6-cycloalkyl-pyrazolopyrimidinones for the treatment of CNS disorders

Information

  • Patent Grant
  • 8912201
  • Patent Number
    8,912,201
  • Date Filed
    Wednesday, August 10, 2011
    12 years ago
  • Date Issued
    Tuesday, December 16, 2014
    9 years ago
Abstract
The invention relates to novel 6-cycloalkyl-pyrazolopyrimidinones according to formula (I)
Description

The invention relates to novel pyrazolopyrimidinones according to formula (I)




embedded image


wherein R1 is a 5 or 6 membered aromatic heteroaryl-group, R2 is an optional substituent, D is optionally substituted cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl or 2-, 3- or 4-pyridyl, m=1 or 2 and n is 0, 1 or 2.


The new compounds are for use as the active entity of medicaments or for the manufacture of medicaments respectively, in particular medicaments for the treatment of conditions concerning deficits in perception, concentration, learning or memory. Such conditions may for example be associated with Alzheimer's disease, schizophrenia and other diseases. The new compounds are also for example for the manufacture of medicaments and/or for use in the treatment of these diseases, in particular for cognitive impairment associated with such disease. The compounds of the invention show PDE9 inhibiting properties.


BACKGROUND OF THE INVENTION

The inhibition of phosphodiesterase 9A (PDE9A) is one of the current concepts to find new access paths to the treatment of cognitive impairments due to CNS disorders like Alzheimer's disease, schizophrenia and other diseases or due to any other neurodegenerative process of the brain. With the present invention, new compounds that follow this concept are presented.


Phosphodiesterase 9A is one member of the wide family of phosphodiesterases. These enzymes modulate the levels of the cyclic nucleotides 5′-3′ cyclic adenosine monophosphate (cAMP) and 5′-3′ cyclic guanosine monophosphate (cGMP). These cyclic nucleotides (cAMP and cGMP) are important second messengers and therefore play a central role in cellular signal transduction cascades. Each of them reactivates inter alia, but not exclusively, protein kinases. The protein kinase activated by cAMP is called protein kinase A (PKA) and the protein kinase activated by cGMP is called protein kinase G (PKG). Activated PKA and PKG are able in turn to phosphorylate a number of cellular effector proteins (e.g. ion channels, G-protein-coupled receptors, structural proteins, transcription factors). It is possible in this way for the second messengers cAMP and cGMP to control a wide variety of physiological processes in a wide variety of organs. However, the cyclic nucleotides are also able to act directly on effector molecules. Thus, it is known, for example, that cGMP is able to act directly on ion channels and thus is able to influence the cellular ion concentration (review in: Wei et al., Prog. Neurobiol., 1998, 56, 37-64). The phosphodiesterases (PDE) are a control mechanism for the activity of cAMP and cGMP and thus in turn for the corresponding physiological processes. PDEs hydrolyse the cyclic monophosphates to the inactive monophosphates AMP and GMP. Currently, 11 PDE families have been defined on the basis of the sequence homology of the corresponding genes. Individual PDE genes within a family are differentiated by letters (e.g. PDE1A and PDE1B). If different splice variants within a gene also occur, then this is indicated by an additional numbering after the letters (e.g. PDE1A1).


Human PDE9A was cloned and sequenced in 1998. The amino acid identity with other PDEs does not exceed 34% (PDE8A) and is never less than 28% (PDE5A). With a Michaelis-Menten constant (Km) of 170 nanomolar (nM), PDE9A has high affinity for cGMP. In addition, PDE9A is selective for cGMP (Km for cAMP=230 micromolar (μM)). PDE9A has no cGMP binding domain, suggesting that the enzyme activity is not regulated by cGMP. It was shown in a Western blot analysis that PDE9A is expressed in humans inter alia in testes, brain, small intestine, skeletal muscle, heart, lung, thymus and spleen. The highest expression was found in the brain, small intestine, kidney, prostate, colon and spleen (Fisher et al., J. Biol. Chem., 1998, 273 (25), 15559-15564; Wang et al., Gene, 2003, 314, 15-27). The gene for human PDE9A is located on chromosome 21q22.3 and comprises 21 exons. 4 alternative splice variants of PDE9A have been identified (Guipponi et al., Hum. Genet., 1998, 103, 386-392). Classical PDE inhibitors do not inhibit human PDE9A. Thus, IBMX, dipyridamole, SKF94120, rolipram and vinpocetine show no inhibition on the isolated enzyme in concentrations of up to 100 micromolar (μM). An IC50 of 35 micromolar (μM) has been demonstrated for zaprinast (Fisher et al., J. Biol. Chem., 1998, 273 (25), 15559-15564).


Murine PDE9A was cloned and sequenced in 1998 by Soderling et al. (J. Biol. Chem., 1998, 273 (19), 15553-15558). This has, like the human form, high affinity for cGMP with a Km of 70 nanomolar (nM). Particularly high expression was found in the mouse kidney, brain, lung and liver. Murine PDE9A is not inhibited by IBMX in concentrations below 200 micromolar either; the IC50 for zaprinast is 29 micromolar (Soderling et al., J. Biol. Chem., 1998, 273 (19), 15553-15558). It has been found that PDE9A is strongly expressed in some regions of the rat brain. These include olfactory bulb, hippocampus, cortex, basal ganglia and basal forebrain (Andreeva et al., J. Neurosci., 2001, 21 (22), 9068-9076). The hippocampus, cortex and basal forebrain in particular play an important role in learning and memory processes. As already mentioned above, PDE9A is distinguished by having particularly high affinity for cGMP. PDE9A is therefore active even at low physiological concentrations, in contrast to PDE2A (Km=10 micromolar (μM); Martins et al., J. Biol. Chem., 1982, 257, 1973-1979), PDE5A (Km=4 micromolar (μM); Francis et al., J. Biol. Chem., 1980, 255, 620-626), PDE6A (Km=17 micromolar (μM); Gillespie and Beavo, J. Biol. Chem., 1988, 263 (17), 8133-8141) and PDE11A (Km=0.52 micromolar (μM); Fawcett et al., Proc. Nat. Acad. Sci., 2000, 97 (7), 3702-3707). In contrast to PDE2A (Murashima et al., Biochemistry, 1990, 29, 5285-5292), the catalytic activity of PDE9A is not increased by cGMP because it has no GAF domain (cGMP-binding domain via which the PDE activity is allosterically increased) (Beavo et al., Current Opinion in Cell Biology, 2000, 12, 174-179). PDE9A inhibitors may therefore lead to an increase in the baseline cGMP concentration.


This outline will make it evident that PDE9A engages into specific physiological processes in a characteristic and unique manner, which distinguishes the role of PDE9A characteristically from any of the other PDE family members.


WO 2004/099210 discloses 6-arylmethyl-substituted pyrazolopyrimidinones which are PDE9 inhibitors.


WO 2004/099211 discloses 6-cyclylmethyl- and 6-alkylmethyl-substituted pyrazolopyrimidines and their use for the improvement of cognition, concentration etc.


DE 102 38 722 discloses the use of PDE9A-inhibitors for the improvement of cognition, concentration.


WO 2004/018474 discloses phenyl-substituted pyrazolopyrimidines and their use for the improvement of perception, concentration learning and/or memory.


WO 2004/026876 discloses alkyl-substituted pyrazolopyrimidines which and their use for the improvement of awareness, concentration learning capacity and/or memory performance.


WO 2004/096811 discloses heterocyclic bicycles as PDE9 inhibitors for the treatment of diabetes, including type 1 and type 2 diabetes, hyperglycemia, dyslipidemia, impaired glucose tolerance, metabolic syndrome and/or cardiovascular disease.


WO2009068617 discloses PDE9 inhibiting compounds derived from pyrazolopyrimidinones with a substituted phenylmethyl- or pyridyl-methyl group in the 6-position.


WO2010112437 discloses PDE9 inhibiting compounds derived from pyrazolopyrimidinones with a phenyl or heteroaryl substituted arylmethyl- or heteroaryl-methyl group in the 6-position.


WO 2009/121919 discloses PDE9 inhibitors derived from pyrazolopyrimidinones with a non-aromatic heterocyclyl group in the 1-position, among which is tetrahydropyranyl.


WO 2010/026214 discloses PDE9 inhibitors derived from pyrazolopyrimidinones with a cycloalkyl or a cycloalkenyl group in the 1-position, among which is 4,4-difluorocyclohexyl.


Some prior art is directed to chemically nucleoside derivatives. As examples it is referred to WO 2002/057425, which discloses nucleoside derivatives, which are inhibitors of RNA-dependent RNA viral polymerase, or WO 2001/060315, which discloses nucleoside derivatives for the treatment of hepatitis C infection or EP679657, which discloses compounds that serve as ribonucleoside analogues or US2002058635, which discloses purine L-nucleoside compounds, in which both the purine rings and the carbohydrate ring (pentose ring) are either modified, functionalized, or both. So the carbohydrate ring for example must show at least one esterified hydroxy group.


WO 2005/051944 discloses oxetane-containing nucleosides, for the treatment of nucleoside analogue related disorders such as disorders involving cellular proliferation and infection.


WO 2006/084281 discloses inhibitors of the E1 activation enzyme that have a sulfonamide moiety.


WO 1998/40384 discloses pyrazolopyrimidinones which are PDE1, 2 and 5 inhibitors and can be employed for the treatment of cardiovascular and cerebrovascular disorders and disorders of the urogenital system.


CH396 924, CH396 925, CH396 926, CH396 927, DE1147234, DE1149013, describe pyrazolopyrimidines which have a coronary-dilating effect and which can be employed for the treatment of disturbances of myocardial blood flow.


U.S. Pat. No. 3,732,225 describes pyrazolopyrimidines which have an anti-inflammatory and blood glucose-lowering effect.


DE2408906 describes styrylpyrazolopyrimidinones which can be employed as antimicrobial and anti-inflammatory agents for the treatment of, for example, oedema.


OBJECTIVE OF THE INVENTION

Changes in the substitution pattern of pyrazolopyrimidinones result in interesting changes concerning biological activity, respectively changes in the affinity towards different target enzymes.


Therefore it is an objective of the present invention to provide compounds as herein described, in particular in the claims, that effectively modulate PDE9A for the purpose of the development of a medicament, in particular in view of diseases or conditions, the treatment of which is accessible via PDE9A modulation.


It is another objective of the present invention to provide compounds that are useful for the manufacture of a medicament for the treatment of CNS disorders.


Yet another objective of the present invention is to provide compounds which show a favourable safety profile.


Another objective of the present invention is to provide compounds that have a favourable selectively profile in favour of PDE9A inhibition over other PDE family members and other pharmacological targets and by this may provide an advantage.


Yet another objective is to provide a medicament that may not only serve for treatment but might also be used for the prevention or modification of the corresponding disease or condition.


The present invention further provides a pharmaceutical composition comprising a compound as herein described, in particular in the claims and a pharmaceutically acceptable carrier.


The present invention further provides a method for the treatment of any of the conditions as described herein in a mammal in need of such treatment, preferably a human, comprising administering to the mammal a therapeutically effective amount of a compound as herein described, in particular in the claims.


The present invention further provides a compound as herein described, in particular in the claims, for use in a method of treatment of the human or animal body by therapy.







DETAILED DESCRIPTION OF THE PRESENT INVENTION

The compounds of the present invention are characterised by general formula (I):




embedded image


wherein


R1: is a 5 or 6 membered heteroaryl-group whereby 1, 2, 3 or 4, preferably 1, 2 or 3, of the ring atoms are heteroatoms that are selected independently of each other from N, O or S,

    • whereby said 5 or 6 membered aromatic heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, HO—, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, NC—, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, NC—, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably 1;


n: is selected from 0, 1 or 2, preferably, 0 or 1, more preferably 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof;


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image


be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


This embodiment is embodiment 1 of the present invention.

Concerning the proviso definition above: it shall be understood that throughout this description this definition of the compound, specifically

    • “the following oxadiazolyl-derivative




embedded image




    • be it in the form of any possible stereoisomer or a mixture of all or some thereof” encompasses the following stereoisomers beside the mixtures of these compounds:







embedded image


Embodiment 2 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a 5 or 6 membered heteroaryl-group whereby 1, 2, 3 or 4, preferably 1, 2 or 3, of the ring atoms are heteroatoms that are selected independently of each other from N, O or S,

    • whereby said 5 or 6 membered aromatic heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from the group consisting 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof;


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image



be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


Embodiment 3 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a 5 membered heteroaryl-group whereby 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 2 or 3 of the ring atoms are heteroatoms that are selected independently of each other from N, O or S,

    • whereby said 5 membered aromatic heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof;


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image



be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


Embodiment 4 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a 6 membered heteroaryl-group whereby 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 2 or 3 of the ring atoms are heteroatoms that are selected independently of each other from N, O or S,

    • whereby said 6 membered aromatic heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof.


Embodiment 5 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a heteroaryl-group selected from the group consisting of thiadiazolyl, oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl, pyridyl and pyrimidinyl, preferably said heteroaryl-group being selected from the group consisting of thiadiazolyl, oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl and pyrimidinyl,

    • whereby said heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof;


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image



be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


Embodiment 6 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a heteroaryl-group selected from the group consisting of thiadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl, pyridyl and pyrimidinyl, preferably said heteroaryl-group being selected from the group consisting of thiadiazolyl, isoxazolyl, thiazolyl, oxazolyl, pyridyl and pyrimidinyl,

    • whereby said heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl,

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents selected from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted independently of one another by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof.


Embodiment 7 of the Present Invention

embodiment 7 of the invention concerns a compound that corresponds in all aspects with embodiment 6, except in that


R1: is a heteroaryl-group selected from the group consisting of thiadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl and pyrimidinyl, preferably said heteroaryl-group being selected from the group consisting of thiadiazolyl, isoxazolyl, thiazolyl, oxazolyl and pyrimidinyl,

    • whereby said heteroaryl-group optionally may be substituted by 1, 2, 3 or 4, preferably 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—.


Embodiment 8 of the Present Invention

Another embodiment of the invention concerns a compound according to general formula (I), wherein


R1: is a heteroaryl-group selected from the group consisting of [1,3,4]thiadiazol-2-yl, isoxazol-5-yl, thiazol-5-yl-, oxazol-2-yl, pyridin-2-yl and pyrimidin-2-yl, preferably said heteroaryl-group being selected from the group consisting of [1,3,4]thiadiazol-2-yl, isoxazol-5-yl, thiazol-5-yl-, oxazol-2-yl and pyrimidin-2-yl,

    • whereby said heteroaryl-group optionally may be substituted by 1 or 2 substituents,
    • whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, CN—, methyl and H2N—;


R2: is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl, preferably fluorine, NC—, F3C— and methyl;


D: is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—; preferably by fluorine;
    • whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2— and methyl;
    • whereby preferably D is selected from the group consisting of 4,4-difluorocyclohex-1-yl, tetrahydropyranyl, thereof preferably tetrahydropyran-4-yl, and 4-methyl-3-pyridyl;


m: is selected from 1 or 2, preferably m is 1;


n: is selected from 0, 1 or 2, preferably n is 0 or 1, more preferably n is 0,

    • whereby if n=2, these two groups R2 are selected independently of one another;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof.


Embodiments 9 to 16 of the Present Invention

In any of the above mentioned embodiments 1 to 8 the preferred compounds are represented by formula (II):


Compounds according to formula (II)




embedded image


with


R1: as defined in any of the aforementioned embodiments 1 to 8;


D being either 4,4-difluorocyclohexyl or tetrahydropyran-4-yl or 4-methyl-3-pyridyl and none of these two groups has further substituents;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof;


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image



be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


The preferred embodiments 9 to 16 according to formula (II) derive from embodiments according to formula (I) in that:


m in formula (I) is 1, so that the corresponding cycloalkyl-group is a cyclobutyl;


n in formula (I) is 0;


D in formula (I) is selected from the group of 4,4-difluorocyclohexyl (without further substituents, i.e. unsubstituted) and tetrahydropyran-4-yl (without further substituents, i.e. unsubstituted) and 4-methyl-3-pyridyl;


R1 in formula (I) is attached to said aforementioned cyclobutyl (m=1) in the 2-position thereof while the 1 position of said cyclobutyl is the attachment point to the 6 position of the D-substituted pyrazolopyrimidinone.


The corresponding embodiments are designated as embodiments 9, 10, 11, 12, 13, 14, 15 and 16 respectively.


Embodiment 9 derives from embodiment 1, embodiment 10 from embodiment 2, embodiment 11 from embodiment 3, embodiment 12 from embodiment 4, embodiment 13 from embodiment 6, embodiment 14 from embodiment 6, embodiment 15 from embodiment 7, embodiment 16 from embodiment 7.


Embodiments 17 to 24 of the Present Invention

Within each of the above mentioned embodiments 1 to 16 more preferred compounds are represented by formula (II):




embedded image


with


R1: as defined in any of the aforementioned embodiments 1 to 8;


D being either 4,4-difluorocyclohexyl or tetrahydropyran-4-yl and none of these two groups has further substituents;


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof,


with the proviso that the compound is not the following oxadiazolyl-derivative




embedded image



be it in the form of any possible stereoisomer or a mixture of all or some thereof or salt thereof or solvate thereof or a solvate of a salt thereof.


For all embodiments 1 to 24: the configuration of the cycloalkyl group at position 6 of the pyrazolopyrimidinones group with respect to said pyrazolopyrimidinones group and the substituent R1 may be cis or trans.


In this respect the compounds of the invention may have the following configurations:














trans configuration 1







embedded image







trans configuration 2







embedded image







cis configuration 1







embedded image







cis configuration 2







embedded image











whereby R1, R2, m, n and D are as defined in any of embodiments 1 to 8.


These stereochemically defined embodiments are a further aspect of the invention.


Embodiment 25 of the Present Invention

Within the context of the present invention one or more compound(s) is (are) preferred that are selected from the group of specifically defined species as listed in the following table. The left column contains a letter code to identify the compound family, which is the group of compounds that have the same general chemical structural formula if no stereochemical properties are considered. Members of these compound families are exemplified in the section Exemplary embodiments.


Table of Species:




















embedded image

  Compound family A, covering example 1,

A









embedded image


Aa









embedded image


Ab









embedded image


Ac









embedded image


Ad









embedded image

  Compound family B, covering example 2,

B









embedded image


Ba









embedded image


Bb









embedded image


Bc









embedded image


Bd









embedded image

  Compound family C, covering example 3,

C









embedded image


Ca









embedded image


Cb









embedded image


Cc









embedded image


Cd









embedded image

  Compound family D, covering example 4,

D









embedded image


Da









embedded image


Db









embedded image


Dc









embedded image


Dd









embedded image

  Compound family E, covering example 5,

E









embedded image


Ea









embedded image


Eb









embedded image


Ec









embedded image


Ed









embedded image

  Compound family F, covering example 6,

F









embedded image


Fa









embedded image


Fb









embedded image


Fc









embedded image


Fd









embedded image

  Compound family G1, covering example 7,

G1









embedded image


G1a









embedded image


G1b









embedded image


G1c









embedded image


G1d









embedded image

  Compound family G2, covering example 8,

G2









embedded image


G2a









embedded image


G2b









embedded image


G2c









embedded image


G2d









embedded image

  Compound family H1, covering examples 9,

H1









embedded image


H1a









embedded image


H1b









embedded image


H1c









embedded image


H1d









embedded image

  Compound family H2, covering examples 10,

H2









embedded image


H2a









embedded image


H2b









embedded image


H2c









embedded image


H2d









embedded image

  Compound family I, covering example 11,

I









embedded image


Ia









embedded image


Ib









embedded image


Ic









embedded image


Id









embedded image

  Compound family J, covering example 12,

J









embedded image


Ja









embedded image


Jb









embedded image


Jc









embedded image


Jd









embedded image

  Compound family K, covering examples 13, 14 and 15,

K









embedded image


Ka









embedded image


Kb









embedded image


Kc









embedded image


Kd









embedded image

  Compound family L, covering example 16,

L









embedded image


La









embedded image


Lb









embedded image


Lc









embedded image


Ld









embedded image

  Compound family M covering examples 17, 18 and 19,

M









embedded image


Ma









embedded image


Mb









embedded image


Mc









embedded image


Md









embedded image

  Compound family N, covering example 20,

N









embedded image


Na









embedded image


Nb









embedded image


Nc









embedded image


Nd









embedded image

  Compound family O, covering example 21,

O









embedded image


Oa









embedded image


Ob









embedded image


Oc









embedded image


Od









embedded image

  Compound family P, covering example 22,

P









embedded image


Pa









embedded image


Pb









embedded image


Pc









embedded image


Pd









embedded image

  Compound family Q, covering examples 23, 24 and 25,

Q









embedded image


Qa









embedded image


Qb









embedded image


Qc









embedded image


Qd









embedded image

  Compound family R, covering examples 29, 30 and 31,

R









embedded image


Ra









embedded image


Rb









embedded image


Rc









embedded image


Rd









embedded image

  Compound family S, covering examples 32, 33 and 34,

S









embedded image


Sa









embedded image


Sb









embedded image


Sc









embedded image


Sd









embedded image

  Compound family T, covering examples 26, 27 and 28,

T









embedded image


Ta









embedded image


Tb









embedded image


Tc









embedded image


Td










and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof.


Within the latter group of compounds, compounds that show trans configuration with respect to the substitution at the cyclobutyl-group may be preferred over compounds with cis configuration. Of the possible trans configured compounds one thereof may show advantages in efficacy. The more efficacious a compound the more it is among the preferred compounds.


Another criterion which may differentiate preferred compounds according to the invention is the balance of efficacy and safety, such as for example selectivity vs. other PDE family members such as PDE1C.


For one pair of trans configured compounds according to the experimental part a single crystal X-ray structure analysis revealed that the absolute stereochemistry of the compound which showed lower efficacy than its enantiomer is R,R. As a consequence thereof absolute stereochemistry of the compound with the higher efficacy is S,S.


For said compound the S,S-configuration is represented by the following structure according to general formula (II):




embedded image


In analogy, one may assume that among the compounds according to embodiment 25, such compounds that show the same absolute stereochemistry might be the more active ones compared with the other members of the same compound family. According to the present invention, within the same compound family the more active compounds are preferred over the less active compounds. The compound family is the group of compounds that differ in their chemical structure only with regard to stereochemical properties.


The different stereoisomers are subject to individual embodiments according to the invention:


embodiment 26 of the present invention concerns a compound according to any one of embodiments 1 to 25, whereby the compound shows the following stereochemical properties if the compound generally can be represented if the compound generally can be represented




embedded image


embodiment 27 of the present invention concerns a compound according to any one of embodiments 1 to 25, whereby the compound shows the following stereochemical properties: if the compound generally can be represented if the compound generally can be represented




embedded image


embodiment 28 of the present invention concerns a compound according to any one of embodiments 1 to 25, whereby the compound shows the following stereochemical properties: if the compound generally can be represented if the compound generally can be represented




embedded image


embodiment 29 of the present invention concerns a compound according to any one of embodiments 1 to 25, whereby the compound shows the following stereochemical properties: if the compound generally can be represented if the compound generally can be represented




embedded image


Embodiment 30 of the Present Invention

Another set of preferred embodiment of the present invention derives from each of the aforementioned embodiments concerning compounds according to formula (I) or (II), inclusively the preferences concerning stereochemical properties thereof, in that


R1 being pyrimidinyl, or pyridyl, preferably pyrimidin-2-yl or pyrdin-2-yl,


m=1,


n=0 and


D is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl

    • whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FHC—; preferably by fluorine;
    • whereby tetrahydrofuranyl, tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;
    • whereby pyridyl optionally may be substituted by 1, 2, 3 or 4, preferably 1, 2 or 3, more preferably 1 or 2, substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2— and methyl;
    • whereby preferably D is selected from the group consisting of 4,4-difluorocyclohex-1-yl, tetrahydropyranyl and 4-methyl-3-pyridyl


and salts, preferably pharmaceutically acceptable salts thereof, solvates thereof and the solvates of the aforementioned salts thereof.


For each of the embodiments 1 to 30: whenever D may be tetrahydrofuranyl, it is preferably tetrahydrofuran-3-yl; whenever D may be tetrahydropyranyl it is preferable tetrahydropyran-3-yl or tetrahydropyran-4-yl, more preferably tetrahydropyran-4-yl.


For each of the embodiments 1 to 30: the heteroaryl-group R1 preferably is bound via a carbon ring atom thereof to the cycloalkyl-group that is attached to the 6-position of the pyrazolopyrimidinone-scaffold. According to general formula (I) said cycloalkyl-group may be a cyclobutyl- or cyclopentyl-group, according to general formula (II) said cycloalkyl-group is a cyclobutyl-group.


TERMS AND DEFINITIONS

Terms not specifically defined herein should be given the meanings that would be given to them by a person skilled in the art in light of the disclosure and the context. Examples include that specific substituents or atoms are presented with their 1 or 2 letter code, like H for hydrogen, N for nitrogen, C for carbon, O for oxygen, S for sulphur and the like. Optionally but not mandatory the letter is followed by a hyphen to indicate a bond. As used in the specification, 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 alkyl radical having 1 to 6 carbon atoms. In general, for groups comprising two or more subgroups, the last named group is the radical attachment point, for example, “(CH3)2N—” means a monovalent radical of the formula (CH3)2N—, which is attached via the nitrogen atom thereof (i.e. a dimethylamino-substituent). If the term of a substituent starts or ends with a minus sign or hyphen, i.e. −, this sign emphasises the attachment point as in the aforementioned example (CH3)2N—, where the N is linked to the group of which the dimethylamino-group is a substituent. Unless otherwise specified below, conventional definitions of terms control and conventional stable atom valences are presumed and achieved in all formulas and groups.


In general, if terms are specifically defined with a given context, such specific definitions shall prevail over the more general definitions as outlined in this paragraph.


In general, all “tautomeric forms and isomeric forms and mixtures”, whether individual geometric isomers or optical isomers or racemic or non-racemic mixtures of isomers, of a chemical structure or compound are intended, unless the specific stereochemistry or isomeric form is specifically indicated in the compound name or structure. Specific definitions prevail.


“Substitution”: The term “substituted” as used herein explicitly or implicitly, means that any one or more hydrogen(s) on the designated atom is replaced with a member of the indicated group of substituents, provided that the designated atom's normal valence is not exceeded. In case a substituent is bound via a double bond, e.g. an oxo substituent, such substituent replaces two hydrogen atoms on the designated atom. The substitution shall result in a stable compound. “Stable” in this context preferably means a compound that from a pharmaceutical point of view is chemically and physically sufficiently stable in order to be used as an active pharmaceutical ingredient of a pharmaceutical composition. If a substituent is not defined, it shall be hydrogen. By the term “optionally substituted” is meant that either the corresponding group is substituted or it is not. A characterisation that substituents of the same group may be “selected independently of one another” shall mean, that the corresponding substituents may be the same or may be different.


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


“pharmaceutically acceptable salt(s)” of the compounds according to the invention are subject of the present invention as well. The term “pharmaceutically acceptable salt(s)” refers to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof, preferably addition salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues/parts of the compounds of the present invention such as aminofunctions; acidic residues/parts within compounds of the present invention may form salts with alkali or organic bases. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid and the like; and the salts prepared from organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, stearic acid, lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, sulfanilic acid, 2-acetoxybenzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethane disulfonic acid, oxalic acid, isethionic acid and the like.


Physiologically acceptable salts with bases also may include salts with conventional bases such as, by way of example and preferably, alkali metal salts (e.g. sodium and potassium salts), alkaline earth metal salts (e.g. calcium and magnesium salts) and ammonia, organic amines having 1 to 16 C atoms, such as, by way of example and preferably, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methyl-morpholine, dehydroabietylamine, arginine, lysine, ethylenediamine and methylpiperidine and the like.


The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound with basic or acidic properties by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.


A “Prodrug” is considered a compound that is designed to release a biologically active compound according to the present invention in-vivo when such prodrug is administered to a mammalian subject. Prodrugs of compounds according to the present invention are prepared by modifying functional groups present in the compound of the invention in such a way that these modifications are retransformed to the original functional groups under physiological conditions. It will be appreciated that prodrugs of the compounds according to the present inventions are subject to the present invention as well.


“Metabolites” are considered derivatives of the compounds according to the present invention that are formed in-vivo. Active metabolites are such metabolites that cause a pharmacological effect. It will be appreciated that metabolites of the compounds according to the present inventions are subject to the present invention as well, in particular active metabolites.


Some of the compounds may form “solvates”. For the purposes of the invention the term “solvates” refers to those forms of the compounds which form, in the solid or liquid state, a complex by coordination with solvent molecules. Hydrates are a specific form of solvates in which the coordination takes place with water. According to the present invention, the term preferably is used for solid solvates, such as amorphous or more preferably crystalline solvates.


“Scaffold”: The scaffold of the compounds according to the present invention is represented by the following core structure. The numeration of the positions of the ring member atoms is indicated in bold:




embedded image


It will be evident for the skilled person in the art, that this scaffold can be described by its tautomeric “enol” form




embedded image


In the context of the present invention both structural representations of the scaffold shall be considered the subject of the present invention, even if only one of the two representatives is presented. Without meant to be limiting or bound, it is believed that for the majority of compounds under ambient conditions and therewith under conditions which are the relevant conditions for a pharmaceutical composition comprising said compounds, the equilibrium of the tautomeric forms lies on the side of the pyrazolopyrimdin-4-one representation. Therefore, all embodiments are presented as pyrazolopyrimdin-4-one-derivatives or more precisely as pyrazolo[3,4-d]pyrimidin-4-one derivatives.


“Bonds”: If within a chemical formula of a ring system or a defined group, a substituent is directly linked to an atom or a group like “RyR” in the formula below, this shall mean that the substituent is only attached to the corresponding atom. If however from another substituent like “RxR” a bond is not specifically linked to an atom of the ring system but drawn towards the center of the ring or group this means that this substituent “RxR” may be linked to any meaningful atom of the ring system/group unless stated otherwise.




embedded image


The bond symbol “−” (=minus sign) or the symbol “−*” (=minus sign followed by an asterisk sign) stands for the bond through which a substituent is bound to the corresponding remaining part of the molecule/scaffold. In cases in that the minus sign does not seem to be sufficiently clear, there may be added an asterisk to the bond symbol “−” in order to determine the point of attachment of said bond with the corresponding main part of the molecule/scaffold.


The term “C1-6-alkyl” denotes a saturated, branched or unbranched hydrocarbon group with 1 to 6 C atoms. Examples of such groups include methyl, ethyl, n-propyl, iso-propyl, butyl, iso-butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl, tert-pentyl, n-hexyl, iso-hexyl. This definition applies for the use of “alkyl” in any reasonable context within the present description in the absence of a further definition.


The term “C3-7-cycloalkyl” denotes a saturated monocyclic group with 3 to 7 C ring atoms. Preferred are 5 or 6 membered cycloalkyl-groups. There are no other ring atoms than carbon atoms. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl. This definition applies for “cycloalkyl” in any reasonable context within the present description in the absence of a further definition.


The term “heteroaryl” used in this application denotes a heterocyclic, monocyclic aromatic ring system which includes within the ring system itself in addition to at least one C atom one or more heteroatom(s) which are independently selected from N, O and/or S. Preferred are heteroaryls with 1 to 3 heteroatoms or 1 to 2 heteroatoms, or 1 heteroatom. Preferred heteroatom is N.


The terms “pyridyl” defines a pyridine-substituent, sometimes also called pyridinyl.


Expressions like “prevention”, “prophylaxis”, “prophylactic treatment” or “preventive treatment” used herein should be understood synonymous and in the sense that the risk to develop a condition mentioned hereinbefore is reduced, especially in a patient having elevated risk for said conditions or a corresponding anamnesis. Thus the expression “prevention of a disease” as used herein means the management and care of an individual at risk of developing the disease prior to the clinical onset of the disease. The purpose of prevention is to combat the development of the disease, condition or disorder and includes the administration of the active compounds to prevent or delay the onset of the symptoms or complications and to prevent or delay the development of related diseases, conditions or disorders. Success of said preventive treatment is reflected statistically by reduced incidence of said condition within a patient population at risk for this condition in comparison to an equivalent patient population without preventive treatment.


The expression “treatment” or “therapy” preferably means therapeutic treatment of (e.g. preferably human) patients having already developed one or more of said conditions in manifest, acute or chronic form, including symptomatic treatment in order to relieve symptoms of the specific indication or causal treatment in order to reverse or partially reverse the condition or to delay the progression of the indication as far as this may be possible, depending on the condition and the severity thereof. Thus the expression “treatment of a disease” as used herein means the management and care of a patient having developed the disease, condition or disorder. The purpose of treatment is to combat the disease, condition, disorder or a symptom thereof. Treatment includes the administration of the active compounds to eliminate or control the disease, condition or disorder as well as to alleviate the symptoms or complications associated with the disease, condition or disorder.


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 for the embodiments of formula (I) if not defined otherwise within the context of the schemes:




embedded image




embedded image














conditions as mentioned in scheme 2
R1
Ra







1.) Reaction with HATU and DIPEA followed by a carboxylic acid hydrazide. 2.) Treatment with Lawesson's reagent in THF at elevated temperatures.


embedded image


H—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with oxalylchloride in THF followed by treatment with trimethylsilydiazomethane followed by hydrochloric acid in dioxane. 2.) Reaction with a thioamide in EtOH


embedded image


H—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with oxalylchloride in THF followed by treatment with trimethylsilydiazomethane followed by hydrochloric acid in dioxane. 2.) Reaction with a thiourea in EtOH.


embedded image


H—, F3C—, HF2C—, FH2C—, methyl





3.) Reaction with oxalylchloride in THF followed by treatment with trimethylsilydiazomethane and hydrochloric acid in dioxane. 4.) Reaction with a thiourea in EtOH.


embedded image


H2N—, (CH3)2N—,





1.) Reaction with TBTU and DIPEA followed by a 2-amino-alkohol. 2.) Oxidation with Dess-Martin- Periodinane in dichloromethane. 3.) Treatment with Burgess-reagent in DME at elevated temperatures.


embedded image


H—, NC—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with TBTU and DIPEA followed by a 2-amino-ketone hydrochloride. 2.) Treatment with Burgess-reagent in DME at elevated temperatures.


embedded image


H—, NC—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with TBTU and DIPHA followed by a 2-amino-alkohol. 2.) Oxidation with Dess-Martin- Periodinane in dichloromethane. 3.) Treatment with Lawesson's reagent in THF at elevated temperatures


embedded image


H—, NC—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with TBTU and DIPEA followed by a 2-amino-ketone hydrochloride. 2.) Treatment with Lawesson's reagent in THF at elevated temperatures.


embedded image


H—, NC—, F3C—, HF2C—, FH2C—, methyl





1.) Reaction with TBTU and DIPEA followed by 1,2-dimethyl- hydroxylamine hydrochloride. 2.) Reaction with a mixture prepared separately from propan-2-one oxime and n-buthyllithium followed by treatment with sulfuric acid in THF/ water.


embedded image








1.) Reaction with TBTU and DIPEA followed by hydrazine hydrate. 2.) Treatment with triethoxymethane at elevated temperatures.


embedded image














embedded image


Further alternative processes for preparing pyrazolo[3,4-d]pyrimidin-4-ones are known in the art and can likewise be employed for synthesizing the compounds of the invention (see, for example: P. Schmidt et al., Helvetica Chimica Acta 1962, 189, 1620 ff.).




embedded image




embedded image


Further information also can be found in:

  • WO 2004/099210 (in particular page 9, last paragraph to page 14, line 8, incorporated by reference),
  • with respect to the general manufacture of compounds with D being tetrahydropyranyl more information can be found in WO2009/121919, particularly on page 120 to 125 and the experimental part thereof (herewith incorporated by reference),
  • with respect to D being 4,4-difluorocyclohexyl more information can be found in WO 2010/026214, particularly on page 59 to 63 and the experimental part thereof (herewith incorporated by reference),
  • and in the experimental part (exemplary embodiments) of this description. The letter in particular with respect to the manufacture of the two building blocks:




embedded image


Method of Treatment


The present invention refers to compounds, which are considered effective in the treatment of diseases. The compounds according to the invention are effective and selective inhibitors of phosphodiesterase 9A and can be used for the development of medicaments. Such medicaments shall preferably be used for the treatment of diseases in which the inhibition of PDE9A can provide a therapeutic, prophylactic or disease modifying effect. Preferably the medicaments shall be used to improve perception, concentration, cognition, learning or memory, like those occurring in particular in situations/diseases/syndromes such as: mild cognitive impairment, 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 Picks 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, epilepsy, temporal lobe epilepsy, schizophrenia, schizophrenia (with dementia), Korsakoff's psychosis or cognitive impairment associated with depression or bipolar disorder.


Another aspect of the present invention may concern the treatment of a disease which is accessible by PDE9A modulation, in particular sleep disorders like insomnia or narcolepsy, bipolar disorder, metabolic syndrome, obesity, diabetes mellitus, including type 1 or type 2 diabetes, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.


Thus the medical aspect of the present invention can be summarised in that it is considered that a compound according to formula (I) or (II) as herein defined, in particular the specifically defined species compounds is used as a medicament.


Such a medicament preferably is for the treatment of a CNS disease.


In an alternative use, the medicament is for the treatment of a CNS disease, the treatment of which is accessible by the inhibition of PDE9.


In an alternative use, the medicament is for the treatment of a disease that is accessible by the inhibition of PDE9, specifically PDE9A.


In the most preferred alternative use, the medicament is for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory, preferably if such cognitive impairment is associated with a disease or condition as described in this section.


In an alternative use, the medicament is 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 Picks 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, epilepsy, temporal lobe epilepsy, schizophrenia, schizophrenia (with dementia), Korsakoff's psychosis or cognitive impairment associated with depression or bipolar disorder.


In an alternative use, the medicament is for the treatment of Alzheimer's disease, schizophrenia or cognitive impairment associated with Alzheimers's disease or associated with schizophrenia.


In an alternative use, the medicament is for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetes mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.


In a further aspect of the invention, the present invention relates to the method of treatment or prevention of a condition or disease selected from the above listed groups of conditions and diseases, whereby the method comprises the administration of a therapeutically effective amount of a compound according to the invention in a human being in need thereof.


Pharmaceutical Compositions


Medicaments for administration, which are also subject to the present invention, comprise a compound according to the present invention in a therapeutically effective amount and a pharmaceutical carrier. By “therapeutically effective amount” it is meant that if the medicament is applied via the appropriate regimen adapted to the patient's condition, the amount of said compound of formula (I) will be sufficient to effectively treat, to prevent or to decelerate the progression of the corresponding disease, or otherwise to ameliorate the state of a patient suffering from such a disease. It may be the case that the “therapeutically effective amount” in a mono-therapy will differ from the “therapeutically effective amount” in a combination therapy with another medicament.


The dose range of the compounds of general formula (I) applicable per day may be usually from 0.1 to 5000 mg, preferably from 0.1 to 1000 mg, preferably from 2 to 500 mg, more preferably from 5 to 250 mg, most preferably from 10 to 100 mg. A dosage unit (e.g. a tablet) preferably may contain between 2 and 250 mg, particularly preferably between 10 and 100 mg of the compounds according to the invention.


The actual pharmaceutically effective amount or therapeutic dosage will depend on factors known by those skilled in the art such as age, weight, gender or other condition of the patient, route of administration, severity of disease and the like.


The compounds according to the invention may be administered by oral, parenteral (intravenous, intramuscular etc.), intranasal, sublingual, inhalative, intrathecal, topical or rectal route. Suitable preparations for administering the compounds according to the present invention include for example patches, tablets, capsules, pills, pellets, dragees, powders, troches, suppositories, liquid preparations such as solutions, suspensions, emulsions, drops, syrups, elixirs, or gaseous preparations such as aerosols, sprays and the like. The content of the pharmaceutically active compound(s) should be in the range from 0.05 to 90 wt.-%, preferably 0.1 to 50 wt.-% of the composition as a whole. Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate. The tablets may also comprise several layers.


Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arabic, talc, titanium dioxide or sugar. To achieve delayed release or prevent incompatibilities the core may also consist of a number of layers. Similarly the tablet coating may consist of a number of layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.


Syrups or elixirs containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavour enhancer, e.g. a flavouring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.


Solutions may be prepared in the usual way, e.g. with the addition of isotonic agents, preservatives such as p-hydroxybenzoates or stabilisers such as alkali metal salts of ethylene-diamine-tetra-acetic acid, optionally using emulsifiers and/or dispersants, while if water shall be used as diluent, for example, organic solvents may optionally be used as solubilisers or dissolving aids and the solutions may be transferred into injection vials or ampoules or infusion bottles.


Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gelatine capsules.


Suitable suppositories may be made for example by mixing with carriers provided for this purpose, such as neutral fats or polyethyleneglycol or the derivatives thereof.


Excipients which may be used include, for example, water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose), emulsifiers (e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone) and lubricants (e.g. magnesium stearate, talc, stearic acid and sodium lauryl sulphate).


For oral use the tablets may contain, in addition to the carriers specified, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additional substances such as starch, preferably potato starch, gelatine and the like. Lubricants such as magnesium stearate, sodium laurylsulphate and talc may also be used to produce the tablets. In the case of aqueous suspensions the active substances may be combined with various flavour enhancers or colourings in addition to the abovementioned excipients.


The dosage of the compounds according to the invention is naturally highly dependent on the method of administration and the complaint which is being treated.


Combinations with Other Active Substances


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


Consequently, a further aspect of the present invention refers to a combination of each of the compounds of the present invention, preferably at least one compound according to the present invention, with another active compound for example selected from the group of beta-secretase inhibitors; gamma-secretase inhibitors; gamma-secretase modulators; amyloid aggregation inhibitors such as e.g. alzhemed; directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants, such as e.g. vitamin E, ginko biloba or ginkolide; anti-inflammatory substances, such as e.g. Cox inhibitors, NSAIDs additionally or exclusively having Aβ (Abeta) lowering properties; HMG-CoA reductase inhibitors, such as statins; acetylcholine esterase inhibitors, such as donepezil, rivastigmine, tacrine, galantamine; NMDA receptor antagonists such as e.g. memantine; AMPA receptor agonists; AMPA receptor positive modulators, AMPkines, glycine transporter 1 inhibitors; monoamine receptor reuptake inhibitors; substances modulating the concentration or release of neurotransmitters; substances inducing the secretion of growth hormone such as ibutamoren mesylate and capromorelin; CB-1 receptor antagonists or inverse agonists; antibiotics such as minocyclin or rifampicin; PDE1, PDE2, PDE4, PDE5 and/or PDE10 inhibitors, GABAA receptor inverse agonists; GABAA alpha5 receptor inverse agonists; GABAA receptor antagonists; nicotinic receptor agonists or partial agonists or positive modulators; alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators; alpha7 nicotinic receptor agonists or partial agonists; histamine receptor H3 antagonists; 5-HT4 receptor agonists or partial agonists; 5-HT6 receptor antagonists; alpha2-adrenoreceptor antagonists, calcium antagonists; muscarinic receptor M1 agonists or partial agonists or positive modulators; muscarinic receptor M2 antagonists; muscarinic receptor M4 antagonists; metabotropic glutamate receptor 5 positive allosteric modulators; metabotropic glutamate receptor 2 antagonists; metabotropic glutamate receptor 2/3 agonists; metabotropic glutamate receptor 2 positive allosteric modulators and other substances that modulate receptors or enzymes in a manner such that the efficacy and/or safety of the compounds according to the invention is increased and/or unwanted side effects are reduced.


This invention further relates to pharmaceutical compositions containing one or more, preferably one active substance. At least one active substance is selected from the compounds according to the invention and/or the corresponding salts thereof. Preferably the composition comprises only one such active compound. In case of more than one active compound the other one can be selected from the aforementioned group of combination partners such as alzhemed, vitamin E, ginkolide, donepezil, rivastigmine, tacrine, galantamine, memantine, ibutamoren mesylate, capromorelin, minocyclin and/or rifampicin. Optionally the composition comprises further ingredients such as inert carriers and/or diluents.


The compounds according to the invention may also be used in combination with immunotherapies such as e.g. active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or antibody fragments for the treatment of the above mentioned diseases and conditions.


The compounds according to the invention also may be combined with Dimebon.


The compounds according to the invention also may be combined with antidepressants like amitriptyline imipramine hydrochloride (TOFRANIL), imipramine maleate (SURMONTIL), lofepramine, desipramine (NORPRAMIN), doxepin (SINEQUAN, ZONALON), trimipramine (SURMONTIL).


Or the compounds according to the invention also may be combined with serotonin (5-HT) reuptake inhibitors such as alaproclate, citalopram (CELEXA, CIPRAMIL) escitalopram (LEXAPRO, CIPRALEX), clomipramine (ANAFRANIL), duloxetine (CYMBALTA), femoxetine (MALEXIL), fenfluramine (PONDIMIN), norfenfluramine, fluoxetine (PROZAC), fluvoxamine (LUVOX), indalpine, milnacipran (IXEL), paroxetine (PAXIL, SEROXAT), sertraline (ZOLOFT, LUSTRAL), trazodone (DESYREL, MOLIPAXIN), venlafaxine (EFFEXOR), zimelidine (NORMUD, ZELMID), bicifadine, desvenlafaxine (PRISTIQ), brasofensme and tesofensine.


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


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


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


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


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


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


EXAMPLES
Pharmaceutical Compositions

Examples which might illustrate possible pharmaceutical formulations, without being meant to be limiting:


The term “active substance” denotes one or more compounds according to the invention including the salts thereof. In the case of one of the aforementioned combinations with one or more other active substances the term “active substance” may also include the additional active substances.


Example A
Tablets Containing 100 mg of Active Substance

Composition: tablet



















active substance
100.0
mg



lactose
80.0
mg



corn starch
34.0
mg



polyvinylpyrrolidone
4.0
mg



magnesium stearate
2.0
mg




220.0
mg










Example B
Tablets Containing 150 mg of Active Substance

Composition: Tablet



















active substance
150.0
mg



powdered lactose
89.0
mg



corn starch
40.0
mg



colloidal silica
10.0
mg



polyvinylpyrrolidone
10.0
mg



magnesium stearate
1.0
mg




300.0
mg










Example C
Hard Gelatine Capsules Containing 150 mg of Active Substance


















active substance
150.0
mg



lactose
87.0
mg



corn starch (dried)
80.0
mg



magnesium stearate
3.0
mg




320.0
mg










Example D
Composition: Suppository

















active substance
150.0 mg



polyethyleneglycol 1500
550.0 mg



polyethyleneglycol 6000
460.0 mg



polyoxyethylene sorbitan
840.0 mg



mono stearate





2000.0 mg 










Example E
Composition: Ampoules Containing 10 mg Active Substance


















active substance
10.0
mg










0.01N hydrochloric acid
q.s.











double-distilled water
ad 2.0
mL










Example F
Composition: Ampoules Containing 50 mg of Active Substance


















active substance
50.0
mg










0.01N hydrochloric acid
q.s.











double-distilled water
ad 10.0
mL










The preparation of any the above mentioned formulations can be done following standard procedures.


Biological Assay


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


PDE9A2 Assay Protocol:


The PDE9A2 enzymatic activity assay was run as scintillation proximity assay (SPA), in general according to the protocol of the manufacturer (GE Healthcare, former Amersham Biosciences, product number: TRKQ 7100).


As enzyme source, lysate (PBS with 1% Triton X-100 supplemented with protease inhibitors, cell debris removed by centrifugation at 13.000 rpm for 30 min) of SF 9 cell expressing the human PDE9A2 was used. The total protein amount included in the assay varied upon infection and production efficacy of the SF9 cells and lay in the range of 0.1-100 ng.


In general, the assay conditions were as follows:


total assay volume: 40 microliter

    • protein amount: 0.1-50 ng
    • substrate concentration (cGMP): 20 nanomolar; ˜1 mCi/1
    • incubation time: 60 min at room temperature
    • final DMSO concentration: 0.2-1%


The assays were run in 384-well format. The test reagents as well as the enzyme and the substrate were diluted in assay buffer. The assay buffer contained 50 mM Tris, 8.3 mM MgCl2, 1.7 mM EGTA, 0.1% BSA, 0.05% Tween 20; the pH of assay buffer was adjusted to 7.5. The reaction was stopped by applying a PDE9 specific inhibitor (e.g. compounds according to WO 2004/099210 or WO 2004/099211, like one of the enantiomeres of example 37, e.g. 1-(2-Chlorophenyl)-6-[(2R)-3,3,3-trifluoro-2-methyl-propyl]-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidine-4-one) in excess.


REFERENCES



  • Wunder F, Tersteegen A, Rebmann A, Erb C, Fahrig T, Hendrix M. Characterization of the first potent and selective PDE9 inhibitor using a cGMP reporter cell line. Molecular Pharmacology. 2005 December; 68(6):1775-81.

  • van der Staay F J, Rutten K, Bärfacker L, Devry J, Erb C, Heckroth H, Karthaus D, Tersteegen A, van Kampen M, Blokland A, Prickaerts J, Reymann K G, Schröder U H, Hendrix M. The novel selective PDE9 inhibitor BAY 73-6691 improves learning and memory in rodents. Neuropharmacology. 2008 October; 55(5):908-18.



PDE1C Assay Protocol:


The assay was run in an analogous manner to the PDE9A2 assay, with the following differences: instead of PDE9A2, PDE1C was used and the assay buffer contained in addition 50 nM Calmodulin, 3 mM CaCl2. The reaction can be stopped by applying the same inhibitor than the one that is outlined above (1-(2-Chlorophenyl)-6-[(2R)-3,3,3-trifluoro-2-methyl-propyl]-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidine-4-one).


Determination of IC50:


IC50 can be calculated with GraphPadPrism or other suited software setting the positive control as 100 and the negative control as 0. For calculation of IC50 dilutions of the test compounds (substrates) are to be selected and tested following the aforementioned protocol.


Data


In the following IC50 values for PDE9A2 inhibition [nanomolar (nM)] illustrate that the compounds according to the present invention inhibit PDE9, specifically PDE9A2. This evidences that the compounds provide useful pharmacological properties. The examples are not meant to be limiting.


The table also provides selectivity values (Selectivity) that show a preference of the compounds for PDE9A versus PDE1C. Selectivity is the ratio (IC50 for PDE1C inhibition [nanomolar (nM)])/(IC50 for PDE9A2 inhibition [nanomolar (nM)]).


The example numbers refer to the final examples as outlined in the section Exemplary embodiments and as defined by the above compound family table (embodiment 25).


All data can be measured according to the procedure described herein. The definition enantiomer 1 or enantiomer 2 is related to the elution orders of enantiomers in chiral SFC and chiral HPLC.















Compound

IC50 PDE9A2



family
Example No.
[nanomolar]
Selectivity


















A
 1*
450
3


B
 2*
5
143


C
 3*
23
34


D
 4*
242
22


E
 5*
60
14


F
 6*
58
15


G1
 7*
31
15


G2
 8*
85
63


H1
 9*
19
46


H2
10*
13
120


I
11*
233
≧43


J
12*
80
38


K
13*
7
328


K
14 (enantiomer 1)
473
4.3


K
15 (enantiomer 2)
4
424


L
16*
5
245


M
17*
16
78


M
18 (enantiomer 1)
5
255


M
19 (enantiomer 2)
1345
0.61


N
20*
31
68


O
21*
433
10


P
22*
21
49


Q
23
23
187


Q
24 (enantiomer 1)
218
8.9


Q
25 (enantiomer 2)
7
197


R
29*
11
117


R
30 (enantiomer 1)
304
4.95


R
31 (enantiomer 2)
7
186


S
32*
7
117


S
33 (enantiomer 1)
4
181


S
34 (enantiomer 2)
388
1.68


T
26*
32
>400


T
27 (enantiomer 1)
11
250


T
28 (enantiomer 2)
360
7





*trans racemic mixture






In-Vivo Effect:


It is believed that the positive in-vitro efficacy results of the compounds of the present invention translate in positive in-vivo efficacy.


The in-vivo effect of the compounds of this invention can be tested in the Novel Object Recognition test according to the procedure of Prickaerts et al. (Neuroscience 2002, 113, 351-361), the social recognition test or the T-maze spontaneous alternation test according to the procedures described by van der Staay et al. (Neuropharmacology 2008, 55, 908-918). For further information concerning biological testing one is also referred to these two citations.


Besides the inhibition property toward the target PDE9, compounds according to the present invention may provide further advantageous pharmacokinetic properties.


E.g. compounds according to the invention may show one or more advantages in the area of safety, balanced metabolism, low risk of causing drug-drug interaction and/or balanced clearance.


Compounds also might show one or more additional or alternative advantages in the area of bioavailability, high fraction absorbed, blood brain transport properties, a favourable (e.g. high mean) residence time (mrt), favourable exposure in the effect compartment and so on.


Chemical Manufacture


Abbreviations:

  • Burgess-reagent (methoxycarbonylsulfamoyl)-triethylammonium-N-betain
  • Lawesson's reagent 2,4-bis-(4-methoxy-phenyl)-[1,3,2,4]dithiadiphosphetane 2,4-disulfide
  • APCI Atmospheric pressure chemical ionization
  • ACN acetonitrile
  • CDI 1,1′-carbonyldiimidazole
  • DEA diethylamine
  • DIPEA diisopropylethylamine
  • DME 1,2-dimethoxyethane
  • DMF dimethylformamide
  • ESI electrospray ionization (in MS)
  • EtOH ethanol
  • Exp. example
  • h hour(s)
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate
  • HPLC high performance liquid chromatography
  • HPLC-MS coupled high performance liquid chromatography-mass spectrometry
  • M molar (mol/L)
  • MeOH methanol
  • min minutes
  • MS mass spectrometry
  • NMP 1-methyl-2-pyrrolidinone
  • Rt retention time (in HPLC)
  • SFC supercrticial fluid chromatography
  • TBTU O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • TLC thin-layer chromatography


LC-MS methods:


Method 1


MS apparatus type: Waters Micromass ZQ; HPLC apparatus type: Waters Alliance 2695, Waters 2996 diode array detector; column: Varian Microsorb 100 C18, 30×4.6 mm, 3.0 μm; eluent A: water+0.13% TFA, eluent B: ACN; gradient: 0.0 min 5% B→0.18 min 5% B→2.0 min 98% B→2.2 min 98% B→2.3 min 5% B→2.5 min 5% B; flow rate: 3.5 mL/min; UV detection: 210-380 nm


Method 2


MS apparatus type: Waters Micromass ZQ; HPLC apparatus type: Waters Alliance 2695, Waters 2996 diode array detector; column: Varian Microsorb 100 C18, 30×4.6 mm, 3.0 μm; eluent A: water+0.13% TFA, eluent B: MeOH; gradient: 0.0 min 5% B→0.35 min 5% B→3.95 min 100% B→4.45 min 100% B→4.55 min 5% B→4.9 min 5% B; flow rate: 2.4 mL/min; UV detection: 210-380 nm.


Method 3


MS apparatus type: Waters Micromass ZQ; HPLC apparatus type: Waters Alliance 2695, Waters 2996 diode array detector; column: Varian Microsorb C18, 20×4.6 mm, 5.0 μm; eluent A: water+0.15% TFA, eluent B: MeOH; gradient: 0.0 min 5% B→0.25 min 5% B→1.90 min 100% B→2.05 min 100% B→2.15 min 5% B→2.25 min 5% B; flow rate: 5.2 mL/min; UV detection: 210-400 nm.


Method 1E hydro


Instrument: LC/MS ThermoFinnigan. Hplc Surveyor DAD, MSQ Quadrupole; column: Synergi Hydro-RP80A, 4 um, 4.60×100 mm; eluent A: 90% water+10% acetonitrile+ammonium formate 10 mM; eluent B=ACN 90%+10% H2O+NH4COOH 10 mM; gradient: A (100) for 1.5 min, then to B (100) in 10 min for 1.5 min; flow rate: 1.2 mL/min; UV


Detection: 254 nm; Ion source: APCI.


Chiral SFC Methods:


Method 4


SFC apparatus type: Berger “Analytix”; column: Daicel IC, 250 mm×4.6 mm, 5.0 μm; eluent: CO2/25% MeOH/0.2% DEA (isocratic); flow rate: 4.0 mL/min, 10 min; temperature: 40° C.; UV detection: 210/220/254 nm.


Method 5


SFC apparatus type: Berger “Analytix”; column: Daicel ADH, 250 mm×4.6 mm, 5.0 μm; eluent: CO2/25% MeOH/0.2% DEA (isocratic); flow rate: 4.0 mL/min, 10 min; temperature: 40° C.; UV detection: 210/220/254 nm.


Chiral HPLC Methods:


Method 6:


HPLC apparatus type: Agilent 1100; column: Daicel chiralcel OJ-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/EtOH80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV Detection: variable (200-500 nm).


Method 6.1:


HPLC apparatus type: Agilent 1100; column: Daicel chiralcel OJ-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/EtOH 85:15; flow rate: 1 mL/min, Temperature: 25° C.; UV Detection: variable (200-500 nm).


Method 7:


HPLC apparatus type: Agilent 1100; column: Chiralpak AD-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/isopropanol 80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV Detection: variable (200-500 nm).


HPLC apparatus type: Agilent 1100; column: Chiralpak AD-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/isopropanol 80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV Detection: variable (200-500 nm).


Microwave Heating:

    • Discover® CEM instruments, equipped with 10 and 35 mL vessels;
    • Biotage Initiator Sixty.


General Comment Concerning the Presentation of the Structures


Compounds with stereogenic centre(s): The structures depicted in the experimental section below will not necessarily show all the stereochemical possibilities of the compounds but only one. However, in such cases a term like “trans-racemic mixture” or “cis-racemic mixture” is added next to the depicted structure in order to indicate the other stereochemical options.


An example is given below. The presented structural formula is




embedded image


The added term “trans-racemic mixture” points to the second stereochemical option:




embedded image


Thus, the manufactured compound is a mixture of




embedded image


This principle applies to other depicted structures as well.


Starting Compounds:


Example 1A
Trans-Racemic Mixture



embedded image


2.00 g (13.9 mmol) trans-cyclobutan-1,2-dicarboxylic acid were mixed with 16 mL EtOH at 0° C. and 2.21 mL (30.5 mmol) thionylchloride were slowly added. The mixture was allowed to warm to room temperature and stirred for 1 h. The solvent was removed under reduced pressure and the product was filtered through a pad of activated basic alumina. 2.71 g (98%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.34 min


MS (ESI pos): m/z=201 (M+H)+


The following example was synthesized in analogy to the preparation of Example 1A, using the corresponding diacid as starting material.




















MS


Example
structure
starting material
Rt [min]
(ESI pos, m/z)







Exp. 1B cis-racemic mixture


embedded image




embedded image


1.12 (Method 3)
201 (M + H)+









Example 2A
Racemic Mixture



embedded image


8.00 g (89.7 mmol) 2-amino-propionic acid were mixed with 88.0 mL (0.93 mol) acetic anhydride and 88.0 mL pyridine. The reaction mixture was stirred at 100° C. for 135 min. The solvent was removed under reduced pressure. Toluene was added to the residue and the solvent was removed under reduced pressure, then 204 mL (816 mmol) HCl (4 M aqueous solution) was added and the mixture was refluxed for 3 h. The solvent was removed under reduced pressure. 1-Butanol (20 mL) was added to the residue and the solvent was removed under reduced pressure. 11.6 g of the title compound were obtained as hydrochloride salt.


MS (ESI pos): m/z=88 (M+H)+


Example 3A
Trans-Racemic Mixture



embedded image


1.00 g (4.09 mmol) 5-Amino-1-(4,4-difluoro-cyclohexyl)-1H-pyrazole-4-carboxylic acid amide (see PCT patent application WO 2010/026214, example 8A) was mixed with 15 mL of anhydrous EtOH, 2.46 g (12.3 mmol) of Example 1A and 0.66 g (16.4 mmol) of sodium hydride (60% suspension in mineral oil) were added. The reaction mixture was heated to 140° C. for 30 min in a microwave oven. The mixture was cooled to room temperature and sodium hydroxide solution (4 M aqueous solution) was added. The solvent was removed under reduced pressure. The residue was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 0.70 g (49%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.24 min


MS (ESI pos): m/z=353 (M+H)+


The following examples were synthesized in analogy to the preparation of Example 3A, using the corresponding amide and ester as starting materials (for starting materials it is referred to PCT patent publications WO 2010/026214, WO 2009/121919 and WO 2004/09921).






















MS





starting

(ESI




starting material:
material:
Rt
pos,


Example
structure
amide
ester
[min]
m/z)







Exp 3B (trans- racemic mixture)


embedded image


5-amino-1- (tetrahydro- pyran-4-yl)-1H- pyrazole-4- carboxylic acid amide (see WO 2009/121919, example 11B)
Exp. 1B
1.07 (Method 3)
319 (M + H)+





Exp. 3C (trans- racemic mixture)


embedded image


5-amino-1-(4- methyl-pyridin- 3-yl)-1H- pyrazole-4- carboxylic acid amide (see WO 2004/099211, example 35A)
Exp. 1A
0.81 (Method 1):
326 (M + H)+









Example 4A
Trans-Racemic Mixture



embedded image


0.200 g (0.568 mmol) Example 3A were mixed with 0.157 mL (1.14 mmol) triethylamine and 5 mL DMF. To the mixture were added 0.237 g (0.624 mmol) HATU, then the reaction mixture was stirred at room temperature for 10 min. To the mixture were added 0.042 g (0.568 mmol) acetic acid hydrazide and the reaction mixture was stirred at room temperature for 1 h. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 30 mg of the product were obtained.


HPLC-MS (Method 1): Rt=1.03 min


MS (ESI pos): m/z=409 (M+H)+


Example 5A
Trans-Racemic Mixture



embedded image


0.150 g (0.426 mmol) of Example 3A were mixed with 2 mL THF. The mixture was cooled to 0° C. and 0.036 mL (0.426 mmol) oxalylchloride and one drop of DMF were added. The reaction mixture was stirred at 0° C. for 1 h. To the reaction mixture were added 2 mL ACN and 0.426 mL (0.851 mmol) trimethylsilyldiazomethane (2 M in hexane). The mixture was stirred for 2 h, then 0.213 mL HCl (4 M in dioxane) was slowly added. The reaction was stirred for 3 h. To the mixture were added ethylacetate and saturated aqueous sodium hydrogen carbonate solution. The organic layer was washed with water and brine and dried over sodium sulfate. The solvents were partially evaporated until volume of approximately 2 mL was reached. The mixture was taken to the next step without further purification.


HPLC-MS (Method 1): Rt=1.40 min


MS (ESI pos): m/z=385/387 (Cl)


The following example was synthesized in analogy to the preparation of Example 5A, using the corresponding acid as starting material.




















MS






(ESI






pos,


Example
structure
starting material
Rt [min]
m/z)







Exp. 5B trans- racemic mixture


embedded image


Exp. 3B
1.12 (Method 1)
351/ 353 (CI)









Example 6A
Trans-Mixture of Stereoisomers



embedded image


0.200 g (0.628 mmol) of Example 3B were mixed with 1 mL DMF. 0.261 mL (1.89 mmol) triethylamine and 0.222 g (0.691 mmol) of TBTU were added. The reaction mixture was stirred at room temperature for 10 min. Then 0.078 g (0.628 mmol) of Example 2A was added and the mixture was stirred at room temperature for 1 h. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 190 mg of the product were obtained.


HPLC-MS (Method 3): Rt=1.03 min


MS (ESI pos): m/z=388 (M+H)+


Example 7A
Trans-Racemic Mixture



embedded image


0.200 g (0.628 mmol) of Example 3B were mixed with 1 mL DMF. 0.174 mL (1.26 mmol) triethylamine and 0.222 g (0.691 mmol) of TBTU were added. The reaction mixture was stirred at room temperature for 10 min. Then 0.066 g (0.628 mmol) 2,2-dimethoxy-ethylamine was added and the mixture was stirred at room temperature for 1 h. Then HCl (2 M aqueous solution) was added and the mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). The residue was mixed with 5 mL acetone and 1 mL HCl (2 M aqueous solution) and stirred overnight under nitrogen. Then the mixture was extracted with DCM. The organic layer was evaporated and purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 170 mg of the product was obtained.


HPLC-MS (Method 3): Rt=1.01 min


MS (ESI pos): m/z=360 (M+H)+


Example 8A
Trans-Mixture of Stereoisomers



embedded image


0.200 g (0.568 mmol) of Example 3A was mixed with 1.0 mL DMF. 0.432 mL (2.84 mmol) DIPEA and 0.200 g (0.624 mmol) TBTU were added. The reaction mixture was stirred at room temperature for 10 min. Then 0.140 g (1.14 mmol) of Example 2A were added and the mixture was stirred at room temperature for 2 h. The mixture was purified by preparative


HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 70 mg (29%) of the product was obtained.


HPLC-MS (Method 1): Rt=1.23 min


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


The following examples were synthesized in analogy to the preparation of Example 8A, using the corresponding nucleophiles as starting materials.




















MS






(ESI




starting

pos,


Example
structure
material
Rt [min]
m/z)







Exp. 8B trans-racemic mixture


embedded image




embedded image


1.31 (method 1)
396 (M + H)+





Exp. 8C trans-mixture of stereoisomers


embedded image




embedded image



410 (M + H)+





Exp. 8D trans-mixture of stereoisomers


embedded image




embedded image


1.12 (method 1)
410 (M + H)+





Exp. 8E trans-racemic mixture


embedded image


hydrazine hydrate
0.99 (method 1)
367 (M + H)+









Example 9A
Trans-Racemic Mixture



embedded image


0.182 g (0.430 mmol) Dess-Martin periodinane were mixed with 2.5 mL DCM. 0.160 g (0.391 mmol) Example 8D in 2.5 mL DCM was added at room temperature. The reaction mixture stirred at room temperature for 30 min and at 30° C. for 30 min To the mixture were added 10 mL sodium thiosulfate solution (10% in water) and 10 mL saturated sodium hydrogen carbonate solution and the mixture was stirred for 20 min. The organic layer was separated and the aqueous layer was extracted with DCM. The organic layer was washed with saturated sodium hydrogen carbonate solution, dried and evaporated. 93 mg (58%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.18 min


MS (ESI pos): m/z=408 (M+H)+


The following example was synthesized in analogy to the preparation of Example 9A, using the corresponding alcohol as starting material.



















starting



Example
structure
material









Exp 9B trans- mixture of stereo- isomers


embedded image


Exp. 8C










Example 10A
Trans-Mixture of Stereoisomers



embedded image


0.450 g of Example 3C was mixed with 3.5 mL DMF and 0.273 g (2.21 mmol) Example 2A. 1.00 mL (6.64 mmol) DIPEA and 0.390 g (1.22 mmol) TBTU were added and the mixture was stirred for 1 h. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 360 mg (83%) of the product was obtained.


HPLC-MS (Method 1): Rt=0.85 min


MS (ESI pos): m/z=395 (M+H)+


Example 11A
Trans-Racemic Mixture



embedded image


300 mg (1.23 mmol) of 5-amino-1-(4,4-difluoro-cyclohexyl)-1H-pyrazole-4-carboxylic acid amide (see WO 2010/026214, example 8A) were mixed with 4 mL anhydrous EtOH, 326 mg (3.07 mmol) trans-cyclobutane-1,2-dicarbonitrile and 0.197 g (4.91 mmol) of sodium hydride (60% suspension in mineral oil) under nitrogen. The reaction mixture was heated to 140° C. for 45 min in a microwave oven. The solvent was removed under reduced pressure. The residue was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 210 mg (51%) of the title compound were obtained.


HPLC-MS (Method 3): Rt=1.19 min


MS (ESI pos): m/z=334 (M+H)+


Example 11B
Trans-Racemic Mixture



embedded image


To a solution of 0.8 g (3.805 mmol) of 5-amino-1-(tetrahydro-pyran-4-yl)-1-H-pyrazole-4-carboxylic acid amide (see PCT patent application WO2010/026214) in 8 mL anhydrous EtOH, 0.457 g (19.6 mmol) of sodium hydride (60% suspension in mineral oil) were added at room temperature under nitrogen. After 1 h under stirring, 1.2 g (11.42 mmol) of trans-cyclobutane-1,2-dicarbonitrile were added and the reaction mixture was heated to 140° C. for 45 min in a microwave oven. The solvent was removed under reduced pressure. The residue was dissolved in DCM, water was added and phases were separated. Organic layers were dried over sodium sulphate and evaporated under reduced pressure. The crude was purified by flash chromatography (Cy/EtOAc from 80/20 to 100%) to obtain the title compound as yellow solid. (0.64 g, 55%)


HPLC-MS (Method 1 Eh): Rt=6.21 min


MS (APCI): m/z=300 (M+H)+


Example 11C
Trans-Racemic Mixture



embedded image


To a solution of 0.85 g (3.91 mmol) of 5-amino-1-(4-methyl-pyridin-3-yl)-1H-pyrazole-4-carboxylic acid amide (see PCT patent application WO 2004/09921) in 10 mL anhydrous EtOH, 0.47 g (11.74 mmol) of sodium hydride (60% suspension in mineral oil) were added at room temperature under nitrogen. After 1 h under stirring, 1.28 g (11.74 mmol) of trans-cyclobutane-1,2-dicarbonitrile were added and the reaction mixture was heated to 140° C. for 45 min in a microwave oven. The reaction mixture was then loaded on SCX cartridge, ammonia fractions were collected and evaporated and the residue was purified by flash chromatography (DCM/MeOH 90:10) to obtain the title compound as white solid. (0.63 g, 52%).


HPLC-MS (Method 1Eh): Rt=5.92 min


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


Example 12A
Trans-Racemic Mixture



embedded image


190 mg (0.570 mmol) of Example 11A were mixed with 0.281 mL toluene and 0.093 mL (2.30 mmol) anhydrous MeOH. 0.103 mL (1.45 mmol) acetylchloride were added slowly at 0° C. The mixture was stirred at room temperature for 12 h. The solvent was removed under reduced pressure. To the residue 0.5 mL MeOH were added. Then 0.407 mL (2.85 mmol) ammonia (7 M in MeOH) were added at 0° C. and the mixture was allowed to warm to room temperature. After 30 min the reaction mixture was treated with water and the pH was adjusted to pH=1 by addition of TFA. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH) yielding 110 mg (42%) of the product were as trifluoroacetic acid salt.


HPLC-MS (Method 3): Rt=1.04 min


MS (ESI pos): m/z=351 (M+H)+


Example 12B
Trans-Racemic Mixture



embedded image


To a mixture of dry EtOH (5 mL) and dry CHCl3 (5 mL) cooled at 0° C., acetylchloride (2.27 mL, 30.82 mmol) was added slowly and mixture left under stirring for 20 min 0° C. A solution of Example 11B (0.410 g, 1.027 mmol) in dry CHCl3 (5 mL) was added dropwise and the mixture stirred at room temperature overnight. Solvents were evaporated under reduced pressure, residue dissolved in dry EtOH (5 mL) and 6.4 mL of a 7.0M solution of ammonia in MeOH (30.82 mmol) were added. The mixture was stirred at room temperature for 12 h. The solvent was removed under reduced pressure. The final product was obtained as hydrochloride and used for the next step without further purification. (0.37 g, content 50% estimated by HPLC-MS).


HPLC-MS (Method 1Eh): Rt=5.38 min


MS (APCI pos): m/z=317 (M+H)+


Example 12C
Trans-Racemic Mixture



embedded image


To a mixture of dry EtOH (4 mL) and dry CHCl3 (10 mL) cooled at 0° C., acetylchloride (4.38 mL, 61.7 mmol) was added slowly and mixture left under stirring for 20 min 0° C. A solution of Example 11C (0.63 g, 2.057 mmol) in dry CHCl3 (5 mL) was added dropwise and the mixture stirred at room temperature overnight. Solvents were evaporated under reduced pressure, residue dissolved in dry MeOH (10 mL) and 10.3 mL of a 7.0M solution of ammonia in MeOH (72 mmol) were added. The mixture was stirred at room temperature for 12 h. The solvent was removed under reduced pressure. The final product, obtained as hydrochloride salt, was used as such in the next step without further purification. (0.85 g, content 84%, estimated by 1H-NMR).


HPLC-MS (Method 1Eh): Rt=5.15 min


MS (APCI pos): m/z=324 (M+H)+


Example 13A
Trans-Racemic Mixture



embedded image


To a solution of 1.6 g (10.24 mmol) of 2-acetyl-cyclobutanecarboxylic acid methyl ester (prepared as described in J. Med. Chem., 25, 109, 1982) in dry EtOH (12 mL), propargylamine (1.4 mL, 20.4 mmol) was added followed by 0.122 g (0.307 mmol) of sodium gold trichloride. The reaction mixture was heated to 140° C. for 45 min in a microwave oven, solid was filtered and the organic evaporated. Crude was purified by flash chromatography (Cy/EtOAc 70:30) to obtain the title compound as yellow green oil. (0.18 g, 9.2%).


HPLC-MS (Method 1Eh): Rt=0.87 min


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


Exemplary Embodiments
Example 1
Trans-Racemic Mixture



embedded image


22.0 mg (0.306 mmol) of propan-2-one oxime were mixed with 2 mL anhydrous THF and 0.471 mL (1.22 mmol) n-butyllithium (2.6 mol/L in toluene) was added carefully to the mixture. The reaction mixture was stirred at room temperature for 30 min 0.110 g (0.278 mmol) of Example 8B in 1 mL anhydrous THF were carefully added during 10 min After 30 min the reaction mixture was added to a mixture of 0.28 mL H2SO4 and 4 mL THF/water (4:1). The mixture was refluxed for 1.5 h. Saturated aqueous sodium hydrogen carbonate solution was added and extracted with ethylacetate. The organic layer was dried and the solvents were evaporated. The residue was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 8 mg (8%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.40 min


MS (ESI pos): m/z=390 (M+H)+


Example 2
Trans-Racemic Mixture



embedded image


0.190 g of Example 6A were mixed with 3 mL DME and 0.273 g (1.14 mmol) Burgess reagent. The reaction mixture was heated to 130° C. for 1 h in a microwave oven. The solvent was evaporated and the residue purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 70 mg (55%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.11 min


MS (ESI pos): m/z=370 (M+H)+


The following examples were synthesized in analogy to the preparation of Example 2, using the corresponding amides as starting materials.




















MS






(ESI




starting

pos,


Example
structure
material
Rt [min]
m/z)







Exp. 3 trans- racemic mixture


embedded image


Exp. 7A
1.17 (Method 3)
342 (M + H)+





Exp. 4 trans- racemic mixture


embedded image


Exp. 4A
1.20 (Method 1)
391 (M + H)+





Exp. 5 trans- racemic mixture


embedded image


Exp. 8A
1.38 (method 1)
404 (M + H)+





Exp. 6 trans- racemic mixture


embedded image


Exp. 9A
1.37 (method 1)
390 (M + H)+





Exp. 7 trans- racemic mixture


embedded image


Exp. 9B
1.42 (method 3)
390 (M + H)+





Exp. 8 trans- racemic mixture


embedded image


Exp. 10A
0.97 (method 1)
377 (M + H)+









Example 9
Trans-Racemic Mixture



embedded image


To a solution of Example 5A, synthesized starting from 0.426 mmol of Example 3A as described above, was added dropwise 0.062 g (0.832 mmol) thioacetamide in 2 mL EtOH. The reaction mixture was stirred overnight. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 62 mg of the title compound were obtained.


HPLC-MS (Method 1): Rt=1.37 min


MS (ESI pos): m/z=406 (M+H)+


The following examples were synthesized in analogy to the preparation of Example 9, using the corresponding starting materials.






















MS





starting

(ESI




starting material:
material:

pos,


Example
structure
nucleophile
chloroketon
Rt [min]
m/z)







Exp. 10 trans- racemic mixture


embedded image


thioacetamide
Exp. 5B
1.21 (Method 3)
372 (M + H)+





Exp. 11 trans- racemic mixture


embedded image


1,1-dimethyl- thiourea
Exp. 5A
1.15 (Method 3)
435 (M + H)+





Exp. 12 trans- racemic mixture


embedded image


thiourea
Exp. 5A
1.15 (Method 3)
407 (M + H)+









Example 13
Trans-Racemic Mixture



embedded image


100 mg (0.215 mmol) of Example 12A were mixed with 1.00 mL (6.07 mmol) 1,1,3,3-tetramethoxypropane. The reaction mixture was heated to 175° C. for 1 h using a microwave oven. The reaction mixture was treated with DCM/MeOH and one drop of triethylamine. The solvents were removed under reduced pressure. The mixture was purified by preparative


HPLC (eluent A: water+0.13% TFA, eluent B: MeOH) yielding 45 mg (54%) of the title compound.


HPLC-MS (Method 3): Rt=1.36 min


MS (ESI pos): m/z=387 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


HPLC apparatus type: Berger Minigram; column: Daicel IC, 5.0 μm, 250 mm×10 mm; method: eluent CO2/30% MeOH/0.2% DEA (isocratic); flow rate: 10 mL/min, Temperature: 40° C.; pressure: 100 bar; UV Detection: 210 nm

















Example
structure
Rt [min]









Exp. 14 trans- enantiomer 1


embedded image


3.15 (Method 4)







Exp. 15 trans- enantiomer 2


embedded image


3.78 (Method 4)










The following example was synthesized in analogy to the preparation of Example 13, using the corresponding dialdehydediacetal as starting material.




















MS






(ESI






pos,


Example
structure
starting material
Rt [min]
m/z)







Exp. 16 trans- racemic mixture


embedded image


1,1,3,3- tetraethoxy-2- methylpropane
1.42 (Method 3)
401 (M + H)+









Example 17
Trans-Racemic Mixture



embedded image


176 mg (0.431 mmol) of Example 4A were mixed with 3 mL THF and 122 mg (0.302 mmol) Lawesson's reagent at room temperature. Then the mixture was stirred for 6 h at 60° C. The reaction mixture was treated with water and diluted with DCM. The mixture was filtered over basic alumina and eluted with DCM and EtOH. The solvents were removed under reduced pressure. The residue was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 45 mg (26%) of the product were obtained.


HPLC-MS (Method 3): Rt=1.37 min


MS (ESI pos): m/z=407 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


HPLC apparatus type: Berger Minigram; column: Daicel ADH, 5.0 μm, 250 mm×10 mm; method: eluent CO2/30% MeOH/0.2% DEA (isocratic); flow rate: 10 mL/min, Temperature: 40° C.; pressure: 100 bar; UV Detection: 210 nm














Example
structure
Rt [min]







Exp. 18 trans- enantiomer 1 (S,S)


embedded image


2.47 (Method 5)





Exp. 19 trans- enantiomer 2 (R,R)


embedded image


2.96 (Method 5)









Single crystals of example 19 have been prepared by recrystallisation from ethylacetate and subjected to X-ray crystal analysis. The data allowed to determine the absolute configuration of example 19 to be (R,R).


Experimental: Data collection and reduction: Data collected on Saturn 944 CCD mounted on AFC11K goniometer, Radiation: Cu Kα from RU200 rotating anode and RIGAKU VARIMAX optics, Temperature: 100K.


Summary of Data Collection Statistics


















Spacegroup
P21



Unit cell dimensions
8.560(2) 6.844(1) 15.603(3)




90.00 98.82(3) 90.00



Resolution range
15.42-0.85 (0.88-0.85) 



Total number of reflections
10857



Number of unique reflections
 1588



Average redundancy
6.84 (2.46)



% completeness
95.7 (79.1)



Rmerge
0.064 (0.118)



Output <I/sigI>
27.7 (7.9) 










Values in ( ) are for the last resolution shell.


Refinement Statistics:


Final Structure Factor Calculation for example 19 in P2,


Total number of 1.s. parameters=255


GooF=S=1.154

Weight=1/[sigma^2(Fo^2)+(0.0421*P)^2+0.38*P] where P=(Max(Fo^2,0)+2*Fc^2)/3


R1=0.0695 for 2207 Fo>4sig(Fo) and 0.0829 for all 2334 data, wR2=0.1646,


Flack×parameter=0.09(3).


Example 20
Trans-Racemic Mixture



embedded image


0.060 g of Example 10A were mixed with 4 mL anhydrous dioxane and 0.074 g (0.180 mmol) Lawesson's reagent. The reaction mixture was heated to 120° C. for 1 h in a microwave oven. The mixture was filtered over basic alumina and eluted with DCM and MeOH. The solvents were removed under reduced pressure. The residue was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 22 mg of the product were obtained as salt with TFA.


HPLC-MS: (Method 1): Rt=0.94 min


MS (ESI pos): m/z=393 (M+H)+


Example 21
Trans-Racemic Mixture



embedded image


0.190 g (0.519 mmol) Example 8E were mixed with 1.38 mL (8.31 mmol) triethoxymethane. The mixture was stirred for 1.5 h at 150° C. The reaction mixture was allowed to cool to room temperature and purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH). 90 mg (46%) of the product were obtained.


HPLC-MS (Method 1): Rt=1.19 min


MS (ESI pos): m/z=377 (M+H)+


Example 22
Trans-Racemic Mixture



embedded image


13 mg (0.10 mmol) CuCl2, 26 mL (0.22 mmol) tert-butyl-nitrite were mixed with ACN. A mixture of 22 mg (0.05 mmol) Example 12 in ACN was carefully added at 0° C. The mixture was stirred for 1 h at 25° C. Additional 9 mg (0.07 mmol) CuCl2 and 13 mL (0.11 mmol) tert-butyl-nitrite was added and stirred another 20 min. The solvents were removed under reduced pressure. The residue was taken up in DCM and extracted with HCl and water. The mixture was purified by preparative HPLC (eluent A: water+0.13% TFA, eluent B: MeOH) yielding 2.1 mg (9%) of the product.


HPLC-MS: (Method 3): Rt=1.46 min


MS (ESI pos): m/z=426/428 (Cl) (M+H)+


Example 23
Trans-Racemic Mixture



embedded image


180 mg (0.26 mmol, content 50%, estimated by HPLC-MS) of Example 12b were mixed with 1.00 mL (6.07 mmol) 1,1,3,3-tetramethoxypropane. The reaction mixture was heated to 175° C. for 1 h using a microwave oven. The reaction mixture was treated with DCM, washed with water. Organic layers were dried over sodiumsulphate and evaporated under reduced pressure. The crude was purified by flash chromatography (Cy/EtOAc from 80/20 to AcOEt/MeOH 96/4) and then with a second flash chromatography (DCM 100% to DCM/EtOH 96/4) to obtain the title compound as beige solid. (0.034 g).


HPLC-MS (Method 1 Eh): Rt=6.57 min


MS (APCI pos): m/z=353 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


Semipreparative Conditions:


HPLC semipreparative system: Waters 600 pump; column: Daicel chiralcel OJ-H, 250 mm×20 mm, 5.0 μm; eluent: hexane/EtOH80:20; flow rate: 15 mL/min, Temperature: 25° C.; UV


Detection: 254 nm














Example
structure
Rt [min]







Exp. 24 trans- enantiomer 1


embedded image


15.604 (Method 6)





Exp. 25 trans- enantiomer 2


embedded image


20.119 (Method 6)









Analytical Conditions


HPLC apparatus type: Agilent 1100; Method 6; column: Daicel chiralcel OJ-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/EtOH80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV


Detection: 254 nm


Example 26
Trans-Racemic Mixture



embedded image


140 mg (content 84%, 0.33 mmol) of Example 12C were mixed with 1.4 mL of 1,1,3,3-tetramethoxypropane and 1.4 mL of NMP. The reaction mixture was heated to 175° C. for 1 h using a microwave oven. The reaction mixture was then diluted with MeOH and loaded on SCX cartridge Ammonia fractions were collected and the residue was purified by flash chromatography (Cy/EtOAc from 90/10 to 100%) to obtain the title compound as white solid (30 mg).


HPLC-MS (Method 1 Eh): Rt=6.72 min


MS (APCIpos): m/z=370 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


Semipreprative Conditions:


HPLC semipreparative system: Waters 600 pump; column: Daicel chiralcel OJ-H, 250 mm×20 mm, 5.0 μm; eluent: hexane/EtOH80:20; flow rate: 15 mL/min, Temperature: 25° C.; UV


Detection: 230 nm

















Example
structure
Rt [min]









Exp. 27 trans- enantiomer 1


embedded image


17.748 (Method 6)







Exp. 28 trans- enantiomer 2


embedded image


20.475 (Method 6)










Analytical Conditions


HPLC apparatus type: Agilent 1100; Method 6; column: Daicel chiralcel OJ-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/EtOH80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV


Detection: 254 nm


Example 29
Trans-Racemic Mixture



embedded image


To a suspension of 0.132 g (0.63 mmol) of 5-amino-1-(tetrahydro-pyran-4-yl)-1-H-pyrazole-4-carboxylic acid amide (see PCT patent application WO2010/026214) in dry EtOH (1.5 mL), 0.066 g (1.66 mmol) of sodium hydride (60% suspension in mineral oil) were added at room temperature under nitrogen. After 10 min, 0.181 mg (0.945 mmol) of Example 13A were added and the reaction mixture was heated to 140° C. for 40 min in a microwave oven (Power 100 W). The reaction mixture was then diluted with DCM, water was added, organics separated and dried over sodiumsulphate. Organics were evaporated under reduced pressure and the crude purified by flash chromatography (DCM/IPA 98:2) to obtain the title compound as a white solid. (54 mg, 32%).


HPLC-MS (Method 1Eh): Rt=8.01 min


MS (APCI pos): m/z=352 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


Semipreprative Conditions: HPLC semipreparative system: Waters 600 pump; column: Daicel chiralcel OJ-H, 250 mm×20 mm, 5.0 μm; eluent: hexane/EtOH85:15; flow rate: 15 mL/min, Temperature: 25° C.; UV Detection: 254 nm














Example
structure
Rt [min]







Exp. 30 trans- enantiomer 1


embedded image


14.754 (Method 6.1)





Exp. 31 trans- enantiomer 2


embedded image


16.834 (Method 6.1)









Analytical Conditions


HPLC apparatus type: Agilent 1100; Method 6.1; column: Daicel chiralcel OJ-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/EtOH85:15; flow rate: 1 mL/min, Temperature: 25° C.; UV


Detection: 254 nm


Example 32
Trans-Racemic Mixture



embedded image


To a suspension of 0.135 g (0.553 mmol) of 5-amino-1-(4,4-difluoro-cyclohexyl)-1-H-pyrazole-4-carboxylic acid amide (see PCT patent application WO2010/026214) in dry EtOH (1.5 mL), 0.066 g (1.66 mmol) of sodium hydride (60% suspension in mineral oil) were added at room temperature under nitrogen. After 10 min, 0.161 mg (0.837 mmol) of Example 13A were added and the reaction mixture was heated to 140° C. for 40 min in a microwave oven (Power 100 W). The reaction mixture was then diluted with DCM, water was added, organics separated and dried over sodium sulphate. Organics were evaporated under reduced pressure and the crude purified by flash chromatography (Cy/EA from 50:50 to 10:90) to obtain the title compound as a white solid. (54 mg, 25%).


HPLC-MS (Method 1Eh): Rt=9.63 min


MS (APCI pos): m/z=386 (M+H)+


The enantiomers of the title compound were separated by HPLC using a chiral stationary phase.


Method for Enantioseparation:


Semipreprative Conditions:


HPLC semipreparative system: Waters 600 pump; Column: Daicel chiralpak AD-H, 250 mm×20 mm, 5.0 μm; eluent: hexane/Isopropanol 80:20; flow rate: 10 mL/min, Temperature: 25° C.; UV Detection: 260 nm

















Example
structure
Rt [min]









Exp. 33 trans- enantiomer 1


embedded image


14.80 (Method 7)







Exp. 34 trans- enantiomer 2


embedded image


20.40 (Method 7)










Analytical Conditions


HPLC apparatus type: Agilent 1100; Method 7; column: Daicel chiralcel AD-H, 250 mm×4.6 mm, 5.0 μm; eluent: hexane/Isopropanol 80:20; flow rate: 1 mL/min, Temperature: 25° C.; UV Detection: 260 nm

Claims
  • 1. A compound of formula (I)
  • 2. The compound according to claim 1, whereinR1 is a 5 or 6 membered heteroaryl-group whereby 1, 2 or 3 of the ring atoms are heteroatoms that are selected independently of each other from N, O or S, whereby said 5 or 6 membered aromatic heteroaryl-group optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;R2 is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl;D is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl 2-, 3- and 4-pyridyl, whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby pyridyl optionally may be substituted by 1, 2, 3 or 4 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;m is selected from 1 or 2;n is selected from 0, 1 or 2, whereby if n=2, two R2 groups are selected independently of one another;or a pharmaceutically acceptable salt thereof;with the proviso that the compound is not
  • 3. The compound according to claim 2, whereinR1 is a heteroaryl-group selected from the group consisting of thiadiazolyl, oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl, pyridyl and pyrimidinyl, whereby said heteroaryl-group optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;or a pharmaceutically acceptable salt thereof.
  • 4. The compound according to claim 1, whereinR1 is a 5 or 6 membered heteroaryl-group selected from the group consisting of thiadiazolyl, 1,2,3-oxadiazolyl,1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, isoxazolyl, thiazolyl, oxazolyl, pyridyl and pyrimidinyl, whereby said heteroaryl-group optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, methyl, H2N— and (CH3)2N—;R2 is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl;D is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, 2-, 3- and 4-pyridyl, whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby pyridyl optionally may be substituted by 1, 2, 3 or 4 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2—, C1-6-alkyl- and C3-7-cycloalkyl;m is selected from 1 or 2;n is selected from 0, 1 or 2, whereby if n=2, two R2 groups are selected independently of one another;or a pharmaceutically acceptable salt thereof.
  • 5. The compound according to claim 1, whereby R1 is not oxadiazolyl, neither unsubstituted nor substituted; or a pharmaceutically acceptable salt thereof.
  • 6. The compound according to claim 4, whereinR1 is a 5 or 6 membered heteroaryl-group selected from the group consisting of [1,3,4]thiadiazol-2-yl, isoxazol-5-yl, thiazol-5-yl-, oxazol-2-yl, pyridin-2-yl and pyrimidin-2-yl, whereby said heteroaryl-group optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, CN—, methyl and H2N—;R2 is selected from the group consisting of fluorine, NC—, F3C—, HF2C—, FH2C— and methyl;D is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, and 2-, 3- and 4-pyridyl, whereby cyclopentyl and cyclohexyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby tetrahydrofuranyl and tetrahydropyranyl optionally may be substituted by 1 or 2 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, F3C—, HF2C— and FH2C—;whereby pyridyl optionally may be substituted by 1, 2, 3 or 4 substituents, whereby said substituents may be selected independently of one another from the group consisting of fluorine, chlorine, bromine, NC—, F3C—, HF2C—, FH2C—, F3C—CH2— and methyl;m is selected from 1 or 2;n is selected from 0, 1 or 2, whereby if n=2, two R2 groups are selected independently of one another;or a pharmaceutically acceptable salt thereof.
  • 7. The compound according to claim 6, wherein R2 is selected from the group consisting of fluorine, NC—, F3C— and methyl;D is selected from the group consisting of cyclopentyl, cyclohexyl, tetrahydrofuran-3-yl, tetrahydropyran-4-yl, 2-pyridyl and 4-pyridyl,m is 1; andn is 0;or a pharmaceutically acceptable salt thereof.
  • 8. The compound according to claim 1, characterized in that it is a compound according to formula (II)
  • 9. The compound according to claim 8, wherein the compound is selected from the group consisting of
  • 10. The compound according to claim 1, whereby the compound is selected from the group consisting of a compound according to formula (Ia), formula (Ib), formula (Ic) and formula (Id)
  • 11. The compound according to claim 8, whereby the compound is selected from the group consisting of a compound according to formula (IIa), formula (IIb), formula (IIc) and formula (IId)
  • 12. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutical carrier.
  • 13. A pharmaceutical composition comprising a combination of a compound according to claim 1 with another active agent selected from the group of beta-secretase inhibitors, gamma-secretase inhibitors, gamma-secretase modulators, amyloid aggregation inhibitors, neuroprotective substances, anti-oxidants, anti-inflammatory substances, HMG-CoA reductase inhibitors, acetylcholine esterase inhibitors, NMDA receptor antagonists, AMPA receptor agonists, AMPA receptor positive modulators, AMPkines, glycine transporter 1 inhibitors, monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmitters, substances inducing the secretion of growth hormone, CB-1 receptor antagonists or inverse agonists, antibiotics, PDE1, PDE2, PDE4, PDE5 and/or PDE10 inhibitors, GABAA receptor inverse agonists, GABAA alpha5 receptor inverse agonists, GABAA receptor antagonists, nicotinic receptor agonists or partial agonists or positive modulators, alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators, alpha7 nicotinic receptor agonists or partial agonists, histamine receptor H3 antagonists, 5-HT4 receptor agonists or partial agonists, 5-HT6 receptor antagonists, alpha2-adrenoreceptor antagonists, calcium antagonists, muscarinic receptor M1 agonists or partial agonists or positive modulators, muscarinic receptor M2 antagonists, muscarinic receptor M4 antagonists, metabotropic glutamate receptor 5 positive allosteric modulators, metabotropic glutamate receptor 2 antagonists, metabotropic glutamate receptor 2/3 agonists and metabotropic glutamate receptor 2 positive allosteric modulators.
  • 14. The compound according to claim 1, wherein the compound is
  • 15. The compound according to claim 1, wherein the compound is
  • 16. The compound according to claim 1, wherein the compound is
  • 17. The compound according to claim 1, wherein the compound is
  • 18. The compound according to claim 1, wherein the compound is
  • 19. The compound according to claim 1, wherein the compound is
  • 20. The compound according to claim 1, wherein the compound is
  • 21. The compound according to claim 1, wherein the compound is
  • 22. The compound according to claim 1, wherein the compound is
  • 23. The compound according to claim 1, wherein the compound is
  • 24. The compound according to claim 1, wherein the compound is
  • 25. The compound according to claim 1, wherein the compound is
Priority Claims (2)
Number Date Country Kind
10172597 Aug 2010 EP regional
11154397 Feb 2011 EP regional
US Referenced Citations (149)
Number Name Date Kind
3165520 Schmidt et al. Jan 1965 A
3169965 Schmidt et al. Feb 1965 A
3211731 Schmidt et al. Oct 1965 A
3244328 Brown Apr 1966 A
3732225 Bruer et al. May 1973 A
3847908 Breuer et al. Nov 1974 A
3884906 Van Der Meer et al. May 1975 A
4602023 Kiely et al. Jul 1986 A
5002949 Peseckis et al. Mar 1991 A
5041449 Belleau et al. Aug 1991 A
5047407 Belleau et al. Sep 1991 A
5053499 Kojima et al. Oct 1991 A
5113855 Newhouse May 1992 A
5201308 Newhouse Apr 1993 A
5239992 Bougamont et al. Aug 1993 A
5256668 Hsu et al. Oct 1993 A
5270315 Belleau et al. Dec 1993 A
5294612 Bacon et al. Mar 1994 A
5341801 Zechner Aug 1994 A
5466806 Belleau et al. Nov 1995 A
5503144 Bacon Apr 1996 A
5541187 Bacon et al. Jul 1996 A
5563049 Kojima et al. Oct 1996 A
5568884 Bruna Oct 1996 A
5634900 Makino et al. Jun 1997 A
5656629 Bacon et al. Aug 1997 A
5684164 Belleau et al. Nov 1997 A
5750673 Martin May 1998 A
5948812 Kraft Sep 1999 A
5969116 Martin Oct 1999 A
5969499 Shaffer Oct 1999 A
5977118 Bacon et al. Nov 1999 A
5977332 Martin Nov 1999 A
6100037 Phillips et al. Aug 2000 A
6174884 Haning et al. Jan 2001 B1
6175008 Belleau et al. Jan 2001 B1
6211158 Seela et al. Apr 2001 B1
6225315 Ellis May 2001 B1
6350753 Belleau et al. Feb 2002 B1
6458796 Haning et al. Oct 2002 B1
6479463 Wang et al. Nov 2002 B1
6831174 Belleau et al. Dec 2004 B2
6903224 Belleau et al. Jun 2005 B2
7022709 Boss et al. Apr 2006 B2
7067507 Pulley et al. Jun 2006 B2
7122693 Belleau et al. Oct 2006 B2
7375213 Deshpande et al. May 2008 B2
7488733 Hendrix et al. Feb 2009 B2
7488766 Peters et al. Feb 2009 B2
7541341 Fushimi et al. Jun 2009 B2
7579449 Eckhardt et al. Aug 2009 B2
7615558 Hendrix et al. Nov 2009 B2
7662790 Himmelsbach et al. Feb 2010 B2
7683160 Eckhardt et al. Mar 2010 B2
7687469 Eckhardt et al. Mar 2010 B2
7708011 Hochrainer et al. May 2010 B2
7713938 Himmelsbach et al. May 2010 B2
7723309 Himmelsbach et al. May 2010 B2
7737156 Boβ et al. Jun 2010 B2
7745414 Eckhardt et al. Jun 2010 B2
7772191 Eckhardt et al. Aug 2010 B2
7772378 Himmelsbach et al. Aug 2010 B2
7776830 Eckhardt et al. Aug 2010 B2
7847074 Eckhardt et al. Dec 2010 B2
7851602 Himmelsbach et al. Dec 2010 B2
7858587 Eckhardt et al. Dec 2010 B2
7870856 Boeck Jan 2011 B2
7879806 Himmelsbach et al. Feb 2011 B2
7879807 Himmelsbach et al. Feb 2011 B2
7984713 Hochrainer et al. Jul 2011 B2
8039441 Himmelsbach et al. Oct 2011 B2
8039477 Hendrix et al. Oct 2011 B2
8044060 Hendrix et al. Oct 2011 B2
8088769 Hendrix et al. Jan 2012 B2
20010041797 Belleau et al. Nov 2001 A1
20010044441 Campbell et al. Nov 2001 A1
20020016348 Simitchieva et al. Feb 2002 A1
20020074774 Hsu et al. Jun 2002 A1
20020086160 Qiu et al. Jul 2002 A1
20020100222 Koenig et al. Aug 2002 A1
20020132754 Boss et al. Sep 2002 A1
20020137903 Ellsworth et al. Sep 2002 A1
20030064935 Gougoutas Apr 2003 A1
20030087918 Belleau et al. May 2003 A1
20030114390 Washburn et al. Jun 2003 A1
20030195205 DeNinno et al. Oct 2003 A1
20040185459 Otsuka et al. Sep 2004 A1
20040187868 Hochrainer et al. Sep 2004 A1
20040220186 Bell et al. Nov 2004 A1
20040254201 Belleau et al. Dec 2004 A1
20040266736 Wunder et al. Dec 2004 A1
20050187168 Eickelmann et al. Aug 2005 A1
20050209251 Linker et al. Sep 2005 A1
20050233982 Himmelsbach et al. Oct 2005 A1
20050263151 Hochrainer et al. Dec 2005 A1
20060009400 Eckhardt et al. Jan 2006 A1
20060025349 Eckhardt et al. Feb 2006 A1
20060074031 Eckhardt et al. Apr 2006 A1
20060100222 Boss et al. May 2006 A1
20060106035 Hendrix et al. May 2006 A1
20060111372 Hendrix et al. May 2006 A1
20070037977 Belleau et al. Feb 2007 A1
20070105876 Hendrix et al. May 2007 A1
20070105881 Hendrix et al. May 2007 A1
20070161662 Hendrix et al. Jul 2007 A1
20070240713 Boeck Oct 2007 A1
20070281940 Dugi et al. Dec 2007 A1
20080255118 Hendrix et al. Oct 2008 A1
20090023913 Eckhardt et al. Jan 2009 A1
20090111838 Hendrix et al. Apr 2009 A1
20090121919 Kihara May 2009 A1
20090194105 Besseler et al. Aug 2009 A1
20090235929 Egen et al. Sep 2009 A1
20090318547 Eckhardt et al. Dec 2009 A1
20090326215 Eckhardt et al. Dec 2009 A1
20100024815 Kladders Feb 2010 A1
20100035900 Hendrix et al. Feb 2010 A1
20100069310 Himmelsbach et al. Mar 2010 A1
20100081625 Wienrich et al. Apr 2010 A1
20100093654 Himmelsbach et al. Apr 2010 A1
20100099641 Himmelsbach et al. Apr 2010 A1
20100179191 Himmelsbach et al. Jul 2010 A1
20100209506 Eisenreich Aug 2010 A1
20100210839 Boss et al. Aug 2010 A1
20100240879 Eckhardt et al. Sep 2010 A1
20100249392 Eckhardt et al. Sep 2010 A1
20100298243 Manuchehri et al. Nov 2010 A1
20100317847 Eckhardt et al. Dec 2010 A1
20110014284 Eisenreich et al. Jan 2011 A1
20110015193 Eickmeier et al. Jan 2011 A1
20110046076 Eickelmann et al. Feb 2011 A1
20110046087 Eickelmann et al. Feb 2011 A1
20110065730 Hendrix et al. Mar 2011 A1
20110065731 Dugi et al. Mar 2011 A1
20110082137 Giovannini et al. Apr 2011 A1
20110098240 Dugi et al. Apr 2011 A1
20110178033 Eckhardt et al. Jul 2011 A1
20110184000 Giovannini et al. Jul 2011 A1
20110203586 Egen et al. Aug 2011 A1
20110207735 Hendrix et al. Aug 2011 A1
20110212960 Heine et al. Sep 2011 A1
20110236477 Schneider et al. Sep 2011 A1
20110237526 Weber et al. Sep 2011 A1
20110237789 Weber et al. Sep 2011 A1
20110294834 Hendrix et al. Dec 2011 A1
20120010224 Hendrix et al. Jan 2012 A1
20120115863 Fuchs et al. May 2012 A1
20120165349 Hendrix et al. Jun 2012 A1
20120202829 Heine et al. Aug 2012 A1
Foreign Referenced Citations (117)
Number Date Country
2090227 Mar 1992 CA
1311201 Dec 1992 CA
2283211 Sep 1998 CA
2238211 Dec 1998 CA
2357146 Jul 2000 CA
2437240 Aug 2002 CA
2438890 Sep 2002 CA
2417631 Jan 2003 CA
2466824 May 2003 CA
2484997 Nov 2003 CA
2496194 Mar 2004 CA
2496292 Apr 2004 CA
2496306 Apr 2004 CA
2496308 Apr 2004 CA
2524900 Nov 2004 CA
2539032 Mar 2005 CA
396923 Aug 1965 CH
396924 Aug 1965 CH
396925 Aug 1965 CH
396926 Aug 1965 CH
396927 Aug 1965 CH
398626 Mar 1966 CH
1147234 Apr 1963 DE
1149013 May 1963 DE
1153023 Aug 1963 DE
1156415 Oct 1963 DE
2408906 Sep 1974 DE
4004558 Sep 1990 DE
4027391 Mar 1992 DE
10156249 May 2003 DE
10238722 Mar 2004 DE
0130735 Jan 1985 EP
0286028 Oct 1988 EP
0496617 Jul 1992 EP
0516510 Dec 1992 EP
0546996 Jun 1993 EP
0626387 Nov 1994 EP
0679657 Nov 1995 EP
0995751 Apr 2000 EP
1460077 Sep 2004 EP
937723 Sep 1963 GB
937724 Sep 1963 GB
937726 Sep 1963 GB
973361 Oct 1964 GB
2001513638 Sep 2001 JP
2001514638 Sep 2001 JP
2002523507 Jul 2002 JP
2004536933 Dec 2004 JP
2005531549 Oct 2005 JP
2006501272 Jan 2006 JP
2006503051 Jan 2006 JP
9414802 Jul 1994 WO
9417803 Aug 1994 WO
9510506 Apr 1995 WO
9628429 Sep 1996 WO
9716456 May 1997 WO
9746569 Dec 1997 WO
9800434 Jan 1998 WO
9810765 Mar 1998 WO
9816184 Apr 1998 WO
9840384 Sep 1998 WO
9941253 Aug 1999 WO
0018758 Apr 2000 WO
0043394 Jul 2000 WO
0160315 Aug 2001 WO
0177075 Oct 2001 WO
0206288 Jan 2002 WO
0209713 Feb 2002 WO
0216348 Feb 2002 WO
02055082 Jul 2002 WO
02057425 Jul 2002 WO
02068423 Sep 2002 WO
02074774 Sep 2002 WO
02086160 Oct 2002 WO
02098864 Dec 2002 WO
03011923 Feb 2003 WO
03011925 Feb 2003 WO
03022859 Mar 2003 WO
03031458 Apr 2003 WO
03037432 May 2003 WO
03037899 May 2003 WO
03041725 May 2003 WO
03072757 Sep 2003 WO
03093269 Nov 2003 WO
03099840 Dec 2003 WO
2004002999 Jan 2004 WO
2004018474 Mar 2004 WO
2004026286 Apr 2004 WO
2004026876 Apr 2004 WO
2004046331 Jun 2004 WO
2004096811 Nov 2004 WO
2004099210 Nov 2004 WO
2004099211 Nov 2004 WO
2004108139 Dec 2004 WO
2004113306 Dec 2004 WO
2005021566 Mar 2005 WO
2005051944 Jun 2005 WO
2005068436 Jul 2005 WO
2006076455 Jul 2006 WO
2006084281 Aug 2006 WO
2006091905 Aug 2006 WO
2006125548 Nov 2006 WO
2007025043 Mar 2007 WO
2007046747 Apr 2007 WO
2008005542 Jan 2008 WO
2008049923 May 2008 WO
2008055959 May 2008 WO
2008100447 Aug 2008 WO
2008104077 Sep 2008 WO
2008139293 Nov 2008 WO
2009068617 Jun 2009 WO
2009121919 Oct 2009 WO
2010026214 Mar 2010 WO
2010092123 Aug 2010 WO
2010092125 Aug 2010 WO
2010112437 Oct 2010 WO
2011018495 Feb 2011 WO
Non-Patent Literature Citations (99)
Entry
Markwalder, J. A. et al; Synthesis and Biological Evaluation of 1-Aryl-4,5-dihydro-1H-pyrazolo[3,4-d]pyrimidin-4-one Inhibitors of Cyclin-Dependent Kinases; J. of Med Chemistry (2004) vol. 47, pp. 5894-5911.
Martins, Timothy, J., et al; Purification and Characterization of a Cyclic GMP-stimulated Cyclic Nucleotide Phosphodiesterase from Bovine Tissues; The Journal of Biological Chemistry (1982) vol. 257, No. 4, pp. 1973-1979.
Merriam-Webster's Collegiate Dictionary, published 1998 by Merriam-Webster Inc. p. 924.
Miki, Takashi, et al; Characterization of the cDNA and Gene Encoding Human PDE3B, the cGIP1 Isoform of the Human Cyclic GMP-Inhibited Cyclic Nucleotide Phosphodiesterase Family; Genomics (1996) vol. 36, pp. 476-485.
Miyashita, A., et al; Studies on Pyrazolo[3,4-d]pyrimidine Derivatives XVIII Facile Preparation of 1H-Pyrazolo[3,4-d] Pyrimidin-4(5H)-Ones; Heterocycles (1990) vol. 31, No. 7, pp. 1309-1314.
Murashima, Seiko., et al; Characterization of Particulate Cyclic Nucleotide Phosphodiesterases from Bovine Brain: Purification of a Distinct cGMP-Stimulated Isoenzyme; Biochemistry (1990) vol. 29, No. 22, pp. 5285-5292.
Obernolte, Rena, et al; The cDNA of a Human Lymphocyte Cyclic AMP Phosphodiesterase (PDE IV) Reveals a Multigene Family; Gene (1993) vol. 129, pp. 239-247.
Podraza, Kenneth F.; Reductive Cyclization of Ketoesters Utilizing Sodium Cyanoborohydride: Synthesis of ?- and ?-Lactones; J. Heterocyclic Chem (1987) vol. 24. pp. 293.
Prickaerts et al; Possible Role of Nitric Oxide-Cyclic GMP Pathway in Object Recognition Memory: Effects of 7 Nitroindazole and Zaprinast; Europ J of Pharmacology (1997) vol. 337, No. 2-3, pp. 125-136.
Prickaerts, J. et al; Effects of Two Selective Phosphodiesterase Type 5 Inhibitors, Sildenafil and Vardenafil, on Object Recognition Memory and Hippocampal Cyclic GMP Levels in the Rat, Neuroscience (2002) vol. 113, No. 2, pp. 351-361.
Puzzo, Daniela, et al; Amyloid-b Peptide Inhibits Activation of the Nitric Oxide/cGMP/cAMP-Responsive Element-Binding Protein Pathway During Hippocampal Synaptic Plasticity; The Journal of Neuroscience (2005) vol. 25, No. 29, pp. 6887-6897.
Reddy, K. Hemender et al; Versatile Synthesis of 6-Alkyl/Aryl-1H-Pyrazolo[3,4-d]Pyrimidin-4[5H]-Ones; Indian Journal of Chemistry (1992) vol. 31B, pp. 163-166.
Reid I. A.; Role of Phosphodiesterase Isozymes in the Control of Renin Secretion: Effects of Selective Enzyme Inhibitors; Current Pharmaceutical Design (1999) vol. 5, No. 9, pp. 725-735.
Related U.S. Appl. No. 12/855,129, filed Aug. 12, 2010.
Related U.S. Appl. No. 12/935,686, filed Sep. 30, 2010.
Related U.S. Appl. No. 13/062,625, filed Mar. 7, 2011.
Related U.S. Appl. No. 13/099,064, filed May 2, 2011.
Rentero, Carles, et al; Identification and Distribution of Different mRNA Variants Produced by Differential Splicing in the Human Phosphodiesterase 9A Gene; Biochemical and Biophysical Research Communications (2003) vol. 301 pp. 686-692.
Reymann, Klaus, et al; The Late Maintenance of Hippocampal LTP: Requirements, Phases, ‘Synaptic Tagging’, ‘Late-Associativity’ and Implications; Neuropharmacology (2007) vol. 52, pp. 24-40.
Roenn, Magnus et al; Palladium (II)-Catalyzed Cyclization Using Molecular Oxygen as Reoxidant; Tetrahedron Letters (1995) vol. 36, No. 42, pp. 7749-7752.
Rosman, Guy, J., et al; Isolation and Characterization of Human cDNSs Encoding a cGMP-Stimulated 3′ ,5′-Cyclic Nucleotide Phosphodiesterase; Gene (1997) vol. 191, pp. 89-95.
Schmidt, Richard, R. et al; Pyrazolo[3, 4-d]Pyrimidin-Nucleoside; Chemische Berichte (1977) vol. 110, pp. 2445-2455.
Schmidt, von P., et al; Heilmittelchemische Studien in der Heterocyclischen Reihe; Helvetica Chimica Acta (1962) vol. 62, No. 189, pp. 1620-1627.
Schousboe, Arne et al; Role of Ca++ and Other Second Messengers in Excitatory Amino Acid Receptor Mediated Neuogeneration: Clinical Perspective; Clinical Neuroscience (1997) vol. 4, pp. 194-198.
Skipper, Howard, E., et al; Structure-Activity Relationships Observed on Screening a Series of Pyrazolopyrimidines Against Experimental Neoplasms; Cancer Research (1957) vol. 17, pp. 579-596.
Soderling, Scott, H. et al; Identification and Characterization of a Novel Family of Cyclic Nucleotide Phosphodiesterases; The Journal of Biological Chemistry (1998) vol. 273, No. 25, pp. 15553-15558.
Soderling, Scott, H. et al; Regulation of cAMP and cGMP signalling: New Phosphodiesterases and New Functions; Current Opinion in Cell Biology (2000) vol. 12, pp. 174-179.
Thomson Innovation Record View, Publication Date: Apr. 18, 1963; English Abstract of DE 1147234B.
Thomson Innovation Record View, Publication Date: Aug. 15, 1965; English Abstract of CH 396 923.
Thomson Innovation Record View, Publication Date: May 22, 1963; English Abstract of DE 1149013B.
Timberlake, J.W. et al; Preparative Procedures: Chemistry of Hydrazo-, Azo-, and Azoxy Groups; Patai (1975) Chapter 4, pp. 69-107.
U.S. Appl. No. 12/545,175, filed Aug. 21, 2009, Inventor: Matthias Eckhardt.
U.S. Appl. No. 12/892,310, filed Sep. 28, 2010. Inventor: Dirk Weber.
U.S. Appl. No. 12/892,326, filed Sep. 28, 2010. Inventor: Dirk Weber.
U.S. Appl. No. 12/894,385, filed Sep. 30, 2010. Inventor: Peter Schneider.
U.S. Appl. No. 13/079,424, filed Apr. 4, 2011. Inventor: Matthias Eckhardt.
U.S. Appl. No. 13/369,596, filed Feb. 9, 2012. Inventor: Niklas Heine.
U.S. Appl. No. 13/369,623, filed Feb. 9, 2012. Inventor: Niklas Heine.
Ugarkar, Bheemarao, et al; Synthesis and antiviral/Antitumor Activities of Certain Pyrazolo[3,4-d]pyrimidine-4(5H)-selone Nucleosides and Related compounds; Journal of Medicinal Chemistry (1984) vol. 27, No. 8, pp. 1026-1030.
Ulrich, Joachim; Crystallization; Kirk-Othmer Encyclopedia of Chem Techn (2002) 7 pages.
Van Der Staay, F. Josef., et al; The Novel Selective PDE9 Inhibitor BAY 73/6691 Improves Learning and Memory in Rodents; Neuropharmacology (2008) vol. 55, pp. 908-916.
Van Staveren, W. C. G., et al; Cloning and localization of the cGMP-specific Phosphodiesterase Type 9 in the Rat Brain; Journal of Neurocytology (2002) vol. 31, pp. 729-741.
Vippagunta, Sudha, R., et al; Crystalline Solids; Advanced Drug Delivery Reviews (2001) vol. 48, pp. 3-26.
Wang, Huanchen, et al; Insight Into Binding of Phosphodiesterase-9-A Selective Inhibitors by Crystal Structures and Mutagenesis; Journal of Medicinal Chemistry (2009) pp. 1-6.
Wang, Peng., et al; Identification and Characterization of a New Human Type 9 cGMP-specific Phosphodiesterase-Splice Variant (PDE9A5) Different Tissue Distribution and Subcellular Localization of PDE9A Variants; Gene (2003) vol. 314, pp. 15-27.
Weeber, Edwin, et al; Molecular Genetics of Human Cognition; Molecular Interventions (2002) vol. 2, No. 6, pp. 376-391.
Wei, Ji-Ye, et al; Molecular and Pharmacological Analysis of Cyclic Nucloeotide-Gated Channel Function in the Central Nervous System; Progress in Neurobiology (1998) vol. 56, pp. 37-64.
West, Anthony, R; Solid Solutions; Department of Chemistry, Univesity of Aberdeen (1988) vol. 10 3 pages.
Wunder, Frank et al; Characterization of the First Potent and Selective PDE9 Inhibitor Using a cGMP Reporter Cell Line; Molecular Pharmacology (2005) vol. 68, No. 6 pp. 1775-1781.
Accessed on Dec. 18, 2008: wikipedia: “Amnesia”, http://www.mentalhealth.org.uk/information/mental-health-a-z/dementia/, last accessed on Dec. 18, 2008.
Accessed on Jun. 30, 2008, Intelihealth: “Alzheimer's Disease,” http://www.intelihealth.com/IH/ihtIH/WSIHW/8303/9117/195703.html?d=dmtHelathAZ.
Accessed on Sep. 22, 2009: Intelihealth: “Dementia,” http://www.intelihealth.com/IH/ihtIH/WSIHW000/244798/00084.html.
Accessed on Sep. 22, 2009: Intelihealth: “Parkinson's Disease”, http://www.intelihealth.com/IH/ihtIH?d=dmtHealthAZ&c=201957.
Andreeva, Svetlana G, et al; “Expression of cGMP-Specific Phosphodiesterase 9A . . . ”, J. of Neuroscience, 2001, Vo. 21, No. 22, pp. 9068-9076.
Bagli, Jehan et al; Chemistry and Positive Inotropic Effect of Pelrinone and related Derivates. A Novel Class of 2-Methylpyrimidones as Inotropic Agents; Journal of Medicinal Chemistry (1988) vol. 31 pp. 814-823.
Barger, Steven, W; Role of Cyclic GMP in the Regulation of Neuronal Calcium and Survival by Secreted Forms of Beta-Amyloid Precursor; Journal of Neurochemistry (1995) vol. 64, No. 5, pp. 2087-2096.
Bernabeu, R., et al; Hippocampal cGMP and cAMP are Differentially Involved in Memory Processing of Inhibitors Avoidance Learning; Neuroreport (1996) vol. 7, No. 2 pp. 585-588.
Byrn, Stephen, R; Solid State Chemistry of Drugs (1999) vol. 2, No. 10, pp. 232-247.
Caligiuri, Maureen, et al; A ProTeome-Wide CDK/CRK-Specific Kinase inhibitor Promotes Tumor Cell Death in the Absence of Cell Cycle Progression; Chemistry & Biology (2005) vol. 12 pp. 1103-1115.
Chem Abstracts Service, Database Accession No. ALB-H01677136, Database Chemcats, 2007, XP002556399.
Cheng, C. C. et al; Potential Purine Antagonists VII. Synthesis of 6-Alkylpyrazolo-[3,4-d]pyrimidinesn Potential Purine Antagonist VII; Gazz. Chim. Ital., (1958) vol. 23, pp. 191-200.
Ciba Geigy AG, “Nucleosides and oligonucleotides and 2′-ether groups,” Data Supplied from the espacenet database, Publication Date: Nov. 30, 1994; English Abstract of EPO 626 387.
DeNinno et al. “The discovery of potent, selective, and orally bioavailable PDE9 . . . ”, Bioorganic & Medicinal Chemistry Letters, 2009, vol. 19, pp. 2537-2541.
Doerwald et al., “Side reactions in organic synthesis,” A Guide to Successful Synthesis Design, 2005, 4 pages.
Ebert et al., “Scopolamine model of demential: electroencephalogram findings and cognitive performance,” Europ J of Clinical Investigation, 1998, vol. 28, No. 11, pp. 944-949.
Farlow, Martin, R; Pharmacokinetic Profiles of Currect Therapies for Alzheimer's Disease: Implications for Switching to Galantamine; Clinical Therapeutics (2001) vol. 23, Suppl. A, pp. A13-A-24.
Fawcett, Lindsay et al; “Molecular Cloning and Characterization of a Distinct Human . . . ”, Proc. Natl. Acad. Science, 2000, vol. 97, No. 7, pp. 3702-3707.
Fischer, Douglas A., et al; “Isolation and Characterization of PDE9A, A Novel . . . ”, J. of Biological Chemistry, 1998, vol. 273, No. 25, pp. 15559-15564.
Fisher, Douglas A, et al; “Isolation and Characterization of PDE8A, a Novel . . . ”, Biochemical and Biophysical Research Communications, 1998, vol. 246, pp. 570-577.
Francis et al; Cortical Pyramidal Neurone Loss May Cause Glutamatergic Hypoativity and Cognitive Impairment in Alzheimer's Disease: investigative and Therapeutic Perspectives; Journal of Neurochemistry (1993) vol. 60, No. 5, pp. 1589-1604.
Francis, Paul T; “Glutamatergic Systems in Alzheimer's Disease” International Journal of Geriatic Psychiatry (2003) vol. 18, pp. S15-S21.
Francis, Sharron H., et al; “Characterization of a Novel cGMP Binding Protein form Rat Lung . . . ”, J. of Biological Chemistry, vol. 255, No. 2, pp. 620-626, (1980).
Fujhishige et al; Cloning and Characterization of a Novel Human Phosphodiesterase That Hydrolyzes Both cAMP and cGMP (PDE10A); Journal of Bilogical Chemistry (1999) vol. 274, No. 26, pp. 18438-18445.
Gielen, Hieke et al; A Novel Approach to Amidines from Esters; Tetrahedron Letters (2002) vol. 43 pp. 419-421.
Gillespie et al; Characterization of a Bovine Cone Photoreceptor Phosphodiesterase Purified by Cyclic Cyclic GMP-Sepharose Chromatography; J. of Biological Chemistry (1988) vol. 263, No. 17, pp. 8133-8141.
Gompper, Rudolf et al; Substituted Dithiocarboxylic Acids and Ketene Thioacetals; Institute for Organic Chemistry Technology (1962) vol. 95, pp. 2861-2870. German & English Translation.
Guipponi, Michel et al; Identification and Characterization of a Novel Cyclic Nucleotide Phosphodiesterase Gene (PDE9A) that Maps to 21q22.3: Alternative Splicing of mRNA Transcripts, Genomic Structure and Sequence; Hum Genet (1998) vol. 103, pp. 386-392.
Harb, A.-F. A., et al; Pyrazoles as Building Blocks in Heterocyclic Synthesis: Synthesis of Some Ne Substituted 1-Triazinylpyrazolo[3,4-d]pyrimidine and 1-Triazinylpyrazolo[3,4-b]pyridine Derivates; Chemical Papers (2005) vol. 59, No. 3, pp. 187-195.
Hendrix et al; “6-cyclymethyl-and 6-alkylmethyl-Substituted Pyrazolopyrimidines,” Publication Date: Nov. 18, 2004, Data Supplied from the espacenet database Worlwide; English Abstract of WO 2004099211.
Hendrix et al; “Use of Pyrazolopyrimidine Against Cardiovascular Disease,” Publication Date: Nov. 30, 2006, Data Supplied from the espacenet database Worldwide; English Abstract or WO 20060125548.
Hetman, J. M., et al; Cloning and Characterization of PDE7B, a cAMP-Specific Phosphodiesterase; Proc, Natl. Acad. Science (2000) vol. 97, No. 1, pp. 472-476.
http://www.nlm.nih.gov/medlineplus/ency/article/000746.htm, last accessed Jul. 15, 2010.
Huettner et al; Primary Culture of Identified Neurons from the Visual Cortex of Postnatal Rats; Journal of Neuroscience (1986) vol. 6, No. 10, pp. 3044-3060.
Hung et al., “A high-yielding synthesis of monalkylhydrazines,” Journal of Organic Chemistry, 1981, vol. 46, pp. 5413-5414.
International Search Report for PCT/EP2008/066350 dated Feb. 23, 2009.
International Search Report for PCT/EP2009/053907 dated May 26, 2009.
International Search Report for PCT/EP2009/061455 dated Mar. 17, 2011.
International Search Report for PCT/EP2010/054050 dated May 27, 2010.
International Search Report for PCT/EP2010/061735 dated Sep. 24, 2010.
International Search Report for PCT/EP2004/006477 dated Oct. 27, 2004.
International Search Report for PCT/EP2004/014872 dated May 19, 2005.
International Search Report of PCT/EP2003/08880 dated Apr. 16, 2004.
International Search Report of PCT/EP2003/08923 dated Dec. 15, 2003.
International Search Report of PCT/EP2003/08979 dated Nov. 25, 2003.
International Search Report of PCT/EP2004/004412 dated Jul. 14, 2004.
International Search Report of PCT/EP2004/004455 dated Sep. 17, 2004.
Loughney, Kate, et al; Isolation and Characterization of cDNAs Corresponsing to Two human Calcium, Calmodulin-regulated, 3′ ,5′-Cyclic Nucleotide Phosphodiesterases; The Journal of Biological Chemistry (1996) vol. 271, No. 2, pp. 796-806.
Loughney, Kate, et al; Isolation and Characterization of cDNAs Encoding PDE5A, a Human cGMP-Bing, cGMP-Specific 3′,5′-Cyclic Nucleotide Phosphodiesterase; Gene (1998) vol. 216, pp. 139-147.
Lugnier, Claire; Cyclic Nucleotide Phosphodiesterase (PDE) Superfamily: A New Target for the Development of Specific Therapeutic Agents; Pharmacology & Therapeutics; (2006) vol. 109, pp. 366-398.
Related Publications (1)
Number Date Country
20120202829 A1 Aug 2012 US