US20140045856A1 - 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes - Google Patents

4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes Download PDF

Info

Publication number
US20140045856A1
US20140045856A1 US13/945,008 US201313945008A US2014045856A1 US 20140045856 A1 US20140045856 A1 US 20140045856A1 US 201313945008 A US201313945008 A US 201313945008A US 2014045856 A1 US2014045856 A1 US 2014045856A1
Authority
US
United States
Prior art keywords
alkyl
group
optionally
substituted
cycloalkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/945,008
Inventor
Riccardo Giovannini
Barbara Bertani
Sara Frattini
Giustino DI ANTONIO
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48906250&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140045856(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Publication of US20140045856A1 publication Critical patent/US20140045856A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems

Definitions

  • the invention relates to 4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene derivatives of general formula (I) which are inhibitors of phosphodiesterase 2 and/or 10, useful in treating central nervous system diseases and other diseases.
  • the invention relates to processes for preparing pharmaceutical compositions as well as processes for manufacture the compounds according to the invention.
  • Cognitive dysfunction plays a role in a number of central nervous system disorders, including neurological disorders, such as Alzheimer's disease (AD), Parkinson disease and dementia, but also psychiatric disorders, such as schizophrenia, depression and bipolar disorders. As world population grows older the number of patients with dementia and AD is growing. Therefore, most people are familiar with the cognitive deficits related to these neurological diseases (Massoud and Gauthier, 2010).
  • Schizophrenia has a heterogeneous symptomatic picture (American Psychiatric Association, 1994) that may be divided into three distinct disease domains: positive symptoms (psychotic episodes of hallucinations, delusions and agitation), negative symptoms (social withdrawal, anhedonia, flattened affect) and cognitive deficits (deficits in executive function, verbal learning and memory, verbal fluency) (Thompson and Meltzer, 1993).
  • ADHD attention deficit/hyperactivity disorder
  • Depression is a severe mental disorder which extremely impairs daily life. Its prevalence is about 10% of the world population with an incidence of 2% according to WHO. Women are more affected than men and elder people more than younger people. The disorder mostly implies a life-long treatment due to the progression of the disease and permanent total disability.
  • Bipolar disorders are characterized by complex symptomatology, including severe symptoms of mood disorders but also manic episodes and cognitive deficits.
  • the Diagnostic and Statistical Manual, 4th edition and International Classification of Mental Disorder recommend subgroups of bipolar disorder based on whether depressive or manic [psychotic] symptoms and episodes are dominating and on the frequency of the episodes (Gaiwani, 2009).
  • Pharmacological agents commonly used in the management of bipolar disorder include lithium; anticonvulsants, such as valproate, carbamazepine and lamotrigine; and recent years have witnessed increasing use of atypical antipsychotics (Altamura et al., 2011).
  • As a problem of current therapy the development of tolerance against anticonvulsant treatment and 30% of treatment refractory cases are described (Post and Weiss, 2010; Gaiwani, 2009).
  • ADHD Attention deficit hyperactivity disorder
  • impulsivity a hyperactivity that is primarily seen in boys.
  • the disease starts at an early age and symptoms are most intense during childhood. After puberty the signs of the disease are more masked and focus on cognitive dysfunction (Jucaite et al. 2005; Turner et al. 2003). Although modern research broadened the understanding of the pathomechanism the exact etiology of the disease remains unclear.
  • the symptoms seen in ADHD are not due to a hyperactivity but a hypoactivity of the so called executive loop of the striatum (Winstanley et al., 2006; Plizska, 2005).
  • the executive loop is responsible for the regulation of cognitive processes such as planning, working memory and attention (Benke et al., 2003; Easton et al., 2007).
  • a dysfunction of the prefrontal cortex or other pathways within the loop induces impulsivity and a loss of the ability to filter stimuli that come from the outside. The latter causes the symptoms of sustained attention and hyperactivity (Roberts and Wallis, 2000; Gonzales et al., 2000).
  • the dopaminergic neurotransmitter system plays a central role in regulating the activity of the executive loop (Jucaite et al., 2005). This conclusion is also supported by the current treatment for ADHD that aims for an activation of the dopaminergic neurotransmitter system (Jucaite et al., 2005).
  • PDE Phosphodiesterases
  • the phosphodiesterases of the different families vary in their substrate selectivity. Thus, some families only hydrolyse cAMP others only cGMP. Some phosphodiesterases, such as phosphodiesterase 2 and 10, inactivate both cAMP and cGMP (Menniti et al., 2006).
  • phosphodiesterase families have different regulatory properties and intracellular location; some are bound to cell membranes and some are dissociated in the cytoplasm, additionally, a division into various intracellular compartments has been reported (Conti and Jin, 1999).
  • PDE2 and PDE10 hydrolyse both, cGMP and cAMP (Menniti et al., 2006; Soderling et al., 1999; Kotera et al., 1999).
  • PDE2 mRNA is mainly distributed in olfactory bulb, olfactory tubercle, cortex, amygdala, striatum, and hippocampus (Lakics et al., 2005; van Staveren et al., 2003).
  • PDE10 PDE10A
  • PDE10A is primarily expressed in the nucleus accumbens and the caudate putamen. Areas with moderate expression are the thalamus, hippocampus, frontal cortex and olfactory tubercle (Menniti et al., 2001).
  • PDE2 in the nucleus accumbens (part of the striatum), the olfactory bulb, the olfactory tubercle and the amygdala and the expression of PDE10 in the nucleus accumbens, the olfactory tubercle and the thalamus supports additional involvement of PDE2 and 10 in the pathophysiology of anxiety and depression (Modell et al., 1990). This is supported by in vivo studies.
  • the selective PDE2 inhibitors BAY60-7550 and ND-7001 are described to be effective in animal models of anxiety and stress-induced behavior (Masood et al., 2008, 2009).
  • PDE10 In addition to the pro-cognitive and antidepressant potential of PDE10 inhibition there is evidence for an additional antipsychotic potential of PDE10 inhibitors.
  • PDE10 In the striatum PDE10 is predominately found postsynaptic in the medium spiny neurons (Xie et al., 2006). By this location, PDE10 may have an important influence on the signal cascade induced by dopaminergic and glutamatergic input on the striatum, two neurotransmitter systems playing a predominate role in the pathomechanism of psychosis.
  • PDE10A inhibitors by up-regulating cAMP and cGMP levels act as D1 agonists and D2 antagonists because the activation of Gs-protein coupled dopamine D1 receptor increases intracellular cAMP, whereas the activation of the Gi-protein coupled dopamine D2 receptor decreases intracellular cAMP levels through inhibition of adenylyl cyclase activity (Mutschler et al., 2001). Accordingly, PDE10 inhibitors are reported to be active in several animal models of schizophrenia (Schmidt et al., 2008; Siuciak et al., 2006; Grauer et al., 2009).
  • PDE10 inhibitors have been disclosed recently in J. Med. Chem., 2011, 54, 7621-7638.
  • CSF cerebrospinal fluid
  • Inhibition of the hERG channel by xenobiotics and subsequent delayed cardiac repolarization is associated with an increased risk for a specific polymorphic ventricular tachyarrhythmia, torsade de pointes, as established by Sanguinetti et al. (1995, Cell, Apr. 21, 81(2):299-307) and a large body of subsequent evidence.
  • low hERG channel inhibition such as that shown by the compounds of the present invention, is highly desirable for therapeutics.
  • Imidazotriazinones are described in WO 2002/068423 for the treatment of e.g. memory deficiency, cognitive disorders, dementia and Alzheimer's disease.
  • Oxindoles are described in WO 2005/041957 for the treatment of dementia.
  • Further inhibitors of PDE2 are known from WO 2007/121319 for the treatment of anxiety and depression, from WO 2013/034761, WO 2012/104293 and WO2013/000924 for the treatment of neurological and psychiatric disorders, from WO 2006/072615, WO 2006/072612, WO 2006/024640 and WO 2005/113517 for the treatment of arthritis, cancer, edema and septic shock, from WO 2005/063723 for the treatment of renal and liver failure, liver dysfunction, restless leg syndrome, rheumatic disorders, arthritis, rhinitis, asthma and obesity, from WO 2005/041957 for the treatment of cancer and thrombotic disorders, from WO 2006/102728 for the treatment of angina pectoris and hypertension from WO 2008/043461 for the treatment of cardiovascular disorders, erectile dysfunction, inflammation and renal failure and from WO 2005/061497 for the treatment of e.g. dementia, memory disorders, cancer and osteoporosis.
  • benzodiazepines are described in WO 2005/063723 for the general treatment of CNS diseases including anxiety, depression, ADHD, neurodegeneration, Alzheimer's disease and psychosis.
  • the compounds of the present invention provide further advantageous pharmacokinetic properties.
  • the compounds of the present invention show high concentration in cerebrospinal fluid (CSF) and have a high CSF to plasma ratio, which translates in lower efficacious doses of the compounds for disease treatment and as a consequence in further potential advantages such as minimization of side effects.
  • CSF cerebrospinal fluid
  • compounds of the present inventions show good metabolic stability and low potential of formation of biologically active metabolite and low hERG potassium channel inhibition.
  • one aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and salts thereof as inhibitors of phosphodiesterase 2 and/or 10.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 and reaching high concentrations in cerebrospinal fluid (CSF) and/or having high CSF to plasma ratio.
  • CSF cerebrospinal fluid
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 and showing good metabolic stability.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 with low hERG channel inhibition.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 with low potential of forming biologically active metabolite.
  • this invention relates to pharmaceutical compositions, containing at least one compound according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof optionally together with one or more inert carriers and/or diluents.
  • a further aspect of the present invention relates to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof for the use in the prevention and/or treatment of disorders associated with PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction.
  • Another aspect of the invention relates to processes of manufacture of the compounds of the present invention.
  • a further aspect of the present invention relates to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof for the use in the prevention and/or treatment of diseases or conditions which can be influenced by inhibition of PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction, such as (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood affective disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) including Autism spectrum disorders; (7) disorders of psychological development, developmental
  • the compounds of the present invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory.
  • the compounds of the present invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis.
  • the compounds of the present invention can be used for the treatment of Alzheimer's disease.
  • compounds of the present invention can be used for the treatment of pain disorders, including but not limited to inflammatory, neuropatic and osteoarthritic pain.
  • the compounds of the present invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.
  • Each R 1x , R 2x , R 3x , R 4x/5x , R 8x , R 7x , R 8x and A x represents a characterized, individual embodiment for the corresponding substituent as described above.
  • preferred individual embodiments of the first aspect of the invention are fully characterized by the term (R 1x , R 2x , R 3x , R 4x/5x , R 6x , R 7x , R 8x and A x ), wherein for each index x an individual figure is given that ranges from “a” to the highest letter given above. All individual embodiments described by the term in parentheses with full permutation of the indices x, referring to the definitions above, shall be comprised by the present invention.
  • Table 1 shows, exemplarily and in the order of increasing preference from the first line to the last line, such embodiments E-1 to E-37 of the invention that are considered preferred. This means that embodiment E-37, represented by the entries in the last row of Table 1, is the most preferred embodiment.
  • aryl-C 1-3 -alkyl- means an aryl group which is bound to a C 1-3 -alkyl group, the latter of which is bound to the core molecule or to the group to which the substituent is attached.
  • core molecule is defined by the following structure:
  • the attachment site of a given residue to another group shall be variable, i.e. any capable atom, bearing hydrogens to be replaced, within this residue may be the linking spot to the group being attached, unless otherwise indicated.
  • An asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule or to the substituent to which it is bound as defined.
  • a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc. . . . ) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound.
  • phrases “pharmaceutically acceptable” or “physiologically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts or “physiologically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts or physiologically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • such salts include salts from ammonia, L-arginine, betaine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine (2,2′-iminobis(ethanol)), diethylamine, 2-(diethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-glucamine, hydrabamine, 1H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, 1-(2-hydroxyethyl)-pyrrolidine, sodium hydroxide, triethanolamine (2,2′,2′′-nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2,2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, be
  • salts can be formed with cations from metals like aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like (also see Pharmaceutical salts, Berge, S. M. et al., J. Pharm. Sci., (1977), 66, 1-19).
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof.
  • Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention e.g. trifluoro acetate salts
  • Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention also comprise a part of the invention.
  • substituted means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's viable valence number is not exceeded, and that the substitution results in a stable compound.
  • partially unsaturated as used herein means that in the designated group or moiety 1, 2, or more, preferably 1 or 2, double bonds are present.
  • the term “partially unsaturated” does not cover fully unsaturated groups or moieties.
  • groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C 1-3 -alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C 1-3 -alkyl-” means that the referenced group may be substituted with 1 to 5 substituents, wherein these substituents can be halogen, HO—, C 1-3 -alkyl-O— which may be optionally fluorinated with 1 to 7 fluoro atoms, and C 1-3 -alkyl-O— which may be optionally fluorinated with 1 to 7 fluoro atoms.
  • halogen generally denotes fluorine, chlorine, bromine and iodine.
  • C 1-n -alkyl wherein n is an integer from 2 to n, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n C atoms.
  • C 1-5 -alkyl embraces the radicals H 3 C—, H 3 C—CH 2 —, H 3 C—CH 2 —CH 2 —, H 3 C—CH(CH 3 )—, H 3 C—CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH(CH 3 )—, H 3 C—CH(CH 3 )—CH 2 —, H 3 C—C(CH 3 ) 2 —, H 3 C—CH 2 —CH 2 —CH 2 —CH 2 —, H 3 C—CH 2 —CH 2 —CH(CH 3 )—, H 3 C—CH 2 —CH(CH 3 )—CH 2 —, H 3 C—CH(CH 3 )—CH 2 —, H 3
  • Carbocyclyl and “carbocycle” as used either alone or in combination with another radical mean, if not mentioned otherwise, a mono- bi- or tricyclic ring structure consisting of 3 to 14 carbon atoms.
  • the terms include the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • C 3-n -cycloalkyl wherein n is an integer from 4 to n, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to n C atoms.
  • C 3-7 -cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • the terms encompass fused, bridged and spirocyclic systems. The terms are intended to include all the possible isomeric forms.
  • the terms include the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • aryl as used herein, either alone or in combination with another radical, denotes a carbocyclic aromatic monocyclic group containing 6 carbon atoms which may be further fused to a second 5- or 6-membered carbocyclic group which may be aromatic, saturated or unsaturated.
  • Aryl includes, but is not limited to, phenyl, indanyl, indenyl, naphthyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl and dihydronaphthyl.
  • heteroaryl is intended to include all the possible isomeric forms.
  • heteroaryl includes the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • the compounds according to the invention may be obtained using methods of synthesis known in principle.
  • the compounds are obtained by the following methods according to the invention which are described in more detail hereinafter.
  • substituted 1,2-pyrido-diamines (A), commercially available or prepared following well known reported procedures, were reacted with the appropriate 1,2-keto ester or acid in EtOH or MeOH as solvents at room temperature, to form the corresponding pyrido[2,3-b]pyrazinone intermediates (B,C).
  • regioisomers were separated by flash chromatography and treated with POCl 3 under heating to obtain the 2-chloro-pyridopyrazines (D).
  • Nucleophilic substitution was performed using hydrazine hydrate in appropriate solvents (e.g. EtOH) at room temperature to form the corresponding pyrido[2,3-b]-pyrazin-3yl-idrazines derivatives (F).
  • Reaction with appropriate acyl chlorides or carboxylic acids in the presence of a coupling agent (e.g. HATU or TBTU) and a base (e.g. TEA or DIPEA) gave the corresponding hydrazides (E) which were submitted to in situ cyclization to triazoles core by heating in appropriate solvent (e.g. cyclohexanol).
  • step 5 preformed hydrazides derivative were reacted with 2-chloro-pyridopyrazines (D) in appropriate solvents (such as DMF) to gave intermediates hydrazides (E) which were then converted to compounds G as described before or heating directly the 2-chloro-pyridopyrazines (D) with the appropriate hydrazides in appropriate solvent such as cyclohexanol at high temperature.
  • appropriate solvents such as DMF
  • R x and R y Examples 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 32a, 33, 34, 35, 35a, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 106, 107, 108a, 108b, 108c, 108d, 109a, 109b, 109c, 109d, 110b, 110d, 111 were prepared following the above described procedures.
  • Examples 70, 71, 72, 73, 74 were prepared following the above described procedure.
  • Examples 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 96a, 96b, 96c, 97 were prepared following the above described procedure.
  • the epoxide obtained by reaction with appropriate trimethyl sulphonium ylide was then subjected to ring opening with hydride and subsequent oxidation with suitable agents such as manganese dioxide in appropriate solvents (DCM).
  • Examples 76, 77, 78, 79 were prepared following the above reported procedure.
  • the omologated derivatives Examples 80, 81 were obtained performing the first step as described in Scheme 9a.
  • Example 103, 103b, 103c were prepared following the above described procedure.
  • the compounds of general formula I according to the invention and the physiologically acceptable salts thereof have valuable pharmacological properties, particularly as inhibitors of phosphodiesterase 2 and/or 10.
  • the in-vitro effect of the active compounds of the invention can be shown with the following biological assays.
  • SF9 lysate containing PDE2A is incubated at room temperature for 1 h with [ 3 H]cAMP and the reaction is terminated by addition of SPA beads in 18 mM zinc sulphate.
  • the [ 3 H]AMP bound to SPA beads is determined after at least 3 hours of sedimentation of the beads, the signal is recorded using the TopCount with a recording time of 3 min/well.
  • PDE 2A protein is expressed upon baculovirus infection in SF9 cells.
  • the cells have been incubated upon infection for ⁇ 3 days and protein production was confirmed by Western Blot.
  • the cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min).
  • the assay conditions were as follows:
  • total assay volume 40 ⁇ l protein amount: 5 ng protein concentration: 500 pg/ ⁇ l substrate concentration: 20 nM; ⁇ 1.08 mCi/l incubation time: 60 min
  • the buffer used for above described assay buffer was:
  • test-compound solution test compound diluted in assay buffer at twofold the desired concentration
  • IC50 are calculated with GraphPadPrism or other suited software setting the positive control as 100 and the negative control as 0. For calculation of IC50 usually 11 dilutions of the test compound are selected.
  • test-compound solution test compound diluted in assay buffer at twofold the desired concentration
  • the signal is recorded using the TopCount with a recording time of 3 min/well.
  • PDE 10A protein is expressed upon baculovirus infection in SF9 cells.
  • the cells have been incubated upon infection for ⁇ 3 days and protein production was confirmed by Western Blot.
  • the cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min).
  • the PDE reaction cleaves cAMP to AMP.
  • the IMAP system (Molecular Device) using fluorescence polarization (FP) as detection principle was used to measure enzyme activity.
  • a fluorescent labeled cAMP was used as substrate for the reaction, generating a labeled AMP.
  • the fluorescent AMP binds specifically to the large M(III)-based nano-particles which reduces the rotational speed of the substrate and thus increases its polarization.
  • the inhibition of PDE 2A or 10 enzyme activity was assessed using IMAP-Phosphodiesterase-cAMP fluorescence labeled substrate (Molecular Devices, Order No. R7506), IMAP TR-FRET screening express (Molecular Devices, Order No. R8160, the TR-FRET component will not be used) and PDE 2A or PDE10 protein expressed upon baculovirus infection in SF9 cells.
  • the cells were incubated after infection for ⁇ 3 days and protein production was confirmed by Western Blot. The cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min). Since SF 9 cells do not express cAMP hydrolyzing enzymes to a high extent, no further purification of the protein was needed.
  • the analysis of the data was performed by calculation of the percentage of inhibition in the presence of test compound compared to the vehicle control samples (100% control, no inhibition) and a low control (0% control, no enzyme).
  • IC50 values are calculated with Assay Explorer or other suited software based on curve fitting of results of at least 8 different compound concentrations.
  • the compound concentrations may vary according to the needed range, but typically cover the range between 10 ⁇ M and 0.1 pM.
  • Test compounds were administered to animals (rat) different routes at doses of 10.0 or 5 ⁇ mol/kg, (both oral and intravenous). After compound administration, the animals were sacrificed in a CO 2 chamber and CSF samples were carefully collected by puncture of the cisterna magna. Immediately after CSF sampling, blood was taken by heart puncture and brains were dissected out. Blood was collected in EDTA-coated microvettes and plasma was prepared by centrifugation. Concentration of the test compounds in plasma, CSF or brain homogenate was determined using HPLC-MS-MS.
  • the compounds of the present invention are not only very potent phosphodiesterase 2 and/or 10 inhibitors but also reach high CSF concentrations and adequate CSF to plasma ratios.
  • the metabolic degradation of the test compound was assayed at 37° C. with pooled liver microsomes from various species.
  • the final incubation volume of 100 ⁇ l per time point contains TRIS buffer pH 7.6 at room temperature (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/mL for human and dog, 0.5 mg/mL for other species) and the test compound at a final concentration of 1 ⁇ M.
  • the reactions were initiated by addition of betanicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM), and terminated by transferring an aliquot into solvent after different time points.
  • HEK (human embryonic kidney) 293 cells were stably transfected with hERG cDNA.
  • Cells were superfused with a bath solution containing (mM): NaCl (137), KCl (4.0), MgCl2 (1.0), CaCl2 (1.8), Glucose (10), HEPES (10), pH 7.4 with NaOH.
  • Patch pipettes were made from borosilicate glass tubing using a horizontal puller and filled with pipette solution containing (mM): K-aspartate (130), MgCl2 (5.0), EGTA (5.0), K2ATP (4.0), HEPES (10.0), pH 7.2 with KOH. Resistance of the microelectrodes was in the range between 2 and 5 M ⁇ .
  • Membrane currents were recorded using an EPC-10 patch clamp amplifier and PatchMaster software. hERG-mediated membrane currents were recorded at 35° C., using the whole-cell configuration of the patch-clamp technique.
  • Transfected HEK293 cells were clamped at a holding potential of ⁇ 60 mV and hERG-mediated inactivating tail currents were elicited using a pulse pattern with fixed amplitudes (activation/inactivation: 40 mV for 2000 ms; recovery: 120 mV for 2 ms; ramp to 40 mV in 2 ms; inactivating tail current: 40 mV for 50 ms) repeated at 15 s intervals.
  • a pulse pattern with fixed amplitudes (activation/inactivation: 40 mV for 2000 ms; recovery: 120 mV for 2 ms; ramp to 40 mV in 2 ms; inactivating tail current: 40 mV for 50 ms) repeated at 15 s intervals.
  • the different concentrations of the test compound were applied sequentially on each of the different cells investigated.
  • a steady state level of baseline current was measured for at least 6 sweeps prior to the application of the first test compound concentration.
  • test compound was dissolved in DMSO to yield a master stock solution which was diluted further in DMSO to stock solutions needed for the lower concentrations.
  • Final dilutions in extracellular buffer were prepared freshly from these stocks by a 1:1000 dilution step each before starting the experiments.
  • Peak current amplitudes were measured 3 ms after the ramp to +40 mV. For baseline and each concentration the peak currents of the three last sweeps before application of the next concentration were averaged. Residual currents ( 1/10) were calculated for each cell as the fraction of actual average peak current and average baseline peak current.
  • the compounds of general formula I according to the invention including the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the hydrates thereof, the solvates and the physiologically acceptable salts thereof, are suitable for the treatment and/or preventative treatment of all those diseases or conditions which can be influenced by inhibition of PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction.
  • compounds according to the invention are particularly suitable for the prevention or treatment of diseases, particularly (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood [affective] disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) and Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders.
  • diseases particularly (1) disorders comprising the symptom
  • the compounds according to the invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory.
  • the compounds according to the invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis.
  • the compounds of the present invention can be used for the treatment of Alzheimer's disease.
  • compounds according toe the invention can be used for the treatment of pain disorders including but not limited to inflammatory, neuropatic and osteoarthritic pain.
  • the compounds according to the invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.
  • the compounds according to the invention are suitable for the treatment of Alzheimer's Disease and for the treatment schizophrenia.
  • the compounds according to the invention are suitable for symptomatic treatment of Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia.
  • the compounds according to the invention are suitable for symptomatic treatment of prodromal and mild-to-moderate Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia.
  • the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of general formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the hydrates thereof, the solvates and the physiologically acceptable salts thereof, to a human being.
  • the dose range of the compounds of general formula I applicable per day is usually from 0.1 to 1000 mg, preferably from 1 to 500 mg by oral route, in each case administered 1 to 4 times a day.
  • Each dosage unit may conveniently contain from 0.1 to 500 mg, preferably 1 to 100 mg.
  • the actual pharmaceutically effective amount or therapeutic dosage will of course depend on factors known by those skilled in the art such as age and weight of the patient, route of administration and severity of disease. In any case the combination will be administered at dosages and in a manner which allows a pharmaceutically effective amount to be delivered based upon patient's unique condition.
  • Suitable preparations for administering the compounds of formula I, including the physiologically acceptable salts thereof, will be apparent to those with ordinary skill in the art and include for example tablets, pills, capsules, suppositories, lozenges, troches, solutions, syrups, elixirs, sachets, injectables, inhalatives, powders, etc.
  • the content of the pharmaceutically active compound(s) should be in the range from 0.1 to 95 wt.-%, preferably 5.0 to 90 wt.-% of the composition as a whole.
  • Suitable tablets may be obtained, for example, by mixing one or more compounds according to formula I with known excipients, for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants.
  • excipients for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants.
  • the tablets may also consist of several layers.
  • the compounds of formula I prepared according to the invention may be formulated, optionally together with other active substances, together with one or more inert conventional carriers and/or diluents, e.g. with corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, citric acid, tartaric acid, water, polyvinylpyrrolidone, water/ethanol, water/glycerol, water/sorbitol, water/polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof.
  • inert conventional carriers and/or diluents e.g. with corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, citric acid, tartaric acid, water, polyvinylpyrrolidone, water/ethanol, water/glycerol, water/sorbitol, water/polyethylene glycol, propylene glycol, cetylstearyl alcohol
  • the compounds according to the invention may also be used in conjunction with other active substances, particularly for the treatment and/or prevention of the diseases and conditions mentioned above.
  • Other active substances which are suitable for such combinations include, for example, BACE inhibitors; amyloid aggregation inhibitors (e.g. ELND-005); directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants (e.g. vitamin E or ginkolide); anti-inflammatory substances (e.g.
  • Cox inhibitors NSAIDs additionally or exclusively having Abeta lowering properties
  • HMG-CoA reductase inhibitors statins
  • acetylcholinesterase inhibitors e.g., donepezil, rivastigmine, tacrine, galantamine
  • NMDA receptor antagonists e.g.
  • AMPA receptor agonists AMPA receptor positive modulators
  • AMPAkines monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmitters
  • substances inducing the secretion of growth hormone e.g., ibutamoren mesylate and capromorelin
  • CB-1 receptor antagonists or inverse agonists antibiotics (e.g., minocyclin or rifampicin); PDE2, PDE4, PDE5, PDE9, PDE10 inhibitors, GABAA receptor inverse agonists, GABAA receptor antagonists, nicotinic receptor agonists or partial agonists or positive modulators, alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators, alpha7 nicotinic receptor agonists or partial agonists or positive modulators; histamine H3 antagonists, 5 HT-4 agonists or partial agonists, 5HT-6 antagonists, alpha2-adrenorecept
  • the compounds according to the invention may also be used in combination with immunotherapies (e.g., active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or nanobodies) for the treatment of the above-mentioned diseases and conditions.
  • immunotherapies e.g., active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or nanobodies
  • the dosage for the combination partners mentioned above is usefully 1/5 of the lowest dose normally recommended up to 1/1 of the normally recommended dose.
  • this invention relates to the use of a compound according to the invention or a physiologically acceptable salt thereof combined with at least one of the active substances described above as a combination partner, for preparing a pharmaceutical composition which is suitable for the treatment or prevention of diseases or conditions which can be affected by inhibitors of phosphodiesterase 2 and/or 10.
  • a pharmaceutical composition which is suitable for the treatment or prevention of diseases or conditions which can be affected by inhibitors of phosphodiesterase 2 and/or 10.
  • These are preferably pathologies related to PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction, particularly one of the diseases or conditions listed above, most particularly prodromal and mild-to-moderate Alzheimer's Disease and cognitive impairment associated with schizophrenia.
  • the use of the compound according to the invention, or a physiologically acceptable salt thereof, in combination with another active substance may take place simultaneously or at staggered times, but particularly within a short space of time. If they are administered simultaneously, the two active substances are given to the patient together; while if they are used at staggered times the two active substances are given to the patient within a period of less than or equal to 12 hours, but particularly less than or equal to 6 hours.
  • this invention relates to a pharmaceutical composition which comprises a compound according to the invention or a physiologically acceptable salt of such a compound and at least one of the active substances described above as combination partners, optionally together with one or more inert carriers and/or diluents.
  • the compound according to the invention, or a physiologically acceptable salt thereof, and the additional active substance to be combined therewith may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts.
  • Instrument GC/MS Thermo Scientific TRACE GC ULTRA, DSQ II MS single quadrupole; column: Agilent DB-5MS, 25 m ⁇ 0.2 5 mmol ⁇ 0.25 ⁇ m; carrier gas: Helium, 1 mL/min costant flow; oven program: 50° C., to 100° C. in 10° C./min, to 200° C. in 20° C./min, to 320° C. in 30° C./min (hold 10 min); detection: DSQ II MS single quadrupole; ion source: EI; scan range: 50-450 amu.
  • the most suitable purification techniques applied for the purification of compounds of the present invention are direct phase silica gel flash chromatography and reverse phase chromatography or preparative HPLC.
  • racemic mixture points to the two stereochemical options and thus the manufactured compounds is a mixture of:
  • single stereoisomer indicates that the absolute configuration is unknown.
  • Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC
  • single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.
  • TRANS-single stereoisomer indicates a relative configuration known (trans) and the planar bond indicates the unknown absolute configuration.
  • Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC
  • single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.
  • Step 1 was performed in analogy to what reported in literature reference: Li, Yuanzhen et al. Organic Letters, 2007, vol. 9, #20 p. 4057-4060, starting from Ester Intermediate 5c.
  • Step 1 was performed in analogy to what reported in the literature reference: Duong, Hung A.; et al. Angewandte Chemie, International Edition, 2011, vol. 50, p. 463-466, starting from commercially available (3-bromo-4-chloro-phenyl)-acetic acid.
  • Step 3 was performed in analogy to Step 2 starting from Intermediate obtained in Step 2.
  • Step 4 was performed in analogy to preparation of Intermediate 5o, starting from Intermediate obtained in Step 3.
  • Step 1 was performed in analogy to what described in Step 1 in the preparation of Intermediate 7a, starting from commercially available 2-Isopropenyl-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane.
  • Chloride Intermediates were prepare in analogy to Chloride Intermediate 9 and purified applying the most suitable purification technique, starting from the corresponding Amide Intermediates. Heating temperature reported in Table.
  • Epoxide Intermediate 29a (4 g) was prepared in analogy to what described in WO2010/47956 A1, starting from methylene cyclobutane (5 g, 73 mmol).
  • Epoxide Intermediate 29b (1 g) was prepared in analogy to what described in WO2013/55577 A1 starting from 3,6-Dihydro-2H-pyran (2 g, 23.78 mmol).
  • Example 1 To a solution of Example 1 (0.2 g, 0.67 mmol) in dry DMF (3 mL), cesium carbonate (0.63 g, 1.92 mmol) and Intermediate 28 (0.41 g, 1.28 mmol) were added. Mixture was heated at 80° C. for 1 h, then solvent evaporated under reduced pressure and the residue dissolved in DOM and washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.21 g of the title compound.
  • Example 1 To a solution of Example 1 (0.4 g, 1.3 mmol) in dry DMF (4 mL), cesium carbonate (0.85 g, 2.62 mmol) and methylbromoacetate (0.152 mL, 1.56 mmol) were added. Mixture was stirred at room temperature overnight, solvent evaporated and the residue treated with EtOAc. A saturated solution of NH 4 Cl was added, phases were separated and evaporated. The crude was purified by flash chromatography (eluent from 50:50 to 0:100 Cy/EtOAc) to obtain 0.29 g of the title compound.
  • Example 12 (0.04 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Intermediate 9f (0.3 g, 0.68 mmol) and Hydrazide Intermediate 4 (0.27, 0.68 mmol).
  • Example 12a (0.15 g) was prepared in analogy to Example 12 and purified applying the most suitable purification technique, starting from the Hydrazide Intermediate 6a (0.23 g, 1.45 mmol).
  • Example 1 To a solution of Example 1 (0.1 g, 0.24 mmol) in dry ACN (3 mL), cesium carbonate (0.2 g) and commercially available 3-chloro-1-propanol (0.04 mL) were added. Mixture was heated at 80° C. for 1 h, then solvent evaporated under reduced pressure and the residue dissolved in DCM and washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evapoarated. The residue was purified by flash chromatography (eluent from 100:0 to 94:6 DCM/EtOH) to obtain 0.02 g of the title compound.
  • Example 1 To a solution of Example 1 (0.05 g, 0.16 mmol) in dry DMF (3 mL), Intermediate 10 (0.05 g, 0.018 mmol) and cesium carbonate (0.08 g, 0.24 mmol) were added and mixture heated at 100° C. for 2 h. Solvent was evaporated, the residue was dissolved with DCM, washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 70:30 to 20:80 Cy/EtOAc) to obtain 0.025 g of the title compound.
  • Example 1 To a solution of Example 1 (0.08 g, 0.16 mmol) in dry ACN (3 mL), commercially available 2-bromo-ethanol (0.65 g, 0.52 mmol) and cesium carbonate (0.5 g, 1.54 mmol) were added and mixture heated at 80° C. for 48 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.04 g of the title compound.
  • Example 1 To a solution of Example 1 (0.05 g, 0.16 mmol) in dry DMF (3 mL), commercially available 1-bromo-2-propanol (0.09 g, 0.64 mmol) and potassium tert-butoxide (0.1 g, 0.96 mmol) were added and mixture heated at 130° C. for 8 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.05 g of the title compound.
  • Example 1 To a solution of Example 1 (0.085 g, 0.27 mmol) in dry DMF (3 mL), commercially available (R)-1-chloro-2-propanol (0.052 g, 0.55 mmol) and potassium tert-butoxide (0.093 g, 0.82 mmol) were added and mixture heated at 130° C. for 48 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.0085 g of the title compound.
  • Example 1 To a solution of Example 1 (0.115 g, 0.37 mmol) in dry DMF (3 mL), cesium carbonate (0.36 g, 1.11 mmol) and Intermediate 11 (0.09 g, 0.37 mmol) were added and mixture heated at 100° C. for 4 h. Solvent was evaporated, the residue was dissolved with DCM, washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 80:20 DCM/EtOH) to obtain 0.025 g of the title compound.
  • Example 20 was prepared as described for Example 19, starting from Example 1 (0.08 g, 0.26 mmol) and Intermediate 13 (0.2 g, 0.78 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.032 g of the title compound.
  • Example 21 was prepared as described for Example 20, starting from Example 1 (0.065 g, 0.21 mmol) and Intermediate 14 (0.12 g, 0.42 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.035 g of the title compound.
  • Example 22 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 15 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.032 g of the title compound.
  • Example 23 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 17 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.02 g of the title compound.
  • Example 24 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 16 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.03 g of the title compound.
  • Example 25 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 18 (0.097 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.03 g of the title compound.
  • Example 26 was prepared as described for Example 20, starting from Example 4 (0.08 g, 0.27 mmol) and commercially available 3-chloro-1-propanol (0.052 g, 0.54 mmol). The crude was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.052 g of the title compound.
  • Example 27 was prepared as described for Example 20, starting from Example 2 (0.133 g, 80% content, 0.32 mmol) and commercially available 3-chloro-1-propanol (0.046 g, 0.49 mmol). The crude was purified by prep-HPLC (Method 11). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM and treated with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.039 g of the title compound.
  • Example 28 was prepared as described for Example 20, starting from Example 2 (0.133 g, 80% content, 0.32 mmol) and commercially available 2-bromo-ethanol (0.061 g, 0.49 mmol). The crude was purified by prep-HPLC (Method 12). Fractions containing the pure compound are combined and evaporated to reduced volume, diluted with DCM and treated with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.025 g of the title compound.
  • Example 2 To a solution of Example 2 (0.07 g, 0.20 mmol) and commercially available 2-bromo-methylether (0.057 g, 0.41 mmol) in dry DMF (3 mL), cesium carbonate (0.199 g, 0.61 mmol) was added and the mixture heated at 100° C. for 1 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 40:60 hexane/EtOAc) to obtain 0.023 g of the title compound.
  • flash chromatography eluent from 80:20 to 40:60 hexane/EtOAc
  • Example 2 To a solution of Example 2 (0.07 g, 0.20 mmol) and Intermediate 14 (0.12 g, 0.41 mmol) in dry DMF (3 mL), cesium carbonate (0.2 g, 0.61 mmol) was added and the mixture heated at 90° C. for 2 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 40:60 hexane/EtOAc) and then further purified by prep-HPLC (Method 14). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.047 g of the title compound.
  • Example 31 was prepared as described for Example 30 starting from Example 2 (0.07 g, 0.20 mmol) and commercially available 1-bromo-3-methoxy-propane (0.063 g, 0.41 mmol). After heating for 3 h at 100° C. in dry DMF (3 mL), solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) to obtain 0.043 g of the title compound.
  • Example 32 was prepared as described for Example 31 starting from Example 2 (0.09 g, 0.26 mmol) and Intermediate 13 (0.20 g, 0.78 mmol). Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) to obtain 0.037 g of the title compound.
  • Example 32a (0.06 g) was prepared in analogy to Example 32 and purified applying the most suitable purification technique, starting from Example 9 (0.08 g, 0.23 mmol).
  • Example 33 was prepared as described for Example 31 starting from Example 2 (0.07 g, 0.20 mmol) and commercially available 1-bromo-3-trifluoro-methoxypropane (0.084 g, 0.41 mmol). Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) and then further purified by prep-HPLC (Method 14). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM.
  • Example 2 To a solution of Example 2 (0.082 g, 0.25 mmol) in dry DMF (3 mL), potassium tert-butoxide (0.084 g, 0.65 mmol) and Intermediate 20 (0.176 g, 0.50 mmol) were added. The resulting solution was heated at 120° C. for 5 h. The mixture was left at room temperature for 24 h. Solvent was evaporated under reduced pressure, the residue dissolved in DCM and treated with a 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 20:80 to 0:100 ACN/H 2 O) to obtain 0.033 g of the title compound.
  • Example 2 To a solution of Example 2 (0.095 g, 0.29 mmol) in dry DMF (3 mL), potassium tert-butoxide (0.074 g, 0.22 mmol and Intermediate 19 (0.204 g, 0.58 mmol) were added. The resulting solution was heated at 120° C. for 5 h. Solvent was evaporated under reduced pressure, the residue dissolved in DCM and treated with a 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 0:100 Cy/EtOAc) to obtain 0.023 g of the title compound.
  • Example 3 To a solution of Example 3 (0.07 g, 0.24 mmol) in dry DMF (3 mL), cesium carbonate (0.23 g, 0.71 mmol) and commercially available 3-chloro-1-propanol (0.024 g, 0.26 mmol) were added and the mixture heated at 100° C. for 3 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by prep-HPLC (Method 11). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM and washed with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.02 g of the title compound.
  • Example 37 was prepared as described for Example 32 starting from Example 1 (0.07 g, 0.22 mmol) and Intermediate 26 (0.22 g, 067 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.046 g of the title compound.
  • Example 38 was prepared as described for Example 32 starting from Example 1 (0.07 g, 0.22 mmol) and Intermediate 27 (0.127 g, 044 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.045 g of the title compound.
  • Example 40 was prepared as described Example 14, starting from Example 1 (0.075 g, 0.24 mmol) and Intermediate 31 (0.12 g, 0.48 mmol). Crude was purified by flash chromatography (eluent from 100:0 to 95:5 DCM/EtOH) to obtain 0.046 g of the title compound.
  • Example 41 (0.05 g) was prepared as described for Example 22 and purified applying the most suitable purification technique, starting from Example 2 (0.1 g, 0.3 mmol).
  • Example 42 (0.06 g) was prepared as described for Example 23 and purified applying the most suitable purification technique, starting from Example 2 (0.09 g, 0.28 mmol).
  • Example 43 (0.03 g) was prepared as described for Example 24 and purified applying the most suitable purification technique, starting from Example 2 (0.09 g, 0.28 mmol).
  • Example 1 (0.07 mg, 0.2 mmol) was dissolved in DMF (3 mL), isobutylene oxide (0.03 mg, 0.43 mmol) and cesium carbonate (208 mg, 0.64 mmol) were added and the reaction mixture was warmed to 100° C. for 2 h. The solvent was removed, dichloromethane was added and the organic phase was washed with a 1M NaOH water solution, dried over sodium sulfate, concentrated under vacuum. The crude product obtained was purified by reverse phase chromatography to give the desired compound (29 mg).
  • Example 44 The following Examples were prepared in analogy to Example 44 and purified applying the most suitable purification technique, starting from the corresponding Starting Example and the suitable epoxides.
  • Example 58 (0.01 g) was prepared in analogy to example 44 starting from Example 1 (0.2 g, 0.61 mmol) and epoxide intermediate 29 (0.07 g, 0.73 mmol) after chiral
  • Example 58b was obtained by chiral HPLC separation of Example 58.
  • HPLC-MS (Method 13): R t 2.95 min
  • the reaction mixture was diluted with ethyl acetate and washed with water.
  • the organic phase was washed with brine, dried over sodium sulfate and concentrated under vacuum.
  • the crude product obtained was purified by flash chromatography (eluent from 100:0 to 80:20 Cy/EtOAc) to obtain the title compound (0.3 g).
  • Example 64 (0.03 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Intermediate 2 (0.1 g, 0.55 mmol) and Hydrazide Intermediate 6i (0.15 g, 0.55 mmol).
  • Example 65 (0.03 mg) was prepared in analogy to the preparation of Intermediate 5i and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30d (0.1 g, 0.25 mmol).
  • Example 70 (0.03 g) was prepared in analogy to the preparation of Intermediate 5j and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30b (0.16 g, 0.38 mmol) and acetone (0.07 mg, 1.13 mmol).
  • step 3 Intermediate from step 3 (0.17 g, 0.38 mmol) was dissolved in DCM (10 mL), manganese dioxide (0.38 g, 3.38 mmol) was added and the suspension was stirred for 1 h at room temperature. The reaction mixture was filtered on a celite pad and the solvent was evaporated. The crude product obtained was purified by flash chromatography (eluent 90:10 DCM/MeOH) to obtain the desired compound (0.14 M.
  • Example 80 (0.02 g) was prepared in step 2, step 3 and step 4 step in analogy to Example 75 and purified applying the most suitable purification technique, starting from the Intermediate obtained in step 1.
  • Step 1 was performed as follow:
  • Step 2 Step 2 and Step 4 were performed in analogy to Example 75.
  • Example 80 was prepared in analogy to Example 80 and purified applying the most suitable purification technique, starting from the corresponding Scaffold Intermediates.
  • Example 82 (0.09 g) was prepared in analogy to Example 1 starting from Chloride Intermediate 2 (0.1 g, 0.61 mmol) and Hydrazide Intermediate 6p (0.2 g, 0.61 mmol).
  • Example 98 (0.026 g) was prepared in analogy to Example 1 starting from Chloride Intermediate 2 (0.3 g, 1.45 mmol) and Hydrazide Intermediate 6q (0.4 g, 1.56 mmol) after semipreparative chiral purification.
  • Example 99 (0.028 g) was obtained via further elution from the column in the semipreparative chiral chromatographic purification of Example 98.
  • Step 1 The intermediate obtained in Step 1 (0.4 g, 0.07 mmol) was dissolved in THF (6 mL) and stirred under nitrogen atmosphere at 0° C. for 5 minutes. Lithiumaluminium hydride (0.5 mL of a 2M solution in THF) was added. The reaction mixture was stirred at 0° C. for 10 minutes then allowed to reach room temperature and stirred for 0.5 h.
  • Example 101 (0.05 g) was prepared in analogy to Example 100 and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30i (0.36 g, 0.9 mmol).
  • Step 1 was performed in analogy to preparation of Example 1 starting from Chloride Intermediate 9 (0.3 g, 1.39 mmol) and Hydrazide Intermediate 6o (0.47 g, 1.39 mmol).
  • step 1 Intermediate from step 1 (0.09 g, 0.26 mmol) and hydrochloric acid (10 mL of a 37% water solution) were dissolved in 10 mL of 1,4-dioxane.
  • the reaction mixture was stirred at room temperature for 20 minutes.
  • a bicarbonate saturated water solution was added, THF was removed in vacuum and the reaction mixture was extracted with DCM.
  • the organic phase was separated, dried over sodium sulfate and concentrated.
  • the crude product obtained was purified by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH) to obtain the title compound (0.03 g).
  • Step 2 was performed in analogy to Step 2 in the preparation of Example 102. 0.02 g of the desired product were obtained after purification by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH).
  • Example 104a was obtained via Chiral HPLC purification of Example 104.
  • Example 104b Further elution from the chiral column gave Example 104b.
  • Example 105 (0.03 g) was prepared in analogy to Example 104, starting from 8-Cyclopropyl-1-[5-(3,6-dihydro-2H-pyran-4-yl)-2-fluoro-phenyl]-4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene (0.05 g, 0.12 mmol) which was obtained as by-product in the preparation of Example 95 and isolated from the same flash chromatography purification.
  • Example 44 The following Examples were prepared in analogy to Example 44 and purified applying the most suitable purification technique, starting from the corresponding Starting Example and the corresponding epoxides.
  • TRANS regioisomeric racemic mixtures were obtained in analogy to preparation of Example 44, starting from epoxide Intermediate 29 and the corresponding Starting Examples or Starting Intermediates.

Abstract

The invention relates to 4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene derivatives of general formula (I) which are inhibitors of phosphodiesterase 2 and/or 10, useful in treating central nervous system diseases and other diseases. In addition, the invention relates to processes for preparing pharmaceutical compositions as well as processes for manufacture the compounds according to the invention.

Description

    FIELD OF THE INVENTION
  • The invention relates to 4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene derivatives of general formula (I) which are inhibitors of phosphodiesterase 2 and/or 10, useful in treating central nervous system diseases and other diseases. In addition, the invention relates to processes for preparing pharmaceutical compositions as well as processes for manufacture the compounds according to the invention.
  • Figure US20140045856A1-20140213-C00001
  • BACKGROUND OF THE INVENTION
  • Cognitive dysfunction plays a role in a number of central nervous system disorders, including neurological disorders, such as Alzheimer's disease (AD), Parkinson disease and dementia, but also psychiatric disorders, such as schizophrenia, depression and bipolar disorders. As world population grows older the number of patients with dementia and AD is growing. Therefore, most people are familiar with the cognitive deficits related to these neurological diseases (Massoud and Gauthier, 2010).
  • However, also in psychiatric disorders cognitive impairment adversely affect the progress and the treatment outcome of the disease. A most prominent example is schizophrenia. Schizophrenia has a heterogeneous symptomatic picture (American Psychiatric Association, 1994) that may be divided into three distinct disease domains: positive symptoms (psychotic episodes of hallucinations, delusions and agitation), negative symptoms (social withdrawal, anhedonia, flattened affect) and cognitive deficits (deficits in executive function, verbal learning and memory, verbal fluency) (Thompson and Meltzer, 1993).
  • Whereas positive symptoms are essentially alleviated by dopamine D2 receptor antagonist and second generation antipsychotics negative symptoms and cognitive deficits are still hardly affected by current treatments. Therefore, research on cognitive deficits in schizophrenia has been intensified over the past years. A worldwide network initiative, called MATRICS, has been founded to characterise the cognitive deficits more deeply and to find novel therapies (Young et al., 2009).
  • However, cognitive impairment is also seen in patients with depression, bipolar disorders (Sachs et al., 2007; Pavuluri et al., 2009) and in many patients with disorders usually first diagnosed in infancy, childhood and adolescence, such as attention deficit/hyperactivity disorder (ADHD) (Jucaite et al., 2005; Turner et al., 2003).
  • Depression is a severe mental disorder which extremely impairs daily life. Its prevalence is about 10% of the world population with an incidence of 2% according to WHO. Women are more affected than men and elder people more than younger people. The disorder mostly implies a life-long treatment due to the progression of the disease and permanent total disability.
  • The most prominent symptoms of the disease are anhedonia, feeling of hopelessness, decreased self esteem, loss of appetite and sleep disturbances. Suicide ideation is also a common symptom of depression and about 10% of depressed patients (Holma et al., 2010) attempt suicide. Depression is often combined with anxiety disorders. Interestingly, it is less known that depression is also regularly associated with various cognitive impairments (Gualtieri et al., 2006; Mandelli et al., 2006). Here, deficits of attentional and executive functions are mostly reported (Paelecke-Habermann et al., 2005). Cognitive deficits are even discussed to be involved in the development of the disease (Beck depression model, Beck, 2008). More recent studies indicate that the severity of the cognitive deficits may predict non-response to certain antidepressant treatment (Dunkin et al., 2000; Gorlyn et al., 2008).
  • Up to now, current antidepressant therapy seems not to be sufficient regarding cognitive deficits. Elder antidepressants are reported to impair memory in animal models of learning and memory probably due to their anticholinergic component (Kumar and Kulkarni, 1996). In contrast, SSRIs, in particular fluoxetine, are described to impair hippocampal-independent but not hippocampal dependent learning in different rodent models (Valluzi and Chan, 2007). At least, in clinic current therapy it is not possible to fully reverse cognitive deficits. Thus, in depressive patients who had been successfully treated cognitive performance could be improved but not normalised (Gualtieri et al., 2006). Therefore, an antidepressant with higher efficacy on cognitive impairment may improve disease outcome.
  • Bipolar disorders are characterized by complex symptomatology, including severe symptoms of mood disorders but also manic episodes and cognitive deficits. The Diagnostic and Statistical Manual, 4th edition and International Classification of Mental Disorder recommend subgroups of bipolar disorder based on whether depressive or manic [psychotic] symptoms and episodes are dominating and on the frequency of the episodes (Gaiwani, 2009). Pharmacological agents commonly used in the management of bipolar disorder include lithium; anticonvulsants, such as valproate, carbamazepine and lamotrigine; and recent years have witnessed increasing use of atypical antipsychotics (Altamura et al., 2011). As a problem of current therapy the development of tolerance against anticonvulsant treatment and 30% of treatment refractory cases are described (Post and Weiss, 2010; Gaiwani, 2009).
  • Attention deficit hyperactivity disorder (ADHD) is a central nervous system disorder that is mainly defined by its clinical signs. ADHD shows a heterogeneous symptom pattern in humans. The most important indicators are attention deficits, impulsivity and a hyperactivity that is primarily seen in boys. The disease starts at an early age and symptoms are most intense during childhood. After puberty the signs of the disease are more masked and focus on cognitive dysfunction (Jucaite et al. 2005; Turner et al. 2003). Although modern research broadened the understanding of the pathomechanism the exact etiology of the disease remains unclear.
  • Interestingly, the symptoms seen in ADHD are not due to a hyperactivity but a hypoactivity of the so called executive loop of the striatum (Winstanley et al., 2006; Plizska, 2005). The executive loop is responsible for the regulation of cognitive processes such as planning, working memory and attention (Benke et al., 2003; Easton et al., 2007). A dysfunction of the prefrontal cortex or other pathways within the loop induces impulsivity and a loss of the ability to filter stimuli that come from the outside. The latter causes the symptoms of sustained attention and hyperactivity (Roberts and Wallis, 2000; Gonzales et al., 2000). The dopaminergic neurotransmitter system plays a central role in regulating the activity of the executive loop (Jucaite et al., 2005). This conclusion is also supported by the current treatment for ADHD that aims for an activation of the dopaminergic neurotransmitter system (Jucaite et al., 2005).
  • Phosphodiesterases (PDE) are expressed in nearly all mammalian cells. To date eleven families of phosphodiesterases have been identified in mammals (Essayan, 2001). It is well established that PDEs are critically involved in cell signalling. Specifically, PDEs are known to inactivate the cyclic nucleotides cAMP and/or cGMP (Soderling and Beavo, 2000). The cyclic nucleotides cAMP and cGMP are synthesised by the adenylyl and guanylyl cyclases and are second messengers that control many key cellular functions. The synthesis of cAMP and cGMP is regulated by different G-protein-coupled receptor types including dopamine D1 and D2 receptors (Mutschler, 2001).
  • The phosphodiesterases of the different families vary in their substrate selectivity. Thus, some families only hydrolyse cAMP others only cGMP. Some phosphodiesterases, such as phosphodiesterase 2 and 10, inactivate both cAMP and cGMP (Menniti et al., 2006).
  • Furthermore, there is a difference in the distribution of the different phosphodiesterases within the organism and additionally, within any particular tissue or organ. For instance, the distribution pattern of the phosphodiesterases within the brain is quite specific (Menniti et al., 2006).
  • Finally, phosphodiesterase families have different regulatory properties and intracellular location; some are bound to cell membranes and some are dissociated in the cytoplasm, additionally, a division into various intracellular compartments has been reported (Conti and Jin, 1999).
  • These differences in the function and location of the different PDE enzyme families suggest that the individual phosphodiesterases are selectively involved in regulating many different physiological processes. Accordingly, selective phosphodiesterase inhibitors may with fine specificity regulate different physiological and pathophysiological processes.
  • PDE2 and PDE10 hydrolyse both, cGMP and cAMP (Menniti et al., 2006; Soderling et al., 1999; Kotera et al., 1999).
  • They are both abundantly expressed in the brain indicating their relevance in CNS function (Bolger et al., 1994; Menniti et al., 2001).
  • PDE2 mRNA is mainly distributed in olfactory bulb, olfactory tubercle, cortex, amygdala, striatum, and hippocampus (Lakics et al., 2005; van Staveren et al., 2003). PDE10 (PDE10A) is primarily expressed in the nucleus accumbens and the caudate putamen. Areas with moderate expression are the thalamus, hippocampus, frontal cortex and olfactory tubercle (Menniti et al., 2001).
  • Although there are certainly fine differences in the function and expression patterns of PDE2 and 10, the expression of PDE2 in the hippocampus, the cortex and in the striatum and the expression of PDE10 in striatum, hippocampus and frontal cortex indicate an involvement in the mechanism of learning and memory/cognition. This is further supported by the fact that increased levels of both cGMP and cAMP are involved in the process of short and long term potentiation (LTP) forming (Blokland et al., 2006; Prickaerts et al., 2002). LTP is regarded as the electrophysiological basis of long term memory (Baddeley, 2003). Boess et al. (2004) showed that PDE2 inhibitors amplify the generation of LTP. Additionally, it is reported that the selective PDE2 inhibitor BAY60-7550 enhances learning and memory in rats and mice in different animal models (Boess et al., 2004; Rutten et al., 2006). Similar pro-cognitive effects are described for selective PDE10 inhibitors, such as papaverine and MP-10. Rodefer et al. (2005) have found that papaverine reverses attentional set-shifting deficits induced by subchronic administration of phencyclidine, an NMDA antagonist, in rats. Grauer et al. (2009) could show a positive effect of papaverine and MP-10 on cognitive deficits in the novel object recognition and in prepulse inhibition of acoustic startle response in rats. These data support the procognitive effect of PDE2 and/or 10 and a synergistic effect of PDE2 and 10 on cognition.
  • Furthermore, the expression of PDE2 in the nucleus accumbens (part of the striatum), the olfactory bulb, the olfactory tubercle and the amygdala and the expression of PDE10 in the nucleus accumbens, the olfactory tubercle and the thalamus supports additional involvement of PDE2 and 10 in the pathophysiology of anxiety and depression (Modell et al., 1990). This is supported by in vivo studies. The selective PDE2 inhibitors BAY60-7550 and ND-7001 are described to be effective in animal models of anxiety and stress-induced behavior (Masood et al., 2008, 2009).
  • In addition to the pro-cognitive and antidepressant potential of PDE10 inhibition there is evidence for an additional antipsychotic potential of PDE10 inhibitors. In the striatum PDE10 is predominately found postsynaptic in the medium spiny neurons (Xie et al., 2006). By this location, PDE10 may have an important influence on the signal cascade induced by dopaminergic and glutamatergic input on the striatum, two neurotransmitter systems playing a predominate role in the pathomechanism of psychosis. Focusing on the dopaminergic input on the medium spiny neurons, PDE10A inhibitors by up-regulating cAMP and cGMP levels act as D1 agonists and D2 antagonists because the activation of Gs-protein coupled dopamine D1 receptor increases intracellular cAMP, whereas the activation of the Gi-protein coupled dopamine D2 receptor decreases intracellular cAMP levels through inhibition of adenylyl cyclase activity (Mutschler et al., 2001). Accordingly, PDE10 inhibitors are reported to be active in several animal models of schizophrenia (Schmidt et al., 2008; Siuciak et al., 2006; Grauer et al., 2009).
  • PDE10 inhibitors have been disclosed recently in J. Med. Chem., 2011, 54, 7621-7638.
  • For drugs with an intended action in the central nervous system (CNS), it is assumed that unbound drug in interstitial spaces in the brain is in direct contact or in equilibrium with the site of action (de Lange and Danhof, 2002).
  • It is commonly accepted that unbound or free drug is the species available for interaction with drug targets within the body, and this is referred to as the free drug hypothesis.
  • Because cerebrospinal fluid (CSF) is in direct contact with the brain tissue, it is assumed to readily equilibrate with brain interstitial fluid concentration (Meineke et al., 2002; Shen et al., 2004) so that CSF concentration is used as a common surrogate measure for drug unbound concentration in clinical pharmacology studies (Bonati et al., 1982; Cherubin et al., 1989; Garver, 1989; Reiter and Doron, 1996; Ostermann et al., 2004). Accordingly, for compounds with an intended action in the central nervous system it is important that they reach a high CSF concentration and a high CSF to plasma ratio in order to have high pharmacological activity in the CNS.
  • Inhibition of the hERG channel by xenobiotics and subsequent delayed cardiac repolarization is associated with an increased risk for a specific polymorphic ventricular tachyarrhythmia, torsade de pointes, as established by Sanguinetti et al. (1995, Cell, Apr. 21, 81(2):299-307) and a large body of subsequent evidence. As such, low hERG channel inhibition, such as that shown by the compounds of the present invention, is highly desirable for therapeutics.
  • Several families of PDE2 inhibitors are known. Imidazotriazinones are described in WO 2002/068423 for the treatment of e.g. memory deficiency, cognitive disorders, dementia and Alzheimer's disease. Oxindoles are described in WO 2005/041957 for the treatment of dementia. Further inhibitors of PDE2 are known from WO 2007/121319 for the treatment of anxiety and depression, from WO 2013/034761, WO 2012/104293 and WO2013/000924 for the treatment of neurological and psychiatric disorders, from WO 2006/072615, WO 2006/072612, WO 2006/024640 and WO 2005/113517 for the treatment of arthritis, cancer, edema and septic shock, from WO 2005/063723 for the treatment of renal and liver failure, liver dysfunction, restless leg syndrome, rheumatic disorders, arthritis, rhinitis, asthma and obesity, from WO 2005/041957 for the treatment of cancer and thrombotic disorders, from WO 2006/102728 for the treatment of angina pectoris and hypertension from WO 2008/043461 for the treatment of cardiovascular disorders, erectile dysfunction, inflammation and renal failure and from WO 2005/061497 for the treatment of e.g. dementia, memory disorders, cancer and osteoporosis.
  • Finally, benzodiazepines are described in WO 2005/063723 for the general treatment of CNS diseases including anxiety, depression, ADHD, neurodegeneration, Alzheimer's disease and psychosis.
  • AIM OF THE INVENTION
  • It has now been found that compounds of the present invention according to general formula I are effective inhibitors of phosphodiesterase 2 and/or 10.
  • Besides the inhibition property toward phosphodiesterase 2 and/or 10 enzymes, the compounds of the present invention provide further advantageous pharmacokinetic properties. For example the compounds of the present invention show high concentration in cerebrospinal fluid (CSF) and have a high CSF to plasma ratio, which translates in lower efficacious doses of the compounds for disease treatment and as a consequence in further potential advantages such as minimization of side effects. Furthermore, compounds of the present inventions show good metabolic stability and low potential of formation of biologically active metabolite and low hERG potassium channel inhibition.
  • Accordingly, one aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and salts thereof as inhibitors of phosphodiesterase 2 and/or 10.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 and reaching high concentrations in cerebrospinal fluid (CSF) and/or having high CSF to plasma ratio.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 and showing good metabolic stability.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 with low hERG channel inhibition.
  • Another aspect of the invention refers to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof as inhibitors of phosphodiesterase 2 and/or 10 with low potential of forming biologically active metabolite.
  • In a further aspect this invention relates to pharmaceutical compositions, containing at least one compound according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof optionally together with one or more inert carriers and/or diluents.
  • A further aspect of the present invention relates to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof for the use in the prevention and/or treatment of disorders associated with PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction.
  • Another aspect of the invention relates to processes of manufacture of the compounds of the present invention.
  • A further aspect of the present invention relates to compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof or pharmaceutical compositions comprising compounds according to formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the physiologically acceptable salts thereof for the use in the prevention and/or treatment of diseases or conditions which can be influenced by inhibition of PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction, such as (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood affective disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) including Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders.
  • In addition, the compounds of the present invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory.
  • In addition, the compounds of the present invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis. In addition, the compounds of the present invention can be used for the treatment of Alzheimer's disease.
  • In addition compounds of the present invention can be used for the treatment of pain disorders, including but not limited to inflammatory, neuropatic and osteoarthritic pain.
  • In addition, the compounds of the present invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.
  • Other aims of the present invention will become apparent to the skilled man directly from the foregoing and following remarks.
  • DETAILED DESCRIPTION
  • In a first aspect the present invention relates to compounds of general formula I
  • Figure US20140045856A1-20140213-C00002
  • wherein
    • A is selected from the group Aa consisting of
  • Figure US20140045856A1-20140213-C00003
      • wherein above mentioned phenyl-, pyridinyl-, pyrimidinyl-, pyridazinyl and pyrazinyl groups are substituted with R1 and R2;
    • R1 is selected from the group R1a consisting of
      • H, halogen, NC—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— with n=0, 1, 2, 3 or 4,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R2 is selected from the group R2a consisting of
      • H, HO—, halogen, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R3 is selected from the group R3a consisting of
      • H, halogen, NC—, C1-4-alkyl- and C3-6-cycloalkyl-,
        • wherein the above mentioned C1-4-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—;
    • R4, R5 are selected independently of each other from the group R4a/R5a consisting of
      • H, halogen, NC—, HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O—,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms, substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—;
    • R6 is selected from the group R6a consisting of
      • H, NC—, C1-6-alkyl-, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl- and C3-8-cycloalkyl-O—
        • wherein above mentioned C1-6-alkyl- groups may optionally be substituted with 1-3 halogen atoms;
    • R7 is selected from the group R7a consisting of
      • H, carbocyclyl, heterocyclyl and heteroaryl,
        • wherein above mentioned carbocyclyl, heterocyclyl and heteroaryl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of HO—, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-O— and halogen;
    • R8 is selected from the group R8a consisting of
      • C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl, wherein above mentioned C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-;
        the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Unless otherwise stated, the groups, residues, and substituents, particularly R1, R2, R3, R4/5, R6, R7, R8 and A are defined as above and hereinafter. If residues, substituents, or groups occur several times in a compound they may have the same or different meanings. Some preferred meanings of groups and substituents of the compounds according to the invention will be given hereinafter.
  • In a further embodiment of the present invention
    • A is selected from the group Ab consisting of
  • Figure US20140045856A1-20140213-C00004
      • wherein above mentioned phenyl- and pyridinyl- groups are substituted with R1 and R2.
  • In a further embodiment of the present invention
    • A is selected from the group Ac consisting of
  • Figure US20140045856A1-20140213-C00005
      • wherein above mentioned phenyl- and pyridinyl- groups are substituted with R1 and R2.
  • In a further embodiment of the present invention
    • A is selected from the group Ad consisting of
  • Figure US20140045856A1-20140213-C00006
  • In a further embodiment of the present invention
    • A is selected from the group Ae consisting of
  • Figure US20140045856A1-20140213-C00007
  • In a further embodiment of the present invention
    • A is selected from the group A1 consisting of
  • Figure US20140045856A1-20140213-C00008
      • wherein above mentioned phenyl-group is substituted with R1 and R2.
  • In a further embodiment of the present invention
    • A is selected from the group Ag consisting of
  • Figure US20140045856A1-20140213-C00009
  • In a further embodiment of the present invention
    • R1 is selected from the group R1b consisting of
      • H, halogen, C1-6-alkyl- and C3-6-cycloalkyl
        • wherein above mentioned C1-6-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R1 is selected from the group R1c consisting of
      • H, H3C—, F3C—, F2HC—, FH2C—, fluorine, chlorine and bromine.
  • In a further embodiment of the present invention
    • R1 is selected from the group R1c1 consisting of
      • H3C—, fluorine and chlorine.
  • In a further embodiment of the present invention
    • R1 is selected from the group R1d consisting of
      • H, fluorine and chlorine.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2a1 consisting of
      • HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2
        • wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups are substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2b consisting of
      • H, HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2 or 3 and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2b1 consisting of
      • HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2 or 3 and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O— groups are substituted with 1 to 3 substituents independently selected from the group consisting of HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl-, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2c consisting of
      • H, HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, R8—(CH2)n—O— with n=0, 1, 2, or 3, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2c1 consisting of
      • HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, R8—(CH2)n—O— with n=0, 1, 2, or 3, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups are substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2d consisting of
      • H, HO—, C1-4-alkyl-, C1-4-alkyl-O—, a saturated 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— with n=0, 1 or 2, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl-, C1-4-alkyl-O—, saturated 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2d1 consisting of
      • HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, a saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— with n=0, 1 or 2, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups are substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl-, C3-6-cycloalkyl-C1-3-alkyl-, saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R2 is selected from the group R2e consisting of
  • Figure US20140045856A1-20140213-C00010
  • In a further embodiment of the present invention
    • R2 is selected from the group R2e1 consisting of
  • Figure US20140045856A1-20140213-C00011
    Figure US20140045856A1-20140213-C00012
  • In a further embodiment of the present invention
    • R2 is selected from the group R2f consisting of
  • Figure US20140045856A1-20140213-C00013
  • In a further embodiment of the present invention
    • R2 is selected from the group R2f1 consisting of
  • Figure US20140045856A1-20140213-C00014
  • In a further embodiment of the present invention
    • R3 is selected from the group R3a1 consisting of
      • C3-6-cycloalkyl-,
        • wherein the above mentioned C3-6-cycloalkyl-group may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3b consisting of
      • H, C1-3-alkyl-, cyclobutyl- and cyclopropyl-,
        • wherein the above mentioned C1-3-alkyl-, cyclobutyl- and cyclopropyl-groups may optionally be substituted with 1 to 7 substituents independently selected from the group consisting of halogen, C1-3-alkyl-O—, NC— and HO—.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3c consisting of
      • H, and H3C— and cyclopropyl-,
        • wherein the above mentioned H3C— and cyclopropyl-groups may optionally be substituted with 1 to 3 fluorine atoms.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3d consisting of
      • H and H3C—,
        • wherein the above mentioned H3C-group may optionally be substituted with 1 to 3 fluorine atoms.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3e consisting of
      • H.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3f consisting of
      • H3C—,
        • wherein the above mentioned H3C-group may optionally be substituted with 1 to 3 fluorine atoms.
  • In a further embodiment of the present invention
    • R3 is selected from the group R3g consisting of
      • cyclopropyl-,
        • wherein the above mentioned cyclopropyl-group may optionally be substituted with 1 to 7 substituents independently selected from the group consisting of halogen, C1-3-alkyl-O—, NC— and HO—.
  • In a further embodiment of the present invention
    • R4, R5 are selected independently of each other from the group R4b/R5b consisting of
      • H, halogen, HO—, H3C—, F3C—, H3C—O—, F2HC—O—, FH2C—O—, F3C—O—, C1-4-alkyl-O—, R7—CH2—O— and R7—(CH2)2—O—,
        • wherein above mentioned C1-4-alkyl-O—, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—.
  • In a further embodiment of the present invention
    • R4, R5 are selected independently of each other from the group R4c/R5c consisting of
      • H, fluorine, chlorine, bromine, HO—, H3C—, F3C—, H3C—O—, F3C—O— and R7—CH2—O—.
  • In a further embodiment of the present invention
    • R4, R5 are selected from the group R4d/R5d consisting of
      • H—.
  • In a further embodiment of the present invention
    • R6 is selected from the group R6b consisting of
      • H, C1-4-alkyl- and cyclopropyl-,
        • wherein above mentioned C1-4-alkyl-group may optionally be substituted with 1-9 fluorine and/or chlorine atoms.
  • In a further embodiment of the present invention
    • R6 is selected from the group R6c consisting of
      • H and C1-2-alkyl-,
        • wherein above mentioned C1-2-alkyl- group may optionally be substituted with 1-5 fluorine and/or chlorine atoms.
  • In a further embodiment of the present invention
    • R6 is selected from the group R6d consisting of
      • H, H3C—, FH2C—, F2HC— and F3C—.
  • In a further embodiment of the present invention
    • R6 is selected from the group R6e consisting of
      • H3C—, FH2C—, F2HC— and F3C—.
  • In a further embodiment of the present invention
    • R6 is selected from the group R6f consisting of
      • H3C—.
  • In a further embodiment of the present invention
    • R7 is selected from the group R7b consisting of
      • H, phenyl, heteroaryl, cycloalkyl and heterocyclyl
        • wherein above mentioned phenyl, heteroaryl, cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of halogen, and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-O—.
  • In a further embodiment of the present invention
    • R7 is selected from the group R7c consisting of
      • H and phenyl,
        • wherein above mentioned phenyl group may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of halogen and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-O—.
  • In a further embodiment of the present invention
    • R7 is selected from the group R7d consisting of
      • H—.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8b consisting of
      • C3-6-cycloalkyl, heterocyclyl and heterocyclyl-C1-3-alkyl-, wherein above mentioned C3-6-cycloalkyl, heterocyclyl and heterocyclyl-C1-3-alkyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8c consisting of
      • C3-6-cycloalkyl and heterocyclyl,
        • wherein above mentioned C3-6-cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8c1 consisting of
      • C3-6-cycloalkyl and a saturated 4 to 6 membered monocyclic heterocycle containing one or two heteroatoms selected from N or O,
        • wherein above mentioned C3-6-cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8d consisting of
  • Figure US20140045856A1-20140213-C00015
      • wherein above mentioned groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 5 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8d1 consisting of
  • Figure US20140045856A1-20140213-C00016
      • wherein above mentioned groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 5 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8e consisting of
  • Figure US20140045856A1-20140213-C00017
      • wherein above mentioned groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8e1 consisting of
  • Figure US20140045856A1-20140213-C00018
      • wherein above mentioned groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-.
  • In a further embodiment of the present invention
    • R8 is selected from the group R8f consisting of
  • Figure US20140045856A1-20140213-C00019
  • In a further embodiment of the present invention
    • R8 is selected from the group R8f1 consisting of
  • Figure US20140045856A1-20140213-C00020
  • Each R1x, R2x, R3x, R4x/5x, R8x, R7x, R8x and Ax represents a characterized, individual embodiment for the corresponding substituent as described above. Thus given the above definitions, preferred individual embodiments of the first aspect of the invention are fully characterized by the term (R1x, R2x, R3x, R4x/5x, R6x, R7x, R8x and Ax), wherein for each index x an individual figure is given that ranges from “a” to the highest letter given above. All individual embodiments described by the term in parentheses with full permutation of the indices x, referring to the definitions above, shall be comprised by the present invention.
  • The following Table 1 shows, exemplarily and in the order of increasing preference from the first line to the last line, such embodiments E-1 to E-37 of the invention that are considered preferred. This means that embodiment E-37, represented by the entries in the last row of Table 1, is the most preferred embodiment.
  • TABLE 1
    Preferred embodiments E-1 to E-37 of the invention
    Ax R1x R2x R3x R4x/R5x R6x R7x R8x
    E-1 Ab R1b R2a R3b R4b/R5b R6b R7b R8a
    E-2 Ac R1b R2a R3b R4b/R5b R6b R7b R8a
    E-3 Ac R1c R2b R3b R4b/R5b R6b R7c R8b
    E-4 Ac R1c R2b R3c R4c/R5c R6b R7c R8c
    E-5 Ac R1c R2b R3c R4c/R5c R6b R7d R8c
    E-6 Ac R1c R2c R3c R4c/R5c R6c R7c R8c
    E-7 Ac R1c R2d R3c R4c/R5c R6c R7c R8d
    E-8 Ad R1c R2d R3d R4d/R5d R6c R8d
    E-9 Ae R1d R2d R3d R4d/R5d R6e R8f
    E-10 Ae R1d R2e R3d R4d/R5d R6f
    E-11 Af R1d R2d R3d R4d/R5d R6e R8f
    E-12 Af R1d R2e R3d R4d/R5d R6f
    E-13 Af R1d R2f R3d R4d/R5d R6e
    E-14 Af R1d R2f R3d R4d/R5d R6f
    E-15 Aa R1a R2a R3a1 R4a/R5a R6a R7a R8a
    E-16 Ab R1c R2c R3a1 R4b/R5b R6b R7b R8b
    E-17 Ad R1c R2d R3a1 R4c/R5c R6b R8c
    E-18 Ae R1c R2d R3a1 R4d/R5d R6b R8d
    E-19 Ag R1c R2d R3a1 R4d/R5d R6b R8f
    E-20 Ad R1c R2d R3g R4d/R5d R6b R8c1
    E-21 Ae R1c R2d R3g R4d/R5d R6b R8d
    E-22 Ag R1c R2d R3g R4d/R5d R6b R8f
    E-23 Aa R1a R2a1 R3a R4a/R5a R6a R7a R8a
    E-24 Ab R1b R2d1 R3a R4c/R5c R6b R7c R8b
    E-25 Ac R1c R2d1 R3a R4d/R5d R6c R8c1
    E-26 Ad R1c R2d R3b R4d/R5d R6c R8d1
    E-27 Ad R1c R2d1 R3c R4d/R5d R6d R8e1
    E-28 Ad R1c1 R2c1 R3c R4d/R5d R6d R8f1
    E-29 Ae R1c R2d1 R3b R4d/R5d R6c R8c1
    E-30 Ae R1c R2d1 R3b R4d/R5d R6d R8d1
    E-31 Ae R1c1 R2e1 R3c R4d/R5d R6e
    E-32 Af R1c R2d1 R3b R4d/R5d R6c R8e1
    E-33 Af R1c1 R2e1 R3c R4d/R5d R6e
    E-34 Af R1c1 R2f1 R3c R4d/R5d R6f
    E-35 Ag R1c R2c1 R3b R4d/R5d R6c R8e1
    E-36 Ag R1c R2d1 R3c R4d/R5d R6e R8f1
    E-37 Ag R1c1 R2f1 R3c R4d/R5d R6f
  • Accordingly, for example E-15 covers compounds of formula I,
  • wherein
    • A is selected from the group Aa consisting of
  • Figure US20140045856A1-20140213-C00021
      • wherein above mentioned phenyl-, pyridinyl-, pyrimidinyl-, pyridazinyl- and pyrazinyl- groups are substituted with R1 and R2;
    • R1 is selected from the group R1a consisting of
      • H, halogen, NC—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— with n=0, 1, 2, 3 or 4,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R2 is selected from the group R2a consisting of
      • H, HO—, halogen, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8— (CH2)m—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R3 is selected from the group R3a1 consisting of
      • C3-6-cycloalkyl-,
        • wherein the above mentioned C3-6-cycloalkyl-group may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—;
    • R4, R5 are selected independently of each other from the group R4a/R5a consisting of
      • H, halogen, NC—, HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O—,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms, substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—;
    • R6 is selected from the group R6a consisting of
      • H, NC—, C1-6-alkyl-, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl- and C3-8-cycloalkyl-O—
        • wherein above mentioned C1-6-alkyl- groups may optionally be substituted with 1-3 halogen atoms;
    • R7 is selected from the group R7a consisting of
      • H, carbocyclyl, heterocyclyl and heteroaryl,
        • wherein above mentioned carbocyclyl, heterocyclyl and heteroaryl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of HO—, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-O— and halogen;
    • R8 is selected from the group R8a consisting of
      • C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl, wherein above mentioned C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-;
        the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Accordingly, for example E-23 covers compounds of formula I,
  • wherein
    • A is selected from the group Aa consisting of
  • Figure US20140045856A1-20140213-C00022
      • wherein above mentioned phenyl-, pyridinyl-, pyrimidinyl-, pyridazinyl- and pyrazinyl- groups are substituted with R1 and R2;
    • R1 is selected from the group R1a consisting of
      • H, halogen, NC—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— with n=0, 1, 2, 3 or 4,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R2 is selected from the group R2a1 consisting of
      • HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2
        • wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups are substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R3 is selected from the group R3a consisting of
      • H, halogen, NC—, C1-4-alkyl- and C3-6-cycloalkyl-,
        • wherein the above mentioned C1-4-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—;
    • R4, R5 are selected independently of each other from the group R4a/R5a consisting of
      • H, halogen, NC—, HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O—,
        • wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms, substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—;
    • R6 is selected from the group R6a consisting of
      • H, NC—, C1-6-alkyl-, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl- and C3-8-cycloalkyl-O—
        • wherein above mentioned C1-6-alkyl- groups may optionally be substituted with 1-3 halogen atoms;
    • R7 is selected from the group R7a consisting of
      • H, carbocyclyl, heterocyclyl and heteroaryl,
        • wherein above mentioned carbocyclyl, heterocyclyl and heteroaryl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of HO—, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-O— and halogen;
    • R8 is selected from the group R8a consisting of
      • C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl, wherein above mentioned C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-;
        the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Accordingly, for example E-29 covers compounds of formula I,
  • wherein
    • A is selected from the group Ae consisting of
  • Figure US20140045856A1-20140213-C00023
    • R1 is selected from the group R1c consisting of
      • H, H3C—, F3C—, F2HC—, FH2C—, fluorine, chlorine and bromine:
    • R2 is selected from the group R2d1 consisting of
      • HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, a saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— with n=0, 1 or 2, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups are substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
        • and
        • wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
        • and
        • wherein above mentioned C3-6-cycloalkyl-, C3-6-cycloalkyl-C1-3-alkyl-, saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— groups may optionally be substituted with 1 to 5, preferably 1 to 3, substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
    • R3 is selected from the group R3b consisting of
      • H, C1-3-alkyl-, cyclobutyl- and cyclopropyl-,
        • wherein the above mentioned C1-3-alkyl-, cyclobutyl-, and cyclopropyl-groups may optionally be substituted with 1 to 7 substituents independently selected from the group consisting of halogen, C1-3-alkyl-O—, NC— and HO—;
    • R4, R5 are selected from the group R4d/R5d consisting of
      • H;
    • R6 is selected from the group R6c consisting of
      • H and C1-2-alkyl-,
        • wherein above mentioned C1-2-alkyl- group may optionally be substituted with 1-5 fluorine and/or chlorine atoms;
    • R8 is selected from the group Fe8c1 consisting of
      • C3-6-cycloalkyl and a saturated 4 to 6 membered monocyclic heterocycle containing one or two heteroatoms selected from N or O,
        • wherein above mentioned C3-6-cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-;
          the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Accordingly, for example E-37 covers compounds of formula I,
  • wherein
    • A is selected from the group Ag consisting of
  • Figure US20140045856A1-20140213-C00024
    • R1 is selected from the group R1c1 consisting of
      • H3C—, fluorine and chlorine;
    • R2 is selected from the group R2f1 consisting of
  • Figure US20140045856A1-20140213-C00025
    • R3 is selected from the group R3c consisting of
      • H, and H3C— and cyclopropyl-,
        • wherein the above mentioned H3C— and cyclopropyl-groups may optionally be substituted with 1 to 3 fluorine atoms;
    • R4, R5 are selected from the group R4d/R5d consisting of
      • H;
    • R6 is selected independently of each other from the group R6f consisting of H3C—;
      the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Further preferred are the following compounds listed in table 2:
  • No. Structure
    I
    Figure US20140045856A1-20140213-C00026
    II
    Figure US20140045856A1-20140213-C00027
    III
    Figure US20140045856A1-20140213-C00028
    IV
    Figure US20140045856A1-20140213-C00029
    V
    Figure US20140045856A1-20140213-C00030
    VI
    Figure US20140045856A1-20140213-C00031
    VII
    Figure US20140045856A1-20140213-C00032
    VIII
    Figure US20140045856A1-20140213-C00033
    IX
    Figure US20140045856A1-20140213-C00034
    X
    Figure US20140045856A1-20140213-C00035
    XI
    Figure US20140045856A1-20140213-C00036
    XII
    Figure US20140045856A1-20140213-C00037
    XIII
    Figure US20140045856A1-20140213-C00038
    XIV
    Figure US20140045856A1-20140213-C00039
    XV
    Figure US20140045856A1-20140213-C00040
    XVI
    Figure US20140045856A1-20140213-C00041
    XVII
    Figure US20140045856A1-20140213-C00042
    XVIII
    Figure US20140045856A1-20140213-C00043
    XIX
    Figure US20140045856A1-20140213-C00044
    XX
    Figure US20140045856A1-20140213-C00045
    XXI
    Figure US20140045856A1-20140213-C00046
    XXII
    Figure US20140045856A1-20140213-C00047
    XXIII
    Figure US20140045856A1-20140213-C00048
    XXIV
    Figure US20140045856A1-20140213-C00049
    XXV
    Figure US20140045856A1-20140213-C00050
    XXVI
    Figure US20140045856A1-20140213-C00051
    XXVII
    Figure US20140045856A1-20140213-C00052
    XXVIII
    Figure US20140045856A1-20140213-C00053
    XXIX
    Figure US20140045856A1-20140213-C00054
    XXX
    Figure US20140045856A1-20140213-C00055
    XXXI
    Figure US20140045856A1-20140213-C00056
    XXXII
    Figure US20140045856A1-20140213-C00057
    XXXIII
    Figure US20140045856A1-20140213-C00058
    XXXIV
    Figure US20140045856A1-20140213-C00059
    XXXV
    Figure US20140045856A1-20140213-C00060
    XXXVI
    Figure US20140045856A1-20140213-C00061
    XXXVII
    Figure US20140045856A1-20140213-C00062
    XXXVIII
    Figure US20140045856A1-20140213-C00063
    XXXIX
    Figure US20140045856A1-20140213-C00064
    XL
    Figure US20140045856A1-20140213-C00065
    XLI
    Figure US20140045856A1-20140213-C00066
    XLII
    Figure US20140045856A1-20140213-C00067
    XLIII
    Figure US20140045856A1-20140213-C00068
    XLIV
    Figure US20140045856A1-20140213-C00069
    XLV
    Figure US20140045856A1-20140213-C00070
    XLVI
    Figure US20140045856A1-20140213-C00071
    XLVII
    Figure US20140045856A1-20140213-C00072
    XLVIII
    Figure US20140045856A1-20140213-C00073
    XLIX
    Figure US20140045856A1-20140213-C00074
    L
    Figure US20140045856A1-20140213-C00075
    LI
    Figure US20140045856A1-20140213-C00076
    LII
    Figure US20140045856A1-20140213-C00077
    LIII
    Figure US20140045856A1-20140213-C00078
    LIV
    Figure US20140045856A1-20140213-C00079
    LV
    Figure US20140045856A1-20140213-C00080
    LVI
    Figure US20140045856A1-20140213-C00081
    LVII
    Figure US20140045856A1-20140213-C00082
    LVIII
    Figure US20140045856A1-20140213-C00083
    LIX
    Figure US20140045856A1-20140213-C00084
    LX
    Figure US20140045856A1-20140213-C00085
    LXI
    Figure US20140045856A1-20140213-C00086
    LXII
    Figure US20140045856A1-20140213-C00087
    LXIII
    Figure US20140045856A1-20140213-C00088
    LXIV
    Figure US20140045856A1-20140213-C00089
    LXV
    Figure US20140045856A1-20140213-C00090
    LXVI
    Figure US20140045856A1-20140213-C00091
    LXVII
    Figure US20140045856A1-20140213-C00092
    LXVIII
    Figure US20140045856A1-20140213-C00093
    LXIX
    Figure US20140045856A1-20140213-C00094
    LXX
    Figure US20140045856A1-20140213-C00095
    LXXI
    Figure US20140045856A1-20140213-C00096
    LXXII
    Figure US20140045856A1-20140213-C00097
    LXXIII
    Figure US20140045856A1-20140213-C00098
    LXXIV
    Figure US20140045856A1-20140213-C00099
    LXXV
    Figure US20140045856A1-20140213-C00100
    LXXVI
    Figure US20140045856A1-20140213-C00101
    LXXVII
    Figure US20140045856A1-20140213-C00102
    LXXVIII
    Figure US20140045856A1-20140213-C00103
    LXXIX
    Figure US20140045856A1-20140213-C00104
    LXXX
    Figure US20140045856A1-20140213-C00105
    LXXXI
    Figure US20140045856A1-20140213-C00106
    LXXXII
    Figure US20140045856A1-20140213-C00107
    LXXXIII
    Figure US20140045856A1-20140213-C00108
    LXXXIV
    Figure US20140045856A1-20140213-C00109
    LXXXV
    Figure US20140045856A1-20140213-C00110
    LXXXVI
    Figure US20140045856A1-20140213-C00111
    LXXXVII
    Figure US20140045856A1-20140213-C00112
    LXXXVIII
    Figure US20140045856A1-20140213-C00113
    LXXXIX
    Figure US20140045856A1-20140213-C00114
    XC
    Figure US20140045856A1-20140213-C00115
    XCI
    Figure US20140045856A1-20140213-C00116
    XCII
    Figure US20140045856A1-20140213-C00117
    XCIII
    Figure US20140045856A1-20140213-C00118
    XCIV
    Figure US20140045856A1-20140213-C00119
    XCV
    Figure US20140045856A1-20140213-C00120
    XCVI
    Figure US20140045856A1-20140213-C00121
    XCVII
    Figure US20140045856A1-20140213-C00122
    XCVIII
    Figure US20140045856A1-20140213-C00123
    XCIX
    Figure US20140045856A1-20140213-C00124
    C
    Figure US20140045856A1-20140213-C00125
    CI
    Figure US20140045856A1-20140213-C00126
    CII
    Figure US20140045856A1-20140213-C00127
    CIII
    Figure US20140045856A1-20140213-C00128
    CIV
    Figure US20140045856A1-20140213-C00129
    CV
    Figure US20140045856A1-20140213-C00130
    CVI
    Figure US20140045856A1-20140213-C00131
    CVII
    Figure US20140045856A1-20140213-C00132
    CVIII
    Figure US20140045856A1-20140213-C00133
    CIX
    Figure US20140045856A1-20140213-C00134
    CX
    Figure US20140045856A1-20140213-C00135
    CXI
    Figure US20140045856A1-20140213-C00136
    CXII
    Figure US20140045856A1-20140213-C00137
    CXIII
    Figure US20140045856A1-20140213-C00138
    CXIV
    Figure US20140045856A1-20140213-C00139
    CXV
    Figure US20140045856A1-20140213-C00140
    CXVI
    Figure US20140045856A1-20140213-C00141
    CXVII
    Figure US20140045856A1-20140213-C00142
    CXVIII
    Figure US20140045856A1-20140213-C00143
    CXIX
    Figure US20140045856A1-20140213-C00144
    CXX
    Figure US20140045856A1-20140213-C00145
    CXXI
    Figure US20140045856A1-20140213-C00146
    CXXII
    Figure US20140045856A1-20140213-C00147
    CXXIII
    Figure US20140045856A1-20140213-C00148
    CXXIV
    Figure US20140045856A1-20140213-C00149
    CXXV
    Figure US20140045856A1-20140213-C00150
    CXXVI
    Figure US20140045856A1-20140213-C00151
    CXXVII
    Figure US20140045856A1-20140213-C00152
    CXXVIII
    Figure US20140045856A1-20140213-C00153
    CXXIX
    Figure US20140045856A1-20140213-C00154
    CXXX
    Figure US20140045856A1-20140213-C00155
    CXXXI
    Figure US20140045856A1-20140213-C00156
    CXXXII
    Figure US20140045856A1-20140213-C00157
    CXXXIII
    Figure US20140045856A1-20140213-C00158
    CXXXIV
    Figure US20140045856A1-20140213-C00159
    CXXXV
    Figure US20140045856A1-20140213-C00160
    CXXXVI
    Figure US20140045856A1-20140213-C00161

    the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the solvates thereof, the hydrates thereof and the salts thereof.
  • Some terms used above and hereinafter to describe the compounds according to the invention will now be defined more closely.
  • Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to. In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example C1-6-alkyl means an alkyl group or radical having 1 to 6 carbon atoms. In general, for groups comprising two or more subgroups, the last named subgroup is the radical attachment point, for example, the substituent “aryl-C1-3-alkyl-” means an aryl group which is bound to a C1-3-alkyl group, the latter of which is bound to the core molecule or to the group to which the substituent is attached.
  • Within the present invention, the term “core molecule” is defined by the following structure:
  • Figure US20140045856A1-20140213-C00162
  • In general, the attachment site of a given residue to another group shall be variable, i.e. any capable atom, bearing hydrogens to be replaced, within this residue may be the linking spot to the group being attached, unless otherwise indicated.
  • In case a compound of the present invention is depicted in form of a chemical name and as a formula in case of any discrepancy the formula shall prevail.
  • An asterisk may be used in sub-formulas to indicate the bond which is connected to the core molecule or to the substituent to which it is bound as defined.
  • Unless specifically indicated, throughout the specification and the appended claims, a given chemical formula or name shall encompass tautomers and all stereo, optical and geometrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc. . . . ) and racemates thereof as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms where such isomers and enantiomers exist, as well as salts, including pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound.
  • The phrase “pharmaceutically acceptable” or “physiologically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.
  • As used herein, “pharmaceutically acceptable salts” or “physiologically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts or physiologically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. For example, such salts include salts from ammonia, L-arginine, betaine, benethamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine (2,2′-iminobis(ethanol)), diethylamine, 2-(diethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-glucamine, hydrabamine, 1H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium hydroxide, 1-(2-hydroxyethyl)-pyrrolidine, sodium hydroxide, triethanolamine (2,2′,2″-nitrilotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2,2-dichloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 2,5-dihydroxybenzoic acid, 4-acetamido-benzoic acid, (+)-camphoric acid, (+)-camphor-10-sulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, decanoic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, ethylenediaminetetraacetic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, D-glucoheptonic acid, D-gluconic acid, D-glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, hexanoic acid, hippuric acid, hydrobromic acid, hydrochloric acid, isobutyric acid, DL-lactic acid, lactobionic acid, lauric acid, lysine, maleic acid, (−)-L-malic acid, malonic acid, DL-mandelic acid, methanesulfonic acid, galactaric acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, octanoic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid (embonic acid), phosphoric acid, propionic acid, (−)-L-pyroglutamic acid, salicylic acid, 4-amino-salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid. Further pharmaceutically acceptable salts can be formed with cations from metals like aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like (also see Pharmaceutical salts, Berge, S. M. et al., J. Pharm. Sci., (1977), 66, 1-19).
  • The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile, or a mixture thereof.
  • Salts of other acids than those mentioned above which for example are useful for purifying or isolating the compounds of the present invention (e.g. trifluoro acetate salts) also comprise a part of the invention.
  • The term “substituted” as used herein means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's viable valence number is not exceeded, and that the substitution results in a stable compound.
  • The term “partially unsaturated” as used herein means that in the designated group or moiety 1, 2, or more, preferably 1 or 2, double bonds are present. Preferably, as used herein, the term “partially unsaturated” does not cover fully unsaturated groups or moieties.
  • Terms like “ . . . optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O— . . . ” or “ . . . optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-” in a section like “ . . . groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-” means that the referenced group may be substituted with 1 to 5 substituents, wherein these substituents can be halogen, HO—, C1-3-alkyl-O— which may be optionally fluorinated with 1 to 7 fluoro atoms, and C1-3-alkyl-O— which may be optionally fluorinated with 1 to 7 fluoro atoms.
  • The term “halogen” generally denotes fluorine, chlorine, bromine and iodine.
  • The term “C1-n-alkyl”, wherein n is an integer from 2 to n, either alone or in combination with another radical denotes an acyclic, saturated, branched or linear hydrocarbon radical with 1 to n C atoms. For example the term C1-5-alkyl embraces the radicals H3C—, H3C—CH2—, H3C—CH2—CH2—, H3C—CH(CH3)—, H3C—CH2—CH2—CH2—, H3C—CH2—CH(CH3)—, H3C—CH(CH3)—CH2—, H3C—C(CH3)2—, H3C—CH2—CH2—CH2—CH2—, H3C—CH2—CH2—CH(CH3)—, H3C—CH2—CH(CH3)—CH2—, H3C—CH(CH3)—CH2—CH2—, H3C—CH2—C(CH3)2—, H3C—C(CH3)2—CH2—, H3C—CH(CH3)—CH(CH3)— and H3C—CH2—CH(CH2CH3)—.
  • The terms “carbocyclyl” and “carbocycle” as used either alone or in combination with another radical, mean, if not mentioned otherwise, a mono- bi- or tricyclic ring structure consisting of 3 to 14 carbon atoms. The terms, if not mentioned otherwise, refers to fully saturated, partially saturated and aromatic ring systems. The terms encompass fused, bridged and spirocyclic systems.
  • Thus, the terms include the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • Figure US20140045856A1-20140213-C00163
  • The term “C3-n-cycloalkyl”, wherein n is an integer from 4 to n, either alone or in combination with another radical denotes a cyclic, saturated, unbranched hydrocarbon radical with 3 to n C atoms. For example the term C3-7-cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • The terms “heterocyclyl” and “heterocycle” as used either alone or in combination with another radical, mean a saturated or unsaturated mono- or polycyclic ring system containing one or more heteroatoms selected from N, O or S(O)r, wherein r=0, 1 or 2, which may contain aromatic rings consisting of, if not mentioned otherwise, 3 to 14 ring atoms wherein none of the heteroatoms is part of an aromatic ring. The terms encompass fused, bridged and spirocyclic systems. The terms are intended to include all the possible isomeric forms.
  • Thus, the terms include the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • Figure US20140045856A1-20140213-C00164
    Figure US20140045856A1-20140213-C00165
    Figure US20140045856A1-20140213-C00166
    Figure US20140045856A1-20140213-C00167
    Figure US20140045856A1-20140213-C00168
  • The term “aryl” as used herein, either alone or in combination with another radical, denotes a carbocyclic aromatic monocyclic group containing 6 carbon atoms which may be further fused to a second 5- or 6-membered carbocyclic group which may be aromatic, saturated or unsaturated. Aryl includes, but is not limited to, phenyl, indanyl, indenyl, naphthyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl and dihydronaphthyl.
  • The term “heteroaryl” means a mono- or polycyclic-ring systems containing one or more heteroatoms selected from N, O or S(O)r, wherein r=0, 1 or 2, consisting of 5 to 14 ring atoms wherein at least one of the heteroatoms is part of an aromatic ring. The term “heteroaryl” is intended to include all the possible isomeric forms.
  • Thus, the term “heteroaryl” includes the following exemplary structures which are not depicted as radicals as each form may be attached through a covalent bond to any atom so long as appropriate valences are maintained:
  • Figure US20140045856A1-20140213-C00169
    Figure US20140045856A1-20140213-C00170
  • Many of the terms given above may be used repeatedly in the definition of a formula or group and in each case have one of the meanings given above, independently of one another.
  • The compounds according to the invention may be obtained using methods of synthesis known in principle. Preferably, the compounds are obtained by the following methods according to the invention which are described in more detail hereinafter.
  • Preparation
  • The following schemes illustrate generally how to manufacture the compounds of the present invention by way of example. The abbreviated substituents may be as defined above if not defined otherwise within the context of the schemes:
  • Figure US20140045856A1-20140213-C00171
  • Scheme 1:
  • In a first step, substituted 1,2-pyrido-diamines (A), commercially available or prepared following well known reported procedures, were reacted with the appropriate 1,2-keto ester or acid in EtOH or MeOH as solvents at room temperature, to form the corresponding pyrido[2,3-b]pyrazinone intermediates (B,C). Where required, regioisomers were separated by flash chromatography and treated with POCl3 under heating to obtain the 2-chloro-pyridopyrazines (D).
  • Nucleophilic substitution was performed using hydrazine hydrate in appropriate solvents (e.g. EtOH) at room temperature to form the corresponding pyrido[2,3-b]-pyrazin-3yl-idrazines derivatives (F). Reaction with appropriate acyl chlorides or carboxylic acids in the presence of a coupling agent (e.g. HATU or TBTU) and a base (e.g. TEA or DIPEA), gave the corresponding hydrazides (E) which were submitted to in situ cyclization to triazoles core by heating in appropriate solvent (e.g. cyclohexanol).
  • Alternatively, in step 5, preformed hydrazides derivative were reacted with 2-chloro-pyridopyrazines (D) in appropriate solvents (such as DMF) to gave intermediates hydrazides (E) which were then converted to compounds G as described before or heating directly the 2-chloro-pyridopyrazines (D) with the appropriate hydrazides in appropriate solvent such as cyclohexanol at high temperature.
  • Further examples of the present invention were prepared following Scheme 2:
  • Figure US20140045856A1-20140213-C00172
  • Further examples of the present invention were prepared by alkylation of the appropriate phenol derivative (obtained as described in Scheme 1) with alkylating agents (e.g. chloride, bromide or tosylates derivatives) in the presence of a suitable base (e.g. tBuOK or Cs2CO3) in a solvent like DMF under heating or by ring opening of the appropriate epoxides in the presence of a suitable base (e.g. Cs2CO3) in a solvent like DMF or DMA under conventional heating or microwave irradiation.
  • Rx and Ry: Examples 13, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 32a, 33, 34, 35, 35a, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 106, 107, 108a, 108b, 108c, 108d, 109a, 109b, 109c, 109d, 110b, 110d, 111 were prepared following the above described procedures.
  • Figure US20140045856A1-20140213-C00173
  • Further examples of the present invention were prepared by aromatic nucleophilic substitution, reacting the appropriate aromatic fluoro derivatives (prepared as described in Scheme 1) with primary or secondary alcohols in the presence of a suitable base (e.g tBuOK or Cs2CO3), in a solvent like DMF under heating. Examples 58a, 58b, 58c were prepared following the above described procedure.
  • Figure US20140045856A1-20140213-C00174
  • Further examples of the present invention were prepared by well-known cross coupling reaction (i.e Sonogashira, see for example Organic Letters, 2007 vol. 9, p. 4057-4060) starting from appropriate aromatic halogen intermediates (X=Br,I). The resulting unsaturated triple bond intermediates were then converted to final compounds by appropriate reduction with Wilkinson's catalyst RhCl(TPP)3 under hydrogen atmosphere in toluene as solvent.
  • Examples 66, 67, 68, 69 were prepared following the above described procedure. Alternatively, the same synthetic approach was applied for the preparation of preformed hydrazydes which were then reacted with intermediate D as described in Scheme 1
  • Figure US20140045856A1-20140213-C00175
  • Examples 64 and 65 were prepared following the above described procedure.
  • Figure US20140045856A1-20140213-C00176
  • Further examples of the present invention were prepared as described in Scheme 6: in a first step the appropriate aromatic alogen derivatives (X=I) was treated with isopropyl magnesium bromide in a suitable solvent (like THF) at low temperature followed by addition of the appropriate ketone or aldehyde derivatives.
  • Examples 70, 71, 72, 73, 74 were prepared following the above described procedure.
  • Alternatively, the same synthetic approach was applied for the preparation of preformed hydrazydes which were then reacted with intermediate D as described in Scheme 1.
  • Figure US20140045856A1-20140213-C00177
  • Examples 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 96a, 96b, 96c, 97 were prepared following the above described procedure.
  • Figure US20140045856A1-20140213-C00178
  • Further examples of the present invention were prepared as described in Scheme 8: in a first step the appropriate aromatic aldehyde is obtained by reaction of alogen derivatives (X=I) with n-BuLi in a suitable solvent (like THF) at low temperature followed by addition of dry DMF in THF. In a second step the suitable commercially available Grignard reagent is added at low temperature.
  • Rz: Examples 59, 61, 62, 63 were prepared following the above reported procedure.
  • Figure US20140045856A1-20140213-C00179
  • Further examples of the present invention were prepared as described in Scheme 9:
  • in a first step the appropriate ketone is obtained by reaction of alogen derivatives (X=1,Br) with tributyl-(1-ethoxyvinyl-)tin in the presence of Pd(PPh3)4. The epoxide obtained by reaction with appropriate trimethyl sulphonium ylide was then subjected to ring opening with hydride and subsequent oxidation with suitable agents such as manganese dioxide in appropriate solvents (DCM).
  • Examples 76, 77, 78, 79 were prepared following the above reported procedure. In analogy, the omologated derivatives Examples 80, 81 were obtained performing the first step as described in Scheme 9a.
  • Figure US20140045856A1-20140213-C00180
  • Figure US20140045856A1-20140213-C00181
  • Further examples of the present invention were prepared as described in Scheme 10: in a first step the appropriate double bond is installed by Suzuki coupling with suitable boronic acids or esters (such as for example 2-isopropenyl-dioxaborolane, Scheme 10). In a second step, the hydratation of the double bond (HCl/dioxane) allowed to introduce hydroxylic function.
  • Example 103, 103b, 103c were prepared following the above described procedure.
  • The compounds according to the invention are advantageously also obtainable using the methods described in the examples that follow, which may also be combined for this purpose with methods known to the skilled man from the literature.
  • As already mentioned, the compounds of general formula I according to the invention and the physiologically acceptable salts thereof have valuable pharmacological properties, particularly as inhibitors of phosphodiesterase 2 and/or 10.
  • BIOLOGICAL EXAMPLES In-Vitro Effect
  • The in-vitro effect of the active compounds of the invention can be shown with the following biological assays.
  • a) Radioactive PDE2 Assay Protocol:
  • For all experiments the Phosphodiesterase [3H]cAMP SPA Enzyme Assay (TRKQ7090, GE Healthcare Europe GmbH) were used. The enzymatic activity of the PDE2 and the inhibitory potency of compounds was measured by the conversion of [3H]cAMP to [3H]AMP. [3H]AMP associate to the scintillator soaked yttrium-silicate beads resulting in an increase in scintillation events. Compounds that inhibit the respective enzymes decrease the generation of [3H]AMP and accordingly the number of counts detected.
  • SF9 lysate containing PDE2A is incubated at room temperature for 1 h with [3H]cAMP and the reaction is terminated by addition of SPA beads in 18 mM zinc sulphate. The [3H]AMP bound to SPA beads is determined after at least 3 hours of sedimentation of the beads, the signal is recorded using the TopCount with a recording time of 3 min/well.
  • PDE 2A protein is expressed upon baculovirus infection in SF9 cells. The cells have been incubated upon infection for ˜3 days and protein production was confirmed by Western Blot. The cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min).
  • The assay conditions were as follows:
  • total assay volume: 40 μl
    protein amount: 5 ng
    protein concentration: 500 pg/μl
    substrate concentration: 20 nM; ~1.08 mCi/l
    incubation time: 60 min
  • The buffer used for above described assay buffer was:
  • 50 mM Tris/HCl, pH 7.4
    8.3 mM MgCl2
    1.7 mM EGTA
    0.1% (w/v) BSA
    0.05% (v/v) Tween 20
  • In detail the protocol is as follows:
  • Into a 384 well plate (OptiPlate, white), the following components are pipetted (either manually or using automated pipetting devices):
  • 20 μl test-compound solution (test compound diluted in assay buffer at twofold the desired concentration)
  • +10 μl PDE 2A preparation (enzyme diluted in assay buffer to a concentration of 0.5 ng/μl protein)
  • +10 μl [3H]cAMP (diluted in assay buffer to a concentration of 80 nM cAMP)
  • Controls included into each experiment:
  • positive control: no inhibitor, DMSO at the concentration as used in the compound dilutions
  • negative control: no PDE 2 (SF9 lysate instead of SF9/PDE 2A lysate)
  • Calculation of % Inhibition:
  • The activity of the positive control (minus the negative control=background) is set to 100% and activity in the presence of compound is expressed relative to these 100%.
  • Calculation of IC50:
  • IC50 are calculated with GraphPadPrism or other suited software setting the positive control as 100 and the negative control as 0. For calculation of IC50 usually 11 dilutions of the test compound are selected.
  • b) Radioactive PDE10 Assay Protocol:
  • For assessing activity of compounds on PDE10 inhibition, the following modifications of the protocol described above are applied:
  • protein amount: 3 ng
  • protein concentration: 75 pg/μl
  • In detail the protocol is as follows:
  • Into a 384 well plate (OptiPlate, white), the following components are pipetted (either manually or using automated pipetting devices):
  • 20 μl test-compound solution (test compound diluted in assay buffer at twofold the desired concentration)
  • +10 μl PDE 10A preparation (enzyme diluted in assay buffer to a concentration of 0.3 ng/μl protein)
  • +10 μl cAMP tracer (diluted in assay buffer to a concentration of 80 nM cAMP, specific activity ˜4 mCi/l)
  • After addition of all 3 components the plates are shortly spun and incubated at room temperature for 60 min. The reaction is stopped by the addition of 20 μl Yttrium SPA PDE Beads containing 18 mM zinc sulphate in water
  • After at least 3 hours sedimentation of the beads, the signal is recorded using the TopCount with a recording time of 3 min/well.
  • PDE 10A protein is expressed upon baculovirus infection in SF9 cells. The cells have been incubated upon infection for ˜3 days and protein production was confirmed by Western Blot. The cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min).
  • In the following table the results of the above described experiments are listed.
  • TABLE 3
    Activity of the examples (Ex) compiled in the experimental part,
    based on above described assays (cAMPA SPA radioactive).
    PDE2 PDE10
    Ex. IC50 [μM] IC50 [μM]
    1 0.016 0.421
    2 0.004 0.040
    3 0.155 0.411
    4 0.138 0.441
    13 0.053 2.190
    14 0.202 3.580
    15 0.040 >10
    16 0.058 1.876
    17 0.098 5.72
    18 0.071 3.01
    19 0.146 3.01
    20 0.028 2.76
    21 0.070 2.82
    22 0.115 >5
    23 0.071 5.10
    24 0.138 6.69
    25 0.055 3.26
    26 0.116 1.38
    27 0.004 0.97
    28 0.009 >10
    29 0.017 2.38
    30 0.027 3.92
    31 0.007 1.90
    32 0.003 1.76
    33 0.004 2.50
    34 0.009 1.89
    35 0.009 1.30
    36 0.219 0.85
    37 0.171 3.08
    38 0.417 1.96
    39 0.914 2.10
    40 0.080 3.47
  • Another, non radioactive, in-vitro assay was performed as follows
  • c) Phosphodiesterase (PDE) 2A and 10 Assay with Fluorescent Substrate
  • Assay Principle:
  • The PDE reaction cleaves cAMP to AMP. The IMAP system (Molecular Device) using fluorescence polarization (FP) as detection principle was used to measure enzyme activity. A fluorescent labeled cAMP was used as substrate for the reaction, generating a labeled AMP. The fluorescent AMP binds specifically to the large M(III)-based nano-particles which reduces the rotational speed of the substrate and thus increases its polarization.
  • Detailed Method:
  • The inhibition of PDE 2A or 10 enzyme activity was assessed using IMAP-Phosphodiesterase-cAMP fluorescence labeled substrate (Molecular Devices, Order No. R7506), IMAP TR-FRET screening express (Molecular Devices, Order No. R8160, the TR-FRET component will not be used) and PDE 2A or PDE10 protein expressed upon baculovirus infection in SF9 cells. The cells were incubated after infection for ˜3 days and protein production was confirmed by Western Blot. The cells were collected by centrifugation and the pellet frozen in liquid nitrogen before it was resuspended in PBS containing 1% Triton X-100 and protease inhibitors. After 45 min incubation on ice, the cell debris was removed by centrifugation (13.000 rpm, 30 min). Since SF 9 cells do not express cAMP hydrolyzing enzymes to a high extent, no further purification of the protein was needed.
  • All reactions were performed in 384 well plates, Perkin Elmer black optiplates and IMAP reaction buffer with 0.1% Tween20 (kit component)
  • Compounds were serial diluted in DMSO. With an intermediate dilution step with reaction buffer DMSO concentration was reduced to achieve 1% DMSO in the assay reaction. Setup of the assay started with 10 μl enzyme (˜10 ng/well, depending on prep. batch), 5 μl compound, reaction was started by addition of 5 μl labeled cAMP (30 nM, final concentration), immediately mixed for 15 seconds on a Eppendorf mixmate (2000 rpm) followed by an incubation at room temperature for 90 minutes in the dark. Reaction is stopped by adding of 60 μl binding buffer for FP/cAMP (kit component). After at least 90 min of further incubation (room temperature, dark) the assay was measured at 485 nm excitation/525 nm emission in an Envision multilabel reader (PerkinElmer).
  • Each assay plate contained wells with vehicle controls (1% DMSO) for the measurement of non-inhibited reaction (=100% control) and wells without enzyme as 0% controls.
  • The analysis of the data was performed by calculation of the percentage of inhibition in the presence of test compound compared to the vehicle control samples (100% control, no inhibition) and a low control (0% control, no enzyme).
  • IC50 values are calculated with Assay Explorer or other suited software based on curve fitting of results of at least 8 different compound concentrations. The compound concentrations may vary according to the needed range, but typically cover the range between 10 μM and 0.1 pM.
  • TABLE 3a
    Activity of the examples (Ex) compiled in the experimental
    part, based on above described assays (IMAP fluorescent).
    PDE2 PDE10
    Ex. IC50 [μM] IC50 [μM]
     1 0.041 0.355
     2 0.003 0.033
     4 0.120 0.256
     5 0.003 0.005
     6 0.003 0.010
     7 0.003 0.016
     8 0.002 0.008
     9 0.001 0.015
    10 0.229 0.254
    11 0.059 0.123
    12 0.009 0.044
     12a 0.036 0.170
    13 0.095 6.363
    15 0.112 1.118
    16 0.098 8.700
    17 0.162 10.200
    18 0.090 8.418
    20 0.077 8.162
    21 0.022 7.047
    22 0.207 9.317
    23 0.216 9.193
    25 0.038 7.333
    27 0.018 0.958
    28 0.020 0.299
    29 0.037 5.097
    30 0.020 2.573
    31 0.008 2.897
    32 0.026 6.703
     32a 0.006 7.390
    33 0.005 3.417
    34 0.017 2.143
    35 0.017 1.477
     35a 0.003 0.822
    40 0.077 8.707
    41 0.050 2.770
    42 0.035 2.520
    43 0.057 3.015
     43a 0.109 4.510
    44 0.156 6.037
    45 0.005 1.488
    46 0.050 1.035
    47 0.008 0.852
    48 0.008 1.520
    49 0.033 3.760
    50 0.194 7.120
    51 0.009 2.150
    52 0.011 1.195
    53 0.140 2.830
    54 0.058 0.798
    55 0.080 5.298
     55a 0.033 2.435
    56 0.002 0.061
     56a 0.008 0.061
     56b 0.001 0.066
    57 0.006 0.045
     57a 0.012 0.167
     57b 0.003 0.091
    58 0.143 0.663
     58a 0.054 0.713
     58b 0.045 0.663
     58c 0.037 0.241
    59 0.008 0.936
    60 0.019 0.285
    61 0.004 0.534
    62 0.011 1.300
    63 0.060 1.710
    64 0.045 3.870
    65 0.214 3.545
    66 0.003 0.674
    67 0.175 3.905
    68 0.002 1.007
    69 0.001 0.891
    70 0.163 8.620
    71 0.109 2.080
    72 0.032 2.865
    73 0.189 10.000
    74 0.131 2.340
    75 0.012 2.403
    76 0.021 1.105
    77 0.004 1.400
    78 0.010 2.845
    79 0.144 0.305
    80 0.010 5.405
    81 0.007 5.384
    82 0.199 5.615
    83 0.004 8.815
    84 0.006 0.565
    85 0.006 4.325
    86 0.434 5.750
    87 0.074 2.594
    88 0.001 0.302
    89 0.018 0.187
    90 0.103 0.485
    91 0.001 0.077
    92 0.002 0.553
    93 0.086 6.107
    94 0.001 2.719
    95 0.005 1.369
    96 0.043 5.945
     96a 0.199 10.000
     96b 0.087 10.000
     96c 0.591 7.460
    97 0.004 2.142
     97a 0.004 5.945
     97b 0.004 0.891
    98 0.139 1.560
    99 0.125 1.510
    100  0.017 3.856
    101  0.047 1.125
    102  0.103 2.265
    103  0.003 1.007
    103b 0.100 10.000
    103c 0.622 7980
    104  0.036 7.715
    104a 0.040 3.229
    104b 0.030 9.765
    105  0.023 1.540
    105a 0.104 3.630
    105b 0.038 3.410
    106  0.160 1.940
    106a 0.429 1.410
    106b 0.050 2.500
    107  0.007 0.022
    111  0.022 3.980
    108a 0.011 0.548
    108b 0.059 0.594
    108c 0.024 0.262
    108d 0.046 0.630
    109a 0.014 1.670
    109b 0.009 1.090
    109c 0.009 1.360
    109d 0.004 1.830
    110b 0.388 >10
    110d 0.467 7.600
  • In-Vivo Effect: Animal Experiments and Sample Analysis (CSF):
  • Test compounds were administered to animals (rat) different routes at doses of 10.0 or 5 μmol/kg, (both oral and intravenous). After compound administration, the animals were sacrificed in a CO2 chamber and CSF samples were carefully collected by puncture of the cisterna magna. Immediately after CSF sampling, blood was taken by heart puncture and brains were dissected out. Blood was collected in EDTA-coated microvettes and plasma was prepared by centrifugation. Concentration of the test compounds in plasma, CSF or brain homogenate was determined using HPLC-MS-MS.
  • TABLE 4
    Plasma, brain and CSF concentration
    conc conc conc
    Time(*) plasma brain c(brain)/ CSF c(CSF)/
    Ex. (h) (nmol/L) (nmol/L) c(plasma) (nmol/L) c(plasma)
    44 0.5 2187 670 0.31 228 0.10
    34 0.5 223 146 0.7 19 0.09
    18 0.5 1225 351 0.3 163 0.13
    20 0.5 338 272 0.82 37 0.11
    45 2 801 183 0.20 25 0.03
    64 2 783 254 0.5 37 0.06
    76 2 463 218 0.5 18 0.04
    75 2 1130 383 0.34 31 0.03
    70 2 2377 695 0.29 276 0.12
    47 2 622 441 0.7 22 0.04
    49 2 239 47 0.2 13 0.06
    (*)Time between administration and CSF sampling
  • For the skilled in the art it is evident from the experimental results shown above that the compounds of the present invention are not only very potent phosphodiesterase 2 and/or 10 inhibitors but also reach high CSF concentrations and adequate CSF to plasma ratios.
  • Metabolic Stability
  • The metabolic stability of the compounds according to the invention has been investigated as follows:
  • The metabolic degradation of the test compound was assayed at 37° C. with pooled liver microsomes from various species. The final incubation volume of 100 μl per time point contains TRIS buffer pH 7.6 at room temperature (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/mL for human and dog, 0.5 mg/mL for other species) and the test compound at a final concentration of 1 μM. Following a short preincubation period at 37° C., the reactions were initiated by addition of betanicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1 mM), and terminated by transferring an aliquot into solvent after different time points. After centrifugation (10000 g, 5 min), an aliquot of the supernatant was assayed by LC10 MS/MS for the amount of parent compound. The half-life was determined by the slope of the semi-logarithmic plot of the concentration-time profile.
  • TABLE 4
    Stability of compounds of the present
    invention in human liver microsomes.
    Ex. Half-life - t½ [min]
     1 130
     5 43
     6 66
     9 41
    12 63
    13 130
    17 130
    18 130
    20 130
    21 130
    30 130
    32 130
     32a 36
    34 130
    35 130
     35a 130
    42 130
    43 130
    44 130
    45 130
    46 130
    47 130
    48 130
    49 130
    50 130
    51 130
    52 130
    54 130
    55 130
     55a 130
    56 120
     56a 110
     56b 62
    57 130
     57a 130
     57b 93
    58 130
     58a 130
     58b 130
     58c 130
    59 49
    61 65
    62 130
    63 130
    64 130
    66 57
    67 130
    68 39
    69 53
    70 130
    71 130
    72 130
    73 130
    75 130
    76 130
    77 90
    78 79
    80 83
    81 26
    82 130
    83 100
    84 39
    85 130
    87 130
    88 53
    89 130
    91 28
    92 23
    93 130
    94 38
    96 110
    97 51
    99 130
    100  12
    101  22
    102  130
    104  130
    104a 130
    104b 130

    hERG (Human Ether-à-Go-Go-Related Gene)-Channel Assay
  • hERG channel inhibition of compounds of the present invention has been investigated as follows:
  • HEK (human embryonic kidney) 293 cells were stably transfected with hERG cDNA. Cells were superfused with a bath solution containing (mM): NaCl (137), KCl (4.0), MgCl2 (1.0), CaCl2 (1.8), Glucose (10), HEPES (10), pH 7.4 with NaOH. Patch pipettes were made from borosilicate glass tubing using a horizontal puller and filled with pipette solution containing (mM): K-aspartate (130), MgCl2 (5.0), EGTA (5.0), K2ATP (4.0), HEPES (10.0), pH 7.2 with KOH. Resistance of the microelectrodes was in the range between 2 and 5 MΩ.
  • Stimulation and Recording:
  • Membrane currents were recorded using an EPC-10 patch clamp amplifier and PatchMaster software. hERG-mediated membrane currents were recorded at 35° C., using the whole-cell configuration of the patch-clamp technique. Transfected HEK293 cells were clamped at a holding potential of −60 mV and hERG-mediated inactivating tail currents were elicited using a pulse pattern with fixed amplitudes (activation/inactivation: 40 mV for 2000 ms; recovery: 120 mV for 2 ms; ramp to 40 mV in 2 ms; inactivating tail current: 40 mV for 50 ms) repeated at 15 s intervals. During each inter-pulse interval 4 pulses scaled down by a factor of 0.2 were recorded for a P/n leak subtraction procedure. R5 compensation was employed up to a level that safely allowed recording devoid of ringing.
  • Compound Preparation and Application:
  • The different concentrations of the test compound were applied sequentially on each of the different cells investigated. A steady state level of baseline current was measured for at least 6 sweeps prior to the application of the first test compound concentration.
  • The test compound was dissolved in DMSO to yield a master stock solution which was diluted further in DMSO to stock solutions needed for the lower concentrations. Final dilutions in extracellular buffer were prepared freshly from these stocks by a 1:1000 dilution step each before starting the experiments.
  • Data analysis and IC50 determination:
  • Peak current amplitudes were measured 3 ms after the ramp to +40 mV. For baseline and each concentration the peak currents of the three last sweeps before application of the next concentration were averaged. Residual currents ( 1/10) were calculated for each cell as the fraction of actual average peak current and average baseline peak current.
  • The logistic concentration-response curve of the following form was fitted to the residual current data using a genetic algorithm:

  • I/I0=1−1/(1+(C/IC50)**p)
  • C: actual concentration of compound (in μM)
    IC50: half-inhibitory concentration (in μM)
    p: Hill slope
  • TABLE 4
    hERG channel inhibition of compounds of the present invention.
    Ex. hERG IC50 [μM]
    13 >10
    18 >10
    20 >10
    21 >10
    27 >10
    28 >10
    32 >10
     32a >10
    34 >10
     35a >10
    45 >10
    47 >10
    48 >10
    49 >10
    55 >10
    64 >10
    66 >10
    68 8.3
    75 >10
    76 >10
    80 >10
    82 >10
    83 >10
    84 6.3
    85 >10
    87 >10
    88 >10
    93 >10
  • In view of their ability to inhibit the activity of phosphodiesterase 2 and/or 10 activity, their CSF values and their metabolic stability and their low inhibition of the hERG channel, the compounds of general formula I according to the invention, including the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the hydrates thereof, the solvates and the physiologically acceptable salts thereof, are suitable for the treatment and/or preventative treatment of all those diseases or conditions which can be influenced by inhibition of PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction. Therefore, compounds according to the invention, including the physiologically acceptable salts thereof, are particularly suitable for the prevention or treatment of diseases, particularly (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood [affective] disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) and Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders.
  • In addition, the compounds according to the invention can be used for the treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory.
  • In addition, the compounds according to the invention can be used for the treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments, age-associated memory losses, vascular dementia, craniocerebral trauma, stroke, dementia occurring after strokes (post stroke dementia), post-traumatic dementia, general concentration impairments, concentration impairments in children with learning and memory problems, Alzheimer's disease, Lewy body dementia, dementia with degeneration of the frontal lobes, including Pick's syndrome, Parkinson's disease, progressive nuclear palsy, dementia with corticobasal degeneration, amyotropic lateral sclerosis (ALS), Huntington's disease, multiple sclerosis, thalamic degeneration, Creutzfeld-Jacob dementia, HIV dementia, schizophrenia with dementia or Korsakoff's psychosis. In addition, the compounds of the present invention can be used for the treatment of Alzheimer's disease.
  • In addition compounds according toe the invention can be used for the treatment of pain disorders including but not limited to inflammatory, neuropatic and osteoarthritic pain.
  • In addition, the compounds according to the invention can be used for the treatment of sleep disorders, bipolar disorder, metabolic syndrome, obesity, diabetis mellitus, hyperglycemia, dyslipidemia, impaired glucose tolerance, or a disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen.
  • Preferably the compounds according to the invention are suitable for the treatment of Alzheimer's Disease and for the treatment schizophrenia.
  • More preferably the compounds according to the invention are suitable for symptomatic treatment of Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia.
  • In particular the compounds according to the invention are suitable for symptomatic treatment of prodromal and mild-to-moderate Alzheimer's Disease and for the treatment of cognitive impairment associated with schizophrenia.
  • In a further aspect of the present invention the present invention relates to methods for the treatment or prevention of above mentioned diseases and conditions, which method comprises the administration of an effective amount of a compound of general formula I, the tautomers thereof, the stereoisomers thereof, the mixtures thereof, the hydrates thereof, the solvates and the physiologically acceptable salts thereof, to a human being.
  • The dose range of the compounds of general formula I applicable per day is usually from 0.1 to 1000 mg, preferably from 1 to 500 mg by oral route, in each case administered 1 to 4 times a day.
  • Each dosage unit may conveniently contain from 0.1 to 500 mg, preferably 1 to 100 mg.
  • The actual pharmaceutically effective amount or therapeutic dosage will of course depend on factors known by those skilled in the art such as age and weight of the patient, route of administration and severity of disease. In any case the combination will be administered at dosages and in a manner which allows a pharmaceutically effective amount to be delivered based upon patient's unique condition.
  • Suitable preparations for administering the compounds of formula I, including the physiologically acceptable salts thereof, will be apparent to those with ordinary skill in the art and include for example tablets, pills, capsules, suppositories, lozenges, troches, solutions, syrups, elixirs, sachets, injectables, inhalatives, powders, etc. The content of the pharmaceutically active compound(s) should be in the range from 0.1 to 95 wt.-%, preferably 5.0 to 90 wt.-% of the composition as a whole.
  • Suitable tablets may be obtained, for example, by mixing one or more compounds according to formula I with known excipients, for example inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or lubricants. The tablets may also consist of several layers.
  • For this purpose, the compounds of formula I prepared according to the invention may be formulated, optionally together with other active substances, together with one or more inert conventional carriers and/or diluents, e.g. with corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, citric acid, tartaric acid, water, polyvinylpyrrolidone, water/ethanol, water/glycerol, water/sorbitol, water/polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof.
  • The compounds according to the invention may also be used in conjunction with other active substances, particularly for the treatment and/or prevention of the diseases and conditions mentioned above. Other active substances which are suitable for such combinations include, for example, BACE inhibitors; amyloid aggregation inhibitors (e.g. ELND-005); directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants (e.g. vitamin E or ginkolide); anti-inflammatory substances (e.g. Cox inhibitors, NSAIDs additionally or exclusively having Abeta lowering properties); HMG-CoA reductase inhibitors (statins); acetylcholinesterase inhibitors (e.g., donepezil, rivastigmine, tacrine, galantamine); NMDA receptor antagonists (e.g. memantine); AMPA receptor agonists; AMPA receptor positive modulators, AMPAkines, monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmitters; substances inducing the secretion of growth hormone (e.g., ibutamoren mesylate and capromorelin); CB-1 receptor antagonists or inverse agonists; antibiotics (e.g., minocyclin or rifampicin); PDE2, PDE4, PDE5, PDE9, PDE10 inhibitors, GABAA receptor inverse agonists, GABAA receptor antagonists, nicotinic receptor agonists or partial agonists or positive modulators, alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators, alpha7 nicotinic receptor agonists or partial agonists or positive modulators; histamine H3 antagonists, 5 HT-4 agonists or partial agonists, 5HT-6 antagonists, alpha2-adrenoreceptor antagonists, calcium antagonists, muscarinic receptor M1 agonists or partial agonists or positive modulators, muscarinic receptor M2 antagonists, muscarinic receptor M4 antagonists, metabotropic glutamate-receptor 5 positive modulators, glycine transporter 1 inhibitors, antidepressants, such as citalopram, fluoxetine, paroxetine, sertraline and trazodone; anxiolytics, such as lorazepam and oxazepam; antiphychotics, such as aripiprazole, clozapine, haloperidol, olanzapine, quetiapine, risperidone and ziprasidone, and other substances that modulate receptors or enzymes in a manner such that the efficacy and/or safety of the compounds according to the invention is increased and/or unwanted side effects are reduced. The compounds according to the invention may also be used in combination with immunotherapies (e.g., active immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or nanobodies) for the treatment of the above-mentioned diseases and conditions.
  • The dosage for the combination partners mentioned above is usefully 1/5 of the lowest dose normally recommended up to 1/1 of the normally recommended dose.
  • Therefore, in another aspect, this invention relates to the use of a compound according to the invention or a physiologically acceptable salt thereof combined with at least one of the active substances described above as a combination partner, for preparing a pharmaceutical composition which is suitable for the treatment or prevention of diseases or conditions which can be affected by inhibitors of phosphodiesterase 2 and/or 10. These are preferably pathologies related to PDE2 and/or 10 hyperactivity and/or cAMP and/or cGMP hypofunction, particularly one of the diseases or conditions listed above, most particularly prodromal and mild-to-moderate Alzheimer's Disease and cognitive impairment associated with schizophrenia.
  • The use of the compound according to the invention, or a physiologically acceptable salt thereof, in combination with another active substance may take place simultaneously or at staggered times, but particularly within a short space of time. If they are administered simultaneously, the two active substances are given to the patient together; while if they are used at staggered times the two active substances are given to the patient within a period of less than or equal to 12 hours, but particularly less than or equal to 6 hours.
  • Consequently, in another aspect, this invention relates to a pharmaceutical composition which comprises a compound according to the invention or a physiologically acceptable salt of such a compound and at least one of the active substances described above as combination partners, optionally together with one or more inert carriers and/or diluents.
  • The compound according to the invention, or a physiologically acceptable salt thereof, and the additional active substance to be combined therewith may both be present together in one formulation, for example a tablet or capsule, or separately in two identical or different formulations, for example as a so-called kit-of-parts.
  • EXAMPLES
  • The following examples are intended to illustrate the invention, without restricting its scope.
  • CHEMICAL MANUFACTURE ABBREVIATIONS
    • ACN acetonitrile
    • APCI Atmospheric pressure chemical ionization
    • d day
    • Cy cyclohexane
    • DCM dichloromethane
    • DIPEA diisopropylethylamine
    • DMF dimethylformamide
    • DMA dimethylacetamide
    • ESI electrospray ionization (in MS)
    • EtOAc ethylacetate
    • EtOH ethanol
    • Ex. 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 minute(s)
    • MS mass spectrometry
    • NMP 1-methyl-2-pyrrolidinone
    • rt room temperature
    • Rt retention time (in HPLC)
    • TBTU O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate
    • TEA triethylamine
    • TFA trifluoroacetic acid
    • THF tetrahydrofuran
    • TLC thin-layer chromatography
    • UPLC-MS ultra performance liquid chromatography—mass spectrometry
  • Methods:
  • UPLC-MS Methods:
  • Method 1
  • Instrument: LC/MS Waters Acquity UPLC System DAD, SQD single quadrupole; column: HSS C18 1.8 μm 2.1×50 mm, Temp 35° C.; mobile phase: A=H2O 90%+10% CH3CN+CF3COOH 0.1%, B=CH3CN 90%+H2O 10%; gradient: 0.0 min 0% B→1.20 min 100% B→1.45 min 100% B→1.55 min 0% B→1.75 min 0% B; flow rate: 0.70 mL/min; detection: UV 254 nm; detection: SQD, single quadrupole; ion source: ES+/ES−; scan range: 90-900 amu.
  • Method 2
  • Instrument: LC/MS Waters Acquity UPLC System DAD, SQD single quadrupole; column: BEH C18 1.7 μm 2.1×50 mm, Temp 35° C.; mobile phase: A=H2O 90%+10% CH3CN+NH4COOH 5 mmol, B=CH3CN 90%+H2O 10%; gradient: 0.0 min 0% B→1.20 min 100% B→1.45 min 100% B→1.55 min 0% B→1.75 min 0% B; flow rate: 0.70 mL/min; detection: UV 254 nm; detection: SQD, single quadrupole; ion source: ES+/ES−; scan range: 90-900 amu.
  • Method 3
  • Instrument: LC/MS Waters Acquity UPLC System DAD, ELSD detector, SQD single quadrupole; column: HSS C18 1.8 μm 2.1×50 mm, Temp 35° C.; mobile phase: A=H2O 90%+10% CH3CN+CF3COOH 0.1%, B=CH3CN 90%+H2O 10%; gradient: 0.0 min 0% B→2.40 min 100% B→2.70 min 100% B→2.80 min 0% B→3.00 min 0% B; flow rate: 0.70 mL/min; detection: UV 254 nm; detection: ELSD detector; detection: SQD, single quadrupole; ion source: ES+/ES−; scan range: 90-900 amu.
  • Method 4
  • Instrument: LC/MS Waters Acquity UPLC System DAD, ELSD detector, SQD single quadrupole; column: HSS C18 1.8 μm 2.1×50 mm, Temp 35° C.; mobile phase: A=H2O 90%+10% CH3CN+NH4COOH 5 mmol, B=CH3CN 90%+H2O 10%; gradient: 0.0 min 0% B→2.40 min 100% B→2.70 min 100% B→2.80 min 0% B→3.00 min 0% B; flow rate: 0.70 mL/min; detection: UV 254 nm; detection: ELSD detector; detection: SQD, single quadrupole; ion source: ES+/ES−; scan range: 90-900 amu.
  • HPLC-MS Methods:
  • Method 5
  • Instrument: LC/MS ThermoFinnigan. Hplc Surveyor DAD, MSQ Quadrupole; column: Synergi Hydro-RP80A, 4 um, 4.60×100 mm; eluent A: 90% water+10% ACN+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.
  • Method 6
  • Instrument: LC/MS ThermoFinnigan HPLC Surveyor DAD, LCQ Fleet Ion Trap; column:
  • Simmetry Shield RPB, 5 μm, 4.6×150 mm; eluent A: 90% water+10% ACN+HCOOH 0.1%; eluent B=ACN 90%+10% H2O+HCOOH 0.1%; gradient: 0.0 min 5% B→1.5 min 5% B→11.5 min 95% B→13.0 min 95% B→13.3 min 5% B→15.0 min 5% B; flow rate: 1.0 mL/min; UV Detection: 254 nm; Detection: Finnigan Fleet, Ion Trap; ion source: ES+; scan range: 100-900 amu.
  • Method 7
  • Instrument: LC/MS ThermoFinnigan. Hplc Surveyor DAD, MSQ Quadrupole; column: Synergi Hydro-RP8, 4 um, 4.60×100 mm; eluent A: 90% water+10% ACN+ammonium formate 10 mM; eluent B=ACN 90%+10% H2O+NH4COOH 10 mM; gradient: 0.0 min 30% B→1.50 min 50% B→8.50 min 100% B→13.50 min 100% B→14.00 min 30% B→15.00 min 30% B; flow rate: 0.85 mL/min; UV Detection: 254 nm; Ion source: ES+.
  • Method 8
  • Instrument: LC/MS ThermoFinnigan. Hplc Surveyor DAD, MSQ Quadrupole; column: Synergi Hydro-RP100A, 2.5 um, 3×50 mm; eluent A: 90% water+10% ACN+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: 0.7 mL/min; UV Detection: 254 nm; Ion source: APCI.
  • Method 9
  • Instrument: LC/MS ThermoFinnigan. Hplc Surveyor DAD, MSQ Quadrupole; column: Synergi Hydro-RP100A, 2.5 um, 3×50 mm; eluent A: 90% water+10% ACN+NH4COOH 5 mM; eluent B=ACN 90%+10% H2O; gradient: A (100), then to B (100) in 4 min for 1.3 min; flow rate: 1.2 mL/min; UV Detection: 254 nm; Ion source: APCI.
  • Method 13
  • Instrument: LC/MS ThermoFinnigan HPLC Surveyor DAD, LCQ Fleet Ion Trap; column: Xselect CSH, 2.5 um, 4.6×50 mm; eluent A: 90% water+10% ACN+HCOOH 0.1%; eluent B=ACN 90%+10% H2O+HCOOH 0.1%; gradient: A (100), then to B (100) in 4 min for 1.3 min; then to A(100) in 1.6 min. flow rate: 1.4 mL/min; UV Detection: 254 nm; Ion source: ESI.
  • Method 16
  • Instrument: LC/MS Waters Alliance 2695 HPLC System DAD, Quattro Micro Triple quadrupole; Column: Atlantis dC18 5 μm 4.6×50 mm, Temp 35° C.: Mobile phase: A=H2O 90%+10% CH3CN+CF3COOH 0.05%; B=CH3CN 90%+10% H2O; flow rate: 1.3 mL/min; UV Detection: 254 nm; Ion source: ESI.
  • Gradient:
  • Time in min % A % B
    0.00 100 0
    0.70 100 0
    4.5 0 100
    5.80 0 100
    6.00 100 0
  • Method 17
  • Instrument: LC/MS ThermoFinnigan HPLC Surveyor DAD, LCQ Fleet Ion Trap; column: Xselect CSH, 2.5 um, 4.6×50 mm; eluent A: 90% water+10% ACN+HCOOH 0.1%; eluent B=ACN 90%+10% H2O+HCOOH 0.1%; flow rate: 1.4 mL/min; UV Detection: 254 nm; Ion source: ESI.
  • Gradient:
  • Time in min: % A % B
    0.00 100 0
    1.00 100 0
    8.50 0 100
    10.0 0 100
    10.2 100 0
    11.0 100 0
  • GC-MS Methods:
  • Method 10 (3A.2)
  • Instrument: GC/MS Thermo Scientific TRACE GC ULTRA, DSQ II MS single quadrupole; column: Agilent DB-5MS, 25 m×0.2 5 mmol×0.25 μm; carrier gas: Helium, 1 mL/min costant flow; oven program: 50° C., to 100° C. in 10° C./min, to 200° C. in 20° C./min, to 320° C. in 30° C./min (hold 10 min); detection: DSQ II MS single quadrupole; ion source: EI; scan range: 50-450 amu.
  • Preparative HPLC Methods:
  • Method 11:
  • Instrument: Waters Autopurification HPLC/MS System; column: Sunfire C18, ODB 5.0 μm, 19×100 mm. MS Zq single quadropole; eluent A: 90% water+0.05% TFA; eluent B=ACN gradient: 0.0 min 20% B→6.5 min 60% B→7.0 min 95% B→8.5 min 20% B flow rate: 40 mL/min; UV Detection: 254 nm; Ion source: ES+.
  • Method 12:
  • Instrument: Waters Autopurification HPLC/MS System; column: Sunfire C18, ODB 5.0 μm, 19×100 mm. MS Zq single quadropole; eluent A: 90% water+0.05% TFA; eluent B=ACN gradient: 0.0 min 50% B→6.5 min 90% B→7.0 min 95% B→8.5 min 50% B flow rate: 40 mL/min; UV Detection: 254 nm; Ion source: ES+.
  • Method 14:
  • Instrument: Waters Autopurification HPLC/MS System; column: Xbridge, C18 5 μm, 19×100 mm. MS Zq single quadropole; eluent A: 90% water+NH4COOH 5 mM, eluent B=ACN gradient: 0.0 min 40% B→6.5 min 80% B→7.0 min 95% B→8.0 min 95% B→8.5 min 40% B flow rate: 40 mL/min; UV Detection: 254 nm; Ion source: ES+.
  • Method 15
  • Instrument: Waters Autopurification HPLC/MS System; column: Xbridge, C18 5 μm, 19×100 mm. MS Zq single quadropole; eluent A: 90% water+NH4COOH 5 mM, eluent B=ACN gradient: 0.0 min 30% B→6.5 min 80% B→6.50 min 70% B→8.0 min 95% B→8.5 min 30% B flow rate: 40 mL/min; UV Detection: 254 nm; Ion source: ES+.
  • Chiral HPLC Methods:
  • Instrument: HPLC Agilent 1100 (DAD equipped; UV Detection: 230 nm); flow rate: 1 mL/min; column temperature: 25° C.
  • Method C1
  • column: Daicel Chiralpack AS-H; eluent: Hexane:EtOH=70:30
  • Method C1a
  • column: Daicel Chiralpack AS-H; eluent: Hexane:EtOH=80:20
  • Method C2
  • column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=70:30
  • Method C2a
  • column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=75:25
  • Method C2b
  • column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=80:20
  • Method C2c
  • column: Daicel Chiralpack AD-H; eluent: Hexane:Isopropanol=60:40
  • Method C3
  • column: Daicel Chiralcel OJ-H; eluent: Hexane:EtOH=80:20
  • The most suitable purification techniques applied for the purification of compounds of the present invention are direct phase silica gel flash chromatography and reverse phase chromatography or preparative HPLC.
  • General Comment Concerning the Presentation of the Structures
  • Compounds with stereogenic centre(s): The structures depicted in the experimental section will not necessarily show all the stereochemical possibilities of the compounds but only one.
  • The structural presentation of the compounds in the experimental section will show a stereochemical bond only in case where the absolute stereochemistry is known.
  • The structural presentation of the compounds in the experimental section with unknown absolute stereochemistry will show a planar bond plus an additional comment that indicates if the described compound is a racemic mixture, a single stereoisomer and where applicable the relative stereochemistry.
  • Two examples are given below.
  • Example 1
  • The presented chemical structure is depicted as:
  • Figure US20140045856A1-20140213-C00182
  • The added term racemic mixture points to the two stereochemical options and thus the manufactured compounds is a mixture of:
  • Figure US20140045856A1-20140213-C00183
  • When racemic mixtures of above depicted structures are separated, the single stereoisomers are depicted as:
  • Figure US20140045856A1-20140213-C00184
  • The added term ‘single stereoisomer’ indicates that the absolute configuration is unknown.
  • Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC, single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.
  • Example 2
  • The presented chemical structure is depicted as:
  • Figure US20140045856A1-20140213-C00185
  • The added term ‘TRANS— racemic mixture’ points to the two stereochemical options and thus the manufactured compounds is a mixture of:
  • Figure US20140045856A1-20140213-C00186
  • The same principles applies to ‘CIS-racemic mixture’.
  • When racemic mixtures of above depicted structures are separated, the single stereoisomers are depicted as:
  • Figure US20140045856A1-20140213-C00187
  • The added term ‘TRANS-single stereoisomer’ indicates a relative configuration known (trans) and the planar bond indicates the unknown absolute configuration.
  • The same principles applies to ‘CIS-single stereoisomer’.
  • Single stereoisomer a is assigned to the first eluting isomer in chiral HPLC, single stereoisomer b is assigned to the second eluting isomer in chiral HPLC.
  • INTERMEDIATES Intermediate 1
  • Figure US20140045856A1-20140213-C00188
  • To a stirred solution of 2-oxo-propanoic acid (157 mL, 2.23 mmol) in dry MeOH (100 mL) at 0° C., pyridine-2,3-diamine (20 g, 18 mmol) was added portionwise. The resulting solution was stirred at room temperature overnight, solvent was evaporated under reduced pressure and the suspension filtered. The solid was washed with MeOH, dried to obtain 14 g of title compound.
  • HPLC-MS (Method 6): Rt=5 min
  • MS: m/z=162 (M+H)+
  • Intermediate 2
  • Figure US20140045856A1-20140213-C00189
  • A mixture of Intermediate 1 (1 g) and phosphorus oxychloride (15 mL) was heated to 90° C. for 2.5 h. After cooling, the reaction mixture was carefully poured on ice, then neutralized with a saturated solution of Na2CO3 and diluted with DCM. Phases were separated, organics washed with a saturated solution of NaCl, dried over sodium sulphate and evaporated to obtain 0.95 g of the title compound.
  • HPLC-MS (Method 8): Rt=4.54 min
  • MS: m/z=180 (M+H)+
  • Intermediate 3
  • Figure US20140045856A1-20140213-C00190
  • To a mixture of commercially available 2-chloro-5-hydroxybenzoic acid (4 g, 23.18 mmol) in dry EtOH (90 mL), concentrated sulphuric acid (1 mL, 17.62 mmol) was added. The mixture was stirred under reflux overnight, then solvent evaporated under reduced pressure and the residue dissolved with DCM. A saturated solution of NaHCO3 was added, phases were separated, organics dried over sodium sulphate, filtered and evaporated to obtain 5 g of the corresponding 2-chloro-5-hydroxybenzoic acid ethyl ester.
  • HPLC-MS (Method 2): Rt=1.0 min
  • MS: m/z=201.6 (M+H)+
  • Intermediate 4
  • Figure US20140045856A1-20140213-C00191
  • Intermediate 3 (5 g, 23.62 mmol) was dissolved in EtOH (100 mL) and hydrazine hydrate (13.0 mL, 262.12 mmol) was added. Mixture was stirred under reflux for 24 h, solvent evaporated and the crude treated with 20 mL of water. The white solid formed was filtered and washed with water and dried for 3 h at 60° C. over P2O5 to obtain 3.0 g of title compound.
  • HPLC-MS (Method 2): Rt=0.53 min
  • MS: m/z=180 (M+H)+
  • Intermediate 5
  • Figure US20140045856A1-20140213-C00192
  • Intermediate 5 was prepared as described for Intermediate 3 starting from commercially available 2-fluoro-5-hydroxy-benzoic acid (1 g, 6.41 mmol) to obtain 1.17 g of the title compound.
  • HPLC-MS (Method 2): Rt=0.94 min
  • MS: m/z=185.16 (M+H)+
  • The following Ester Intermediates were prepared in analogy to Intermediate 3 starting from commercially available benzoic acids.
  • Starting
    benzoic acid Ester Intermediate MS m/z Rt (min) Method
    Figure US20140045856A1-20140213-C00193
    5a
    Figure US20140045856A1-20140213-C00194
    180 [M]+  9.73 10
    Figure US20140045856A1-20140213-C00195
    5b
    Figure US20140045856A1-20140213-C00196
    203 [M + H]+ 1.2  2
    Figure US20140045856A1-20140213-C00197
    5c
    Figure US20140045856A1-20140213-C00198
    263 [M]+  9.90 10
    Figure US20140045856A1-20140213-C00199
    5d
    Figure US20140045856A1-20140213-C00200
    310 [M]+ 10.47 10
    Figure US20140045856A1-20140213-C00201
    5e
    Figure US20140045856A1-20140213-C00202
    294 [M]+  9.52 10
    Figure US20140045856A1-20140213-C00203
    5f
    Figure US20140045856A1-20140213-C00204
    242 [M]+  9.42 10
    Figure US20140045856A1-20140213-C00205
    5g
    Figure US20140045856A1-20140213-C00206
    246 [M]+  8.84 10
    Figure US20140045856A1-20140213-C00207
    5h
    Figure US20140045856A1-20140213-C00208
    290 [M]+ 10.15 10
  • Intermediate 5i
  • Figure US20140045856A1-20140213-C00209
  • Step 1:
  • Step 1 was performed in analogy to what reported in literature reference: Li, Yuanzhen et al. Organic Letters, 2007, vol. 9, #20 p. 4057-4060, starting from Ester Intermediate 5c.
  • Step 2:
  • Intermediate obtained from Step 1 (0.5 g), was dissolved in toluene (10 mL). Wilkinson's catalyst RhCl(TPP)3 (0.1 g) was added and the reaction mixture stirred at 60° C. under hydrogen atmosphere for 18 h. The reaction mixture was filtered on a celite pad and the solvent was removed under vacuum to obtain the desired product (0.4 g).
  • GC-MS (Method 10) Rt=11.55 min
  • MS: m/z=256 [M]+
  • Intermediate 5j
  • Figure US20140045856A1-20140213-C00210
  • Under nitrogen atmosphere, starting Ester Intermediate 5e (5.0 g, 15.3 mmol) was dissolved in THF (40 mL), the reaction mixture was cooled to −50° C. and stirred for 10 minutes. Isopropyl-magnesium-chloride (11 mL of a 2M solution in THF) was added dropwise and the reaction mixture was stirred at −50° C. for 2 h. The reaction mixture was cooled to −78° C. and a solution of cyclobutanone (2.15 g, 30.6 mmol) in THF (10 mL) was added dropwise. The reaction mixture was stirred at −78° C. for 1 h, then warmed to room temperature and stirred for 18 h. The solvent was removed under vacuum, dichloromethane was added and the reaction mixture was washed with a saturated solution of NaHCO3, The organic phase was concentrated under vacuum and the crude product obtained was purified by flash chromatography (eluent from 90:10 to 80:20 Cy/EtOAc) to obtain the desired product (3.0 g).
  • GC-MS (Method 10): Rt=10.91 min
  • MS: m/z=210 (M−28)+
  • The following Ester Intermediates were prepared in analogy to Intermediate 5j and purified applying the most suitable purification technique, starting from the corresponding Starting Esters Intermediates and commercially available ketones.
  • Starting
    Ester Starting Ester intermediate Rt
    intermediate ketone Structure MS m/z (min) Method
    5d
    Figure US20140045856A1-20140213-C00211
    5k
    Figure US20140045856A1-20140213-C00212
    255 [M + H]+  4.23 16
    5d
    Figure US20140045856A1-20140213-C00213
    5l
    Figure US20140045856A1-20140213-C00214
    284 [M]+ 12.58 10
    5d
    Figure US20140045856A1-20140213-C00215
    5m
    Figure US20140045856A1-20140213-C00216
    243 [M + H]+  1.04  2
    5e
    Figure US20140045856A1-20140213-C00217
    5n
    Figure US20140045856A1-20140213-C00218
    268 [M]+ 11.97 10
    5h
    Figure US20140045856A1-20140213-C00219
     5na
    Figure US20140045856A1-20140213-C00220
    264 [M]+ 12.18 10
    5d
    Figure US20140045856A1-20140213-C00221
    5q
    Figure US20140045856A1-20140213-C00222
    253 [M + H]+  2.84 13
    5e
    Figure US20140045856A1-20140213-C00223
    5r
    Figure US20140045856A1-20140213-C00224
    237 [M − 17]+  2.68 13
  • Intermediate 5o
  • Figure US20140045856A1-20140213-C00225
  • To a solution of Ester Intermediate 5m (0.37 g, 1.52 mmol) in dichloromethane (9 mL), pyridinium-para-toluenesulfonate (0.06 mg, 0.23 mmol) and 3,4-dihydro-2H-pyran (0.2 mL, 2.29 mmol) were added and the reaction mixture was stirred at room temperature for 4 h. The reaction mixture was washed with water and the organic phase was concentrated under vacuum. Crude was purified by flash chromatography (eluent from 100:0 to 80:20 Cy/EtOAc) to obtain the desired product (0.4 g).
  • GC-MS (Method 10): Rt=12.52 min
  • MS: m/z=326 (M)+
  • Intermediate 5p
  • Figure US20140045856A1-20140213-C00226
  • Step 1:
  • Step 1 was performed in analogy to what reported in the literature reference: Duong, Hung A.; et al. Angewandte Chemie, International Edition, 2011, vol. 50, p. 463-466, starting from commercially available (3-bromo-4-chloro-phenyl)-acetic acid.
  • Step 2:
  • Under nitrogen atmosphere, a solution of intermediate from Step 1 (0.24 g) in THF (10 mL) was stirred at −60° C., methylmagnesium bromide was added dropwise and the reaction mixture was stirred at −78° C. to r.t. for 2 h. The reaction mixture was quenched with saturated ammonium chloride water solution and extracted with ethyl acetate twice, dried over sodium sulfate and concentrated under vacuum to get the crude product as colour-less oil.
  • Step 3:
  • Step 3 was performed in analogy to Step 2 starting from Intermediate obtained in Step 2.
  • Step 4:
  • Step 4 was performed in analogy to preparation of Intermediate 5o, starting from Intermediate obtained in Step 3.
  • Step 5:
  • To a solution of the Intermediate obtained from Step 4 (15 g) in M($!EB80BDF4-E19E-4FD6-9A46-54BC1C095AB53)e0H (150 mL), [1,1′-bis(diphenylphosphino)ferrocene]clichloropalladium(II)] (3.088 g) and triethylamine($!9DC53CF4-BE29-4563-8B63-CD95B9951 D7A!riethylamine (22 g, 21.4 mL) were added and the reaction mixture was stirred at 80° C. under CO atmoshere for 36 h.
  • The solvent was removed under vacuum and the crude product obtained was purified by flash chromatography to get the desired product.
  • Intermediate 6
  • Figure US20140045856A1-20140213-C00227
  • Intermediate 6 (4.4 g) was prepared as described for Intermediate 4 and purified applying the most suitable purification technique, starting from Intermediate 5 (5.84 g, 0.03 mmol).
  • HPLC-MS (Method 2): Rt=0.42 min
  • MS: m/z=171.2 (M+H)+
  • The following Hydrazide Intermediates were prepared in analogy to Intermediate 4 and purified applying the most suitable purification technique, starting from the corresponding Ester Intermediates.
  • Starting
    Ester Hydrazide Intermediate
    intermediate Structure MS m/z Rt (min) Method
    5a 6a
    Figure US20140045856A1-20140213-C00228
    167 [M + H]+  0.47 17
    5b 6b
    Figure US20140045856A1-20140213-C00229
    188 [M]+  9.47 10
    5c 6c
    Figure US20140045856A1-20140213-C00230
    249 [M + H]+  1.98 13
    5d 6d
    Figure US20140045856A1-20140213-C00231
    296 [M]+ 11.78 10
    5e 6e
    Figure US20140045856A1-20140213-C00232
    281 [M + H]+  1.83  9
    5f 6f
    Figure US20140045856A1-20140213-C00233
    229 [M + H]+  1.80  9
    5g 6g
    Figure US20140045856A1-20140213-C00234
    233 [M + H]+  1.55  9
    5h 6h
    Figure US20140045856A1-20140213-C00235
    277 [M + H]+  1.94  9
    5i 6i
    Figure US20140045856A1-20140213-C00236
    256 [M + H]+  0.68  2
    5j 6j
    Figure US20140045856A1-20140213-C00237
    225 [M + H]+  0.56  2
    5k 6k
    Figure US20140045856A1-20140213-C00238
    241 [M + H]+  0.59  2
    5l 6l
    Figure US20140045856A1-20140213-C00239
    271 [M + H]+  0.57  2
    5m 6m
    Figure US20140045856A1-20140213-C00240
    228 [M]+ 11.79 10
    5n 6n
    Figure US20140045856A1-20140213-C00241
    255 [M + H]+  0.43  2
     5na  6na
    Figure US20140045856A1-20140213-C00242
    251 [M + H]+  0.57  2
    5o 6o
    Figure US20140045856A1-20140213-C00243
    313 [M + H]+  0.85  2
    5p 6p
    Figure US20140045856A1-20140213-C00244
    327 [M + H]+  0.90  2
    5q 6q
    Figure US20140045856A1-20140213-C00245
    257 [M + H]+  0.49 13
    5r 6r
    Figure US20140045856A1-20140213-C00246
    241 [M + H]+  0.44  2
  • Intermediate 7
  • Figure US20140045856A1-20140213-C00247
  • Commercially available 2-amino-6-methyl-3-nitropyridine (12 g, 78.36 mmol) was dissolved in a mixture of dry THF/MeOH (1:1, 60 mL) and hydrogenated in the presence of Pd/C (10%, 1.2 g) under 5 bar hydrogen atmosphere for 5 h. Catalyst was filtered, solvent evaporated under reduced pressure to obtain 7 g of the title compound.
  • HPLC-MS (Method 2): Rt=0.77 min
  • MS: m/z=154 (M+H)+
  • Intermediate 7a
  • Figure US20140045856A1-20140213-C00248
  • Step 1:
  • 2-Amino-6-chloro-3-nitro-pyridine (20 g, 115 mmol), cyclopropylboronic acid (12.8 g, 150 mmol), tricyclohexylphosphine (3.2 g, 11.5 mmol), 2,6-ditert-butyl-4-methylphenol (5.08 g, 23 mmol), palladium acetate (1.3 g, 5.7 mmol) and tri-potassium-phosphate (73 g, 346 mmol) were suspended in toluene (40 mL) and water (2 mL). The reaction mixture was stirred at reflux for 18 h, diluted with ethyl acetate and washed with water. The organic phase was separated, concentrated under vacuum and dried over sodium sulfate. The crude product obtained was purified by flash chromatography (eluent from 90:10 to 45:55 Cy/EtOAc) to obtain the desired product (10.6 g).
  • GC-MS (Method 10): Rt=10.54 min
  • MS: m/z=179 (M)+
  • Step 2:
  • To a solution of Intermediate from Step 1 (6 g, 19 mmol) in absolute EtOH (120 mL), zinc (13.7 g, 209 mmol) and ammonium chloride (30.8 g, 573 mmol) were added and the reaction mixture was stirred at room temperature for 18 h. The reaction mixture was filtered on a celite pad, the solvent was removed under vacuum and the crude product obtained was purified by flash chromatography (eluent 90:10 dichloromethane/MeOH) to obtain the desired product (2.4 g).
  • HPLC-MS (Method 9): Rt=1.42 min
  • MS: m/z=150 (M+H)+
  • Intermediate 7b
  • Figure US20140045856A1-20140213-C00249
  • Step 1:
  • Step 1 was performed in analogy to what described in Step 1 in the preparation of Intermediate 7a, starting from commercially available 2-Isopropenyl-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane.
  • Step 2:
  • Intermediate from Step 1 (5 g, 26.5 mmol) was dissolved in 20 mL of methanol, Pd/C (0.25 g) was added and the reaction mixture was stirred under hydrogen atmosphere (4 bar) for 2 h at room temperature. The reaction mixture was filtered on a celite pad and the solvent was removed under vacuum to give the desired product (3 g).
  • GC-MS (Method 10): Rt=8.91 min
  • MS: m/z=151 (M)+
  • Intermediate 7c
  • Figure US20140045856A1-20140213-C00250
  • Intermediate 7c (3.99 g) was prepared in analogy to Intermediate 7 and purified applying the most suitable purification technique, starting from commercially available 2-amino-3-nitro-5-picoline (5.0 g, 32.65 mmol).
  • Intermediate 7d
  • Figure US20140045856A1-20140213-C00251
  • Intermediate 7d (4 g) was prepared in analogy to Intermediate 7b and purified applying the most suitable purification technique, starting from commercially available 2-amino-5-chloro-3-nitro-pyridine (5 g, 26.5 mmol).
  • HPLC-MS (Method 2): Rt=0.6 min
  • MS: m/z=152 (M+H)+
  • Intermediate 8
  • Figure US20140045856A1-20140213-C00252
  • To stirred solution of 2-oxo-propanoic acid (40.8 mL, 587 mmol) in dry MeOH (130 mL) at 0° C., 6-methyl-pyridine-2,3-diamine (Intermediate 7, 6.8 g, 55.2 mmol) dissolved in dry MeOH (40 mL) was added dropwise. The solution was stirred at room temperature overnight then solvent was evaporated under reduced pressure and the resulting suspension filtered. The solid was washed with MeOH, dried to obtain 1.35 g of title compound.
  • HPLC-MS (Method 9): Rt=1.31 min
  • MS: m/z=176 (M+H)+
  • Intermediate 8a
  • Figure US20140045856A1-20140213-C00253
  • Step 1:
  • Intermediate 7a (2.4 g, 15.2 mmol) was dissolved in absolute EtOH and stirred at 0° C., Dimethylacetylenedicarboxylate (2.4 g, 16.8 mmol) was added dropwise and the reaction mixture was allowed to react room temperature for 18 h. The solid formed was filtered, washed with EtOH, Ethyl Ether and dried in vacuum to give the desired product (3 g).
  • HPLC-MS (Method 13): Rt=3.16 min
  • MS: m/z=260 (M+H)+
  • Step 2:
  • Intermediate obtained from Step 1 (3 g 0.01 mol) was dissolved in a sodium hydroxide water solution (1.36 g, 0.03 mol in 50 mL) and the reaction mixture was reflux for 2 h. pH was adjusted to 2-3 and the reaction mixture was extracted with ethyl acetate. The organic phase was dried over sodium sulfate, concentrated under vacuum. The crude product obtained was purified by flash chromatography (eluent from 80:20 to 20:80 Cy/EtOAc) to obtained the desired product (2.0 g).
  • HPLC-MS (Method 13): Rt=2.46 min
  • MS: m/z=202 (M+H)+
  • The following Amide Intermediates were prepared in analogy to Amide Intermediate 8a and purified applying the most suitable purification technique, starting from the corresponding Di-amine Intermediates.
  • Starting
    Di-amine Amide Intermediate Rt
    Intermediate Structure MS m/z (min) Method
    7b 8b
    Figure US20140045856A1-20140213-C00254
    204 [M + H]+ 2.48 13
    7c 8c
    Figure US20140045856A1-20140213-C00255
    176 [M + H]+ 1.88 13
    7d 8d
    Figure US20140045856A1-20140213-C00256
    204 [M + H]+ 0.78  2
    6-(1- Methyl- cyclo- propyl)- pyridine- 2,3- diamine (commer- cially available) 8e
    Figure US20140045856A1-20140213-C00257
    216 [M + H]+ 4.96 17
    6- trifluoro- methyl- pyridine- 2,3- diamine (commer- cially available) 8f
    Figure US20140045856A1-20140213-C00258
    229 [M]+ 9.42 10
  • Intermediate 9
  • Figure US20140045856A1-20140213-C00259
  • A mixture of Amide intermediate 8 (1.35 g, 7.71 mmol) and phosphorus oxychloride (16 mL) was heated to 90° C. for 3 h. After cooling, solvent was reduced under reduced pressure, the reaction mixture was carefully poured on ice and neutralized with a saturated solution of Na2CO3. DCM was added, phases were separated, organics washed with a saturated solution of NaCl, dried over sodium sulphate and evaporated to obtain 1.39 g of the title compound.
  • HPLC-MS (Method 8): Rt=0.84 min
  • MS: m/z=194.6 (M+H)+
  • The following Chloride Intermediates were prepare in analogy to Chloride Intermediate 9 and purified applying the most suitable purification technique, starting from the corresponding Amide Intermediates. Heating temperature reported in Table.
  • Chloride
    Starting Intermediate Heating Rt
    Intermediate Structure Temperature MS m/z (min) Method
    8a 9a
    Figure US20140045856A1-20140213-C00260
    55° C. 220 [M + H]+ 1.01  2
    8b 9b
    Figure US20140045856A1-20140213-C00261
    80° C. 222 [M + H]+ 3.18 13
    8c 9c
    Figure US20140045856A1-20140213-C00262
    90° C. 194 [M + H]+ 2.52 13
    8d 9d
    Figure US20140045856A1-20140213-C00263
    55° C. 222 [M + H]+ 3.19 13
    8e 9e
    Figure US20140045856A1-20140213-C00264
    80° C. 234 [M + H]+ 1.15  2
  • Intermediate 9f
  • Figure US20140045856A1-20140213-C00265
  • Intermediate 8f (1.35 g, 5.9 mmol) and N,N-diisopropylethylamine (2.28 g, 3 mmol) were dissolved in DMF (10 mL). The reaction mixture was cooled to 0° C. and methanesulfonylchloride (0.74 g, 6.48 mmol) was added dropwise. The reaction mixture was allowed to reach room temperature and stirred for 4 h. Ammonium chloride saturared water solution was added and the reaction mixture was extracted with dichloromethane. The organic phase was washed with citric acid (10% water solution), dried over sodium sulfate and concentrated under vacuum, to give the desired product (1.5 g).
  • HPLC-MS (Method 13): Rt=3.09 min
  • MS: m/z=308 (M+H)+
  • The following Intermediates were prepared based on well known reported procedures starting from commercially available starting materials.
  • Intermediate Structure Reference
    10 Toluene-4-sulfonic acid 3-hydroxy-3-methyl- butyl ester
    Figure US20140045856A1-20140213-C00266
    Bioorganic & Medicinal Chemistry Letters (2007), 17(22), 6164- 6168
    11 Toluene-4-sulfonic acid 3-hydroxy-butyl ester
    Figure US20140045856A1-20140213-C00267
    Bioorganic & Medicinal Chemistry, 18(4), 1665-1675; 2010
    12 Toluene-4-sulfonic acid 3,3,3-trifluoro-propyl ester
    Figure US20140045856A1-20140213-C00268
    US 2009-564132
    13 Toluene-4-sulfonic acid tetrahydropyran-4-yl ester
    Figure US20140045856A1-20140213-C00269
    WO 2011-20767
    14 Toluene-4-sulfonic acid 4,4-difluoro-cyclohexyl ester
    Figure US20140045856A1-20140213-C00270
    WO 2008-119663
    15 Toluene-4-sulfonic acid tetrahydro-furan-3-yl ester
    Figure US20140045856A1-20140213-C00271
    WO 2010-023594
    16 Toluene-4-sulfonic acid (S)-(tetrahydro-furan-3- yl) ester
    Figure US20140045856A1-20140213-C00272
    WO 2011-061590
    17 Toluene-4-sulfonic acid (R)-(tetrahydro-furan-3- yl) ester
    Figure US20140045856A1-20140213-C00273
    WO 2010-102512
    18 Toluene-4-sulfonic acid tetrahydro-furan-3- ylmethyl ester
    Figure US20140045856A1-20140213-C00274
    WO 2011-084402
    19 tert-Butyl-(R)-2-chloro- 1-methyl-ethoxy)- diphenyl-silane
    Figure US20140045856A1-20140213-C00275
    Tetrahedron Letters (1982), 23(25), 2543-6.
    20 tert-Butyl-(S)-2-chloro- 1-methyl-ethoxy)- diphenyl-silane
    Figure US20140045856A1-20140213-C00276
    Tetrahedron Letters (1982), 23(25), 2543-6.
    21 2-((S)-2-Chloro-1- methyl-ethoxy)- tetrahydro-pyran
    Figure US20140045856A1-20140213-C00277
    Tetrahedron Letters (1982), 23(25), 2543-6.
    26 Toluene-4-sulfonic acid 3-methyl-oxetan-3- ylmethyl ester
    Figure US20140045856A1-20140213-C00278
    Synthetic Communications (2011), 41(17), 2539- 2543
    27 Toluene-4-sulfonic acid 1-(tetrahydro-pyran-4- yl)-ethyl ester
    Figure US20140045856A1-20140213-C00279
    Journal of Organometallic Chemistry (1978), 150(2), 179-85
    28 Toluene-4-sulfonic acid 1,4-dioxa- spiro[4.5]dec-8-yl ester
    Figure US20140045856A1-20140213-C00280
    WO 2010-006086
    29 Toluene-4-sulfonic acid 3-hydroxy-cyclopentyl ester
    Figure US20140045856A1-20140213-C00281
    US 2007-0049537
  • Intermediate 29a
  • Figure US20140045856A1-20140213-C00282
  • Literature Reference: Journal of Organic Chemistry USSR (English Translation), 1989, vol. 25, #6.1 p. 1019-1025).
  • Epoxide Intermediate 29a (4 g) was prepared in analogy to what described in WO2010/47956 A1, starting from methylene cyclobutane (5 g, 73 mmol).
  • GC-MS (Method 10) Rt=2.70 min
  • [M]+=83
  • Intermediate 29b
  • Figure US20140045856A1-20140213-C00283
  • Epoxide Intermediate 29b (1 g) was prepared in analogy to what described in WO2013/55577 A1 starting from 3,6-Dihydro-2H-pyran (2 g, 23.78 mmol).
  • GC-MS (Method 10) Rt=8.10 min
  • [M]+=99
  • Intermediate 29c
  • Figure US20140045856A1-20140213-C00284
  • To a solution of Example 1 (0.2 g, 0.67 mmol) in dry DMF (3 mL), cesium carbonate (0.63 g, 1.92 mmol) and Intermediate 28 (0.41 g, 1.28 mmol) were added. Mixture was heated at 80° C. for 1 h, then solvent evaporated under reduced pressure and the residue dissolved in DOM and washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.21 g of the title compound.
  • HPLC-MS (Method 2): Rt=1.12 min
  • MS: m/z=452 (M+H)+
  • Intermediate 30
  • Figure US20140045856A1-20140213-C00285
  • To a solution of Intermediate 29c (0.21 g, 0.46 mmol) in acetone (10 mL), a 4M solution of HCl (1 mL) was added and mixture stirred at room temperature for 2 h. Solvent was evaporated under reduced pressure, the residue treated with DCM, washed with a saturated solution of NaHCO3. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 90:10 to 50:50 Cy/EtOAc) to obtain 0.18 g of the title compound.
  • HPLC-MS (Method 2): Rt=1.09 min
  • MS: m/z=410 (M+H)+
  • The following intermediates were prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from the corresponding Chloride and Idrazide intermediates:
  • Chloride
    Inter- MS Rt
    mediate Hydrazide Intermediate Structure m/z (min) Method
    9  6c 30a
    Figure US20140045856A1-20140213-C00286
    388 [M + H]+ 3.58 13
    2  6d 30b
    Figure US20140045856A1-20140213-C00287
    422 [M + H]+ 1.07  2
    2  6h 30c
    Figure US20140045856A1-20140213-C00288
    402 [M + H]+ 3.00  9
    2  6e 30d
    Figure US20140045856A1-20140213-C00289
    406 [M + H]+ 3.02  9
    9a 6e 30e
    Figure US20140045856A1-20140213-C00290
    446 [M + H]+ 3.78 13
    2  6f 30f
    Figure US20140045856A1-20140213-C00291
    354 [M + H]+ 3.35 13
    9a 6f 30g
    Figure US20140045856A1-20140213-C00292
    394 [M + H]+ 3.69 13
    9a 6d 30h
    Figure US20140045856A1-20140213-C00293
    462 [M + H]+ 3.81 13
    9a 6g 30i
    Figure US20140045856A1-20140213-C00294
    398 [M + H]+ 3.72 13
    9  6d 30j
    Figure US20140045856A1-20140213-C00295
    436 [M + H]+ 3.64 13
    9b 6d 30k
    Figure US20140045856A1-20140213-C00296
    464 [M + H]+ 3.99 13
    9b 6f 30l
    Figure US20140045856A1-20140213-C00297
    396 [M + H]+ 3.85 13
    9e 6e 30m
    Figure US20140045856A1-20140213-C00298
    460 [M + H]+ 1.29  2
    9d 6f 30n
    Figure US20140045856A1-20140213-C00299
    397 [M + H]+ 1.27  2
    9a 6b 30o
    Figure US20140045856A1-20140213-C00300
    354 [M + H]+ 3.53 13
    9f 6d 30r
    Figure US20140045856A1-20140213-C00301
    490 [M + H]+ 1.25  2
    9f 6g 30s
    Figure US20140045856A1-20140213-C00302
    427 [M + H]+ 1.15  2
  • Intermediate 30p
  • Figure US20140045856A1-20140213-C00303
  • Intermediate 30p (0.26 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Chloride Intermediate 9d (0.25 g, 1.01 mmol) and Hydrazide Intermediate 4 (0.19 g, 1.01 mmol).
  • HPLC-MS (Method 2): Rt=1.01 min
  • MS: m/z=354 (M+H)+
  • Intermediate 31
  • Figure US20140045856A1-20140213-C00304
  • To a solution of Example 1 (0.4 g, 1.3 mmol) in dry DMF (4 mL), cesium carbonate (0.85 g, 2.62 mmol) and methylbromoacetate (0.152 mL, 1.56 mmol) were added. Mixture was stirred at room temperature overnight, solvent evaporated and the residue treated with EtOAc. A saturated solution of NH4Cl was added, phases were separated and evaporated. The crude was purified by flash chromatography (eluent from 50:50 to 0:100 Cy/EtOAc) to obtain 0.29 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.06 min
  • MS: m/z=384 (M+H)+
  • Intermediate 32
  • Figure US20140045856A1-20140213-C00305
  • Intermediate 32 (0.5 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Intermediate 9f (0.9 g, 2.52 mmol) and Hydrazide Intermediate 6 (0.47 g, 2.77 mmol).
  • HPLC-MS (Method 13) Rt=3.11 min
  • MS: m/z=380 (M+H)+
  • EXEMPLARY EMBODIMENTS Example 1
  • Figure US20140045856A1-20140213-C00306
  • Intermediate 2 (0.60 g, 3.34 mmol) and Intermediate 4 (0.62 g, 3.34 mmol) were suspended in cyclohexanol (5 mL) and heated under reflux for 2 h. The resulting solution was cooled and solvent evaporated under reduced pressure and the residue purified by flash chromatography (eluent 95:5 DCM/EtOH) to obtain 0.95 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.17 min
  • MS: m/z=312 (M+H)+
  • The following Examples were prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from the corresponding Chloride Intermediates and Hydrazide Intermediates:
  • Starting Chloride Starting Hydrazide MS m/z Rt
    Intermediate Intermediate Ex. Structure [M + H]+ (min) Method
    9  4  2
    Figure US20140045856A1-20140213-C00307
    326 0.93  2
    9  3-hydroxy- benzhydrazide (commercially available) 3
    Figure US20140045856A1-20140213-C00308
    292 2.62 13
    2  6  4
    Figure US20140045856A1-20140213-C00309
    296 0.79 13
    9a 6  5
    Figure US20140045856A1-20140213-C00310
    336 2.97 13
    9a 6a 6
    Figure US20140045856A1-20140213-C00311
    226 2.93 13
    9b 6a 7
    Figure US20140045856A1-20140213-C00312
    334 3.16 13
    9b 6  8
    Figure US20140045856A1-20140213-C00313
    338 3.14 13
    9a 4  9
    Figure US20140045856A1-20140213-C00314
    352 2.59  9
    2  6a 10
    Figure US20140045856A1-20140213-C00315
    292 2.03 13
    9c 4  11
    Figure US20140045856A1-20140213-C00316
    326 2.96 13
  • Example 12
  • Figure US20140045856A1-20140213-C00317
  • Example 12 (0.04 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Intermediate 9f (0.3 g, 0.68 mmol) and Hydrazide Intermediate 4 (0.27, 0.68 mmol).
  • HPLC-MS (Method 13) Rt=3.11 min
  • MS: m/z=380 (M+H)+
  • Example 12a
  • Figure US20140045856A1-20140213-C00318
  • Example 12a (0.15 g) was prepared in analogy to Example 12 and purified applying the most suitable purification technique, starting from the Hydrazide Intermediate 6a (0.23 g, 1.45 mmol).
  • HPLC-MS (Method) Rt=3.00 min
  • MS: m/z=360 (M+H)+
  • Example 13
  • Figure US20140045856A1-20140213-C00319
  • To a solution of Example 1 (0.1 g, 0.24 mmol) in dry ACN (3 mL), cesium carbonate (0.2 g) and commercially available 3-chloro-1-propanol (0.04 mL) were added. Mixture was heated at 80° C. for 1 h, then solvent evaporated under reduced pressure and the residue dissolved in DCM and washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evapoarated. The residue was purified by flash chromatography (eluent from 100:0 to 94:6 DCM/EtOH) to obtain 0.02 g of the title compound.
  • HPLC-MS (Method 8) Rt=5.23 min
  • MS: m/z=370 (M+H)+
  • Example 14
  • Figure US20140045856A1-20140213-C00320
  • To a solution of Example 1 (0.05 g, 0.16 mmol) in dry DMF (3 mL), Intermediate 10 (0.05 g, 0.018 mmol) and cesium carbonate (0.08 g, 0.24 mmol) were added and mixture heated at 100° C. for 2 h. Solvent was evaporated, the residue was dissolved with DCM, washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 70:30 to 20:80 Cy/EtOAc) to obtain 0.025 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.57 min
  • MS: m/z=398 (M+H)+
  • Example 15
  • Figure US20140045856A1-20140213-C00321
  • To a solution of Example 1 (0.08 g, 0.16 mmol) in dry ACN (3 mL), commercially available 2-bromo-ethanol (0.65 g, 0.52 mmol) and cesium carbonate (0.5 g, 1.54 mmol) were added and mixture heated at 80° C. for 48 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.04 g of the title compound.
  • HPLC-MS (Method 6): Rt=8.75 min
  • MS: m/z=356 (M+H)+
  • Example 16
  • Figure US20140045856A1-20140213-C00322
  • To a solution of Example 1 (0.05 g, 0.16 mmol) in dry DMF (3 mL), commercially available 1-bromo-2-propanol (0.09 g, 0.64 mmol) and potassium tert-butoxide (0.1 g, 0.96 mmol) were added and mixture heated at 130° C. for 8 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.05 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.23 min
  • MS: m/z=370 (M+H)+
  • Example 17
  • Figure US20140045856A1-20140213-C00323
  • To a solution of Example 1 (0.085 g, 0.27 mmol) in dry DMF (3 mL), commercially available (R)-1-chloro-2-propanol (0.052 g, 0.55 mmol) and potassium tert-butoxide (0.093 g, 0.82 mmol) were added and mixture heated at 130° C. for 48 h. Solvent was evaporated, the residue was dissolved with DCM, washed with 1N solution of NaOH and then with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.0085 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.22 min
  • MS: m/z=370 (M+H)+
  • Example 18
  • Figure US20140045856A1-20140213-C00324
  • Example 18 was prepared as described for Example 17 starting from Example 1 (0.085 g, 0.27 mmol) and commercially available (S)-1-chloro-2-propanol. The residue was purified by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH) to obtain 0.045 g of the title compound. HPLC-MS (Method 9): Rt=2.19 min
  • MS: m/z=370 (M+H)+
  • Example 19
  • Figure US20140045856A1-20140213-C00325
  • To a solution of Example 1 (0.115 g, 0.37 mmol) in dry DMF (3 mL), cesium carbonate (0.36 g, 1.11 mmol) and Intermediate 11 (0.09 g, 0.37 mmol) were added and mixture heated at 100° C. for 4 h. Solvent was evaporated, the residue was dissolved with DCM, washed with a saturated solution of NaCl. Phases were separated, organics dried over sodium sulphate and evaporated. The residue was purified by flash chromatography (eluent from 100:0 to 80:20 DCM/EtOH) to obtain 0.025 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.4 min
  • MS: m/z=384 (M+H)+
  • Example 20
  • Figure US20140045856A1-20140213-C00326
  • Example 20 was prepared as described for Example 19, starting from Example 1 (0.08 g, 0.26 mmol) and Intermediate 13 (0.2 g, 0.78 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.032 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.65 min
  • MS: m/z=396 (M+H)+
  • Example 21
  • Figure US20140045856A1-20140213-C00327
  • Example 21 was prepared as described for Example 20, starting from Example 1 (0.065 g, 0.21 mmol) and Intermediate 14 (0.12 g, 0.42 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.035 g of the title compound.
  • HPLC-MS (Method 9): Rt=3.32 min
  • MS: m/z=430 (M+H)+
  • Example 22
  • Figure US20140045856A1-20140213-C00328
  • Example 22 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 15 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.032 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.03 min
  • MS: m/z=382 (M+H)+
  • Example 23
  • Figure US20140045856A1-20140213-C00329
  • Example 23 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 17 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.02 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.07 min
  • MS: m/z=382 (M+H)+
  • Example 24
  • Figure US20140045856A1-20140213-C00330
  • Example 24 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 16 (0.093 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.03 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.05 min
  • MS: m/z=382 (M+H)+
  • Example 25
  • Figure US20140045856A1-20140213-C00331
  • Example 25 was prepared as described for Example 20, starting from Example 1 (0.1 g, 0.32 mmol) and Intermediate 18 (0.097 g, 0.38 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.03 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.18 min
  • MS: m/z=396 (M+H)+
  • Example 26
  • Figure US20140045856A1-20140213-C00332
  • Example 26 was prepared as described for Example 20, starting from Example 4 (0.08 g, 0.27 mmol) and commercially available 3-chloro-1-propanol (0.052 g, 0.54 mmol). The crude was purified by flash chromatography (eluent from 100:0 to 97:3 DCM/EtOH) to obtain 0.052 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.71 min
  • MS: m/z=354 (M+H)+
  • Example 27
  • Figure US20140045856A1-20140213-C00333
  • Example 27 was prepared as described for Example 20, starting from Example 2 (0.133 g, 80% content, 0.32 mmol) and commercially available 3-chloro-1-propanol (0.046 g, 0.49 mmol). The crude was purified by prep-HPLC (Method 11). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM and treated with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.039 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.46 min
  • MS: m/z=384 (M+H)+
  • Example 28
  • Figure US20140045856A1-20140213-C00334
  • Example 28 was prepared as described for Example 20, starting from Example 2 (0.133 g, 80% content, 0.32 mmol) and commercially available 2-bromo-ethanol (0.061 g, 0.49 mmol). The crude was purified by prep-HPLC (Method 12). Fractions containing the pure compound are combined and evaporated to reduced volume, diluted with DCM and treated with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.025 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.35 min
  • MS: m/z=370 (M+H)+
  • Example 29
  • Figure US20140045856A1-20140213-C00335
  • To a solution of Example 2 (0.07 g, 0.20 mmol) and commercially available 2-bromo-methylether (0.057 g, 0.41 mmol) in dry DMF (3 mL), cesium carbonate (0.199 g, 0.61 mmol) was added and the mixture heated at 100° C. for 1 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 40:60 hexane/EtOAc) to obtain 0.023 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.82 min
  • MS: m/z=384 (M+H)+
  • Example 30
  • Figure US20140045856A1-20140213-C00336
  • To a solution of Example 2 (0.07 g, 0.20 mmol) and Intermediate 14 (0.12 g, 0.41 mmol) in dry DMF (3 mL), cesium carbonate (0.2 g, 0.61 mmol) was added and the mixture heated at 90° C. for 2 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 40:60 hexane/EtOAc) and then further purified by prep-HPLC (Method 14). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.047 g of the title compound.
  • HPLC-MS (Method 9): Rt=3.56 min
  • MS: m/z=444 (M+H)+
  • Example 31
  • Figure US20140045856A1-20140213-C00337
  • Example 31 was prepared as described for Example 30 starting from Example 2 (0.07 g, 0.20 mmol) and commercially available 1-bromo-3-methoxy-propane (0.063 g, 0.41 mmol). After heating for 3 h at 100° C. in dry DMF (3 mL), solvent was evaporated under reduced pressure, the residue dissolved with DCM and treated with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) to obtain 0.043 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.55 min
  • MS: m/z=398 (M+H)+
  • Example 32
  • Figure US20140045856A1-20140213-C00338
  • Example 32 was prepared as described for Example 31 starting from Example 2 (0.09 g, 0.26 mmol) and Intermediate 13 (0.20 g, 0.78 mmol). Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) to obtain 0.037 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.95 min
  • MS: m/z=410 (M+H)+
  • Example 32a
  • Figure US20140045856A1-20140213-C00339
  • Example 32a (0.06 g) was prepared in analogy to Example 32 and purified applying the most suitable purification technique, starting from Example 9 (0.08 g, 0.23 mmol).
  • HPLC-MS (Method 16): Rt=4.51 min
  • MS: m/z=436 (M+H)+
  • Example 33
  • Figure US20140045856A1-20140213-C00340
  • Example 33 was prepared as described for Example 31 starting from Example 2 (0.07 g, 0.20 mmol) and commercially available 1-bromo-3-trifluoro-methoxypropane (0.084 g, 0.41 mmol). Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with water. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) and then further purified by prep-HPLC (Method 14). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.047 g of the title compound. chromatography (eluent from 88:12 to 0:100 Cy/EtOAc) to obtain 0.037 g of the title compound.
  • HPLC-MS (Method 13): Rt=1.06 min
  • MS: m/z=452 (M+H)+
  • Example 34
  • Figure US20140045856A1-20140213-C00341
  • To a solution of Example 2 (0.082 g, 0.25 mmol) in dry DMF (3 mL), potassium tert-butoxide (0.084 g, 0.65 mmol) and Intermediate 20 (0.176 g, 0.50 mmol) were added. The resulting solution was heated at 120° C. for 5 h. The mixture was left at room temperature for 24 h. Solvent was evaporated under reduced pressure, the residue dissolved in DCM and treated with a 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 20:80 to 0:100 ACN/H2O) to obtain 0.033 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.98 min
  • MS: m/z=384 (M+H)+
  • Example 35
  • Figure US20140045856A1-20140213-C00342
  • To a solution of Example 2 (0.095 g, 0.29 mmol) in dry DMF (3 mL), potassium tert-butoxide (0.074 g, 0.22 mmol and Intermediate 19 (0.204 g, 0.58 mmol) were added. The resulting solution was heated at 120° C. for 5 h. Solvent was evaporated under reduced pressure, the residue dissolved in DCM and treated with a 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 0:100 Cy/EtOAc) to obtain 0.023 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.98 min
  • MS: m/z=384 (M+H)+
  • Example 35a
  • Figure US20140045856A1-20140213-C00343
  • To a suspension of Intermediate 9a (0.08 g, 0.23 mmol) in dry DMF (4 mL), t-BuOK (0.076 g, 0.68 mmol) and Intermediate 21 (0.09 g, 0.45 mmol) were added and the reaction mixture was heated at 90° C. for 5 h. Dry DMF was then added and mixture heated at 110° C. overnight. Solvent was evaporated under reduced pressure, the residue dissolved in DCM and treated with a 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by flash chromatography (eluent from 80:20 to 0:100 Cy/EtOAc) to obtain 0.055 g of the protected intermediate 1-{2-chloro-5-[(S)-2-(tetrahydro-pyran-2-yloxy)-propoxy]-phenyl}-8-cyclopropyl-4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene.
  • The compound (0.055 g, 0.11 mmol) was then dissolved in dry MeOH (1 mL) and p-toluenesulfonic acid monohydrate (0.012 g, 0.06 mmol) was added. Mixture was stirred at room temperature for 1 h, solvent evaporated under reduced pressure and the residue purified by reverse phase flash chromatography (eluent ACN/H2O from 10:100 to 100:0) and then by silica flash chromatography (eluent DCM/EtOH 90:10) to obtain 0.03 g of the title compound.
  • HPLC-MS (Method 9): Rt=2.69 min
  • MS: m/z=384 (M+H)+
  • Example 36
  • Figure US20140045856A1-20140213-C00344
  • To a solution of Example 3 (0.07 g, 0.24 mmol) in dry DMF (3 mL), cesium carbonate (0.23 g, 0.71 mmol) and commercially available 3-chloro-1-propanol (0.024 g, 0.26 mmol) were added and the mixture heated at 100° C. for 3 h. Solvent was evaporated under reduced pressure, the residue dissolved with DCM and washed with 1N solution of NaOH. Phases were separated, dried over sodium sulphate and the crude purified by prep-HPLC (Method 11). Fractions containing the pure compound were combined and evaporated to reduced volume, diluted with DCM and washed with a saturated solution of sodium carbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.02 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.71 min
  • MS: m/z=350 (M+H)+
  • Example 37
  • Figure US20140045856A1-20140213-C00345
  • Example 37 was prepared as described for Example 32 starting from Example 1 (0.07 g, 0.22 mmol) and Intermediate 26 (0.22 g, 067 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.046 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.15 min
  • MS: m/z=396 (M+H)+
  • Example 38
  • Figure US20140045856A1-20140213-C00346
  • Example 38 was prepared as described for Example 32 starting from Example 1 (0.07 g, 0.22 mmol) and Intermediate 27 (0.127 g, 044 mmol). The residue was purified by flash chromatography (eluent from 100:0 to 50:50 Cy/EtOAc) to obtain 0.045 g of the title compound.
  • HPLC-MS (Method 13): Rt=3.45 min
  • MS: m/z=424 (M+H)+
  • Example 39
  • Figure US20140045856A1-20140213-C00347
  • To a solution of Intermediate 30 (0.18 g, 0.44 mmol) in absolute EtOH (10 mL) sodiumborohydride (0.008 g, 0.22 mmol) was added and the mixture stirred at room temperature for 30 min. Solvent was evaporated under reduced pressure, the residue treated with DCM and washed with a saturated solution of sodiumcarbonate. Phases were separated, dried over sodium sulphate and evaporated under reduced pressure to obtain 0.047 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.96 min
  • MS: m/z=410 (M+H)+
  • Example 40
  • Figure US20140045856A1-20140213-C00348
  • Example 40 was prepared as described Example 14, starting from Example 1 (0.075 g, 0.24 mmol) and Intermediate 31 (0.12 g, 0.48 mmol). Crude was purified by flash chromatography (eluent from 100:0 to 95:5 DCM/EtOH) to obtain 0.046 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.87 min
  • MS: m/z=396 (M+H)+
  • Example 41
  • Figure US20140045856A1-20140213-C00349
  • Example 41 (0.05 g) was prepared as described for Example 22 and purified applying the most suitable purification technique, starting from Example 2 (0.1 g, 0.3 mmol).
  • HPLC-MS (Method 13): Rt=3.21 min
  • MS: m/z=396 (M+H)+
  • Example 42
  • Figure US20140045856A1-20140213-C00350
  • Example 42 (0.06 g) was prepared as described for Example 23 and purified applying the most suitable purification technique, starting from Example 2 (0.09 g, 0.28 mmol).
  • (HPLC-MS (Method 13): Rt=3.21 min
  • MS: m/z=396 (M+H)+
  • Example 43
  • Figure US20140045856A1-20140213-C00351
  • Example 43 (0.03 g) was prepared as described for Example 24 and purified applying the most suitable purification technique, starting from Example 2 (0.09 g, 0.28 mmol).
  • HPLC-MS (Method 13): Rt=3.21 min
  • MS: m/z=396 (M+H)+
  • Example 43a
  • Figure US20140045856A1-20140213-C00352
  • To a solution of ethylmagnesium bromide (0.7 mL of a 3M solution in diethyl ether) in dry DCM (2 mL) at −75° C., titanium(IV) isopropoxide (0.2 mL, 0.7 mmol) was added. Mixture was stirred for 10 min, then a solution of Intermediate 31 (0.28 g, 0.73 mmol) in dry DCM (8 mL) was added dropwise. Mixture was allowed to reach room temperature and stirred at room temperature overnight. Mixture was diluted with EtOAc, hydrochloric acid (1 N solution) was added and phases separated. Organics were dried over sodium sulphate and evaporated under reduced pressure and the crude purified by silica flash chromatography (eluent AcOEt/EtOH 90:10) followed by a reverse phase chromatography to obtain 0.006 g of the title compound.
  • HPLC-MS (Method 13): Rt=2.93 min
  • MS: m/z=382 (M+H)+
  • Example 44
  • Figure US20140045856A1-20140213-C00353
  • Example 1 (0.07 mg, 0.2 mmol) was dissolved in DMF (3 mL), isobutylene oxide (0.03 mg, 0.43 mmol) and cesium carbonate (208 mg, 0.64 mmol) were added and the reaction mixture was warmed to 100° C. for 2 h. The solvent was removed, dichloromethane was added and the organic phase was washed with a 1M NaOH water solution, dried over sodium sulfate, concentrated under vacuum. The crude product obtained was purified by reverse phase chromatography to give the desired compound (29 mg).
  • HPLC-MS (Method 13): Rt=2.93 min
  • MS: m/z=386 (M+H)+
  • The following Examples were prepared in analogy to Example 44 and purified applying the most suitable purification technique, starting from the corresponding Starting Example and the suitable epoxides.
  • MS
    Starting Example or m/z Rt
    Starting Intermediate Starting Epoxide Ex. # Structure [M + H]+ (min) Method
     9
    Figure US20140045856A1-20140213-C00354
    45
    Figure US20140045856A1-20140213-C00355
    424 2.88 9
     2
    Figure US20140045856A1-20140213-C00356
    46
    Figure US20140045856A1-20140213-C00357
    398 3.01 9
    5
    Figure US20140045856A1-20140213-C00358
    47
    Figure US20140045856A1-20140213-C00359
    408 3.19 13
     6
    Figure US20140045856A1-20140213-C00360
    48
    Figure US20140045856A1-20140213-C00361
    404 3.19 13
     1 29a 49
    Figure US20140045856A1-20140213-C00362
    396 3.04 13
    10 29a 50
    Figure US20140045856A1-20140213-C00363
    376 2.88 13
     7
    Figure US20140045856A1-20140213-C00364
    51
    Figure US20140045856A1-20140213-C00365
    406 3.32 13
     8
    Figure US20140045856A1-20140213-C00366
    52
    Figure US20140045856A1-20140213-C00367
    410 3.32 13
     4 29a 53
    Figure US20140045856A1-20140213-C00368
    380 2.89 13
    30p
    Figure US20140045856A1-20140213-C00369
    54
    Figure US20140045856A1-20140213-C00370
    426 3.56 13
     1
    Figure US20140045856A1-20140213-C00371
    55 TRANS- racemic mixture
    Figure US20140045856A1-20140213-C00372
    398 2.74 13
     9
    Figure US20140045856A1-20140213-C00373
    56 TRANS- racemic mixture
    Figure US20140045856A1-20140213-C00374
    438 3.10 13
     6 57 TRANS- racemic mixture
    Figure US20140045856A1-20140213-C00375
    418 2.94 13
  • The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding racemic mixture.
  • Starting MS Rt (min) Rt (min)
    Racemic m/z [LC-MS [Chiral HPLC
    Mixture Ex. # Structure [M + H]+ Method] Method]
    55 TRANS- racemic mixture 55a TRANS- single stereoisomer a
    Figure US20140045856A1-20140213-C00376
    398 2.63 [13] 8.23 [C1]
    56 TRANS- racemic mixture 56a TRANS- single stereoisomer a
    Figure US20140045856A1-20140213-C00377
    438 3.03 [13] 12.13 [C2]
    56 TRANS- racemic mixture 56b TRANS- single stereoisomer b
    Figure US20140045856A1-20140213-C00378
    438 3.03 [13] 16.56 [C2]
    57 TRANS- racemic mixture 57a TRANS- single stereoisomer a
    Figure US20140045856A1-20140213-C00379
    418 2.18 [13] 17.66 [C2]
    57 TRANS- racemic mixture 57b TRANS- single stereoisomer b
    Figure US20140045856A1-20140213-C00380
    418 2.81 [13] 19.81 [C2]
  • Example 58 TRANS—Single Stereoisomer a
  • Figure US20140045856A1-20140213-C00381
  • Example 58 (0.01 g) was prepared in analogy to example 44 starting from Example 1 (0.2 g, 0.61 mmol) and epoxide intermediate 29 (0.07 g, 0.73 mmol) after chiral
  • HPLC separation HPLC-MS (Method 13): Rt=2.63 min
  • MS: m/z=412 (M+H)+
  • Chiral HPLC (Method C2): Rt=16.84 min
  • Example 58a CIS—Racemic Mixture
  • Figure US20140045856A1-20140213-C00382
  • To a solution of Intermediate 300 (0.13 g, 0.37 mmol) in dry DMF (3 mL), potassium tert.butoxide (0.083 g, 0.73 mmol) and commercially available 1,4-anhydroerythritol (0.06 mL, 0.73 mmol) were added and mixture heated at 90 C for 6 h. Solvent was evaporated, DCM and water added. Organics were separated, washed with a saturated solution of ammonium chloride, evaporated. The crude was purified by flash chromatography (eluent Cy/EtOAc 90:10 to 0:100) to give the desired compound (0.055 g).
  • HPLC-MS (Method 13): Rt=2.94 min
  • MS: m/z=438 (M+H)+
  • Example 58b CIS—Single Stereoisomer a
  • Figure US20140045856A1-20140213-C00383
  • Example 58b was obtained by chiral HPLC separation of Example 58. HPLC-MS (Method 13): Rt=2.95 min
  • MS: m/z=438 (M+H)+
  • Chiral HPLC (Method C1): Rt=7.65 min
  • Example 58c CIS—Single Stereoisomer b
  • Figure US20140045856A1-20140213-C00384
  • Further elution from the chiral column in the purification of Example 58 gave Example 58c
  • HPLC-MS (Method 13): Rt=2.95 min
  • MS: m/z=438 (M+H)+
  • Chiral HPLC (Method C1): Rt=9.54 min
  • Example 59 and Example 60
  • Figure US20140045856A1-20140213-C00385
  • Step 1:
  • Scaffold Intermediate 30a (1.4 g, 3.5 mmol) was suspended in THF (40 mL) and stirred at −78° C. under nitrogen atmosphere. N-Butyllithium (3 mL of a 2.5M solution in hexane) was added dropwise and the reaction mixture was stirred at −78° C. for 0.5 h. A solution of N,N.dimethylformamide (1.43 mL, 17.6 mmol) in THF (15 mL) was added dropwise and the reaction mixture was stirred at −78° C. for 30 minutes. Ammonium chloride saturated water solution (20 mL) was added and the reaction mixture was allowed to reach room temperature. The reaction mixture was diluted with ethyl acetate and washed with water. The organic phase was washed with brine, dried over sodium sulfate and concentrated under vacuum. The crude product obtained was purified by flash chromatography (eluent from 100:0 to 80:20 Cy/EtOAc) to obtain the title compound (0.3 g).
  • Step 2:
  • Intermediate obtained from Step 1 (0.07 g, 0.21 mmol) was suspended in THF (2 mL) and stirred at 0° C. A 2M solution of isopropylmagnesium chloride in THF (0.1 mL) was added an the reaction mixture was allowed to reach room temperature in 2 h. Ammonium chloride saturated water solution (20 mL) was added and the reaction mixture was allowed to reach room temperature. The reaction mixture was diluted with ethyl acetate and washed with water. The organic phase was washed with brine, dried over sodium sulfate and concentrated under vacuum. The crude product obtained was purified by flash chromatography (eluent 50:50 Cy/EtOAc) to obtain Example 60 (0.02 g).
  • HPLC-MS (Method 9): Rt=2.85 min
  • MS: m/z=382 (M+H)+
  • Further elution from the column (eluent from 50:50 to 0:100 Cy/EtOAc) gave Example 60 (0.03 g)
  • HPLC-MS (Method 9): Rt=2.28 min
  • MS: m/z=340 (M+H)+
  • The following examples were prepared in analogy to Example 59 and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30a and the corresponding commercially available Grignard reagents.
  • MS m/z Rt
    Grignard reagent Ex. # Structure (M + H)+ (min) Method
    Figure US20140045856A1-20140213-C00386
    61
    Figure US20140045856A1-20140213-C00387
    382 2.89  9
    Figure US20140045856A1-20140213-C00388
    62
    Figure US20140045856A1-20140213-C00389
    368 3.12 13
    —Mg—Cl 63
    Figure US20140045856A1-20140213-C00390
    354 2.93 13
  • Example 64
  • Figure US20140045856A1-20140213-C00391
  • Example 64 (0.03 g) was prepared in analogy to Example 1 and purified applying the most suitable purification technique, starting from Intermediate 2 (0.1 g, 0.55 mmol) and Hydrazide Intermediate 6i (0.15 g, 0.55 mmol).
  • HPLC-MS (Method 9): Rt=2.52 min
  • MS: m/z=382 (M+H)+
  • Example 65
  • Figure US20140045856A1-20140213-C00392
  • Example 65 (0.03 mg) was prepared in analogy to the preparation of Intermediate 5i and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30d (0.1 g, 0.25 mmol).
  • HPLC-MS (Method 9): Rt=2.45 min
  • MS: m/z=366 (M+H)+
  • The following examples were prepared in analogy to Example 65 and purified applying the most suitable purification technique, starting from the corresponding Scaffold Intermediates.
  • Starting MS m/z Rt
    Intermediate Ex. # Structure (M + H)+ (min) Method
    30e 66
    Figure US20140045856A1-20140213-C00393
    406 3.29 13
    30f 67
    Figure US20140045856A1-20140213-C00394
    362 3.83 16
    30g 68
    Figure US20140045856A1-20140213-C00395
    402 3.22 13
    30h 69
    Figure US20140045856A1-20140213-C00396
    422 3.33 13
  • Example 70
  • Figure US20140045856A1-20140213-C00397
  • Example 70 (0.03 g) was prepared in analogy to the preparation of Intermediate 5j and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30b (0.16 g, 0.38 mmol) and acetone (0.07 mg, 1.13 mmol).
  • HPLC-MS (Method 13): Rt=2.67 min
  • MS: m/z=354 (M+H)+
  • The following examples were prepared in analogy to Example 70 and purified applying the most suitable purification technique, starting from the corresponding Scaffold Intermediates and commercially available ketons.
  • Starting MS m/z Rt
    Intermediate Ketone Ex. # Structure (M + H)+ (min) Method
    30j
    Figure US20140045856A1-20140213-C00398
    71
    Figure US20140045856A1-20140213-C00399
    368 2.95 13
    30m
    Figure US20140045856A1-20140213-C00400
    72
    Figure US20140045856A1-20140213-C00401
    392 3.32 13
    30c
    Figure US20140045856A1-20140213-C00402
    73
    Figure US20140045856A1-20140213-C00403
    346 2.77 13
    30b
    Figure US20140045856A1-20140213-C00404
    74
    Figure US20140045856A1-20140213-C00405
    382 2.61 13
  • Example 75
  • Figure US20140045856A1-20140213-C00406
  • Step 1:
  • Under N2 atmosphere, to a solution of Scaffold Intermediate 30 g (1.0 g, 2.54 mmol) in toluene (35 mL), tributyl-(1-ethoxyvinyl-)tin (1.3 mL, 3.04 mmol) and Pd tetrakis(triphenylphosphine) (580 mg, 0.51 mmol). The solution was stirred at 105° C. for 18 h. The reaction mixture was cooled at room temperature, concentrated in vacuo and the crude product obtained was dissolved in THF. A 2N water solution of HCl 2N (5 ml) was added and the reaction mixture was stirred for 30 mins. After dilution with ethyl acetate, phases were separated and organic layer was dried over sodium sulfate and concentrated. The crude product obtained was purified by flash chromatography (eluent from 90:10 to 75:25 Cy/EtOAc) to obtain the desired compound (0.8 g).
  • HPLC-MS (Method 13): Rt=3.09 min
  • MS: m/z=358 (M+H)+
  • Step 2:
  • To a solution of the intermediate from Step 1 (0.8 g, 2.2 mmol) in DCM (20 mL) 50% NaOH/H2O solution (0.8 mL) was added and the reaction mixture was stirred for 10 minutes. Trimethylsulfonium methyl sulfate (0.4 g, 2.8 mmol) was added and the reaction mixture was refluxed for 2 h. The reaction mixture was diluted with DCM and washed with water. The organic layer was separated, dried over sodium sulfate and concentrated. The crude product obtained was purified by flash chromatography (eluent 85:15 Cy/EtOAc) to obtain the desired compound (0.5 g).
  • HPLC-MS (Method 13): Rt=3.33 min
  • MS: m/z=372 (M+H)+
  • Step 3:
  • To a solution of lithiumaluminiumhydride (0.15 mL of a 2M solution in THF) in 15 mL of THF stirred at 0° C., the intermediate from step 2 (0.4 g, 0.4 mmol) was added slowly. The reaction mixture was stirred for 1 h and allowe to reach room temperature. Water (2 mL) and NaOH (1 mL of a 2M water solution) were added and the reaction mixture was diluted with ethyl acetate. Organic phase was separated, dried over sodium sulfate and concentrated to give the desired product (0.28 g).
  • HPLC-MS (Method 13): Rt=3.0 min
  • MS: m/z=376 (M+H)+
  • Step 4:
  • Intermediate from step 3 (0.17 g, 0.38 mmol) was dissolved in DCM (10 mL), manganese dioxide (0.38 g, 3.38 mmol) was added and the suspension was stirred for 1 h at room temperature. The reaction mixture was filtered on a celite pad and the solvent was evaporated. The crude product obtained was purified by flash chromatography (eluent 90:10 DCM/MeOH) to obtain the desired compound (0.14 M.
  • HPLC-MS (Method 13): Rt=3.04 min
  • MS: m/z=374 (M+H)+
  • The following examples were prepared in analogy to Example 75 and purified applying the most suitable purification technique, starting from the corresponding Intermediates.
  • Starting MS m/z Rt
    Intermediate Ex. Structure (M + H)+ (min) Method
    30e 76
    Figure US20140045856A1-20140213-C00407
    378 3.12 13
    30k 77
    Figure US20140045856A1-20140213-C00408
    396 3.37 13
    30l 78
    Figure US20140045856A1-20140213-C00409
    376 3.3  13
    30n 79
    Figure US20140045856A1-20140213-C00410
    376 3.31 13
  • Example 80
  • Figure US20140045856A1-20140213-C00411
  • Example 80 (0.02 g) was prepared in step 2, step 3 and step 4 step in analogy to Example 75 and purified applying the most suitable purification technique, starting from the Intermediate obtained in step 1.
  • Step 1 was performed as follow:
  • Scaffold Intermediate 30i (1.35 g, 3.39 mmol) tri-n-butylmethoxytin (1.51 mL, 5.08 mmol), isopropenyl acetate (5.6 mL, 51 mmol), tri-o-tolylphosphine (0.07 g, 0.25 mmol) and palladium acetate (0.03 g) were suspended in toluene (30 mL) under nitrogen atmosphere and stirred for 2 h at room temperature, then at 110° C. for 20 minutes. Water (10 mL) and a bicarbonate saturated water solution (3 mL) were added, toluene was removed under vacuum and DCM was added. The organic phase was separated, and concentrated. The crude product obtained was purified by flash chromatography (eluent from 30:70 to 0:100 cyclohexane/AcOEt) to obtain the desired compound (0.5 g).
  • Step 2, Step 3 and Step 4 were performed in analogy to Example 75.
  • HPLC-MS (Method 16): Rt=3.13 min
  • MS: m/z=392 (M+H)+
  • The following example was prepared in analogy to Example 80 and purified applying the most suitable purification technique, starting from the corresponding Scaffold Intermediates.
  • MS m/z Rt
    Intermediate Ex. # Structure (M + H)+ (min) Method
    30g 81
    Figure US20140045856A1-20140213-C00412
    388 5.75 17
  • Example 82
  • Figure US20140045856A1-20140213-C00413
  • Example 82 (0.09 g) was prepared in analogy to Example 1 starting from Chloride Intermediate 2 (0.1 g, 0.61 mmol) and Hydrazide Intermediate 6p (0.2 g, 0.61 mmol).
  • (HPLC-MS (Method 13): Rt 2.80=min
  • MS: m/z=368 (M+H)+
  • The following examples were prepared in analogy to Example 82 and purified applying the most suitable purification technique, starting from the corresponding Chloride and Hydrazide Intermediates.
  • Starting Starting
    Chloride Hydrazide MS m/z Rt
    Intermediate Intermediate Ex. # Structure (M + H)+ (min) Method
    9a 6p 83 
    Figure US20140045856A1-20140213-C00414
    408 3.24 13
    9a 6j 84 
    Figure US20140045856A1-20140213-C00415
    390 3.18 13
    9a 6o 85 
    Figure US20140045856A1-20140213-C00416
    394 3.22 13
    2  6j 86 
    Figure US20140045856A1-20140213-C00417
    350 2.76 13
    2  6k 87 
    Figure US20140045856A1-20140213-C00418
    366 2.93 13
    9a 6k 88 
    Figure US20140045856A1-20140213-C00419
    406 3.33 13
    9  6k 89 
    Figure US20140045856A1-20140213-C00420
    380 3.05 13
    9  6j 90 
    Figure US20140045856A1-20140213-C00421
    364 2.91 13
    9b 6k 91 
    Figure US20140045856A1-20140213-C00422
    408 3.45 13
    9b 6j 92 
    Figure US20140045856A1-20140213-C00423
    392 3.34 13
    2  6l 93 
    Figure US20140045856A1-20140213-C00424
    396 2.96 13
    9a 6l 94 
    Figure US20140045856A1-20140213-C00425
    436 3.08 13
    9a 6n 95 
    Figure US20140045856A1-20140213-C00426
    420 2.9  13
    9f 6l 96 
    Figure US20140045856A1-20140213-C00427
    464 3.04 13
    9f 6n 96a
    Figure US20140045856A1-20140213-C00428
    448 2.98 13
    9f 6na 96b
    Figure US20140045856A1-20140213-C00429
    444 2.94 13
    9f 6r 96c
    Figure US20140045856A1-20140213-C00430
    434 2.91 13
    9a 6q 97 
    Figure US20140045856A1-20140213-C00431
    422 2.90 13
  • The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding racemic mixture.
  • Rt (min)
    Starting Rt (min) [Chiral
    Racemic MS m/z [LC-MS HPLC
    Mixture Ex. # Structure [M + H]+ Method] Method]
    97 Racemic mixture 97a Single stereo- isomer b
    Figure US20140045856A1-20140213-C00432
    422 2.89 [13] 15.75 [C2a]
    97 Racemic mixture 97b Single stereo- isomer a
    Figure US20140045856A1-20140213-C00433
    422 2.89 [13] 13.32 [C2a]
  • Example 98 Single Stereoisomer a
  • Figure US20140045856A1-20140213-C00434
  • Example 98 (0.026 g) was prepared in analogy to Example 1 starting from Chloride Intermediate 2 (0.3 g, 1.45 mmol) and Hydrazide Intermediate 6q (0.4 g, 1.56 mmol) after semipreparative chiral purification.
  • HPLC-MS (Method 13): Rt=2.56 min
  • MS: m/z=382 (M+H)+
  • Chiral HPLC (Method C1a): Rt=9.55 min
  • Example 99 Single Stereoisomer b
  • Figure US20140045856A1-20140213-C00435
  • Example 99 (0.028 g) was obtained via further elution from the column in the semipreparative chiral chromatographic purification of Example 98.
  • HPLC-MS (Method 13): Rt=2.52 min
  • MS: m/z=382 (M+H)+
  • Chiral HPLC (Method C1a): Rt=11.03 min
  • Example 100
  • Figure US20140045856A1-20140213-C00436
  • Step 1:
  • Scaffold Intermediate 30 g (0.5 g, 1.27 mmol), 1-methoxy-2-methyl-1-(trimethylsililoxy)propene (1.9 g, 11.4 mmol), bis(dibenzylideneacetone)palladium(0) (0.14 g, 0.25 mmol) and zinc fluoride (0.19 g, 1.9 mmol) were suspended in DMF (25 mL) and stirred under nitrogen atmosphere for 10 minutes. Tri-tert-butyl-phosphine (0.06 mL, 0.25 mmol) was added and the reaction mixture was stirred at 115° C. for 18 h. The reaction mixture was concentrated, an ammonium chloride water saturated solution was added and the reaction mixture was extracted with DCM. The organic phase was separated, dried over sodium sulfate and concentrated. The crude product obtained was purified by flash chromatography (eluent from 100:0 to 50:50 cyclohexane/AcOEt) to obtain the desired compound (0.4 g).
  • Step 2:
  • The intermediate obtained in Step 1 (0.4 g, 0.07 mmol) was dissolved in THF (6 mL) and stirred under nitrogen atmosphere at 0° C. for 5 minutes. Lithiumaluminium hydride (0.5 mL of a 2M solution in THF) was added. The reaction mixture was stirred at 0° C. for 10 minutes then allowed to reach room temperature and stirred for 0.5 h.
  • The solvent was removed, the crude product was dissolved in DCM and the reaction mixture was washed with water (4 mL), bicarbonate saturated water solution (2 mL) and brine (2 mL). The organic phase was separated, dried over sodium sulfate and manganese dioxide (0.15 g, 1.73 mmol) was added. The reaction mixture was stirred for 0.5 h, filtered on a celite pad and concentrated. The crude product obtained was purified by flash chromatography (eluent from 100:0 to 95:5 DCM/MeOH) to obtain the desired compound (0.1 g).
  • (HPLC-MS (Method 13): Rt=3.29 min
  • MS: m/z=388 (M+H)+
  • Example 101
  • Figure US20140045856A1-20140213-C00437
  • Example 101 (0.05 g) was prepared in analogy to Example 100 and purified applying the most suitable purification technique, starting from Scaffold Intermediate 30i (0.36 g, 0.9 mmol).
  • HPLC-MS (Method): Rt=3.3 min
  • MS: m/z=392 (M+H)+
  • Example 102
  • Figure US20140045856A1-20140213-C00438
  • Step 1:
  • Step 1 was performed in analogy to preparation of Example 1 starting from Chloride Intermediate 9 (0.3 g, 1.39 mmol) and Hydrazide Intermediate 6o (0.47 g, 1.39 mmol).
  • Step 2:
  • Intermediate from step 1 (0.09 g, 0.26 mmol) and hydrochloric acid (10 mL of a 37% water solution) were dissolved in 10 mL of 1,4-dioxane. The reaction mixture was stirred at room temperature for 20 minutes. A bicarbonate saturated water solution was added, THF was removed in vacuum and the reaction mixture was extracted with DCM. The organic phase was separated, dried over sodium sulfate and concentrated. The crude product obtained was purified by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH) to obtain the title compound (0.03 g).
  • HPLC-MS (Method 13): Rt=2.95 min
  • MS: m/z=360 (M+H)+
  • Example 103
  • Figure US20140045856A1-20140213-C00439
  • Step 1:
  • Under nitrogen atmosphere, Scaffold Intermediate 30 g (0.3 g, 0.61 mmol), potassium carbonate (0.34 g, 2.4 mmol) and [1,1′Bis(diphenylphosphino)ferrocene]clichloropalladium(II) (0.05 g) were suspended in toluene (150 mL) and stirred at room temperature for 10 minutes. 3,6-dihydro-2H-pyran-4-boronic acid pinacol ester (0.17 g, 0.79 mmol) was added and the reaction mixture was stirres at 100° C. for 4 h. Water (30 mL) was added and the reaction mixture was extracted with AcOEt. The organic phase was separated and concentrated. The crude product obtained was purified by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH) to obtain the title compound (0.02 g).
  • Step 2:
  • Step 2 was performed in analogy to Step 2 in the preparation of Example 102. 0.02 g of the desired product were obtained after purification by flash chromatography (eluent from 100:0 to 90:10 DCM/MeOH).
  • (HPLC-MS (Method 13): Rt=2.88 min
  • MS: m/z=416 (M+H)+
  • The following examples were prepared in analogy to Example 103 and purified applying the most suitable purification technique, starting from the corresponding Scaffold Intermediate and the boronic ester 2-Isopropenyl-4,4,5,5-tetramethyl-[1,3,2]dioxaborolane.
  • Starting MS m/z Rt
    Intermediate Ex. # Structure (M + H)+ (min) Method
    30r 103b
    Figure US20140045856A1-20140213-C00440
    422 3.25 13
    30s 103c
    Figure US20140045856A1-20140213-C00441
    406 3.17 13
  • Example 104 CIS-Racemic Mixture
  • Figure US20140045856A1-20140213-C00442
  • Intermediate obtained from Step 1 in preparation of Example 103 (50 mg, 0.13 mmol) was dissolved in DCM (5 mL). Osmium tetroxide (0.1 mL, 0.01 mmol) and 4-methyl-morpholine-4-oxide (0.07 g, 0.63 mmol) were added and the reaction mixture was stirred at room temperature for 5 h. The solvent was removed and the crude product obtained was purified by flash chromatography (eluent from 100:0 to 80.20 DCM/MeOH) to obtain the title compound (0.03 g).
  • HPLC-MS (Method 13): Rt=2.73 min
  • MS: m/z=432 (M+H)+
  • Example 104a CIS-Single Stereoisomer a
  • Figure US20140045856A1-20140213-C00443
  • Example 104a was obtained via Chiral HPLC purification of Example 104.
  • HPLC-MS (Method 13): Rt=2.65 min
  • MS: m/z=432 (M+H)+
  • Chiral HPLC (Method C2b): Rt=18.70 min
  • Example 104b CIS-Single Stereoisomer b
  • Figure US20140045856A1-20140213-C00444
  • Further elution from the chiral column gave Example 104b.
  • HPLC-MS (Method 13): Rt=2.65 min
  • MS: m/z=432 (M+H)+
  • Chiral HPLC (Method C2b): Rt=21.04 min
  • Example 105 CIS-Racemic Mixture
  • Figure US20140045856A1-20140213-C00445
  • Example 105 (0.03 g) was prepared in analogy to Example 104, starting from 8-Cyclopropyl-1-[5-(3,6-dihydro-2H-pyran-4-yl)-2-fluoro-phenyl]-4-methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalene (0.05 g, 0.12 mmol) which was obtained as by-product in the preparation of Example 95 and isolated from the same flash chromatography purification.
  • HPLC-MS (Method 13): Rt=2.68 min
  • MS: m/z=436 (M+H)+
  • The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding racemic mixture.
  • Rt (min)
    Starting Rt (min) [Chiral
    Racemic MS m/z [LC-MS HPLC
    Mixture Ex. # Structure [M + H]+ Method] Method]
    105 CIS- Racemic mixture 105a CIS- Single stereo- isomer a
    Figure US20140045856A1-20140213-C00446
    436 2.69 [13] 15.05 [C2b]
     97 CIS- Racemic mixture 105b CIS- Single stereo- isomer b
    Figure US20140045856A1-20140213-C00447
    436 2.68 [13] 18.47 [C2b]
  • The following Examples were prepared in analogy to Example 44 and purified applying the most suitable purification technique, starting from the corresponding Starting Example and the corresponding epoxides.
  • Starting Example or Starting MS m/z Rt
    Starting Intermediate Epoxide Ex. # Structure (M + H)+ (min) Method
    Intermediate 32
    Figure US20140045856A1-20140213-C00448
    106 TRANS- Racemic mixture
    Figure US20140045856A1-20140213-C00449
    450 2.97 13
     5
    Figure US20140045856A1-20140213-C00450
    107 TRANS- Racemic mixture
    Figure US20140045856A1-20140213-C00451
    422 3.88 16
    12
    Figure US20140045856A1-20140213-C00452
    111 TRANS- Racemic mixture
    Figure US20140045856A1-20140213-C00453
    466 3.09 13
  • The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding racemic mixture.
  • Rt (min)
    Starting Rt (min) [Chiral
    Racemic MS m/z [LC-MS HPLC
    Mixture Ex. # Structure [M + H]+ Method] Method]
    106 TRANS- Racemic mixture 106a TRANS- single stereo- isomer a
    Figure US20140045856A1-20140213-C00454
    450 2.95 [13]  7.77 [C2c]
    106 TRANS- Racemic mixture 106b TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00455
    450 2.98 [13] 11.58 [C2c]
  • The following examples were obtained as single stereoisomers by chiral HPLC separation of the corresponding TRANS regioisomeric racemic mixtures. TRANS regioisomeric racemic mixtures were obtained in analogy to preparation of Example 44, starting from epoxide Intermediate 29 and the corresponding Starting Examples or Starting Intermediates.
  • Rt (min)
    Rt (min) [Chiral
    Starting Example or MS m/z [LC-MS HPLC
    Starting Intermediate Ex. # Structure [M + H]+ Method] Method]
    5 108a TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00456
    436 2.91 [13] 31.54 [C3]
    5 108c TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00457
    436 2.91 [13] 44.59 [C2c]
    5 108b TRANS- single stereo- isomer a
    Figure US20140045856A1-20140213-C00458
    436 2.92 [13] 27.61 [C3]
    5 108d TRANS- single stereo- isomer a
    Figure US20140045856A1-20140213-C00459
    436 2.91 [13] 22.06 [C2c]
    9 109a TRANS- single stereo- isomer a
    Figure US20140045856A1-20140213-C00460
    452 3.03 [13] 23.62 [C2]
    9 109c TRANS- single stereo- isomer a
    Figure US20140045856A1-20140213-C00461
    452 3.03 [13] 40.91 [C2]
    9 109b TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00462
    452 3.03 [13] 89.29 [C2]
    9 109d TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00463
    452 3.03 [13] 25.61 [C2]
    Intermediate 32 110b TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00464
    464 3.00 [13] 21.99 [C2]
    Intermediate 32 110d TRANS- single stereo- isomer b
    Figure US20140045856A1-20140213-C00465
    464 3.02 [13] 62.95 [C2]

Claims (22)

1. A compound of formula I
Figure US20140045856A1-20140213-C00466
wherein
A is selected from the group Aa consisting of
Figure US20140045856A1-20140213-C00467
wherein above mentioned phenyl-, pyridinyl-, pyrimidinyl-, pyridazinyl and pyrazinyl groups are substituted with R1 and R2;
R1 is selected from the group R1a consisting of
H, halogen, NC—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— with n=0, 1, 2, 3 or 4,
wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl and R8—(CH2)n—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
R2 is selected from the group R2a consisting of
H, HO—, halogen, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2,
wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8— (CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-;
R3 is selected from the group R3a consisting of
H, halogen, NC—, C1-4-alkyl- and C3-6-cycloalkyl-,
wherein the above mentioned C1-4-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—;
R4, R5 are selected independently of each other from the group R4a/R5a consisting of
H, halogen, NC—, HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O—,
wherein above mentioned C1-6-alkyl-, C1-6-alkyl-O—, C3-8-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl-, C3-8-cycloalkyl-O—, heterocyclyl-O—, heterocyclyl, heteroaryl, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms, substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—;
R6 is selected from the group R6a consisting of
H, NC—, C1-6-alkyl-, Cm-cycloalkyl-, C3-8-cycloalkyl-C1-3-alkyl- and C3-8-cycloalkyl-O—
wherein above mentioned C1-6-alkyl- groups may optionally be substituted with 1-3 halogen atoms;
R7 is selected from the group R7a consisting of
H, carbocyclyl, heterocyclyl and heteroaryl,
wherein above mentioned carbocyclyl, heterocyclyl and heteroaryl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of HO—, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-, optionally with 1 to 3 halogen atoms substituted C1-4-alkyl-O— and halogen;
R8 is selected from the group R8a consisting of
C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl,
wherein above mentioned C3-6-cycloalkyl, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl and pyridyl groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-;
or a tautomer or salt thereof.
2. A compound according to claim 1, wherein
R2 is selected from the group R2a1 consisting of
HO—, C1-6-alkyl-, C1-6-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2, 3 or 4, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0, 1 or 2 and o=0, 1 or 2
wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups are substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
and
wherein above mentioned C1-6-alkyl- and C1-6-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
and
wherein above mentioned C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
3. A compound according to claim 1, wherein
R3 is selected from the group R3a1 consisting of
C3-6-cycloalkyl-,
wherein the above mentioned C3-6-cycloalkyl-group may optionally be substituted with 1 to 9 substituents independently selected from the group consisting of halogen, NC—, HO—, C1-3-alkyl- and C1-3-alkyl-O—.
4. A compound according to claim 1, wherein
A is selected from the group Ab consisting of
Figure US20140045856A1-20140213-C00468
wherein above mentioned phenyl- and pyridinyl- groups are substituted with R1 and R2.
5. A compound according to claim 1, wherein
A is selected from the group Ae consisting of
Figure US20140045856A1-20140213-C00469
6. A compound according to claim 1, wherein
R1 is selected from the group R1b consisting of
H, halogen, C1-6-alkyl- and C3-6-cycloalkyl
wherein above mentioned C1-6-alkyl- and C3-6-cycloalkyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
7. A compound according to claim 1, wherein
R1 is selected from the group R1c consisting of
H, H3C—, F3C—, F2HC—, FH2C—, fluorine, chlorine and bromine.
8. A compound according to claim 1, wherein
R2 is selected from the group R2b1 consisting of
HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, phenyl, heteroaryl, R8—(CH2)n—O— with n=0, 1, 2 or 3 and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O— groups are substituted with 1 to 3 substituents independently selected from the group consisting of HO— and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
and
wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
and
wherein above mentioned C3-6-cycloalkyl-, C3-6-cycloalkyl-C1-3-alkyl-, heterocyclyl, heterocyclyl-C1-3-alkyl-, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O-groups may optionally be substituted with 1 to 3 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
9. A compound according to claim 1, wherein
R2 is selected from the group R2d1 consisting of
HO—, C1-4-alkyl-, C1-4-alkyl-O—, C3-6-cycloalkyl, C3-6-cycloalkyl-C1-3-alkyl-, a saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— with n=0, 1 or 2, and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— with m=0 or 1 and o=0 or 1
wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups are substituted with 1 to 5 substituents independently selected from the group consisting of HO—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—,
and
wherein above mentioned C1-4-alkyl- and C1-4-alkyl-O-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-,
and
wherein above mentioned C3-6-cycloalkyl-, C3-6-cycloalkyl-C1-3-alkyl-, saturated 5 or 6 membered monocyclic heterocycle containing one heteroatom selected from N or O, R8—(CH2)n—O— and R8—(CH2)m—(CH)(CH3)—(CH2)o—O— groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, HO—, optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-O—, and optionally with 1 to 7 fluorine atoms substituted C1-3-alkyl-.
10. A compound according to claim 1, wherein
R3 is selected from the group R3b consisting of
H, C1-3-alkyl-, cyclobutyl- and cyclopropyl-,
wherein the above mentioned C1-3-alkyl-, cyclobutyl- and cyclopropyl-groups may optionally be substituted with 1 to 7 substituents independently selected from the group consisting of halogen, C1-3-alkyl-O—, NC— and HO—.
11. A compound according to claim 1, wherein
R3 is selected from the group R3c consisting of
H, and H3C— and cyclopropyl-,
wherein the above mentioned H3C— and cyclopropyl-groups may optionally be substituted with 1 to 3 fluorine atoms.
12. A compound according to claim 1, wherein
R4, R5 are selected independently of each other from the group R4b/R5b consisting of
H, halogen, HO—, H3C—, F3C—, H3C—O—, F2HC—O—, FH2C—O—, F3C—O—, C1-4-alkyl-O—, R7—CH2—O— and R7—(CH2)2—O—,
wherein above mentioned C1-4-alkyl-O—, R7—CH2—O— and R7—(CH2)2—O-groups, may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of halogen, optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-, and optionally with 1 to 5 halogen atoms substituted C1-2-alkyl-O—.
13. A compound according to claim 1, wherein
R4, R5 are selected from the group R4d/R5d consisting of H—.
14. A compound according to claim 1, wherein
R6 is selected from the group R6b consisting of
H, C1-4-alkyl- and cyclopropyl-,
wherein above mentioned C1-4-alkyl-group may optionally be substituted with 1-9 fluorine and/or chlorine atoms.
15. A compound according to claim 1, wherein
R6 is selected from the group R6c consisting of
H and C1-2-alkyl-,
wherein above mentioned C1-2-alkyl- group may optionally be substituted with 1-5 fluorine and/or chlorine atoms.
16. A compound according to claim 1, wherein
R6 is selected from the group R6e consisting of
H3C—, FH2C—, F2HC— and F3C—.
17. A compound according to claim 1, wherein
R7 is selected from the group R7b consisting of
H, phenyl, heteroaryl, cycloalkyl and heterocyclyl
wherein above mentioned phenyl, heteroaryl, cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of halogen, and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-O—.
18. A compound according to claim 1, wherein
R7 is selected from the group R7c consisting of
H and phenyl,
wherein above mentioned phenyl group may optionally be substituted with 1 to 4 substituents independently selected from the group consisting of halogen and optionally with 1 to 3 halogen atoms substituted C1-3-alkyl-O—.
19. A compound according to claim 1, wherein
R8 is selected from the group R8b consisting of
C3-6-cycloalkyl, heterocyclyl and heterocyclyl-C1-3-alkyl-,
wherein above mentioned C3-6-cycloalkyl, heterocyclyl and heterocyclyl-C1-3-alkyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-.
20. A compound according to claim 1, wherein
R8 is selected from the group R8b1 consisting of
C3-6-cycloalkyl and a saturated 4 to 6 membered monocyclic heterocycle containing one or two heteroatoms selected from N or O,
wherein above mentioned C3-6-cycloalkyl and heterocyclyl-groups may optionally be substituted with 1 to 5 substituents independently selected from the group consisting of HO—, fluorine and optionally with 1 to 7 halogen atoms substituted C1-3-alkyl-.
21. A pharmaceutical composition containing a compound according to claim 1 or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier.
22. A method for treating (1) disorders comprising the symptom of cognitive deficiency; (2) organic, including symptomatic, mental disorders, dementia; (3) mental retardation; (4) mood [affective] disorders; (5) neurotic, stress-related and somatoform disorders including anxiety disorders; (6) behavioural and emotional disorders with onset usually occurring in childhood and adolescence, attention deficit hyperactivity syndrome (ADHD) and Autism spectrum disorders; (7) disorders of psychological development, developmental disorders of scholastic skills; (8) schizophrenia and other psychotic disorders; (9) disorders of adult personality and behaviour; (10) mental and behavioural disorders due to psychoactive substance use; (11) extrapyramidal and movement disorders; (12) episodic and paroxysmal disorders, epilepsy; (13) Systemic atrophies primarily affecting the central nervous system, ataxia; (14) Behavioural syndromes associated with physiological disturbances and physical factors; (15) sexual dysfunction comprising excessive sexual drive; (16) factitious disorders; (17) Treatment, amelioration and/or prevention of cognitive impairment being related to perception, concentration, cognition, learning or memory; (18) Treatment amelioration and/or prevention of cognitive impairment being related to age-associated learning and memory impairments; (19) Age-associated memory losses; (20) Vascular dementia; (21) Craniocerebral trauma; (22) Stroke; (23) Dementia occurring after strokes (post stroke dementia); (24) Post-traumatic dementia; (25) General concentration impairments; (26) Concentration impairments in children with learning and memory problems; (27) Alzheimer's disease; (28) Lewy body dementia; (29) Dementia with degeneration of the frontal lobes; including Pick's syndrome; (30) Parkinson's disease; (31) Progressive nuclear palsy; (32) Dementia with corticobasal degeneration; (33) Amyotropic lateral sclerosis (ALS); (34) Huntington's disease; (35) Multiple sclerosis; (36) Thalamic degeneration; (37) Creutzfeld-Jacob dementia; (38) HIV dementia; (38) Schizophrenia with dementia or Korsakoff's psychosis; (39) Sleep disorders; (40) Bipolar disorder; (41) Metabolic syndrome; (42) Obesity; (43) Diabetis mellitus; (44) Hyperglycemia; (45) Dyslipidemia; (46) Impaired glucose tolerance; (47) Disease of the testes, brain, small intestine, skeletal muscle, heart, lung, thymus or spleen and (48) pain disorders, which method comprises administering a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, to a patient suffering from such condition.
US13/945,008 2012-07-31 2013-07-18 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes Abandoned US20140045856A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12178713 2012-07-31
EP12178713.9 2012-07-31

Publications (1)

Publication Number Publication Date
US20140045856A1 true US20140045856A1 (en) 2014-02-13

Family

ID=48906250

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/945,008 Abandoned US20140045856A1 (en) 2012-07-31 2013-07-18 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes
US13/951,531 Active US9085584B2 (en) 2012-07-31 2013-07-26 Substituted pyrido[3,2-E][1,2,4]-triazolo[4,3-A]pyrazines for the treatment of central nervous system disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/951,531 Active US9085584B2 (en) 2012-07-31 2013-07-26 Substituted pyrido[3,2-E][1,2,4]-triazolo[4,3-A]pyrazines for the treatment of central nervous system disorders

Country Status (28)

Country Link
US (2) US20140045856A1 (en)
EP (1) EP2882749B1 (en)
JP (1) JP6356671B2 (en)
KR (1) KR20150037950A (en)
CN (1) CN104428302B (en)
AP (1) AP2015008202A0 (en)
AR (1) AR091939A1 (en)
AU (1) AU2013298621B2 (en)
BR (1) BR112015002118A2 (en)
CA (1) CA2880447A1 (en)
CL (1) CL2015000096A1 (en)
CO (1) CO7310531A2 (en)
EA (1) EA028958B1 (en)
EC (1) ECSP15006871A (en)
GE (1) GEP201706707B (en)
HK (1) HK1208440A1 (en)
IL (1) IL236494A0 (en)
MA (1) MA37798B1 (en)
MX (1) MX2015000965A (en)
NZ (1) NZ703797A (en)
PE (1) PE20150706A1 (en)
PH (1) PH12015500211A1 (en)
SG (1) SG11201500772VA (en)
TN (1) TN2015000022A1 (en)
TW (1) TW201418255A (en)
UA (1) UA115561C2 (en)
UY (1) UY34955A (en)
WO (1) WO2014019979A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9540379B2 (en) 2011-01-31 2017-01-10 Boehringer Ingelheim International Gmbh (1,2,4)triazolo[4,3-A]quinoxaline derivatives as inhibitors of phosphodiesterases
US10479794B2 (en) 2015-10-13 2019-11-19 Boehringer Ingelheim International Gmbh Cyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013034761A1 (en) * 2011-09-09 2013-03-14 H. Lundbeck A/S Pyridine compounds and uses thereof
US20140045856A1 (en) * 2012-07-31 2014-02-13 Boehringer Ingelheim International Gmbh 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes
WO2016145614A1 (en) 2015-03-17 2016-09-22 Merck Sharp & Dohme Corp. Triazolyl pyrimidinone compounds as pde2 inhibitors
US10287269B2 (en) 2015-03-26 2019-05-14 Merck Sharp & Dohme Corp. Pyrazolyl pyrimidinone compounds as PDE2 inhibitors
EP3291817B1 (en) 2015-05-05 2019-10-02 Merck Sharp & Dohme Corp. Heteroaryl-pyrimidinone compounds as pde2 inhibitors
WO2016183741A1 (en) 2015-05-15 2016-11-24 Merck Sharp & Dohme Corp. Pyrimidinone amide compounds as pde2 inhibitors
WO2016184313A1 (en) * 2015-05-20 2016-11-24 南京明德新药研发股份有限公司 Hydroxyl purine compounds and use thereof
WO2016191935A1 (en) 2015-05-29 2016-12-08 Merck Sharp & Dohme Corp. 6-alkyl dihydropyrazolopyrimidinone compounds as pde2 inhibitors
WO2016192083A1 (en) 2015-06-04 2016-12-08 Merck Sharp & Dohme Corp. Dihydropyrazolopyrimidinone compounds as pde2 inhibitors
EP3313852B1 (en) 2015-06-25 2021-01-20 Merck Sharp & Dohme Corp. Substituted pyrazolo/imidazolo bicyclic compounds as pde2 inhibitors
WO2017000276A1 (en) 2015-07-01 2017-01-05 Merck Sharp & Dohme Corp. Bicyclic heterocyclic compounds as pde2 inhibitors
WO2017000277A1 (en) 2015-07-01 2017-01-05 Merck Sharp & Dohme Corp. Substituted triazolo bicycliccompounds as pde2 inhibitors
AU2017246334C1 (en) * 2016-04-04 2022-11-17 Sinopia Biosciences, Inc. Treating extrapyramidal syndrome using Trapidil
AR112457A1 (en) * 2017-08-02 2019-10-30 Lilly Co Eli DERIVATIVES OF [1,2,4] TRIAZOLO [4,3-A] PIRAZIN-6 (5H) -ONE
EP3713572A1 (en) 2017-11-23 2020-09-30 Oslo University Hospital HF Treatment of tachycardia

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000319278A (en) * 1999-05-11 2000-11-21 Ono Pharmaceut Co Ltd Condensed pyrazine compound and medicinal agent having the same as active ingredient
JP2000319277A (en) * 1999-05-11 2000-11-21 Ono Pharmaceut Co Ltd Condensed pyrazine compound and medicinal agent having the same as active ingredient
DE10108752A1 (en) 2001-02-23 2002-09-05 Bayer Ag New substituted imidazotriazinones
WO2004085439A1 (en) 2003-03-27 2004-10-07 Pfizer Products Inc. Substituted 4-amino[1,2,4]triazolo[4,3-a]quinoxalines
WO2005041957A1 (en) 2003-10-29 2005-05-12 Pfizer Products Inc. Oxindole derivatives and their use as phosphodiesterase type 2 inhibitors
US20070135457A1 (en) 2003-12-16 2007-06-14 Pfizer Inc. Pyrido[2,3-d]pyrimidine-2,4-diamines as pde2 inhibitors
EP1548011A1 (en) 2003-12-23 2005-06-29 Neuro3D Benzo[1,4]diazepin-2-one derivatives as phosphodiesterase PDE2 inhibitors, preparation and therapeutic use thereof
FR2865331A1 (en) 2004-01-21 2005-07-22 France Telecom Removable digital medium e.g. CD-ROM, stores single policy enforcement point identifier intended to be transmitted to policy decision point server in order to be stored in server corresponding to network address
FR2870539B1 (en) 2004-05-19 2006-08-04 Greenpharma Sa Sa NEW METHODS AND MEDICINES
US7851472B2 (en) 2004-09-02 2010-12-14 Nycomed Gmbh Triazolophthalazines
MX2007008137A (en) 2005-01-05 2007-07-19 Altana Pharma Ag Triazolophthalazines.
EP1874775B1 (en) 2005-01-05 2012-10-10 Nycomed GmbH Triazolophthalazines as pde2-inhibitors
BRPI0502411A (en) 2005-03-31 2006-11-28 Univ Minas Gerais process of developing substances as potent and selective inhibitors of phosphodiesterase isoforms of types 1 to 5 (pde1, pde2, pde3, pde4, pde5) based on diocleine, fluranol or analogs and their pharmaceutical compositions for the study and treatment of cardiovascular disease and associated products
WO2007087250A2 (en) 2006-01-23 2007-08-02 Amira Pharmaceuticals, Inc. Tricyclic inhibitors of 5-lipoxygenase
WO2007121319A2 (en) 2006-04-14 2007-10-25 University Of California Compostions and methods for determining and predicting treatment responses for depression and anxiety
TW200815436A (en) 2006-05-30 2008-04-01 Elbion Ag 4-amino-pyrido[3,2-e]pyrazines, their use as inhibitors of phosphodiesterase 10, and processes for preparing them
TW200817400A (en) 2006-05-30 2008-04-16 Elbion Ag Pyrido [3,2-e] pyrazines, their use as inhibitors of phosphodiesterase 10, and processes for preparing them
US20080090834A1 (en) 2006-07-06 2008-04-17 Pfizer Inc Selective azole pde10a inhibitor compounds
DE102006048693A1 (en) 2006-10-14 2008-04-17 Bayer Healthcare Ag Inhibition of PDE2A
ES2599054T3 (en) * 2006-12-13 2017-01-31 Aska Pharmaceutical Co., Ltd. Quinoxaline derivatives
WO2009070583A1 (en) * 2007-11-30 2009-06-04 Wyeth Pyrido[3,2-e]pyrazines, process for preparing the same, and their use as inhibitors of phosphodiesterase 10
TWI501965B (en) 2008-06-20 2015-10-01 Lundbeck & Co As H Novel phenylimidazole derivatives as pde10a enzyme inhibitors
AR073574A1 (en) 2008-09-10 2010-11-17 Alcon Res Ltd HETEROCICLIC INHIBITORS OF HISTAMINE RECEPTORS FOR THE TREATMENT OF A DISEASE
US8415386B2 (en) 2008-10-08 2013-04-09 Bristol-Myers Squibb Company Azolopyrrolone melanin concentrating hormone receptor-1 antagonists
US20100120762A1 (en) 2008-11-07 2010-05-13 Wyeth Triazine derivatives as inhibitors of phosphodiesterases
US8614221B2 (en) * 2009-03-11 2013-12-24 Merck Sharp & Dohme Corp. Inhibitors of Akt activity
EP2266985A1 (en) * 2009-06-26 2010-12-29 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Tricyclic Pyrimidine Derivatives as Wnt antagonists
CN103619841B (en) 2011-01-11 2017-03-29 桑诺维恩药品公司 Heteroaryl compound and its using method
US9540379B2 (en) 2011-01-31 2017-01-10 Boehringer Ingelheim International Gmbh (1,2,4)triazolo[4,3-A]quinoxaline derivatives as inhibitors of phosphodiesterases
MX344600B (en) 2011-06-27 2016-12-20 Janssen Pharmaceutica Nv 1-ARYL-4-METHYL-[1,2,4]TRIAZOLO[4,3-a]QUINOXALINE DERIVATIVES.
WO2013034761A1 (en) 2011-09-09 2013-03-14 H. Lundbeck A/S Pyridine compounds and uses thereof
WO2013034758A1 (en) 2011-09-09 2013-03-14 H. Lundbeck A/S Substituted triazolopyrazines and uses thereof
AR088082A1 (en) 2011-10-13 2014-05-07 Lilly Co Eli ANDROGEN RECEIVER SELECTIVE MODULATORS
US9669035B2 (en) 2012-06-26 2017-06-06 Janssen Pharmaceutica Nv Combinations comprising PDE 2 inhibitors such as 1-aryl-4-methyl-[1,2,4]triazolo-[4,3-A]]quinoxaline compounds and PDE 10 inhibitors for use in the treatment of neurological of metabolic disorders
US20140045856A1 (en) * 2012-07-31 2014-02-13 Boehringer Ingelheim International Gmbh 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9540379B2 (en) 2011-01-31 2017-01-10 Boehringer Ingelheim International Gmbh (1,2,4)triazolo[4,3-A]quinoxaline derivatives as inhibitors of phosphodiesterases
US10479794B2 (en) 2015-10-13 2019-11-19 Boehringer Ingelheim International Gmbh Cyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide
US10875867B2 (en) 2015-10-13 2020-12-29 Boehringer Ingelheim International Gmbh Cyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide
US11691977B2 (en) 2015-10-13 2023-07-04 Boehringer Ingelheim International Gmbh Cyclic ether derivatives of pyrazolo[1,5-A]pyrimidine-3-carboxyamide

Also Published As

Publication number Publication date
AU2013298621A1 (en) 2015-02-05
PH12015500211B1 (en) 2015-03-16
CA2880447A1 (en) 2014-02-06
SG11201500772VA (en) 2015-02-27
CL2015000096A1 (en) 2015-06-19
EA028958B1 (en) 2018-01-31
CN104428302A (en) 2015-03-18
US9085584B2 (en) 2015-07-21
HK1208440A1 (en) 2016-03-04
TN2015000022A1 (en) 2016-06-29
PE20150706A1 (en) 2015-06-03
EA201500182A1 (en) 2015-07-30
US20140045857A1 (en) 2014-02-13
PH12015500211A1 (en) 2015-03-16
KR20150037950A (en) 2015-04-08
ECSP15006871A (en) 2015-12-31
NZ703797A (en) 2017-09-29
MX2015000965A (en) 2015-04-10
TW201418255A (en) 2014-05-16
AU2013298621B2 (en) 2017-12-21
JP2015523406A (en) 2015-08-13
CO7310531A2 (en) 2015-06-30
UY34955A (en) 2014-01-31
MA37798B1 (en) 2018-11-30
UA115561C2 (en) 2017-11-27
EP2882749B1 (en) 2018-04-04
WO2014019979A1 (en) 2014-02-06
CN104428302B (en) 2017-03-22
JP6356671B2 (en) 2018-07-11
AR091939A1 (en) 2015-03-11
AP2015008202A0 (en) 2015-01-31
EP2882749A1 (en) 2015-06-17
BR112015002118A2 (en) 2017-07-04
GEP201706707B (en) 2017-07-25
IL236494A0 (en) 2015-02-26

Similar Documents

Publication Publication Date Title
US9085584B2 (en) Substituted pyrido[3,2-E][1,2,4]-triazolo[4,3-A]pyrazines for the treatment of central nervous system disorders
RU2603140C2 (en) Imidazotriazinone compounds
JP6740249B2 (en) Imidazotriazinones as PDE1 inhibitors
JP5349056B2 (en) Indropyridines as Eg5 kinesin modulators
US10875867B2 (en) Cyclic ether derivatives of pyrazolo[1,5-a]pyrimidine-3-carboxyamide
EP2855455B1 (en) Triazolo compounds as pde10 inhibitors
IE912126A1 (en) Polycyclic guanine derivatives
EP3057970A1 (en) Heterocyclic compounds and methods of use
CN109890824B (en) [1,2,4] triazolo [1,5-a ] pyrimidine compounds as PDE2 inhibitors
OA17160A (en) 4-methyl-2,3,5,9,9b-pentaza-cyclopenta[a]-naphthalenes
US9006226B2 (en) Dihydropteridinones I

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION