and C1-2-alkyl-, wherein the above mentioned C1-2-alkyl-group may optionally be substituted with 1 to 5 F—,or R5/R6 together form a group O═,or a pharmaceutically acceptable salt thereof.
  • 27. The method of claim 15, wherein the compound of formula (I) is selected from the group consisting of
Priority Claims (1)
Number Date Country Kind
15189600.8 Oct 2015 EP regional
RELATED APPLICATIONS

This application is a Divisional of co-pending U.S. application Ser. No. 15/865,695, filed Jan. 9, 2018, which is a Divisional of U.S. application Ser. No. 15/291,316, filed Oct. 10, 2016, now U.S. Pat. No. 10,023,575, which claims priority to EP Application No. 15189600.8 filed Oct. 13, 2015, the content of each of which is incorporated herein by reference in its entirety.

Divisions (2)
Number Date Country
Parent 15865695 Jan 2018 US
Child 16600965 US
Parent 15291316 Oct 2016 US
Child 15865695 US