WO2011051478A1 - Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors - Google Patents

Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors Download PDF

Info

Publication number
WO2011051478A1
WO2011051478A1 PCT/EP2010/066537 EP2010066537W WO2011051478A1 WO 2011051478 A1 WO2011051478 A1 WO 2011051478A1 EP 2010066537 W EP2010066537 W EP 2010066537W WO 2011051478 A1 WO2011051478 A1 WO 2011051478A1
Authority
WO
WIPO (PCT)
Prior art keywords
oxime
chromen
alkyl
pyrimidin
ethoxy
Prior art date
Application number
PCT/EP2010/066537
Other languages
French (fr)
Inventor
Stephan Schann
Stanislas Mayer
Christophe Morice
Bruno Giethlen
Original Assignee
Domain Therapeutics
Prestwick Chemical, Inc.
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
Application filed by Domain Therapeutics, Prestwick Chemical, Inc. filed Critical Domain Therapeutics
Priority to ES10781455.0T priority Critical patent/ES2522827T3/en
Priority to CN201080059805.3A priority patent/CN102812014B/en
Priority to DK10781455.0T priority patent/DK2493871T3/en
Priority to JP2012535861A priority patent/JP5806672B2/en
Priority to CA2779073A priority patent/CA2779073C/en
Priority to US13/505,235 priority patent/US8962627B2/en
Priority to AU2010311321A priority patent/AU2010311321B2/en
Priority to EP10781455.0A priority patent/EP2493871B1/en
Publication of WO2011051478A1 publication Critical patent/WO2011051478A1/en
Priority to IL219149A priority patent/IL219149A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • C07D311/68Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4 with nitrogen atoms directly attached in position 4
    • 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
    • A61P25/14Drugs 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
    • A61P25/22Anxiolytics
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D471/02Heterocyclic 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 two hetero rings
    • C07D471/04Ortho-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
    • C07D487/02Heterocyclic 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 two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides new oxime derivatives of the general formula (I), pharmaceutical compositions containing them and their use for the treatment and/or prophylaxis of conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of giutamate level or signalling in mammals.
  • This invention further provides new oxime derivatives of the general formula (! consisting of modulators of nervous system receptors sensitive to giutamate, which makes them particularly suitable for the treatment and/or prophylaxis of acute and chronic neurological and/or psychiatric disorders.
  • the new oxime derivatives of the invention are modulators of metabotropic giutamate receptors (mGluRs).
  • the invention further provides positive allosteric modulators of mGluRs and more specifically positive allosteric modulators of mGluR4.
  • Glutamatergic pathways have been shown to be clearly involved in the physiopathology of a number of neuronal damages and injuries.
  • Many nervous system disorders including epilepsy and chronic or acute degenerative processes such as for example Alzheimer's disease, Huntington's disease, Parkinson's disease and amyotrophic lateral sclerosis (Mattson MP., Neuromolecular Med., 3(2), 65-94, 2003), but also A!DS-tnduced dementia, multiple sclerosis, spinal muscular atrophy, retinopathy, stroke, ischemia, hypoxia, hypoglycaemia and various traumatic brain injuries, involve neuronal cell death caused by imbalanced levels of giutamate.
  • compounds modulating glutamate level or signalling may be of great therapeutic value for diseases and/or disorders not directly mediated by glutamate !eveis and/or glutamate receptors malfunctioning, but which cou!d be affected by alteration of glutamate levels or signaling.
  • L-glutamate is the main excitatory neurotransmitter and is referred to as an excitatory amino-acid (EAA), and gamma- aminobutyric acid (GABA) is the main inhibitory neurotransmitter.
  • EAA excitatory amino-acid
  • GABA gamma- aminobutyric acid
  • Glutamate is ubiquitously distributed in the nervous system in high concentrations, especially in the brain and spina! cord of mammals, where it is working at a variety of excitatory synapses being thereby involved in virtually all physiological functions such as motor control, vision, central control of heart, processes of learning and memory.
  • excitotoxicity This combination of neuroexcitatory activities and neurotoxic properties is called excitotoxicity.
  • Glutamate operates through two classes of receptors (Brauner-Osborne H et al., J. Med. Chem. 43(14), 2609-45, 2000).
  • the first class of glutamate receptors is directly coupled to the opening of cation channels in the cellular membrane of the neurons. Therefore they are called ionotropic glutamate receptors (IGluRs).
  • IGluRs ionotropic glutamate receptors
  • the !GluRs are divided in three subtypes, which are named according to the depolarizing action of their selective agonists: N-methyl-D-aspartate (N DA), ct-amino-3-hydroxy-5- methylisoxazole-4-propionic acid (AMPA), and kainic acid (KA).
  • the second class of glutamate receptor consists of G-protein coupled receptors (GPCRs) called metabotropic glutamate receptors (mGluRs). These mGluRs are localized both pre- and post-synaptically. They are coupled to multiple second messenger systems and their role is to regulate the activity of the ionic channels or enzymes producing second messengers via G-proteins binding the GTP (Conn PJ and Pin JP. ; Annu. Rev. Pharmacol. Toxicol., 37, 205-37, 1997).
  • GPCRs G-protein coupled receptors
  • mGluRs metabotropic glutamate receptors
  • mGluRs modulate the efficacy of the synapses by regulating either the post-synaptic channels and their receptors, or the pre-synaptic release or recapture of glutamate. Therefore, mGluRs play an important role in a variety of physiological processes such as long-term potentiation and long- term depression of synaptic transmission, regulation of baroreceptive reflexes, spatial learning, motor learning, and postural and kinetic integration.
  • Group ! is constituted of mGluRI and mGiuRS, group I! of mGluR2 and mGiuR3 and group I II of mGluR4, mGiuR6, mGluR7 and mGluR8 (Pin JP and Acher F. , Curr. Drug Targets CNS Neurol. Disord. , 1 (3), 297-317, 2002; Schoepp DD et al. , Neuropharmacology, 38(10), 1431 -76, 1999).
  • mGluRs modulators can be classified in two families depending on their site of interaction with the receptor (see Brauner-Osbome H et al. , J. Med. Chern. 43(14), 2609-45, 2000 for review).
  • the first family consists in orthosteric modulators (or competitive modulators) able to interact with the glutamate binding-site of the mGluRs, which is localized in the large extra-celiular N-terminal part of the receptor (about 560 amino acids). Therefore, they are glutamate analogs and constitute a highly polar family of ligand.
  • orthosteric modulators examples include S-DHPG or LY-367385 for group I mG!uRs, LY-354740 or (2R-4R)-APDC for group II mGluRs and ACPT-I or L- AP4 for group Mi mGluRs.
  • the second family of mGluRs modulators consists in allosteric modulators that interact with a different site from the extracellular active site of the receptor (see Bridges TM et al. , ACS Chem Biol, 3(9), 530-41 , 2008 for review). Their action results in a modulation of the effects induced by the endogenous ligand glutamate.
  • examples of such allosteric modulators are Ro-674853, MPEP or JNJ16259685 for group ! mGluRs and CBiPES, LY181837 or LY487379 for group II mGluRs.
  • Group III mGluR modulators showed positive results in several animal models of schizophrenia (Paiucha-Poniewiera A et al., Neuropharmacology, 55(4), 517-24, 2008) and chronic pain (Goudet C et ai., Pain, 137(1 ), 1 12-24, 2008; Zhang HM et a!., Neuroscience, 158(2), 875-84, 2009).
  • mGluR were also shown to exert the excitotoxic actions of homocysteine and homocysteic acid contributing to the neuronal pathology and immunosenescence that occur in Alzheimer Disease (Boldyrev AA and Johnson P, J. Alzheimers Dis. 1 (2), 219-28, 2007).
  • group III mGluR modulators showed promising results in animal models of Parkinson and neurodegeneration (Conn J et al., Nat Rev. Neuroscience, 6(10), 787-98, 2005 for review; Vernon AC et al., J. Pharmacol. Exp. Then, 320(1 ), 397-409, 2007; Lopez S et al., Neuropharmacology, 55(4), 483-90, 2008; Vernon AC et al., Neuroreport, 19(4), 475-8, 2008). It was further demonstrated with selective ligands that the mGluR subtype implicated in these antiparkinsonian and neuroprotective effects was mGluR4 (Marino MJ et al., Proc. Natl. Acad. Sci.
  • mGluR4 modulators were also shown to exert anxiolytic activity (Stachowicz K et al., Eur. J. Pharmacol., 498(1-3), 153-6, 2004) and anti-depressive actions (Palucha A et ai., Neuropharmacology 46(2), 151-9, 2004; Klak K et al., Amino Acids 32(2), 169-72, 2006).
  • mGluR4 were also shown to be involved in glucagon secretion inhibition (Uehara S., Diabetes 53(4), 998-1006, 2004). Therefore, orthosteric or positive allosteric modulators of mGluR4 have potential for the treatment of type 2 diabetes through its hypoglycemic effect.
  • mGluR4 was shown to be expressed in prostate cancer cell-line (Pessimissis N et al., Anticancer Res. 29(1 ), 371-7, 2009) or colorectal carcinoma (Chang HJ et a!., CIL Cancer Res. 1 1 (9), 3288-95, 2005) and its activation with PHCCC was shown to inhibit growth of medul!oblastomas (iacoveili L et al., J. Neurosci. 26(32) 8388-97, 2006), mGluR4 modulators may therefore have also potential role for the treatment of cancers.
  • mGluR4 modulators may also be useful as taste agents, flavour agents, flavour enhancing agents or food additives.
  • Chromone-derived core structures for pharmaceutically active compounds were described in the patent application WO 2004/092154. In the latter application, they are disclosed as inhibitors of protein kinases.
  • EP-A-0 787 723 relates to specific cyclopropachromencarboxylic acid derivatives which are said to have mGluR antagonistic activity.
  • the -present invention relates to compounds of the general formula (I):
  • R 1 , R 2 , R 3 , and R 4 each independently represent a -L-R group.
  • L represents a bond, a C C-io alkylene, a C 2 -C 10 alkenylene, or a C2-C 10 atkynylene, wherein said alkylene, said alkenylene or said alkynylene is optionally substituted with one or more groups independently selected from halogen, -CF 3 , -CN, -OH, or -NH 2 , and further wherein one or more -CH 2 - units comprised in said alkyiene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -0-, -NR 11 -, -CO-, -S-, -SO-, or -S0 2 -.
  • R is selected from hydrogen, C C 10 alkyl, halogen, optionaily substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, -NR 11 R 12 , -OR 11 , -SR , -SOR 11 , -S0 2 R 11 , -CF 3 , or -CN, wherein said optionaily substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyl may be substituted with one or more groups independently selected from C C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -NH 2 , -NH(C C 4 alkyl), -N(C C 4 alkyS)(C C 4 alkyl) wherein the two C -C 4 alkyl moieties of said -N(C
  • R 1 and R 12 are each independently selected from hydrogen, optionally substituted C Cio alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionaily substituted heterocycloalkyl, or -CF 3 , wherein said optionally substituted alkyl, said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyl may be substituted with one or more groups independently selected from C C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C r C 4 alkyl), -NH 2s -NH(C C 4 alkyl), or -N(C 1 -C 4 alkyl)(C-i-C 4 alkyl), wherein the two C C 4 alkyl moieties of said -N(C C 4 alkyl)(d-C 4 alkyl) are optionally mutually
  • L 1 Is selected from a bond, C C 10 alkyiene, C 2 ⁇ C 10 alkenylene, or C 2 -C 10 alkynylene, wherein one or two -CH 2 - units comprised in said alkyiene, said alkenylene or said alkynylene are each optionaily replaced by a group independently selected from -0-, -NH-, -N(C C 4 alkyl)-, -CO-, -S-, -SO- or -S0 2 -.
  • R 13 is selected from hydrogen, C r C 4 alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycioalkyi, optionally substituted heterocycloalkyi, -NH 2 , -NH ⁇ d-d alkyl), -N(C r C 4 alkyl)(d-d alkyl), -OH, -0(d-d alkyl), -SH, -S(d-C 4 a!kyl), -CF 3 , or -CN, wherein said optionally substituted phenyl, said optionally substituted heteroaryl, said optionally substituted cycioalkyi, or said optionally substituted heterocycloalkyi may be substituted with one or more groups independently selected from C d alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -NH 2 , -NH(d-d al
  • s is selected from hydrogen, C C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -COOH, -COO(C C 4 alkyl), -CONH 2 , -CONH(C C 4 alkyl), -CON(C d alkyl)(d-d alkyl), wherein the two d-d alkyl moieties of said -CON(d-d alkyl)(C C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -NH 2 , -NH(d-C 4 alkyl), or -N(C C 4 alkyI)(C C 4 alkyl), wherein the two d-d alkyl moieties of said -N(d-C alkyi)(d-C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to
  • A is a bicyclic moiety corresponding to formula (II):
  • n 0 or 1 ;
  • X 1 to X 6 are each independently selected from N, N(R x1 ), C(R x2 ), C(R x2 )(R x3 ), O, S, S(O), S(0) 2 , or C(O);
  • X 7 is N or N(R x );
  • any of groups X 1 to X 7 containing a nitrogen atom may form an N-oxide group
  • X s and X 9 are each independently selected from N, C, or C(R 2 );
  • each R x is independently selected from hydrogen, d ⁇ C 4 alkyl, -OH, -0(C C 4 alkyl), or -(C C 4 alkytene)-phenyl;
  • each R 2 and each R 3 is independently selected from hydrogen, C r C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -COOH, -COO(C-rC 4 alkyl), -CONH 2 , -CONH(C C 4 a!kyi), -CON ⁇ C r C 4 alkyl)(C 1 -C alkyl), -NH 2 , -NH(C C 4 alkyl), or -N(C r C alkyl)(C C 4 alkyl), wherein the two C C alkyl moieties of said -N(C C 4 alkyl)(C C 4 alkyl) or of said -CON(C C 4 alkyI)(C C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
  • bicyclic ring system of formula (I) including the O-heteroatom and the oxime group, to which the substituents R 1 to R 5 and A are attached is also referred to as "chromone moiety".
  • the present invention relates to new oxime derivatives of the general formula (I) as described and defined herein and pharmaceutically acceptable salts, solvates and prodrugs thereof, pharmaceutical compositions containing any of the aforementioned entities and their use for the treatment and/or prophylaxis of conditions associated with altered giutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling in mammals. It further relates to a method of treating and/or preventing conditions associated with altered giutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling in a mammal.
  • the present invention provides a method of treating and/or preventing a disease or disorder, in particular a condition associated with altered giutamatergic signalling and/or functions, and/or a condition which can be affected by alteration of glutamate level or signalling, the method comprising the administration of a compound of the general formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities, to a subject (preferably, a mammal; more preferably, a human) in need of such treatment or prevention.
  • a subject preferably, a mammal; more preferably, a human
  • the compounds of the general formula (!) are modulators of mGluRs of the nervous system, in preferred embodiments the compounds of the invention are allosteric modulators of the mGluRs and in a most preferred embodiment they are positive allosteric modulators of mGluR4.
  • the conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling, and which can be treated and/or prevented with the compounds or the pharmaceutical compositions according to the invention include in particular: epilepsy, including newborn, infantile, childhood and adult syndromes, partial (localization-related) and generalized epilepsies, with partial and generalized, convulsive and non-convulsive seizures, with and without impairment of consciousness, and status epiiepticus; Dementias and related diseases, including dementias of the Alzheimer's type (DAT), Alzheimer's disease, Pick's disease, vascular dementias, Lewy-body disease, dementias due to metabolic, toxic and deficiency diseases (including alcoholism, hypothyroidism, and vitamin B12 deficiency), AIDS-dementia complex, Creutzfeld- Jacob disease and atypical subacute spongiform encephalopathy; Parkinsonism and movement disorders, including Parkinson's disease, multiple system atrophy,
  • Dementias and related diseases including dementias of the Alzheimer's type (DAT), Alzheimer's disease, Pick's disease, vascular dementias, Lewy-body disease, dementias due to metabolic, toxic and deficiency diseases (including alcoholism, hypothyroidism, and vitamin B12 deficiency), AlDS-dementia complex, Creutzfetd-Jacob disease and atypical subacute spongiform encephalopathy; Parkinsonism and movement disorders, including Parkinson's disease, multiple system atrophy, progressive supranuclear palsy, coriicobasal degeneration, hepatolenticular degeneration, chorea (including Huntington's disease and hemiballismus), athetosis, dystonias ⁇ including spasmod
  • the present invention further provides a method for identifying an agent that binds to metabotropic glutamate receptor 4 (mGiuR4), or in other words for determining the capability of one or more test agent(s) to bind to the receptor, comprising the following steps: (a) contacting mG!uR4 with a compound of the present invention which is labeled, preferably radio-labeled or fluorescence-labeled, under conditions that permit binding of the compound to mGluR4, thereby generating a bound, labeled compound; (b) detecting a signal that corresponds to the amount of the bound, labeled compound in the absence of test agent; (c) contacting the bound, labeled compound with a test agent; (d) detecting a signal that corresponds to the amount of the bound labeled compound in the presence of test agent; and (e) comparing the signal detected in step (d) to the signal detected in step (b) to determine whether the test agent binds to mGiuR4.
  • a substantially unchanged signal detected in step (d) in comparison with the signal detected in step (b) indicates that the test agent does not bind to the receptor, or binds to the receptor less strongly than the compounds according to the invention.
  • a decreased or increased signal detected in step (d) in comparison with the signal detected in step (b) indicates that the test agent binds to the receptor.
  • agents that bind to mGiuR4 can be identified among the test agents employed in the above method. It will further be understood that it is preferred to remove unbound labeled compounds, e.g. in a washing step, before carrying out steps (b) and (d).
  • the rrsGluR4 which is used in the above method may be a human form (F!or PJ, Lukic S, Ruegg D, Leonhardt T, Knopfel T, Kuhn R. 1995. Neuropharmacology. 34: 149-155. akoff A, Lelchuk R, Oxer M, Harrington K, Emson P. 1996. Brain Res. Mol. Brain Res. 37:239-248. Wu S, Wright RA, Rockey PK, Burgett SG, Arnold JS, Rosteck PR Jr, Johnson BG, Schoepp DD, Belagaje RM. 1998. Brain Res. Mot. Brain Res. 53:88- 97.), e.g.
  • a non-human form including e.g. a mouse form or rat form (Tanabe Y, Masu M, !shii T, Shigemoto R, Nakanishi S. 1992. Neuron. 8:169-179.), or a
  • Said mutain can preferably be obtained by substitution, insertion, addition and/or deletion of one or more (such as, e.g., 1 to 20, including 1 to 10 or 1 to 3) amino acid residues of said aforementioned entitites.
  • the mGluR4 used in the above method may also be a functional fragment of any of the aforementioned entitites (including said muteins), i.e. a fragment which retains the mGluR4 activity of the respective aforementioned entity or, in other words, a fragment having essentially the same biological activity (i.e., at least about 60% activitiy, preferably at least about 70% activity, more preferably at least about 80% activity, even more preferably at least about 90% activity) as the respective aforementioned entity.
  • the mGluR4 used in the above method may also be a compound comprising any one or more of the aforementioned entitites (including, without limitation, a protein of the accession number NPJ300832, a protein having at least 80% amino acid identity to said protein of the accession number NP_000832, or a functional fragment thereof), wherein the mGluR4 activity is retained.
  • the mGluR4 used in the above method is a human form.
  • R , R 2 , R 3 , and R 4 each independently represent a -L-R group.
  • L represents a bond, CrC 10 alkylene, C 2 -C 0 alkenylene, or C 2 -C 10 alkynyiene.
  • Said alkylene, said alkenylene or said alkynyiene is optionally substituted with one or more (such as, e.g., one, two, three, or four) groups independently selected from halogen, -CF 3 , -CN, -OH, or -NH 2 .
  • one or more (such as, e.g., one, two, three, or four), preferably one or two, more preferably one, -CH 2 - units comprised in said alkylene, said alkenylene or said aikynylene are each optionally replaced by a group independently selected from: -0-, -NR 11 -, -CO-, -S-, -SO-, or -S0 2 ⁇ .
  • no -CH 2 - unit, one -CH 2 - unit or two -CH 2 - units are each replaced by a group independently selected from -0-, -NR 11 -, -CO-, or -S-. More preferably, no -CH 2 - unit is replaced or one -CH 2 - unit or two -CH 2 - units are each replaced by a group independently selected from -0-, -NR 11 -, or -S-. Even more preferably, no -CH 2 - unit is replaced or one or two -CH 2 - units are each replaced by -0-,
  • L is a bond, Ci-C 6 alkylene, C 2 -C 6 alkenylene, or C 2 -C 6 aikynylene, wherein one or two -CH 2 - units comprised in said alkylene, said alkenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NR 11 -, -CO-, or -S-.
  • L is a bond, Ci-C 3 alkylene, C 2 -C 6 alkenylene, or C 2 -C 6 aikynylene, wherein one or two -CH 2 ⁇ units comprised in said alkylene, said alkenylene, or said aikynylene are each optionally replaced by -0-.
  • L is a bond or C C 6 alkylene, wherein one or two -CH 2 - units comprised in said alkylene are each optionally replaced by -0-.
  • R is selected from hydrogen, C C 0 alkyl, halogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloaikyi, optionally substituted heterocycioalkyi, -NR 11 R 12 , -OR 11 , -SR 11 , -SOR 11 , -S0 2 R 11 , -CF 3 , or -CN, wherein said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloaikyi, or said optionally substituted heterocycioalkyi may be substituted with one or more (such as, e.g., one, two, three, or four), preferably one or two, substituent groups independently selected from C C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -NH 2 , -NH(C C 4 alkyl), -N(C C 4
  • Said substituent groups are preferably selected from C-1-C4 alkyl, -(C C 4 alkylene)-OH, ⁇ C C alkylene)-0-(Ci-C 4 alkyl), -OH, -0 ⁇ d-C 4 alkyl), -0-(C r C 4 alkylene)-0-(C C 4 alkyl), -NH 2 , -NH(Ci-C 4 alkyl), -N(C C 4 alkyl)(C C 4 alkyl) wherein the two C C 4 alkyl moieties of said -N(C 1 -C 4 alkyl) ⁇ C C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -CF 3
  • R is selected from: hydrogen; optionally substituted aryl; optionally substituted heieroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or and the other ring atoms are carbon atoms; opttonaliy substituted heterocycioalkyl having 3 to 10 ring atoms, wherein one or more (such as, e.g., one, two, or three) ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -NH(C C 4 alkyl); -N(CTC 4 alkyl)(Ci-C 4 alkyl); or -0(C C alkyl).
  • Said aryl may, for example, be a phenyl.
  • Said heteroaryl may, for example, be selected from pyridinyl, thienyi, furyl, pyrrolyl, imidazolyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, isoxazoiyl, or furazanyl.
  • Said heterocycioalkyl may, for example, be selected from tetrahydrofuranyl, piperidinyl, piperazinyl, pyrrolidinyl, imidazolidinyl, morpholinyi, pyrazolidinyl, tetrahydrothienyl, oxazolidinyi, isoxazolidinyl, aziridinyl, azetidinyl, octahydroquinolinyl, octahydroisoquinolinyl, azepanyl, diazepanyl, oxazepanyi or 2-oxa-5-aza- bicyclo[2.2.1]hept-5-yl.
  • R is selected from: hydrogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -0(C C alkyl); or optiona!ly substituted heterocycioalkyl having 3 to 10 ring atoms, wherein one or more ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms.
  • R is selected from: hydrogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independentiy selected from O, S, or N and the other ring atoms are carbon atoms; or optionally substituted heterocycloalkyl having 5 to 7 ring atoms, wherein one or two ring atoms are each independentiy selected from N or O and the other ring atoms are carbon atoms.
  • preferred combinations -L-R are those wherein L is a bond, C-
  • Said optionally substituted phenyl, said optionally substituted heteroaryl having 5 or 6 ring atoms, or said optionally substituted heterocycloalkyl having 5 to 7 ring atoms may be substituted with one or more, preferably one or two, groups independently selected from C C 4 alkyl, -(C r C 4 aikylene)-OH, -(C C 4 aikylene)-0-(C 1 -C 4 alkyl), -OH, -0 ⁇ CrC 4 alkyl), -0-(C C 4 alkylene)-0- ⁇ C C 4 alkyl), -NH 2 , -NH(C r C 4 alkyl), or -N(C C 4 a!kyS)(C C 4 alkyl) wherein the two C C 4 alkyl moieties of said -N(C T C 4 alkyl)(C 1 -C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they
  • Exemplary preferred -L-R groups encompassed by the above preferred definition are independently selected from: hydrogen; (VC 4 alkyl; -(C C 3 alkyIene)-0 ⁇ ((VC 2 alkyl); -(C 2 -C 4 alkyny!ene)-phenyl; -OH; -0(d-C 4 alkyl); -0(C C 2 alkylene)-0-(C C 2 alkyl); -(C C 4 alky!ene)-morpholinyl; -0(C C 4 alkylene)-phenyl; -0(C C A alkylene)-imidazolyl; -0(C C 4 alkylene)-pyrrolidinyl; -0(CVC 4 alkylene)-piperidinyl; -0(C C 4 alkylene)-morpholinyl; -0(C C 4 alkylene)-pyridinyl; -0(C ⁇ -C 4 a!kylene)-o
  • R to R 4 are all hydrogen, or one of R 1 to R 4 is a -L-R group other than hydrogen as defined above and the other ones of R 1 to R 4 are hydrogen.
  • the -L-R group other than hydrogen is in particular one of R 2 and R 3 , and more preferably R 2 .
  • R 1 and R 4 are each hydrogen, one of R 2 and R 3 (preferably R 3 ) is hydrogen, and the other one of R 2 and R 3 (preferably R 2 ) is selected from: hydrogen; d-d alkyl; ⁇ (d ⁇ d a!kylene)-0-(d-C 2 alkyl); -(d-d alkynylene)-phenyl; -OH; -0(C d alkyl); -0(C C 2 alky!ene)-0-(d-C 2 alkyl); -(d-d alkylene)-morpholinyl; -0(C d alkylene)-phenyl; ⁇ 0 ⁇ d-d alkylene)-imidazolyl; -0(C d alkylene)-pyrrolidinyl; -0(d-d alkylene)-piperidinyl; -0(d-d alkylene)-morphoiinyl; -0(d-d alkyl
  • R 11 and R 12 are each independently selected from hydrogen, optionally substituted C C 10 alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionaliy substituted heterocycloalkyi, or -CF 3 , wherein said optionally substituted alkyl, said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyi may be substituted with one or more, preferably one or two, more preferably one, groups independently selected from C C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 a!kyl), -NH 2 , -NH(C C 4 alkyi), or -N(C r C 4 alkyl)(C C 4 alkyl) wherein the two C C 4 alkyl moieties of said - ⁇ ((_ ⁇ 4 alkylXd-C
  • L 1 is selected from a bond, C C 0 alkylene, C 2 -C 10 aikenylene, or C 2 -C 10 aikynylene, wherein one or two -CH 2 - units comprised in said alkylene, said aikenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NH-, -N(C C 4 alkyl)-, -CO-, -S-, -SO- or -S0 2 -. This includes the option that the replacing group -NH- or the replacing group -N(C C 4 alkyl)- is combined with the replacing group -CO-, in any order, to form an amide group. Otherwise, if more than one -CH 2 - unit is replaced, it is preferred that these -CH 2 - units are non-adjacent.
  • L 1 is selected from a bond, C C 6 alkylene, C 2 -C 6 aikenylene, or C 2 -C 6 aikynylene, wherein one or two -CH 2 - units comprised in said alkylene, said aikenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NH-, -CO-, or -S-.
  • L 1 is selected from a bond, C C 5 alkylene, C 2 -C 6 aikenylene, or C 2 -C 5 alkynylene, wherein one or two -CH 2 - units comprised in said aikylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -0-, -NH-, or -S-.
  • L 1 is a bond, C C 6 aikylene, C 2 -C 6 alkenylene, or C 2 -C 6 alkynylene, wherein one or two -CH 2 - units comprised in said aikylene, said alkenylene or said alkynylene are each optionally replaced by -0-.
  • L is a bond or C C s aikylene, wherein one or two -CH 2 - units comprised in said aikylene are each optionally replaced by -0-.
  • R 13 is selected from hydrogen, d-C alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycloa!kyl, optionally substituted heterocycloalkyl, -NH 2 , -NH(C C 4 alkyl), -N(C C 4 alkyl)(C C 4 alkyl), -OH, -0(C C 4 alkyl), -SH, -S(C C 4 alkyl), -CF 3 , or -CN, wherein said optionally substituted phenyl, said optionally substituted heteroaryl, said optionally substituted cycloalkyi, or said optionally substituted heterocycloalkyl may be substituted with one or more, preferably one or two, more preferably one, groups independently selected from C r C 4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -NH 2 , -NH(C
  • R 13 is selected from hydrogen, C C alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycloalkyi, optionally substituted heterocycloalkyl, -NH(C C 4 alkyl), -N(C C 4 alkyl)(C C 4 alkyl), -0 ⁇ C C 4 alkyl), -S(C-,-C 4 alkyl), -CF 3 , or -CN.
  • R 13 is selected from: hydrogen; Ci-C 4 alkyl; halogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; cycloalkyi having 3 to 7 ring atoms; optionally substituted heterocycloalkyl having 3 to 10 ring atoms, wherein one or more (such as, e.g., one, two, or three) ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -NH(C r C 4 alkyl); -N(CrC 4 aIkyl)(C 1 -C 4 alkyl); -0(C C 4 alkyl); -S(C C 4 alkyl); -CF 3 ; or -CN.
  • R 13 is selected from: hydrogen; -0(CrC 4 alkyl); optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; or optionally substituted heterocycloaikyl having 3 to 10 ring atoms, wherein one or more ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms.
  • R 6 is selected from hydrogen, C C 4 alky!, halogen, -CF 3 , -CN, -OH, -0(C C 4 alky!), -COOH, -000(0,-04 alkyl), -CONH 2 , -CONH(C C 4 alkyi), -CON(Ci-C 4 alkyl)(C 1 -C 4 alkyl) wherein the two C C 4 alkyl moieties of said -CON(C-i-C 4 aikyt)(CrC 4 alkyl) are optionally mutually linked to form a ring (such as, e.g., a pyrroiidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to, -NH 2 , -NH ⁇ Ci-C 4 alkyl), or -N(CrC 4 alkyl)(C f C 4 alkyl) wherein the two Ci-C 4 alkyl moi
  • Said halogen may, for example, be selected from fluoro, chloro, bromo, or iodo.
  • R 5 is selected from hydrogen, Ci-C 4 alkyl, halogen, -CF 3 , -CN, -OH, or -0(C C 4 alkyl). More preferably, R 5 is selected from hydrogen or C C 4 alkyl. Even more preferably, R s is hydrogen.
  • A is a bicyclic moiety corresponding to formula (II):
  • n 0 or 1 .
  • X 1 to X s are each independently selected from N, N(R x1 ), C(R x2 ), C(R x2 )(R x3 ), O, S, S(0), S(0) 2 , or C(O).
  • X 1 to X 6 are each independently selected from N, N(R* 1 ), C(R* 2 ) ; C(R x2 )(R x3 ), S or O.
  • X 7 is N or N(R x1 ). Included is furthermore the option that any of groups X 1 to X 7 containing a nitrogen atom can form an N-oxide group.
  • X 8 and X 9 are each independently selected from N, C, or C(R* 2 ).
  • Each R x1 is independently selected from hydrogen, C C 4 alkyi, -OH, -0 ⁇ C r C 4 alkyi), or -(C1-C4 alkyiene)-phenyl.
  • each R x1 is independently selected from hydrogen, or d-d alkyi.
  • Each R x2 and each R 3 is independently selected from hydrogen, d-d aikyl, halogen, -CF 3 , -CN, -OH, -0(CrC 4 alkyi), -COOH, -COO(C C 4 alkyi), -CONH 2 , -CONH(C C 4 alkyi), -CON(C C 4 alkyl)(d-C 4 alkyi), -NH 2 , -NH(C C 4 aikyl), or -N(C C alkyl)(d-C 4 alkyi), wherein the two C C 4 aikyl moieties of said -N(C T C 4 alkyl)(C 1 -C 4 alkyi) or of said -CON(C d aikyl)(C C 4 aikyl) are optionally mutually linked to form a ring (such as, e.g.
  • each R xZ and each R x3 is independently selected from hydrogen, d-C 4 aikyl, halogen, -CF 3 , -CN, -OH, -0(C d alkyi), -NH 2 , -NH(d-C 4 aikyl), or -N(d-d alkyl)(C r d alkyi) wherein the two d-d alkyi moieties of said -N(d-C 4 alkyl)(d-d aikyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
  • each R x2 and each R x3 is independently selected from hydrogen, - alkyi, halogen, -CF 3 , -OH, or -0(C C 4 alkyi).
  • the bicyciic moiety corresponding to formula (II) is linked to the remainder of the compound of general formula (I) by the interrupted bond shown in formula (II), i.e.
  • the absence or the presence of one or two substituents R x1 , R x2 and R x3 in the groups X 1 to X 9 will depend on the presence and the position of double bonds in the bicyciic ring system of formula (I I).
  • one or two of the groups X 1 to X 6 , X s and X 9 are each independently selected from N, N(R ), O, S, S(O), or S(0) 2 (preferably, selected from N, N(R x1 ), S or O) in the case of X 1 to X s and from N in the case of X 8 and X 9 , respectively, and the remaining groups X 1 to X 6 , X s and X 9 are each independently selected from C(R x2 ) or C(R x2 )(R 3 ) in the case of X 1 to X 6 and from C or C(R xZ ) in the case of X 8 and X 9 , respectively.
  • one or two of the groups X 1 to X 6 are each independently selected from N, N(R ), O, or S, the remaining groups X 1 to X 6 are each independently selected from C(R 2 ) or C(R 2 )(R 3 ), and X 8 and X 9 are each independently selected from C or C(R* 2 ); alternatively, in another embodiment one or none of the groups X 1 to X 6 is selected from N, N(R ), O, or S, the remaining groups X 1 to X 6 are each independently selected from C(R x2 ) or C(R x2 )(R 3 ), one of X 8 and X 9 is N, and the other one of X 8 and X 9 is C or C(R" 2 ).
  • one or two of the groups X 1 to X 6 , X s and X 9 are each independently selected from N, N(R x1 ) or S in the case of X 1 to X 6 and from N in the case of X s and X 9 , respectively, and the remaining groups X 1 to X 6 , X s and X 9 are each independently selected from C ⁇ R x2 ) or C(R x2 )(R 3 ) in the case of X 1 to X 6 and from C or C(R 2 ) in the case of X 8 and X s , respectively.
  • one or two of X 1 to X 6 are each independently selected from N, N(R 1 ) or S, the remaining groups X 1 to X 6 are each independently selected from C(R x2 ) or C(R x2 )(R x3 ), and X s and X 9 are each independently selected from C or C(R x2 ); alternatively, in another embodiment one or none of the groups X 1 to X 6 is selected from N, N(R x1 ) or S, the remaining groups X 1 to X s are each independently selected from C(R x2 ) or C(R x2 )(R x3 ), one of X 8 and X 9 is N, and the other one of X 8 and X 9 is C or C(R x2 ).
  • A is a bicyclic moiety corresponding to formula (II) as described and defined herein above, wherein the first ring of the bicyclic moiety (i.e. the ring which is linked to the remainder of the compound of general formula (I)) is aromatic, and it is more preferred that A is a bicyclic moiety corresponding to formula (II) as described and defined herein above, wherein the two rings of the bicyclic moiety form an aromatic ring system.
  • A is one of the following groups:
  • each of which may optionally be substituted on ring carbon atoms with one or more groups independently selected from C ⁇ -C 4 aikyl, halogen, -CF 3 , -CN, -OH, -0(Ci-C aikyl), -NH 2 , -NH(C C 4 alkyl), or -N(C C 4 alkyl)(C C 4 alkyl) wherein the two d-C 4 alkyl moieties of said -N(d-C 4 aikyl)(C C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from C C 4 alkyl, halogen, -OH, or -0(C C aikyl).
  • each R x1 is independently selected from hydrogen, C C 4 aikyl, -OH, or -0(C C 4 alkyl), and preferably selected from hydrogen or d-d alkyl.
  • Each R x4 is independently selected from hydrogen or Cj-C 4 aikyl, or the two groups R 4 (if present) are each independently C1-C4 alkyl and are mutually linked to form a ring (such as, e.g., a pyrrolidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to. It is preferred that the above depicted optionaiiy substituted groups are unsubstituted and R 1 (if present) is hydrogen. .
  • A is one of the following groups: each of which may optional!y be substituted on ring carbon atoms with one or more groups independently selected from C1-C4 alkyl, halogen, -CF 3 , -CN, -OH, -0(C C 4 alkyl), -NH 2 , -NH(d-d alkyl), or -N(d-C 4 aikyi)(d-C 4 alkyl) wherein the two C C 4 alky!
  • moieties of said -N(d-d alky!(d-C 4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from d-d alkyl, halogen, -OH, or -0 ⁇ C-
  • A is one of the following groups:
  • each of which may optionally be substituted on ring carbon atoms with one or more groups independently selected from d- alkyl, halogen, -CF 3l -CN, -OH, -0(d-d alkyl), -NH 2 , -NH(C alkyl), or -N(d-C 4 alkyl)(C C 4 alkyl) wherein the two - alkyl moieties of said -N(d ⁇ d alkyl)(C d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from d-d alkyl, halogen, -OH, or -0(C C 4 aikyf). It is preferred that the above depicted optionally substituted groups are unsubstituted.
  • the compound of the general formula (I) is selected from:
  • Compounds of the present invention are characterized in that they provide mGluR4 activity due to two specific features: 1) the presence of a bicyciic ring on position 2 of the chromone ring (i.e. as group A in formula (I)), and 2) the presence of a nitrogen atom on the first ring of that bicyciic ring and at the position adjacent to the carbon linked to the chromone moiety but not adjacent to any of the two ring atoms which form part of both rings condensed to said bicyciic ring, as shown in the scheme below. It will be understood that this is a schematic representation, and that the N atom in the bicyciic system can be bound via a double bond in the ring structure or can carry a substituent as defined above:
  • Example 1 is a positive allosteric modulator (PAM) of mGluR4 having an EC 50 lower than 1 ⁇ .
  • Example 74 reference
  • Example 79 reference
  • Example 79 reference
  • Example 79 reference
  • Example 79 is the isomer of example 1 with a nitrogen atom on the other position adjacent to the carbon linked to the chromone moiety. Comparison of activities of these three molecules demonstrates that a nitrogen atom on the position X 7 of Formula (II) is required for the mGluR4 PAM activity.
  • example 63 is an mGluR4 PAM having an EC 50 lower than 1 ⁇ .
  • Example 80 (reference), that has no PAM activity on mGluR4 up to 100 ⁇ , is an analog of example 63 with a monocyclic heteroaryl. Comparison of activities of these two molecules demonstrates that a bicyclsc ring is required as moiety A in formula (!) for the mGluR4 PAM activity.
  • stereoisomers it embraces the racemic forms and the isolated optical isomers.
  • the racemic forms can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates using conventional methods, such as, e.g., salt formation with an optically active counterpart followed by crystallization.
  • a!kyl represents a straight or branched chain saturated hydrocarbon residue which does not comprise any carbon-to-carbon double bonds or carbon-to- carbon triple bonds.
  • alkenyl represents a straight or branched chain unsaturated hydrocarbon residue comprising at least one carbon-to-carbon double bond.
  • alkynyl represents a straight or branched chain unsaturated hydrocarbon residue comprising at least one carbon-to-carbon triple bond.
  • alkylene represents a straight or branched chain alkanediyl group which does not comprise any carbon-to-carbon double bonds or carbon-to-carbon triple bonds.
  • alkenylene represents a straight or branched chain alkenediyl group comprising at least one carbon-to-carbon double bond.
  • alkyny!ene represents a straight or branched chain alkynediyl group comprising at least one carbon-to-carbon triple bond.
  • aryl represents a 6-10 membered aromatic hydrocarbon ring, including bridged ring or fused ring systems containing at least one aromatic ring.
  • Aryl may, for example, refer to phenyl or naphthyl.
  • heteroaryl represents a 5-14 membered aromatic ring, including bridged ring or fused ring systems containing at least one aromatic ring, comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein one or more of said S ring atoms (if present) and/or one or more of said N ring atoms (if present) may optionally be oxidized.
  • a “heteroaryl”, as defined herein above, preferably represents a 5-14 membered aromatic ring, including bridged ring or fused ring systems containing at least one aromatic ring, comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N.
  • Heteroaryl may, for example, refer to thienyl (thiophenyi), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (furany!), isobenzofuranyl, chromenyl, xanthenyl, phenoxathiiny!, pyrrolyl (including, without limitation, 2H-pyrroiyl), imidazolyl, pyrazoiyl, pyridyl (pyridinyl; including, without limitation, 2-pyridyl, 3-pyridyl, and 4-pyridyl), pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, indolyl (including, without limitation, 3H-indolyl), indazolyl, purinyi, isoquinolyl, quino!y!, phthalaziny
  • cyc!oalkyi represents a 3-10 membered saturated hydrocarbon ring, including bridged ring or fused ring systems.
  • Cycloalkyl may, for example, refer to cyclopropy!, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl.
  • heterocycloalky represents a 3-10 membered saturated ring, including bridged ring, spiro ring or fused ring systems, containing one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein one or more of said S ring atoms (if present) and/or one or more of said N ring atoms (if present) may optionally be oxidized.
  • a “heterocycloalkyl”, as defined herein above, preferably represents a 3-10 membered saturated ring, including bridged ring or fused ring systems, containing one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N.
  • Heterocycloalkyl may, for example, refer to tetrahydrofuranyi, piperidinyi, piperazinyl, aziridinyl, azetidinyl, pyrrolidinyl, imidazoiidinyl, morpholinyl, pyrazolidinyi, tetrahydrothienyl, octahydroquinolinyl, octahydroisoquinolinyl, oxazolidiny!, isoxazolidinyl, azepanyl, diazepanyl, oxazepanyl or 2-oxa-5-aza-bicyclo[2.2, 1]hept-5-yl.
  • halogen represents fiuoro, chloro, bromo. or iodo, and in particular fiuoro, chloro, or bromo.
  • the scope of the invention embraces ail pharmaceutically acceptable salt forms of the compounds of the general formula (I) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of a carboxylic acid group with a physiologically acceptable cation as they are well known in the art.
  • Exemplary base addition salts comprise, for example, alkaii metal salts such as sodium or potassium salts; alkaline-earth metai salts such as calcium or magnesium salts; ammonium salts; aliphatic amine salts such as trimethyiamine, triethylamine, dicyc!ohexylamine, ethanolamine, diethanolamine, triethano!amine, procaine salts, meglumine salts, diethanol amine salts or ethylenediamine salts; aralkyl amine salts such as ⁇ , ⁇ -dibenzylethylenediamine salts, benetamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoHne salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethy!ammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyl
  • Exemplary acid addition salts comprise, for example, mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts, nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts or perchlorate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, undecanoate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, nicotinate, benzoate, salicylate or ascorbate salts; sulfonate salts such as methanesulfonate, ethanesulfonate, 2-hydroxyethanesulfonate, benzenesulfonate
  • the scope of the invention embraces solid forms of the compounds of the general formula (I) in any solvated form, including e.g. solvates with water, for example hydrates, or with organic solvents such as, e.g., methanol, ethano! or acetonitri!e, i.e. as a methanolate, ethanolate or acetonitrilate, respectively; or in the form of any polymorph.
  • prodrugs of compounds that can be used in the present invention are derivatives which have chemically or metabolically cieavable groups and become, by solvo!ysis or under physiological conditions, the compounds used in the present invention which are pharmaceuticaily active in vivo.
  • Prodrugs of compounds that can be used in the present invention may be formed in a conventional manner with a functional group of the compounds such as with an amino, hydroxy or carboxy group.
  • the prodrug derivative form often offers advantages of solubility, tissue compatibility or delayed release in a mamma!ian organism (see, Bundgaard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985).
  • Prodrugs include acid derivatives well known to the person skilled in the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine.
  • an ester derivative prepared by reacting the carboxyl group with a suitable alcohol or an amide derivative prepared by reacting the carboxyl group with a suitable amine is exemplified as a prodrug.
  • An especially preferred ester derivative as a prodrug is methylester, ethyiester, n-propylester, isopropylester, n-butylester, isobutylester, tert-butylester, morpholinoethylester or ⁇ , ⁇ -diethylg!ycoSamidoester.
  • a compound employed in the present invention has a hydroxy group
  • an acyloxy derivative prepared by reacting, the hydroxy! group with a suitable acylhalide or a suitable acid anhydride is exemplified as a prodrug.
  • an amide derivative prepared by reacting the amino group with a suitable acid halide or a suitable mixed anhydride is exemplified as a prodrug.
  • O-acyl-oxime or acyloxy derivative
  • the oxime -OH group of the compounds of the general formula (I) may also be in the form of O-alkyl-oxime such as, e.g., -0-CH 3 , -0-C 2 H 5 , -O-C3H7 or -O-(tert-butyl).
  • compositions comprising as an active ingredient one ore more compounds of the general formula (I), as defined herein above.
  • the pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, or antioxidants.
  • the pharmaceutical compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in Remington's Pharmaceutical Sciences, 20 th Edition.
  • the pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, rectal, nasal, topical, aerosol or vaginal administration.
  • Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersibie powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration.
  • Dosage forms for recta! and vaginal administration include suppositories and ovula.
  • Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler.
  • Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
  • the compounds according to the invention in particular the compounds of the general formula (I), or the above described pharmaceutical compositions comprising one ore more compounds of the general formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripheraily or at the site of desired action, including but not limited to one or more of: oral (e.g. as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e. g., using injection techniques or infusion techniques, and including, for example, by injection, e.g.
  • oral e.g. as a tablet, capsule, or as an ingestible solution
  • topical e.g., transdermal, intranasal, ocular, buccal, and sublingual
  • parenteral e. g., using injection techniques or infusion techniques, and including, for example, by injection, e.g.
  • implant of a depot for example, subcutaneously or intramuscularly
  • pulmonary e.g., by inhalation or insufflation therapy using, e.g., an aerosol, e.g. through mouth or nose
  • examples of such administration include one or more of: intravenously, intraarterially, intraperitoneal ⁇ , intrathecal ⁇ , intraventricularly, intraurethra!ly, intrasternally, intracranial ⁇ , intramuscularly or subcutaneously administering the compounds pharmaceutical compositions, and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmeilose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropy!methyicellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmeilose sodium and certain complex silicates,
  • Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diiuents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or it may be appiied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermaily or transdermaily administered, for example, by the use of a skin patch.
  • Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route.
  • they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride.
  • a preservative such as a benzylalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
  • a proposed, yet non-limiting dose of the compounds of the general formula (1) for administration to a human may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose.
  • the unit dose may be administered, for example, 1 to 4 times per day.
  • the dose will depend on the route of administration. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the subject or patient may be a eukaryote, an animal, a vertebrate animal, a mammal, a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), a murine (e.g. a mouse), a canine (e.g. a dog), a feline (e.g. a cat), an equine (e.g. a horse), a primate, a simian (e.g. a monkey or ape), a monkey (e.g. a marmoset, a baboon), an ape (e. g.
  • Non-limiting examples of agronomica!ly important animals are sheep, cattle and pig, while, for example, cats and dogs may be considered as economically important animals.
  • the subject/patient is a mammal; more preferably, the subject/patient is a human.
  • treatment of a disorder or disease is well known in the art. 'Treatment of a disorder or disease” implies that a disorder or disease is suspected or has been diagnosed in a patient/subject.
  • a patient/subject suspected of suffering from a disorder or disease typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e. diagnose a disorder or disease).
  • Treatment of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease (e.g. no deterioration of symptoms) or a delay in the progression of the disorder or disease (in case the halt in progression is of a transient nature only).
  • Treatment of a disorder or disease may also lead to a partial response (e.g. amelioration of symptoms) or complete response (e.g. disappearance of symptoms) of the subject/patient suffering from the disorder or disease.
  • “Amelioration" of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease or a delay in the progression of the disorder or disease. Such a partial or complete response may be followed by a relapse. It is to be understood that a subject/patient may experience a broad range of responses to a treatment (e.g. the exemplary responses as described herein above).
  • Treatment of a disorder or disease may, inter alia, comprise curative treatment (preferably leading to a complete response and eventually to healing of the disorder or disease) and palliative treatment (including symptomatic relief).
  • prophylaxis of a disorder or disease is well known in the art.
  • a patient/subject suspected of being prone to suffer from a disorder or disease as defined herein may, in particular, benefit from a prophylaxis of the disorder or disease.
  • Said subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition.
  • Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators.
  • a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in said patient/subject (for example, said patient/subject does not show any clinical or pathological symptoms).
  • the term “prophylaxis” comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician.
  • the terms “prophylaxis” and “prevention” are used herein interchangeably.
  • the test agent may, for example, be selected from nucleic acids, DNA, RNA, PNA, oligonucleotides, aptamers (Gold, Ann. Rev. Biochem.
  • RNAzymes see e.g., EP-B1 0 291 533, EP-A1 0 321 201 , EP-B1 0 360 257), antisense DNA, antisense oligonucleotides, antisense RNA, siRNA, RNAi, shRNA, amino acids, peptides, polypeptides, proteins, glycoproteins, lipoproteins, nucleoproteins, antibodies (Harlow and Lane “Antibodies, A Laboratory Manual", CSH Press, Cold Spring Harbor, 1988), monocloncal antibodies, polyclonal antibodies, immunoglobulins, affibodies (Hansson, Immunotechnology 4 (1999), 237-252; Henning, Hum Gene Ther.
  • anticalins EP-B-1 1 017 814
  • hormones peptide and protein hormones, non-peptide hormones, steroids, interieukins, interferons, cytokines, neurotransmitters, toxins, enzymes, polysaccharides, carbohydrates, lipids, lipopolysaccharides, vitamins, crown ethers, cyclodextrins, cryptands, calixarenes, aldehydes, thiols, amines, drugs, drugs of abuse, therapeutic agents, medicaments, pharmaceuticals, substrates, fragments, portions, components or products of microorganisms, metabolites of or antibodies to any of the above substances and the like.
  • EXPERIMENTAL PROCEDURES EXPERIMENTAL PROCEDURES
  • the 2-heterocycfic substituted chromones of the invention may be prepared using a common synthetic scheme illustrated in examp!e 1A, differing only in the starting phenol and heieroaryl ester (or heterocycloaikyl ester) compounds. (J. Med. Chem, 1999, 42 (1 1), 1881-1893).
  • a substituted 2'-hydroxyphenyl ketone of Formula I wherein R 1 , R 2 , R 3 , R 4 , and R s are as described and defined herein above, is reacted with an appropriate heteroaryl ester or heterocycloaikyl ester of Formula 11, wherein R is methyl or ethyl and A is as described and defined herein above, in a solvent such as pyridine and in presence of a strong base, preferentially sodium hydride, to yield the corresponding diketone III.
  • the diketone III is treated in strong acidic conditions especially with sulfuric acid (H 2 S0 4 ) in acetic acid (AcOH) to give the desired chromone IV.
  • 2-heterocyclic substituted chromones IV can also be prepared using a typical 3 steps procedure well known in the art for flavone derivatives synthesis (eg. J. Med. Chem. 2004, 47, 6466-6475 ; J. Org. Chem. 1993, 68, 7903-7905 ; US04065467).
  • Substituted 2 ! -hydroxyacetophenone I is reacted with a heieroaryl carboxylic acid or a heterocycloaikyl carboxylic acid of formula II, wherein R is hydrogen and A is as described and defined herein above, preferentially activated as acid chloride in pyridine to yield the corresponding ester of Formula V.
  • the ester is treated with a strong base, preferentially potassium hydroxide, in a suitable solvent such as pyridine to perform the mixed Claisen rearrangement and to give the corresponding diketone of formula ill.
  • a strong base preferentially potassium hydroxide
  • a suitable solvent such as pyridine
  • the diketone III is then treated in strong acidic conditions especially with sulfuric acid in acetic acid to give the desired chromone of formula IV.
  • the chromone oximes of the invention of formula VI in particular the compounds of the general formula (1), may be prepared by the foliowing procedure illustrated in example 73.
  • the chromone derivative of formula IV which may be prepared according to method A or method B as described herein above, is treated with hydroxyiamine hydrochloride in pyridine or methanol under microwave irradiation to yield directly the ox ' sme compound of formula VI.
  • the chromone oximes of the invention of formula VI may also be obtained using the following two steps procedure illustrated in example 1 B and 1 C:
  • the appropriate chromone of formuia IV which may be prepared according to method A or method B as described herein above, is reacted with ferf-butyi hydroxyiamine hydrochloride in a suitable solvent such as methanol and under microwave irradiation to yield the protected ie/f-buty!oxime product of formula VII (step 1).
  • the protected oxime VII is treated with a Lewis acid such as titanium tetrachloride in an inert solvent, preferentially dich!oromethane, to give the desired free oxime of formuia VI (WO2004/52S69) (step 2).
  • a Lewis acid such as titanium tetrachloride in an inert solvent, preferentially dich!oromethane
  • the compounds of the present invention can be radio-labeled by carrying out their synthesis using precursors comprising at least one atom which is a radioisotope.
  • precursors comprising at least one atom which is a radioisotope.
  • radioisotopes of carbon atoms, hydrogen atoms, sulfur atoms, or iodine atoms are employed, such as, e.g., 4 C, 3 H, 35 S, or 125 1.
  • Compounds labeled with 3 H can also be prepared by subjecting a compound of the invention to a hydrogen exchange reaction such as, e.g., a platinum-catalyzed exchange reaction in tritiated acetic acid (i.e., acetic acid comprising 3 H instead of 1 H), an acid-cata!yzed exchange reaction in tritiated trifluoroacetic acid, or a heterogeneous-catalyzed exchange reaction with tritium gas.
  • a hydrogen exchange reaction such as, e.g., a platinum-catalyzed exchange reaction in tritiated acetic acid (i.e., acetic acid comprising 3 H instead of 1 H), an acid-cata!yzed exchange reaction in tritiated trifluoroacetic acid, or a heterogeneous-catalyzed exchange reaction with tritium gas.
  • a hydrogen exchange reaction such as, e.g., a platinum-catalyzed exchange reaction in tritiated
  • Figure 2 Effect of the compound of Example 85 in the marble burying test in the mouse, administered p.o. at 100 or 300 mg/kg (see Example 173). The figure shows the mean number of marbles burying in each group of animals, as also explained in Example 173.
  • Example 1C 2 ⁇ isoquinolin-3-yl-chromen-4 ⁇ one oxime
  • Example 2A 7-bromo-2-isoquinolin-3-yl ⁇ 7 ⁇ bromo-chromen ⁇ 4-one wasprepared using the method A illustrated in example 1A. 4 > -Bromo-2'- hydroxyacetophenone (627 mg, 2.91 mmol) and methy!-2-isoquinoline-3-carboxylate (600 mg, 3.21 mmo!), was treated first with sodium hydride (60% in mineral oil, 350 mg, 8.73 mmol) in dry pyridine (2x6 ml) at 90°C for 15 min. and then with sulfuric acid (73 ⁇ ) in acetic acid (15 ml) at 100°C for 45 min.
  • Example 28 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared using the method D (step 1) illustrated in example 1 B.
  • 7-bromo- 2-isoquinoiin-3-yl-chromen-4-one (682 mg, 194 mmol) was treated with O-ferf-butyl- hydroxylamine hydrochloride (486 mg, 3.87 mmol) in methanol (9 ml) under microwave irradiation to yield 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-feri-butyl oxime (629 mg, 77%) as a yellow solid after purification by flash chromatography over silica gel (cyc!ohexane/ethyl acetate : 50/50).
  • Example 2C 7-bromo-2-isoquinolin-3-yl-chromen-4-one oxime was prepared using the method D, (step 2) illustrated in example 1C.
  • 7-bromo-2- isoquinoiin-3-yl-chromen-4-one O-ferf-butyl oxime 150 mg, 0.35 mmol
  • 7-bromo-2-isoquinoiin-3-yl-chromen-4-one O-ferf-butyl oxime 150 mg, 0.35 mmol
  • 7-bromo-2-isoquinoiin-3-yl-chromen-4-one O-ferf-butyl oxime 150 mg, 0.35 mmol
  • 7-bromo-2-isoquinoiin-3-y chromen-4-one oxime 56 mg, 37%) as a beige solid after recrystal!ization in hot chioroform.
  • the product was isolated as a 90/10 mixture of Z/E oxime isomers.
  • Example 3A 7-bromo ⁇ 2 ⁇ isoquinolin-3-yl ⁇ 6 ⁇ methyl-chromen-4 ⁇ one was prepared using the method A illustrated in example 1A. 4'- ⁇ -5'- ⁇ 1-2'- hydroxyacetophenone (556 mg, 2.42 mmol) and methyi-2-isoquinoiine-3-carboxySate (500 mg, 2.67 mmol), was treated with sodium hydride (60% in mineral oil, 291 mg, 7.28 mmol) in dry pyridine (13 ml) at 90°C for 15 min. and then with sulfuric acid (100 ⁇ ) in acetic acid (15 ml) at 100°C for 30 min. to yield 7-bromo-2-isoquinolin-3-yl-6- methyl-chromen-4-one (738 mg, 83%) as beige solid.
  • sodium hydride 50% in mineral oil, 291 mg, 7.28 mmol
  • dry pyridine 13 ml
  • sulfuric acid 100 ⁇
  • acetic acid 15
  • Example 3B 7-bromo-2-isoquinolin-3-yl-6-methyl-chromen-4-one O-tert- butyl oxime was prepared using the method D (step 1 ) illustrated in example 1 B.
  • 7- bromo-2-isoquinoltn-3-yl-6-methyi-chromen-4-one (730 mg, 1.98 mmol) was treated with O-ie f-butyl-hydroxylamine hydrochioride (500 mg, 3.98 mmol) in methanol (13 ml) under microwave irradiation to yieid 7-bromo-2-isoquinolin-3-yl-chromen-4-one O- ferf-butyl oxime (383 mg, 44%) as a yellow solid after purification by flash chromatography over silica gei (cyclohexane/Chloroform/ethyl acetate : 96/4/0; 50/50/0; 0/100/0 0/
  • Example 3C 7 ⁇ bromo-2-isoquinolin ⁇ 3-yl-6-methyl-chromen-4 ⁇ one oxime was prepared using the method D, (step 2) illustrated in example 1 C.
  • 7-bromo-2- isoquinolin-3-yl-6-methyl-chromen-4-one O-tert-butyl oxime (100 mg, 0.22 mmol) was treated with a 1 M solution of titanium tetrachloride (0.68 ml, 0.68 mmol) in dfchloromethane (4 ml) to yield 7-bromo-2-isoquinolin-3-yi-6-methyl-chromen-4-one oxime (79 mg, 94%) as a yellow solid after recrystailization in hot chloroform.
  • the compound was isolated as a 88/12 mixture of 2/E oxime isomers.
  • Example 4A 6-bromo ⁇ 2-isoquinolin-3-yl-chromen-4-one was prepared using the method A illustrated in example 1A. 5'-Bromo-2'-hydroxyacetophenone (1.56 g, 7.28 mmoi) and methyl-2-isoquinoline-3-carboxyiate (1.50 g, 8.0 mmol), was first treated with sodium hydride (60% in mineral oil, 960 mg, 24.0 mmol) in dry pyridine (40 ml) at 90°C for 15 min.
  • Example 4B 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared using the method D (step 1) illustrated in example B. 6-bromo- 2-isoquinolin-3-yl-chromen-4-one (600 mg, 1.7 mmol) was treated with O-ferf-butyl- hydroxyfamine hydrochloride (427 mg, 3.4 mmol) in methanol (10 mi) under microwave irradiation to yield 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ierf-butyl oxime (546 mg, 76%) as a yellow solid.
  • Example 4C 6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime was prepared using the method D, (step 2) illustrated in example 1C.
  • 6-bromo-2- isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime 50 mg, 0.12 mmol
  • 6-bromo-2- isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime 50 mg, 0.12 mmol
  • yieid 6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime 37 mg, 85%
  • the compound was isolated as a 98/2 mixture of Z E oxime isomers.
  • Example 7 6, 8-difluoro-2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 31% overall yield as a beige solid and as a 96/4 mixture of Z/E oxime isomers.
  • Example 9 4-fiuoro-2-isoquinolin-3-yl-chromen-4-one-(Z)- oxime was isolated in 54% overall yield as a yellow solid.
  • Example 10 2-isoquinolin-3-yl-6-trifluoromethoxy'Chromen-4-one oxime was isofated in 15% overall yield as a yellow powder and as a 95/5 mixture of Z/E oxime isomers.
  • Example 11 2-isoquinolin-3-yl-6-trifluoromethyl-chromen-4-one oxime was isolated in 10% overall yield starting from 2'-hydroxy-5'-trif!uoromethyl- aceiophenone (example 1 1A) and methyl isoquinoline-3-carboxylate. The yellow solid product was a 95/5 mixture of Z/E oxime isomers.
  • Example 11 A 2'-hydroxy-5 ! -trifluoromethyl-acetophenone was prepared using the following process (JACS, 2004, 126(3), 712-713). To a cooled solution of 2'- methoxy-5'-trifluoromethy!-acetophenone (650 mg, 2.98 mmol) at -78°C in dry dichloromethane (40 ml) was slowly added a 1 M solution of boron trichloride in dichloromethane (7.5 ml, 7.5 mmol) keeping the internal temperature below -70°C. The brown-orange solution was slowly warmed up to room temperature within 2 hours.
  • Example 11 B 2'-methoxy-5'-trifluoromethyl-acetophenone was prepared as follow ⁇ US2003/0109574): in a water bath at room temperature, to a solution of trifluoromethane sulfonic acid (5 ml, 56.7 mmol) under argon was slowly added a solution of 4-trifiuoromethyl-anisol (2.0 g, 1 1.35 mmol) in acetic anhydride (2.15 ml, 22.7 mmol). The resulting dark mixture was stirred for 3 hours at room temperature then poured onto ice water (26 ml). The product was extracted several times with ether.
  • Example 12 2-(7-fluoro-isoquinolin-3 ⁇ yl)-chromen-4-one oxime was isolated in 11% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 7-fiuoro-isoquinoline-3-carboxylate (example 12A) as a yellow solid and a 95/5 mixture of Z/E oxime isomers.
  • Example 12A methyl 7-fluoro-isoquinoline-3 ⁇ carboxylate: A sealed tube was charged with 2-bromo-5-fluorobenzaidehyde (300 mg, 1.47 mmol), dry dimethylformamide (1.5 ml), triethylamine (0.7 ml, 5.02 mmol), methyl 2- acetamidoacryiate (273 mg, 1.91 mmol), fris-o-toiyi-phosphine (89 mg, 0.29 mmol) and palladium acetate (33 mg, 0.14 mmol). The solution was degassed with Argon for 10 min. and then warmed at 110°C for 4.5 hours.
  • Example 13 2-(7-methoxy-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 3% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 7-fluoro-isoquinoline-3-carboxyiate (example 12A).
  • the brown solid product was a 80/20 mixture of Z/E oxime isomers.
  • Example 14 2-(6.7-dimethoxy-isoquinolin-3-yl)'Chromen-4-one oxime was isolated in 8% overai! yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 6,7-dimethoxy-isoquinoline-3-carboxylate (US054777252) as a yellow solid and a 95/5 mixture of ZJE oxime isomers.
  • Example 15 2-(6-methyl-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 35% overall yield using method A and D starting from 2 ! -hydroxy- acetophenone and methyl 6-methyl-isoquinoline-3-carboxyiate (example 15A) as a yellow solid and a 95/5 mixture of ZJE oxime isomers.
  • Example 15A methyl 6-methyl ⁇ isoqumolm " e ⁇ 3 ⁇ carboxylate was prepared using the procedure described in example 12A: 2-bromo-4-methytbenzaldehyde was reacted with methyl 2-acetamidoacrylate at 1 10°C for 18 hours to yieid methyl 6- methyi-isoquinoline-3-carboxylate (50%) as brown solid.
  • Example 16 2-(7-chloro-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 20% overall yield using method A and D starting from 2'-hydroxy- acetophenone and methyl 7-chloro-isoquinoline-3-carboxylate (example 16A) as pale a yellow solid and a 90/10 mixture of Z/E oxime isomers.
  • Example 16A methyl 7-chloro-isoquinoline-3-carboxy!ate was prepared using the procedure described in example 12A: 2-bromo-5-chlorobenzaldehyde was reacted with methyl 2-acetamidoacrylate at 110°C for 24 hours to yield methyl 7- chloro-isoquinoiine-3-carboxylate (25%) as brown solid.
  • Example 17 2-(5-bromO"isoquinolin-3 ⁇ yl) ⁇ chromen-4-one oxime was isolated in 15% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 5-bromo-isoquinoiine-3-carboxylate (example 17A) as a yellow solid and a 90/10 mixture of Z/E oxime isomers.
  • Example 17A methy-5-bromo-isoquinoline-3-carboxylate was prepared using the foilowing procedure describe in US054777252 for methyl-6,7-dimethoxy- isoquinoiine-3-carboxylate: 5-bromo-1 ,2,3,4-tetrahydroisoquinoline-3-carboxylic acid methyl ester (880 mg, 3.26 mmo! was treated with 2,3-Dichloro-5,6- dicyanobenzoquinone (1.62 mg, 7.19 mmol) in dry tetrahydrofuran (19 ml) at reflux temperature for 18 hours. The cooled dark mixture was filtered and the solid washed with dichloromethane.
  • the filtrate was treated with a 1 M aqueous sodium hydroxide solution and the aqueous layer was extracted twice with dichloromethane. The organic layer was washed with water, brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-100%) to yield methyl 5-bromo- isoquinoiine-3-carboxylaie (793 mg, 91 %) as beige solid.
  • Example 18 2-(5-hydroxy-isoquinotin-3-yl)-chromen-4-one oxime was isolated in 14% yield using method D (step 2), starting from 2-(5-hydroxy-isoquinolin- 3-yi)-chromen-4-one O-tert-butyl oxime (example 8A) as orange solid and as a 80/20 mixture of 27 E oxime isomers.
  • Example 18A 2-(5-hydroxy-isoquinolin-3-yl)-chromen-4-one O-tert-butyl oxime was prepared using a palladium coupling procedure describes by K. W. Anderson (JACS, 2006, 128, 10694-10695):
  • reaction mixture was diluted with water and extracted with ethyl acetate. The organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-40%) to yield 2-(5-hydroxy-isoquinolin-3-yl)- chromen-4-one O-ierf-butyi oxime (230 mg, 73%) as a yellow solid.
  • Example 19 2-(5-methoxy-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 94% yield using method D (step 2), starting from 2-(5-methoxy-isoquinolin- 3-yl)-chromen-4-one O-ferf-butyS oxime (example 19A) as a yellow solid and as a 90/ 0 mixture of Z/E oxime isomers.
  • Example 21 2 ⁇ isoquinolin-3-yl ⁇ 7-((E)-styryl) ⁇ chromen ⁇ 4"
  • One oxime was isolated in 55% yield using method D (step 2), starting from 2-!soquinolin-3-yl-7-((E)- styryl)-chromen-4-one O-ferf-butyl oxime (example 21 A) as a pale yellow solid and as a 90/10 mixture of Z/E oxime isomers
  • Example 21A 2-isoquinolin-3-yl-7-((E)styryl) ⁇ chromen ⁇ 4-one O-tert-butyl oxime:
  • the vial was sealed and the mixture was heated under microwave irradiation to 150°C for 5 min. After cooling, the solution was diluted with dichioromethane and absorbed over silica gel for purification by flash chromatography (gradient cyclohexane/dichloromethane: 0-80 to yield 2-lsoquinolin-3-yl-7-((E)-styryl)-chromen-4-one O-ferf-butyl oxime (160 mg, 76%) as a yellow solid.
  • Example 22 2-isoquinolin-3-yl-7-phenethyl ⁇ chromen-4-one oxime was isolated using method D (step 2), starting from 2-lsoquinolin-3-yl-7-phenethyl- chromen-4-one O-ferf-butyl oxime (example 22A) as a yellow solid and as a 95/5 mixture of Z/E oxime isomers
  • Example 22A 2-isoquinolin -3-yl-7-phenethyl-chromen-4-one O-tert-butyl oxime:
  • Example 23 7-ethynyl ⁇ 2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 72% yield using method D (step 2), starting from 7-ethynyl-2-isoquinoiin-3- yl-chrornen-4-one O-re f-butyl oxime (example 23A) as a yellow solid and as a 90/10 mixture of Z E oxime isomers.
  • Example 23 was purified by preparative HPLC (gradient 35-20% Water/acetonitrile+0.05% trifluoroacetic acid)
  • Example 24 2'isoquinolin-3-yl'7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime was isolated in 42% yield using method D (step 2), starting from 2-isoquinolin- 3-yl-7-(pyridin-2-yl-ethynyl)-chromen-4-one O-ferf-butyl oxime (example 24A) as a yel!ow solid after recrysta!lization in hot chloroform.
  • Example 25 2-isoquinolin-3-y!-7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime was prepared in 18% overall yield using the method described in example 23, starting from 7-bromo-2-isoquinoiin-3-y[-6-methyl-chromen-4-one O-ferf-butyl oxime (example 3B) and phenyiacetylene.
  • the titie compound was isolated as a yellow solid and as a 95/5 mixture of Z E oxime isomers.
  • Example 26 2-isoquinolin-3-yl-7-(pyridin ⁇ 4-yl)ethynyl ⁇ chromen-4-one oxime was prepared in 6% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (exampie 2B) and 4-eihynylpyridine. The title compound was isolated as a yellow solid and as a 95/5 mixture of Z/E oxime isomers.
  • Example 27 7 ⁇ (4 ⁇ dimethylamin ophenyl) ethynyl- 2-isoquinoliti-3-yl- chromen-4-one oxime was prepared in 8% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (example 2B) and 4-dimethylaminophenylacetyiene. The title compound was isolated as an orange solid and as a 95/5 mixture of Z/E oxime isomers.
  • Example 28 2-iso quin olin-3-yl- 7-( 3-methoxyphenyl) ethynyl-chrom en-4- one oxime was prepared in 22% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyi oxime (example 2B) and 3-methoxyphenyiacetyiene. The title compound was isolated as an orange solid and as a 93/7 mixture of Z/E oxime isomers.
  • Example 29 7-( 3-aminoph enyl) ethynyl-2-iso quin olin-3-yl-chrom en-4-one oxime was prepared in 17% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyi oxime (example 2B) and 3-aminophenylacetylene. The title compound was isolated as a ye!low solid and as a 95/5 mixture of Z/E oxime isomers.
  • Example 30 7-(3-hydroxyphenyl)ethynyl-2-isoquinolin-3-yl-chromen-4- one oxime was prepared in 28% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one Oferf-buty! oxime (example 2B) and 3-hydroxyphenylacetyiene. The title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers.
  • Example 31 2-isoquinolin-3-yl-7-(4-methoxyphenyl)ethynyl-chromen ⁇ 4 ⁇ one oxime was prepared in 9% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-fert-butyl oxime (example 2B) and 4-methoxyphenyiacetylene.
  • the title compound was purified by preparative HPLC (gradient 35-20% Water/acetonitrile+0.05% trifluoroacetic acid) and isolated as a yellow solid and as a 95/05 mixture of Z/E oxime isomers.
  • Example 32 7-( 2-chlorophenyl) ethynyi-2-isoquin olin-3-yl-chrom en-4-one oxime was prepared in 10% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3 ⁇ yl-chromen-4-one O-iert-butyl oxime (example 2B) and 2-chiorophenylacetylene. The title compound was isolated as an orange solid and as a 80/20 mixture of Z/E oxime isomers.
  • Example 33 7-( 3- im ethylamino-prop- 1-ynyl) -2-iso quin olin-3-yl-chrom en- 4-one oxime was prepared in 20% overaii yield using the method described in example 24, starting from 7-bromo-2-isoquino!in-3-yl-chromen-4-one O-ierf-butyl oxime (example 2B) and N,N-dimethylamino-2-propyne.
  • the titie compound was purified by preparative HPLC (gradient 70 - 55% Water/acetonitrile + 0.05% trifluoroacetic acid) and isolated as an orange solid and as a 85/15 mixture of Z/E oxime isomers.
  • Example 34 6-cyclopropyl-2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 60% yield using method D (step 2), starting from 6-cyclopropyl-2- isoquinolin-3-yl-chromen -4-one O-tert-butyl oxime (example 34A) as a yellow solid after purification by preparative HPLC (gradient 50 - 35% Water/acetonitrile + 0.05% trifluoroacetic acid). The title compound was isolated as a 85/15 mixture of Z/E oxime isomers.
  • Example 34 6'Cyclopropy!-2-isoquinolin-3-yl-chromen-4-one O-tert- butyl oxime
  • Example 35 2-isoquinolin-3-yl-6-(pyrroiidin-1-yl)'Chromen-4-one oxime was isolated in 63% yield using method D (step 2), starting from 2-isoquinolin-3-yl-6- (pyrrolidin-1-y!)-chromen-4-one O-ferf-butyl oxime (example 35A) as an orange solid after trituraiing in hot chloroform. The title compound was isolated as a 97/3 mixture of Z/E oxime isomers.
  • Example 36 2 ⁇ isoquinolin-3-yl ⁇ 6-(vinyl)-chromen ⁇ 4-one oxime was isolated in 40% yield using method D (step 2), starting from 2-isoquinolin-3-yl-6- (vinyl)-chromen-4-one O-terf-butyl oxime (example 36A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate : 0- 40%)
  • Example 36A 2-isoquinoiin-3-yl-6-(vinyl)-chromen-4-one O-tert-butyl
  • Tributyi(vinyl)tin (76 ⁇ , 0.26 mmol) and tetrakis(t phenylphosphine)palladium 13 mg, 0.012 mmol
  • Example 37 6-ethyl ⁇ 2 ⁇ isoquinolin-3-yl-chromen-4-one oxime was isolated in 50% yield using method D (step 2), starting from 6-ethyl-2-isoquino!in-3-yi-chromen- 4-one O-ferf-butyl oxime (example 37 A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate : 0-20%)
  • Example 37 6-ethyl ⁇ 2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime
  • Example 38 6 ⁇ cyano-2-isoquinoIin-3-yI ⁇ chromen-4-one oxime was prepared in 63% yield using method D (step 2), starting from 6-cyano-2-isoquinolin-3- yl-chromen-4-one O-ferf-butyl oxime (example 38A) as a paie yellow solid after recrystailization in hot chloroform. The title compound was isolated as a 90/10 mixture of Z/E oxime isomers.
  • Example 38 A 6-cyano-2-isoquinolin-3-yl-chromen ⁇ 4-one O-tert-butyl oxime
  • Example 39 6-dimethylamino-2-isoquinolin-3-yl-chromen-4-one oxime was prepared in 51 % overall yield following the method described in example 35, starting from 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime and dimethy!amine (2.0 M in tetrahydrofuran). The title compound was isolated as a pale yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-60%).
  • Example 40 2-iso quin olin-3-yl-6-( morpholin-4-yl-m ethyl)-chrom en-4-one oxime was prepared in 30% yield using method D (step 2), starting from 2-isoquinolin- 3-yl-6-(morpholin-4-yl-methyl)-chromen-4-one O-ferf-butyi oxime (example 40A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-100%). The title compound was isolated as a 70/30 mixture of Z/E oxime isomers.
  • Example 40A 2-isoquinolin-3-yt-6-(morpholin ⁇ 4-yl ⁇ methyl) ⁇ chromen-4- one O-tert-butyl oxime
  • the mixture was diluted with ethyl acetate, washed with water, a saturated aqueous solution of ammonium chloride, and extracted three times with a 1 N solution of hydrochloric acid.
  • the aqueous layers were neutralized with a 6N solution of sodium hydroxide and extracted with ethyl acetate.
  • the organic layers were dried over sodium sulfate and concentrated to dryness to yield 2-isoquinolin-3-yl-6-(morpholin-4-yl-methyl)-chromen- 4-one O-ferf-butyl oxime (10 mg, 10%) as a brown residue that was used without further purification.
  • Example 41 6-hydroxy-2-isoquinolin-3-yl ⁇ chromen-4-one oxime was prepared in 56% yield using method D (step 2), starting from 6-hydroxy-2-isoquinolin- 3-yl-chromen-4-one O-ferf-butyl oxime (example 41 A) as a yellow solid after purification by flash chromatography over silica gel (cyc!ohexane/ethyl acetate: 0- 80%). The title compound was isolated as a 95/5 mixture of Z/E oxime isomers.
  • Example 41A 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared in 89% yieid following the method describe in example 18A, starting from 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0- 20%). The title compound was isolated as a yellow solid.
  • Example 42 2-isoqum " oHn-3-yl-6-methoxy-chromen-4 ⁇ one oxime was prepared in 62% yield using method D (step 2), starting from 2-isoqutnoiin-3-yl-6- methoxy-chromen-4-one O-ferf-butyl oxime (example 42A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0- 80%, then dichioromethane/methanol: 0-10%). The title compound was isolated as a 90/10 mixture of Z/E oxime isomers.
  • Example 42A 2-isoquinolin-3-yl-6-methoxy-chromen-4-one O-tert-buty! oxime was prepared in 67% yield following the method described in example 19A, starting from 6-hydroxy ⁇ 2-isoquinolin-3-yl-chromen-4-one O-ierf-butyl oxime (example 41 A). The title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-20%):
  • Example 43 2-isoquinolin-3-yl- 6-( 2-methoxy-ethoxy)-chrom en-4-one oxime was prepared in 67% overall yield using the method described in example 42 and 42A, starting from 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (example 41A) and bromoethylmethylether instead of iodomethane.
  • the title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-80%), and as a 85/15 mixture of Z E oxime isomers.
  • Example 44 6-(2-dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4- one oxime was prepared in 66% yield using method D (step 2), starting from 6-(2- dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4-one O-iert-butyl oxime (example 44A) as an orange solid and as a 90/10 mixture of Z/E oxime isomers.
  • Example 44A 6-(2-dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4- one O-tert-butyl oxime
  • Example 45 2-isoquinolin-3-yl- 6-[3-(4-methyl-piperazin- 1-yl)- propylamino]-chromen-4 ⁇ one oxime was prepared in 85% overall yield using the method described in examples 35 and 35A, starting from 6-bromo-2-isoquinolsn-3-yl- chromen-4-one O-ferf-butyl oxime (example 4B) and 3-(4-methyl-piperazin-1 -yl)- propytamine. The title compound was isolated as a yellow solid and as a 95/5 mixture of Z E oxime isomers.
  • Example 46 2-isoquinolin-3-yl-6-[2-(4-methyl-piperazin-1-yl)-ethoxyJ- chromen-4-one oxime was prepared in 49% overall yield using the method described in examples 44 and 44A, starting from 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O- fe/ -butyl oxime (example 41 A) and 2-(4-methyl-piperaztn-1-yl)-ethanoi. The title compound was isolated as a yellow solid and as a 90/10 mixture of ZE oxime isomers.
  • Example 47 2-isoquinolin-3-yl-7-phenyl-chromen-4-one oxime was isolated in 30% yield using method D (step 2), starting from 2-lsoquinolin-3-yl-7- phenyl-chromen-4-one O-iert-butyl oxime (example 47A) as a pale yellow solid after recrystallization in hot chloroform.
  • Example 47A 2-isoquinolin-3-yl-7-phenyI-chromen-4-one O-tert-butyl oxime
  • reaction mixture was poured onto a saturated solution of ammonium chloride and extracted with ethyl acetate.
  • the extracts were washed with brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography ⁇ gradient cydohexane/dichloromethane: 0-80%) to yield 2-lsoquinolin-3-yl-7-phenyl-chromen-4- one O-ferf-butyl oxime (135 mg, 91 %) as a yellow solid.
  • Example 48 7-(4-biphenyl) ⁇ 2-i$oquinoiin ⁇ 3-yl-chromen-4-one oxime was isolated in 26% overall yield using the method described in examples 47 and 47A, starting from 7-bromo-2-lsoquinolin-3-yl-chromen-4-one O-terf-butyl oxime and 4- biphenylboronic acid. The title compound was isolated as a beige solid after triturating in hot chloroform and as a 80/20 mixture of Z/E oxime isomers.
  • Example 49 2-isoquinoiin-3-yl-7-[3-(4-methyl-piperazin-1-yl)- propylaminoJ'Chromen-4-one oxime was prepared in 19% overall yield using the method described in examples 45, starting from 7-bromo-2-isoquinolin-3-yl-chromen- 4-one O-terf-butyl oxime (example 2B) and 3-(4-methyl-piperazin-1-yl)-propylamine. The title compound was isolated as a yellow solid.
  • Example 50 2-lsoquinolin-3-yl-7- ⁇ 3-[2-(4 ⁇ methyl-piperazin-1-yl)-ethoxy]- phenylethynyl ⁇ -chromen-4-one oxime was prepared in 9% overall yield Using the method described in examples 44 and 44A, starting from 7-(3-hydroxyphenyl)ethynyl- 2-isoquinoiin-3-yl-chromen-4-one O-terf-butyl oxime (te/f-buiyl protected oxime of example 30) and 2-(4-methyi-piperazin-1-yl)-ethanol. The title compound was isolated as a yellow solid and as a 90/10 mixture of 2 E oxime isomers after Preparative HPLC purification (gradient 65-50% Water/aceionitrile+0.05% trifluoroacetic acid).
  • Example 51 2-lsoquinolin-3-yl-7- ⁇ 3-methylaminophenylethynyl ⁇ - chromen-4-one oxime was prepared using method D (step 2) starting from 2- isoquinolin-3-yl-7- ⁇ 3-methylaminophenyiethynyl ⁇ -chromen-4-one O-tert-butyi oxime (example 51 A).
  • the title compound was isolated as a yellow solid and as a 90/10 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 50-35% Wa ⁇ er/aceionitrile ⁇ 0.05% trifluoroacetic acid).
  • Example 51 A 2-!s oquinolin-3-yl-7- ⁇ 3-methylamin oph enylethynyl ⁇ - chromen-4-one O-tert-butyl oxime.
  • a solution of 7-(3-aminophenylethynyl)-2- (lsoquinoiin-3-yl)-chromen-4-one O-fert-butyl oxime (iert-butyi protected oxime of example 29) (127 mg, 0.286 mmol) in dimethy!formamide (3 ml) was added to a cold suspension of sodium hydride (60% in mineral oii, 24 mg, 0.608 mmol) in DMF (1 ml).
  • Example 52 7-(4-Hydroxy-b ut-1-ynyl) -2-isoquinolin-3-yl-chrom en-4-on e oxime was prepared in 10% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyS oxime (example 2B) and 3-butyn-1 -ol. The title compound was isolated as a yellow solid and as a 70/30 mixture of Z/E oxime isomers.
  • Example 53 2-!soquinolin-3-yl- 7-[3-(2-methoxy-ethoxy)'phenylethynyl] ⁇ chromen-4-one oxime was prepared in 48% overall yield using the methods described in examples 42 and 42A, starting from 7-(3-hydroxyphenyl)ethynyl-2- isoquinolin-3-yl-chromerv4-one O-fert-butyl oxime (ferf-butyl protected oxime of example 30) and bromoethylmethylether instead of iodomethane.
  • the title compound was isolated as a yeiiow solid and as a 90/10 mixture of Z/E oxime isomers after purification by flash chromatography over silica gel (gradient cyciohexane/ethyl acetate: 0-50%).
  • Example 54 2-ls oquinolin-3-yl- 7-[3-(2-meth oxy-ethoxy)-prop- 1-ynylJ- chromen-4-one oxime was prepared in 7% overall yield using the methods described in examples 42 and 42A, starting from 7-(3-hydroxy-prop-1 -ynyl)-2-lsoquinolin-3-yl- chromen-4-one O-ierf-buty! oxime (example 54A) and bromoethylmethyiether. The title compound was isolated as a beige solid and as a 90/10 mixture of Z E oxime isomers after triturating in diethyl ether.
  • Example 54 A 7-(3-hydroxy-prop-1-ynyl)-2-isoquinolin-3-yl-chromen-4- one O-tert-butyl-oxime was prepared in 58% overall yield using the method described in example 23A, starting from 7-bromo-2-isoquinoiin-3-yl-chromen-4-one O- ferf-butyl oxime (example 2B) and 2-propyn-1 -o!.
  • Example 55 7-but-3-en-1-ynyl-2-lsoquinolin-3-yl-chromen-4-one oxime was prepared using method D (step 2) starting from 7-(but-3-en-1-ynyl)-2-lsoquinolin- 3-yi-chromen-4-one O-ferf-butyl oxime (example 55A). The title compound was isolated in 93% yield as a yellow solid and as a 90/10 mixture of Z/E oxime isomers.
  • Example 55A 7-(but-3-en-1-ynyl)-2-lsoquinolin-3-yl-chromen-4-one O- tert-butyl oxime.
  • 7-(4-Hydroxy-but-1 -ynyl)-2-isoquinolin-3-yl- chromen-4-one O-ferf-butyl oxime (f erf-butyl protected oxime of example 52) 200 mg, 0.485 mmol
  • triethylamine (0.15 ml, 1 .067 mmol
  • dichloromethane 3 ml
  • the methansulfonic ester was dissolved in dimethyiformamtde (4 ml) and treated with potassium carbonate (201 mg, 146 mmol) and methyl piperazine (73 mg, 0.728 mmol) and the mixture was stirred at 40°C for 20 hours. The mixture was poured onto a saturated solution of sodium hydrogenocarbonate.
  • reaction mixture was extracted with dichloromethane, washed with water, brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography (gradient cyclohexane/dichioromethane; 0-45%) to yield 7-(but-3-en-1 -yny!-2-lsoquinolin-3-yi ⁇ chromen-4-one O-rert-butyl oxime (156 mg, 82%) as a yellow solid.
  • Example 56 2"isoquinolin ⁇ 3-yl-7-methoxy-chromen-4-one oxime was prepared in 18% overall yield using the methods described in examples 41 and 42, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-rerf-butyl oxime (example 2B). The title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers.
  • Example 57 2-isoquinolin-3-yl-7-(2-methoxy-ethoxy) ⁇ chromen-4 ⁇ one oxime was prepared in 22% overall yield using the methods described in examples 41 and 43, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ie/ -butyl oxime (example 2B).
  • the title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers after purification by flash chromatography over silica ge! (gradient cyclohexane/ethyl acetate: 20-50%).
  • Example 58 7-cyano-2-isoquinolin-3-yl-chromen-4-one oxime was prepared in 8% overall yield using the methods described in examples 38 and 38A, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-iert-butyl oxime (example 2B). The title compound was isolated as a beige solid after purification by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-60%).
  • Example 59 2-lsoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-yl)-prop-1 ⁇ ynyl] ⁇ chromen-4-one oxime was prepared in 67% yield using method D (step 2) starting from 2-isoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-y!-prop-1-ynyl]-chromen-4-one O- ferf-butyl oxime (example 59A). The title compound was isolated as a ye!low solid and as a 95/5 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 75-60% Water/acetonttrile+0.05% trifluoroacetic acid).
  • Example 59A 2-lsoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-yl)-prop-1- ynyl]-chromen -4-one O-tert-butyl oxime.
  • 7-(3-Hydroxy-prop-1- ynyl)-2-isoquinolin-3-yl-chromen-4-one O-terf-butyi oxime (example 54A) (230 mg, 0.577 mmoi) and triethylamine (0.18 ml, 1.27 mmol) in dichioromethane (3 m!) was added dropwise methanesuSfonyl chloride (90 ⁇ , 1.15 mmo!).
  • the reaction mixture was stirred for 4 hours at 0°C and poured onto a saturated solution of ammonium chloride.
  • the product was extracted with dichioromethane, washed with brine, dried over sodium sulfate and concentrated to dryness.
  • the residue methansulfonic ester was dissolved in acetone (3 ml) and treated with potassium carbonate (239 mg, 1.73 mmoi) and methyl piperazine (87 mg, 0.866 mmol) and the mixture was stirred at 25°C for 20 hours.
  • Example 60 2-(7-hydroxy-isoquinolin ⁇ 3-yl)-chromen-4-one oxime was prepared in 51 % yield using the method described in example 18, starting from 2-(7- chloro-isoquinolin-3-yi) ⁇ chromen-4-one oxime (example 16) instead of 2-(5-bromo- isoquinolin-3-yl)-chromen-4-one O-fert-butyl oxime. The title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-80%).
  • Example 61 2-[2,6]Naphthyridin-3-yl-chromen-4-one oxime was isolated using methods A and D starting from 2'-hydroxy-acetophenone and [2,6]Naphthyridine-3-carboxylic acid methyl ester (example 61A) as a yellow solid after preparative HPLC purification (gradient 75-60% Water/acetonitrile+0.05% trifluoroacetic acid).
  • Example 62 2-[1,6]Naphthyridin-3-yl-chromen-4-one oxime was isolated in 7% overall yield using methods A and D starting from 2'-hydroxy-ace ⁇ ophenone and [1 ,6]Naphthyridine-3-carboxylic acid methyl ester (example 62A) as a yellow solid and a 80/20 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 45-30% Water/MethanoS+0.05% trifluoroacetic acid).
  • Example 62A methyl 6-methyl-isoquinoline-3-carboxylate was prepared in 43 % yield according to the method described in example 61 A, starting from 3- Dimethoxymethyl-pyridine-2-carbaldehyde instead of 4-Dime ⁇ hoxymethy!-pyridfne-3- carbaidehyde.
  • Example 63 2-Pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime was isolated in 43% overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Pyrrolo[1 ,2-c]pyrimidine-3-carboxyIic acid methyl ester as a green- yellow solid after recrystallization in 1-butanol.
  • Example 64 2-(5, 7-dimethyl-Pyrrolo[ 1,2-c]pyrimidin-3-yl)-chromen-4 ⁇ one oxime was isolated as a yeliow solid in 19% overall yield using method A and D starting from 2'-hydroxy-acetophenone and 5,7-dimethyl-Pyrrolo[1,2-cjpyrimidine-3- carboxylic acid methyl ester.
  • Example 65 2-(6-bromo-Pyrro!o[1,2-c]pyrimidin-3-yI)-chromen-4-one oxime was isolated as a yellow solid using method A and D starting from 2'-hydroxy- acetophenone and 6-bromo-PyrroIo[1,2-c]pyrimidine-3-carboxylic acid methyl ester.
  • Example 66 6-bromo-2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4-one oxime was prepared in 34% overall yield using methods A and D, starting from 5'- bromo-2 ! -hydroxy-acetopbenone and pyrrolo[1 ,2-c]pyrimidine-3-carboxylic acid methyl ester.
  • the title compound was isolated as a yellow solid and a 80/20 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 45-30% Water/acetonitrsle ⁇ 0.05% trifluoroacetic acid).
  • Example 67 6-methoxyethoxy-2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4- one oxime was prepared in 23% overall yield using the methods described in examples 41 and 43, starting from 6-bromo-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4- one O-ferf-butyl oxime (tert-butyl protected oxime of example 66). The title compound was isolated as an orange solid.
  • Example 68 2-(1-B enzyl- 1 H-imidazo[ 4, 5-c]pyridin-6-yl)-chrom en-4-one oxime was prepared in 20% overall yield using methods A and D, starting from 2'- hydroxy-acetophenone and 1-Benzyl-1 H-imidazo[4,5-c]pyridine-6-carboxylic acid methyl ester (example 68A). The title compound was isolated as a ye!low solid and a 90/10 mixture of Z E oxime isomers.
  • Example 69 2-Thieno[2,3-c]pyridin-5-yl-chromen-4-one ox/me was prepared in 21 % overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Thieno[2,3-c]pyridin-5-carboxylic acid methyl ester (J. Med. Chem. 2006, 49, 4425-4436). The title compound was isolated as a beige solid and a 95/5 mixture of Z/E oxime isomers.
  • Example 70 2-Thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime was prepared in 43% overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Thieno[3,2-c]pyridin-6-carboxylic acid methyl ester (J. Med. Chem. 2006, 49, 4425-4436). The title compound was isolated as a yellow solid.
  • Example 71A 2-lso quin olin-3-yl-3-m ethyl-chromen-4-on e was prepared in 76% yield using method A, starting from 2 ! -hydroxy-propiophenone and methyl isoquino!ine-3-carboxylate.
  • Example 72 3- ⁇ 4-[(E)-Hydroxyimino]-4H-chromen-2-yl ⁇ -2H-isoquinolin-1- one was prepared in 9% overall yield using methods A and D, starting from 2'- hydroxy-acetophenone and 1-oxo-1 ,2-dihydro-isoquinotine-3-carboxylic acid methyl ester. The title compound was isolated as a pale yellow solid and a 90/10 mixture of Z/E oxime isomers.
  • Example 73 2-lmidazo[1,2-c]pyrimidin-7-yl-chromen-4-one oxime was prepared in 0.3% overall yield using methods A and C, starting from 2'-hydroxy- acetophenone and !midazo 1 ,2-c]pynrnsdine-7-carboxyi!c acid ethyl ester (US2003/236264). The title compound was isolated as a yellow solid and a 60/40 mixture of Z E oxime isomers after purification by silica gel thin layer chromatography (dichloromethane/methanol : 96/4).
  • Example 74 (Reference): 2-Naphthalen-2-yl-chromen-4-one oxime was isolated in 69% yield using method D starting from 2-Naphthalen-2-yl-chromen-4-one (compound 74A) as a yellow solid and a 95/5 mixture of Z/E oxime isomers.
  • Method B To a cold suspension of 2-naphtoic acid (2.0 g, 1 1.6 mmol) in dtchloromethane (60 ml) were added oxalyl chloride (1.12 ml, 12.7 mmol) and then dimethySformamide (10 drops). The mixture was stirred at room temperature for 2 hours and concentrated to dryness to give the crude 2-naphtoic acid chloride (2.5 g). The crude product was dissolved in dry pyridine (50 ml) under argon, cooled to 0°C and treated with 2'-hydroxy-acetophenone (1.43 g, 10.5 mmol).
  • the reaction mixture was heated to 60°C for 2 hours and poured into ice-cold water (150 ml).
  • the solution was acidified to pH 1 with concentrated hydrochloric acid and the precipitate isolated by filtration, washed with water to yield Naphthalene-2-carboxylic acid 2'-acetyl-phenyl ester (2.7 g, 80%).
  • the ester was dissolved in dimeihylsulfoxide (30 ml) under argon and treated with freshly crushed potassium hydroxide (1 .4 g, 25.8 mmol) and stirred at room temperature for 14 hours.
  • the mixture was poured into ice-cold water and acidified to pH 3-4 with a 6N aqueous hydrochloric acid solution.
  • Examples 75 and 76 were prepared simultaneously by the following procedure:
  • the catalyst was removed by filtration ant the filtrate purified by silica gei chromatography (cyciohexane/ethy! acetate 0-20%) to give a 70/30 mixture of 2-(1 H-Pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one O-tert-butyl-oxsme and 2- (1-Hydroxy-1 H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one 0-tert-butyl-oxime (165 mg, 70%).
  • Example 75A 2 ⁇ (4 ⁇ Methyl ⁇ 5 ⁇ nitro ⁇ yridin-2-yl) ⁇ chromen-4-one O-tert- butyl-oxime
  • Examples 77 and 78 were prepared simultaneously by the following procedure:
  • Example 79 2-Quinolin ⁇ 2 ⁇ yl ⁇ chromen-4 ⁇ one oxime was prepared in 24% overall yield using method B and D as described in compound A, starting from quinaldyl chloride. The title compound was isolated as a yellow solid,
  • Example 80 2-Pyrimidin-4-yl-chromen-4-one oxime was prepared in 61 % overall yield using method A and D, starting from 2'- hydroxyacetophenone and pyrimidine-4-carboxylic acid methy! ester. The title compound was isolated as a yellow solid.
  • Example 81 6-Hydroxy-2 ⁇ pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4-one oxime was prepared using method D (step 2) starting from 6-Hydroxy-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 81A) and isolated as a yellow powder.
  • Example 81 A 6-Hydroxy-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen'4-one O- tert-butyl-oxime was prepared in 59% yield using the procedure described in example 18A, starting from 6-bromo-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one O- ferf-butyl oxime (tert-butyl protected oxime of example 66). The title compound was isolated as a yellow solid.
  • Example 82 2-Thiazolo[5,4-c]pyridin-6-yl-chromen-4-one oxime was prepared in 0.2% overall yield using the method described in example 75A, starting from Thiazolo[5,4-c]pyridine-6-carboxylic acid ethyl ester (US6342606).
  • Example 83 6-(3-Methoxy-propyl)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime was prepared in 32% yield using method D (step 2) starting from 6-(3-Methoxy-propyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyl- oxime (example 83A) and isolated as a ye!low solid and a 95/5 mixture of Z/E isomers.
  • Example 83 A 6-( 3-Methoxy-propyl)-2-pyrrolo[1, 2-c]py midin- chromen-4-one O-tert-butyl-oxime
  • Example 83 B 6 ⁇ ( 3-Meth oxy-prop- 1 ⁇ ynyl)-2-pyrrolo[ 1, 2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 39% yield using the procedure described in example 23A, starting from 6-bromo-2-pyrrolo[1 ,2-cjpyrimidin-3-yl- chromen-4-one O-ferf-butyl oxime (tert-butyl protected oxime of example 66) and 3- Methoxy-propyne. The title compound was isolated as a yellow solid.
  • Example 84 6-(3-Dimethylamino-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime hydrochloride
  • oxime was prepared in 22% overall yield using the procedure described in example 42A, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-fert-butyl oxime (example 81A) and (3-Chioro-propyl)-dimethy!-amine hydrochloride.
  • the compound was treated with a 1 .25 N solution of hydrogen chloride in isopropanol to yield 6-(3-Dimethylamino-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime hydrochloride as an orange solid.
  • Example 85 6-(3-Morpholin-4-yt-ethoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime hydrochloride was prepared in 65% overall yield using method D (step 2), starting from 6-(3-Morpholin-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin- 3-yl-chromen ⁇ 4-one O-terf-butyl oxime (example 85A). The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanol.
  • Example 85 A 6-(3-Morpholin-4-yl-ethoxy)-2-pyrrolo[1, 2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime
  • Example 86 A 6-(2,3-Dihydroxy-propoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime was prepared in 74% yield using the method described in example 85A, starting from 6-Hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (example 81 A) and 3-chloro-1 ,2-propandiol. The title compound was isolated as a yellow solid.
  • Example 87 6-(2-Pyrrolidin-1-yl-ethoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 37% yield using method D (step 2), starting from 6-(2-Pyrrolidin-1 -yl-ethoxy)-2-pyrroio[1 ,2-c]pyrimidtn-3-yl- chromen-4-one O-tert-butyi oxime (example 87A). The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1 .25 M solution of hydrogen chloride in isopropanol.
  • Example 87 A 6-(2 ⁇ PyrroUdin- 1 -yl-ethoxy)-2 ⁇ pyrrolo[ 1, 2-cJpyrimidin-3-yl- chromen-4-one O-tert-butyl oxime
  • Example 87B 6-(2-chloro-ethoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen- 4-one O-tert-butyl oxime was prepared in 60% yield using the method described in example 85A, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O- tert-butyl-oxime (example 81 A) and 1 ,2-dichforoethane in dimethyiformamide. The title compound was isolated as a yeflow solid.
  • Example 88 6 ⁇ (2 ⁇ Piperidin ⁇ 1-yl'ethoxy)-2 ⁇ pyrrolo[ 1 ,2 ⁇ c]pyrimidm-3-yl- chromen-4-one oxime, hydrochloride was prepared in 27% overalf yield using the method described in example 85, starting from 6-hydroxy-2-pyrroio[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyl oxime (example 81A) and 1-(2-chloroethyl)-piperidine hydrochloride .
  • Example 89 6-(2-dimethylamino-ethoxy)-2 ⁇ pyrrolo[1,2-c]pyrimidin-3 ⁇ yl- chromen-4-one oxime, hydrochloride was prepared in 39% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyl oxime (example 81 A) and 1-(2-chloroethyl)- dimethyiamine hydrochlonde.
  • Example 90 6-(2-diethylamino-ethoxy)-2-pyrrolo[ 1, 2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen- 4-one 0-£er£-buiyl oxime (example 87B) and 1-(2-chloroethyl)-diethylamine hydrochloride.
  • Example 91 2-Pyrrolo[1,2-a]pyrazin-3-yl-chromen-4-one oxime was prepared in 41 % yield using methods A and D, starting from 2'-hydroxyacetophenone and Pyrrolo[1 ,2-a]pyrazine-3-carboxyiic acid methyl ester (example 91 A). The title compound was isolated as a pale yellow solid.
  • Example 92 6 ⁇ [2-(2-methyl-pyrrolidin-1-yl)-ethoxy]-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 22 % overall yield using the method described in example 87, starting from 6-(2-chioro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 2-methylpyrrolidine.
  • Example 93 6-[2-(4-methyI-piperazin-1-yl)-ethoxy]-2-pyrroIo[ 1,2-c] pyrimidin-3-yl-chromen-4-one oxime, dihydrochlo de was prepared in 26 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrro!o[1 ,2-c]pyrimldin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 4-methyl-piperazine.
  • Example 94 6-[2-(4-methyl-[ 1,4Jdiazepan-1-yl)-ethoxy]-2-pyrrolo[ 1,2-c] pyrimidin-3-yl-chromen"4-one oxime, dihydrochloride was prepared in 13 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrroio[ ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 1-methy!-[1 ,4]diazepane.
  • Example 95 6-[2-((S)-2 ⁇ hydroxym ethylpyrrolidin- 1-yl)-eth oxy]-2- pyrrolo[1,2-c] pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 8 % overall yieid using the method described in example 87, starting from 6-(2- chioro-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4-one O-rert-butyl oxime (example 87B) and 2-((S)-2-hydroxymethylpyrrolidine.
  • Example 96 6-[2-((S)-2-methoxymethylpyrrolidin- 1-yl)-ethoxy]-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 40 % overall yieid using the method described in example 87, starting from 6-(2- chloro-ethoxy)-2-pyrroio[1 T 2-c]pyrimidin-3-yl-chromen-4-one O-ferf-buty! oxime (example 87B) and 2-((S)-2-methoxymethylpyrrolidine.
  • Example 97 6-[2-((R)-2-methoxymethyipyrroiidin-1-yi)-ethoxy]-2- pyrrolof 1, 2-c]pyrimidin-3-yl"Chromen-4-one oxime, hydrochloride was prepared in 33 % overall yield using the method described in example 87, starting from 6-(2- chioro-ethoxy)-2-pyrrolo[ ,2-c]pyrimidin-3-yi-chromen-4-one O-terf-butyl oxime (example 87B) and 2-((R)-2-meihoxymethylpyrrolidine.
  • Example 98 6-[2-(3-hydroxy-pyrrolidin- 1-yl)-ethoxy]-2-pyrroIo[ 1, 2-c] pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 23 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 3- pyrrolidinoi.
  • Example 99 6-[2-(4-dimethylammo-piperidin-1-yl)-ethoxyJ-2-pyrrolo[1,2-c] pyrimidin-3-yl-chromen-4-one oxime, dihydrochloride was prepared in 30 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-buty! oxime (example 87B) and 4-dsmethylarninopiperidine,
  • Example 100 6 ⁇ (2 ⁇ cyclopentylamino-ethoxy)-2-pyrrolo[ 1,2 ⁇ c]pyrimidin-3- yl-chromen-4-one oxime, hydrochloride was prepared in 17 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2- c]pyrimsdin-3-yl-chromen-4-one O-fert-butyl oxime (example 87 B) and cyclopentylamine.
  • Example 101 6-(3-morpholin-4-yl-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one ox/me, hydrochloride was prepared in 29 % overall yield using the method described in example 87, starting from 6-(3-chloro-propoxy)-2-pyrrolo[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-iert-butyl oxime (example 101A) and morphoiine.

Abstract

The present invention provides new oxime derivatives of the general formula (I), pharmaceutical compositions containing them and their use for the treatment and/or prophylaxis of conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling in mammals. This invention further provides new oxime derivatives of the general formula (I) consisting of modulators of nervous system receptors sensitive to glutamate, which makes them particularly suitable for the treatment and/or prophylaxis of acute and chronic neurological and/or psychiatric disorders. In particular embodiments, the new oxime derivatives of the invention are modulators of metabotropic glutamate receptors (mGluRs). The invention further provides positive allosteric modulators of mGluRs and more specifically positive alSosteric modulators of mGiuR4.

Description

NOVEL OXIME DERIVATIVES AND THEIR USE AS ALLOSTERIC MODULATORS OF METABOTROPIC GLUTAMATE RECEPTORS
The present invention provides new oxime derivatives of the general formula (I), pharmaceutical compositions containing them and their use for the treatment and/or prophylaxis of conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of giutamate level or signalling in mammals. This invention further provides new oxime derivatives of the general formula (!) consisting of modulators of nervous system receptors sensitive to giutamate, which makes them particularly suitable for the treatment and/or prophylaxis of acute and chronic neurological and/or psychiatric disorders. In particular embodiments, the new oxime derivatives of the invention are modulators of metabotropic giutamate receptors (mGluRs). The invention further provides positive allosteric modulators of mGluRs and more specifically positive allosteric modulators of mGluR4.
Glutamatergic pathways have been shown to be clearly involved in the physiopathology of a number of neuronal damages and injuries. Many nervous system disorders including epilepsy and chronic or acute degenerative processes such as for example Alzheimer's disease, Huntington's disease, Parkinson's disease and amyotrophic lateral sclerosis (Mattson MP., Neuromolecular Med., 3(2), 65-94, 2003), but also A!DS-tnduced dementia, multiple sclerosis, spinal muscular atrophy, retinopathy, stroke, ischemia, hypoxia, hypoglycaemia and various traumatic brain injuries, involve neuronal cell death caused by imbalanced levels of giutamate. It has also been shown that drug-induced neurotoxicity, for example neurotoxic effects of methamphetamine (METH) on striatal dopaminergic neurons, could actually be mediated by over-stimulation of the giutamate receptors (Stephans SE and Yamamoto BK, Synapse 17(3), 203-9, 1994). Antidepressant and anxiolytic-like effects of compounds acting on giutamate have also been observed on mice, suggesting that glutamatergic transmission is implicated in the pathophysiology of affective disorders such as major depression, schizophrenia and anxiety (Palucha A et a!., Pharmacol. Ther. 1 15(1), 1 16-47, 2007; Cryan JF et al., Eur. J. Neurosc. 17(1 1), 2409- 17, 2003; Conn PJ et a!., Trends Pharmacol. Sci. 30(1 ), 25-31 , 2009). Consequently, any compound able to modulate glutamatergic signalling or function would constitute a promising therapeutic compound for many disorders of the nervous system.
Moreover, compounds modulating glutamate level or signalling may be of great therapeutic value for diseases and/or disorders not directly mediated by glutamate !eveis and/or glutamate receptors malfunctioning, but which cou!d be affected by alteration of glutamate levels or signaling.
In the central nervous system (CNS), L-glutamate (Glu) is the main excitatory neurotransmitter and is referred to as an excitatory amino-acid (EAA), and gamma- aminobutyric acid (GABA) is the main inhibitory neurotransmitter. The balance between excitation and inhibition is of utmost importance to CNS functions, and dysfunctions of either of the two can be related to various neurological disorders.
Glutamate is ubiquitously distributed in the nervous system in high concentrations, especially in the brain and spina! cord of mammals, where it is working at a variety of excitatory synapses being thereby involved in virtually all physiological functions such as motor control, vision, central control of heart, processes of learning and memory. However, a large number of studies have established that cellular communication involving glutamate can also lead to a mechanism of cell destruction. This combination of neuroexcitatory activities and neurotoxic properties is called excitotoxicity.
Glutamate operates through two classes of receptors (Brauner-Osborne H et al., J. Med. Chem. 43(14), 2609-45, 2000). The first class of glutamate receptors is directly coupled to the opening of cation channels in the cellular membrane of the neurons. Therefore they are called ionotropic glutamate receptors (IGluRs). The !GluRs are divided in three subtypes, which are named according to the depolarizing action of their selective agonists: N-methyl-D-aspartate (N DA), ct-amino-3-hydroxy-5- methylisoxazole-4-propionic acid (AMPA), and kainic acid (KA). The second class of glutamate receptor consists of G-protein coupled receptors (GPCRs) called metabotropic glutamate receptors (mGluRs). These mGluRs are localized both pre- and post-synaptically. They are coupled to multiple second messenger systems and their role is to regulate the activity of the ionic channels or enzymes producing second messengers via G-proteins binding the GTP (Conn PJ and Pin JP.; Annu. Rev. Pharmacol. Toxicol., 37, 205-37, 1997). Although they are generally not directly involved in rapid synaptic transmission, the mGluRs modulate the efficacy of the synapses by regulating either the post-synaptic channels and their receptors, or the pre-synaptic release or recapture of glutamate. Therefore, mGluRs play an important role in a variety of physiological processes such as long-term potentiation and long- term depression of synaptic transmission, regulation of baroreceptive reflexes, spatial learning, motor learning, and postural and kinetic integration.
To date, eight mGluRs have been cloned and classified in three groups according to their sequence homologies, pharmacological properties and signal transduction mechanisms. Group ! is constituted of mGluRI and mGiuRS, group I! of mGluR2 and mGiuR3 and group I II of mGluR4, mGiuR6, mGluR7 and mGluR8 (Pin JP and Acher F. , Curr. Drug Targets CNS Neurol. Disord. , 1 (3), 297-317, 2002; Schoepp DD et al. , Neuropharmacology, 38(10), 1431 -76, 1999).
mGluRs modulators can be classified in two families depending on their site of interaction with the receptor (see Brauner-Osbome H et al. , J. Med. Chern. 43(14), 2609-45, 2000 for review). The first family consists in orthosteric modulators (or competitive modulators) able to interact with the glutamate binding-site of the mGluRs, which is localized in the large extra-celiular N-terminal part of the receptor (about 560 amino acids). Therefore, they are glutamate analogs and constitute a highly polar family of ligand. Examples of orthosteric modulators are S-DHPG or LY-367385 for group I mG!uRs, LY-354740 or (2R-4R)-APDC for group II mGluRs and ACPT-I or L- AP4 for group Mi mGluRs. The second family of mGluRs modulators consists in allosteric modulators that interact with a different site from the extracellular active site of the receptor (see Bridges TM et al. , ACS Chem Biol, 3(9), 530-41 , 2008 for review). Their action results in a modulation of the effects induced by the endogenous ligand glutamate. Examples of such allosteric modulators are Ro-674853, MPEP or JNJ16259685 for group ! mGluRs and CBiPES, LY181837 or LY487379 for group II mGluRs.
For groups Mi mGluRs, examples of allosteric modulators were so far described for the mGluR subtype 4 (mGiuR4). PHCCC, MPEP and S1B1893 (Maj M et al. , Neuropharmacology, 45(7), 895-903, 2003; Mathiesen JM et al., Br. J, Pharmacol. 138(6), 1026-30, 2003) were the first ones described in 2003. More recently, more potent positive allosteric modulators were reported in the literature (Niswender CM et al., Mol. Pharmacol. 74(5), 1345-58, 2008; Niswender CM et al. , Bioorg. Med. Chem. Lett 18(20), 5626-30, 2008; Williams R et al. , Bioorg. Med. Chem. Lett. 19(3), 962-6, 2009; Engers DW et al., J. Med. Chem. May 27 2009) and in two patent publications describing families of amido and heteroarornatic compounds (WO 2009/010454 and WO 2009/010455).
Numerous studies have already described the potentiai applications of mGiuR modulators in neuroprotection (see Bruno V et al., J. Cereb. Blood Flow Metab., 21 (9), 1013-33, 2001 for review). For instance, antagonist compounds of group I mGluRs showed interesting results in animal models for anxiety and postischemic neuronal injury (Pile A et al., Neuropharmacology, 43(2), 181 -7, 2002; Meli E et ai., Pharmacol. Biochem. Behav., 73(2), 439-46, 2002), agonists of group II mG!uRs showed good results in animal models for Parkinson and anxiety (Konieczny J et al., Naunyn- Schmlederbergs Arch. Pharmacol., 358(4), 500-2, 1998).
Group III mGluR modulators showed positive results in several animal models of schizophrenia (Paiucha-Poniewiera A et al., Neuropharmacology, 55(4), 517-24, 2008) and chronic pain (Goudet C et ai., Pain, 137(1 ), 1 12-24, 2008; Zhang HM et a!., Neuroscience, 158(2), 875-84, 2009).
Group II! mGluR were also shown to exert the excitotoxic actions of homocysteine and homocysteic acid contributing to the neuronal pathology and immunosenescence that occur in Alzheimer Disease (Boldyrev AA and Johnson P, J. Alzheimers Dis. 1 (2), 219-28, 2007).
Moreover, group III mGluR modulators showed promising results in animal models of Parkinson and neurodegeneration (Conn J et al., Nat Rev. Neuroscience, 6(10), 787-98, 2005 for review; Vernon AC et al., J. Pharmacol. Exp. Then, 320(1 ), 397-409, 2007; Lopez S et al., Neuropharmacology, 55(4), 483-90, 2008; Vernon AC et al., Neuroreport, 19(4), 475-8, 2008). It was further demonstrated with selective ligands that the mGluR subtype implicated in these antiparkinsonian and neuroprotective effects was mGluR4 (Marino MJ et al., Proc. Natl. Acad. Sci. USA 100(23), 13668-73, 2003; Battaglia G et al., J. Neurosci. 26(27), 7222-9, 2006; Niswender CM et al., Mol. Pharmacol. 74(5), 1345-58, 2008).
mGluR4 modulators were also shown to exert anxiolytic activity (Stachowicz K et al., Eur. J. Pharmacol., 498(1-3), 153-6, 2004) and anti-depressive actions (Palucha A et ai., Neuropharmacology 46(2), 151-9, 2004; Klak K et al., Amino Acids 32(2), 169-72, 2006).
In addition, mGluR4 were also shown to be involved in glucagon secretion inhibition (Uehara S., Diabetes 53(4), 998-1006, 2004). Therefore, orthosteric or positive allosteric modulators of mGluR4 have potential for the treatment of type 2 diabetes through its hypoglycemic effect.
Moreover, mGluR4 was shown to be expressed in prostate cancer cell-line (Pessimissis N et al., Anticancer Res. 29(1 ), 371-7, 2009) or colorectal carcinoma (Chang HJ et a!., CIL Cancer Res. 1 1 (9), 3288-95, 2005) and its activation with PHCCC was shown to inhibit growth of medul!oblastomas (iacoveili L et al., J. Neurosci. 26(32) 8388-97, 2006), mGluR4 modulators may therefore have also potential role for the treatment of cancers.
Finally, receptors of the umami taste expressed in taste tissues were shown to be variants of the rnGiuR4 receptor (Eschle BK., Neuroscience, 155(2), 522-9, 2008). As a consequence, mGluR4 modulators may also be useful as taste agents, flavour agents, flavour enhancing agents or food additives.
Chromone-derived core structures for pharmaceutically active compounds were described in the patent application WO 2004/092154. In the latter application, they are disclosed as inhibitors of protein kinases.
EP-A-0 787 723 relates to specific cyclopropachromencarboxylic acid derivatives which are said to have mGluR antagonistic activity.
The -present invention relates to compounds of the general formula (I):
Figure imgf000006_0001
and pharmaceutically acceptable salts, solvates and prodrugs thereof.
R1, R2, R3, and R4 each independently represent a -L-R group.
L represents a bond, a C C-io alkylene, a C2-C10 alkenylene, or a C2-C10 atkynylene, wherein said alkylene, said alkenylene or said alkynylene is optionally substituted with one or more groups independently selected from halogen, -CF3, -CN, -OH, or -NH2, and further wherein one or more -CH2- units comprised in said alkyiene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -0-, -NR11-, -CO-, -S-, -SO-, or -S02-.
R is selected from hydrogen, C C10 alkyl, halogen, optionaily substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, -NR11R12, -OR11, -SR , -SOR11, -S02R11, -CF3, or -CN, wherein said optionaily substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyl may be substituted with one or more groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(C C4 alkyl), -N(C C4 alkyS)(C C4 alkyl) wherein the two C -C4 alkyl moieties of said -N(CrC4 alkyl){C C alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, or -L1-R13
R1 and R12 are each independently selected from hydrogen, optionally substituted C Cio alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionaily substituted heterocycloalkyl, or -CF3, wherein said optionally substituted alkyl, said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyl may be substituted with one or more groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(CrC4 alkyl), -NH2s -NH(C C4 alkyl), or -N(C1-C4 alkyl)(C-i-C4 alkyl), wherein the two C C4 alkyl moieties of said -N(C C4 alkyl)(d-C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
L1 Is selected from a bond, C C10 alkyiene, C2~C10 alkenylene, or C2-C10 alkynylene, wherein one or two -CH2- units comprised in said alkyiene, said alkenylene or said alkynylene are each optionaily replaced by a group independently selected from -0-, -NH-, -N(C C4 alkyl)-, -CO-, -S-, -SO- or -S02-.
R13 is selected from hydrogen, CrC4 alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycioalkyi, optionally substituted heterocycloalkyi, -NH2, -NH{d-d alkyl), -N(CrC4 alkyl)(d-d alkyl), -OH, -0(d-d alkyl), -SH, -S(d-C4 a!kyl), -CF3, or -CN, wherein said optionally substituted phenyl, said optionally substituted heteroaryl, said optionally substituted cycioalkyi, or said optionally substituted heterocycloalkyi may be substituted with one or more groups independently selected from C d alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(d-d alkyl), or -N(C C4 aikyl)(C C4 alkyl), wherein the two C C4 alkyl moieties of said -N(C C4 alkyl)(Ci-C4 aikyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to. s is selected from hydrogen, C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -COOH, -COO(C C4 alkyl), -CONH2, -CONH(C C4 alkyl), -CON(C d alkyl)(d-d alkyl), wherein the two d-d alkyl moieties of said -CON(d-d alkyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -NH2, -NH(d-C4 alkyl), or -N(C C4 alkyI)(C C4 alkyl), wherein the two d-d alkyl moieties of said -N(d-C alkyi)(d-C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
A is a bicyclic moiety corresponding to formula (II):
Figure imgf000008_0001
which may be saturated or unsaturated, and wherein:
n is 0 or 1 ;
X1 to X6 are each independently selected from N, N(Rx1), C(Rx2), C(Rx2)(Rx3), O, S, S(O), S(0)2, or C(O);
X7 is N or N(Rx );
any of groups X1 to X7 containing a nitrogen atom may form an N-oxide group;
Xs and X9 are each independently selected from N, C, or C(R 2);
each Rx is independently selected from hydrogen, d~C4 alkyl, -OH, -0(C C4 alkyl), or -(C C4 alkytene)-phenyl; and
each R 2 and each R 3 is independently selected from hydrogen, CrC4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -COOH, -COO(C-rC4 alkyl), -CONH2, -CONH(C C4 a!kyi), -CON<CrC4 alkyl)(C1-C alkyl), -NH2, -NH(C C4 alkyl), or -N(CrC alkyl)(C C4 alkyl), wherein the two C C alkyl moieties of said -N(C C4 alkyl)(C C4 alkyl) or of said -CON(C C4 alkyI)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
In this description, the bicyclic ring system of formula (I) including the O-heteroatom and the oxime group, to which the substituents R1 to R5 and A are attached, is also referred to as "chromone moiety".
Thus, the present invention relates to new oxime derivatives of the general formula (I) as described and defined herein and pharmaceutically acceptable salts, solvates and prodrugs thereof, pharmaceutical compositions containing any of the aforementioned entities and their use for the treatment and/or prophylaxis of conditions associated with altered giutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling in mammals. It further relates to a method of treating and/or preventing conditions associated with altered giutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling in a mammal. Accordingly, the present invention provides a method of treating and/or preventing a disease or disorder, in particular a condition associated with altered giutamatergic signalling and/or functions, and/or a condition which can be affected by alteration of glutamate level or signalling, the method comprising the administration of a compound of the general formula (I) as described and defined herein, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising any of the aforementioned entities, to a subject (preferably, a mammal; more preferably, a human) in need of such treatment or prevention. In further embodiments the compounds of the general formula (!) are modulators of mGluRs of the nervous system, in preferred embodiments the compounds of the invention are allosteric modulators of the mGluRs and in a most preferred embodiment they are positive allosteric modulators of mGluR4. The conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling, and which can be treated and/or prevented with the compounds or the pharmaceutical compositions according to the invention, include in particular: epilepsy, including newborn, infantile, childhood and adult syndromes, partial (localization-related) and generalized epilepsies, with partial and generalized, convulsive and non-convulsive seizures, with and without impairment of consciousness, and status epiiepticus; Dementias and related diseases, including dementias of the Alzheimer's type (DAT), Alzheimer's disease, Pick's disease, vascular dementias, Lewy-body disease, dementias due to metabolic, toxic and deficiency diseases (including alcoholism, hypothyroidism, and vitamin B12 deficiency), AIDS-dementia complex, Creutzfeld- Jacob disease and atypical subacute spongiform encephalopathy; Parkinsonism and movement disorders, including Parkinson's disease, multiple system atrophy, progressive supranuclear palsy, corticobasal degeneration, hepatolenticular degeneration, chorea (including Huntington's disease and hemiballismus), athetosis, dystonias (including spasmodic torticollis, occupational movement disorder, Gilles de la Tourette syndrome), tardive or drug induced dyskinesias, tremor and myoclonus; Motor neuron disease or amyotrophic lateral sclerosis (ALS); Other neurodegenerative and/or hereditary disorders of the nervous system, including spinocerebrellar degenerations such as Friedrich's ataxia and other hereditary cerebellar ataxias, predominantly spinal muscular atrophies, hereditary neuropathies, and phakomatoses; Disorders of the peripheral nervous system, including trigeminal neuralgia, facial nerve disorders, disorders of the other cranial nerves, nerve root and plexus disorders, mononeuritis such as carpal tunnel syndrome and sciatica, hereditary and idiopathic peripheral neuropathies, inflammatory and toxic neuropathies; Multiple sclerosis and other demyelinating diseases of the nervous system; Infantile cerebral palsy (spastic), monoplegic, paraplegic or tetraplegic; Hemiplegia and hemiparesis, flaccid or spastic, and other paralytic syndromes; Cerebrovascular disorders, including subarachnoid hemorrhage, intracerebral hemorrhage, occlusion and stenosis of precerebral arteries, occlusion of cerebral arteries including thrombosis and embolism, brain ischemia, stroke, transient ischemic attacks, atherosclerosis, cerebrovascular dementias, aneurysms, cerebral deficits due to cardiac bypass surgery and grafting; Migraine, including classical migraine and variants such as cluster headache; Headache; Myoneural disorders inciuding myasthenia gravis, acute muscle spasms, myopathies including muscular dystrophies, mytoionias and familial periodic paralysis; Disorders of the eye and visual pathways, including retinal disorders, and visual disturbances; Intracranial trauma/injury and their sequels; Trauma/injury to nerves and spinal cord and their sequels; Poisoning and toxic effects of nonmedicinal substances; Accidental poisoning by drugs, medicinal substances and biologicals acting on the central, peripheral and autonomic system; Neurological and psychiatric adverse effects of drugs, medicinal and biological substances; Disturbance of sphincter control and sexual function; Mental disorders usually diagnosed in infancy, childhood or adolescence, including: mental retardation, learning disorders, motor skill disorders, communication disorders, pervasive developmental disorders, attention deficit and disruptive behaviour disorders, feeding and eating disorders, TIC disorders, elimination disorders; Delirium and other cognitive disorders; Substance related disorders including: alcohol-related disorders, nicotine-related disorders, disorders related to ***e, opioids, cannabis, hallucinogens and other drugs; Schizophrenia and other psychotic disorders; Mood disorders, including depressive disorders and bipolar disorders; Anxiety disorders, including panic disorders, phobias, obsessive-compulsive disorders, stress disorders, generalized anxiety disorders; Eating disorders, including anorexia and bulimia; Sleep disorders, including dyssomnias (insomnia, hypersomnia, narcolepsy, breathing related sleep disorder) and parasomnias; Medication-induced movement disorders (including neuroleptic-induced parkinsonism and tardive dyskinesia); Endocrine and metabolic diseases including diabetes, disorders of the endocrine glands, hypoglycaemta; Acute and chronic pain; Nausea and vomiting; Irritable bowel syndrome; or cancers.
(n particular, the conditions associated with altered glutamatergic signalling and/or functions, and/or conditions which can be affected by alteration of glutamate level or signalling to be treated and/or prevented by the compounds or the pharmaceutical compositions according to the invention, include: Dementias and related diseases, including dementias of the Alzheimer's type (DAT), Alzheimer's disease, Pick's disease, vascular dementias, Lewy-body disease, dementias due to metabolic, toxic and deficiency diseases (including alcoholism, hypothyroidism, and vitamin B12 deficiency), AlDS-dementia complex, Creutzfetd-Jacob disease and atypical subacute spongiform encephalopathy; Parkinsonism and movement disorders, including Parkinson's disease, multiple system atrophy, progressive supranuclear palsy, coriicobasal degeneration, hepatolenticular degeneration, chorea (including Huntington's disease and hemiballismus), athetosis, dystonias {including spasmodic torticollis, occupational movement disorder, Gilles de la Tourette syndrome), tardive or drug induced dyskinesias, tremor and myoclonus; Acute and chronic pain; Anxiety disorders, inc!uding panic disorders, phobias, obsessive-compulsive disorders, stress disorders and generalized anxiety disorders; Schizophrenia and other psychotic disorders; Mood disorders, including depressive disorders and bipolar disorders; Endocrine and metabolic diseases including diabetes, disorders of the endocrine glands and hypoglycaemia; or cancers.
The present invention further provides a method for identifying an agent that binds to metabotropic glutamate receptor 4 (mGiuR4), or in other words for determining the capability of one or more test agent(s) to bind to the receptor, comprising the following steps: (a) contacting mG!uR4 with a compound of the present invention which is labeled, preferably radio-labeled or fluorescence-labeled, under conditions that permit binding of the compound to mGluR4, thereby generating a bound, labeled compound; (b) detecting a signal that corresponds to the amount of the bound, labeled compound in the absence of test agent; (c) contacting the bound, labeled compound with a test agent; (d) detecting a signal that corresponds to the amount of the bound labeled compound in the presence of test agent; and (e) comparing the signal detected in step (d) to the signal detected in step (b) to determine whether the test agent binds to mGiuR4. As will be understood, a substantially unchanged signal detected in step (d) in comparison with the signal detected in step (b) indicates that the test agent does not bind to the receptor, or binds to the receptor less strongly than the compounds according to the invention. A decreased or increased signal detected in step (d) in comparison with the signal detected in step (b) indicates that the test agent binds to the receptor. Thus, agents that bind to mGiuR4 can be identified among the test agents employed in the above method. It will further be understood that it is preferred to remove unbound labeled compounds, e.g. in a washing step, before carrying out steps (b) and (d).
The rrsGluR4 which is used in the above method may be a human form (F!or PJ, Lukic S, Ruegg D, Leonhardt T, Knopfel T, Kuhn R. 1995. Neuropharmacology. 34: 149-155. akoff A, Lelchuk R, Oxer M, Harrington K, Emson P. 1996. Brain Res. Mol. Brain Res. 37:239-248. Wu S, Wright RA, Rockey PK, Burgett SG, Arnold JS, Rosteck PR Jr, Johnson BG, Schoepp DD, Belagaje RM. 1998. Brain Res. Mot. Brain Res. 53:88- 97.), e.g. a protein of the accession number NPJD00832 or a protein having at [east 80% (preferably, at least 90%; more preferably, at least 95%; even more preferably, at least 99%) amino acid identity to said protein of the accession number NP__000832, or a non-human form, including e.g. a mouse form or rat form (Tanabe Y, Masu M, !shii T, Shigemoto R, Nakanishi S. 1992. Neuron. 8:169-179.), or a homolog thereof found in a different species (e.g. in a different mammalian species), or a mutein of any of the aforementioned entitites which mutein retains the mGluR4 activity. Said mutain can preferably be obtained by substitution, insertion, addition and/or deletion of one or more (such as, e.g., 1 to 20, including 1 to 10 or 1 to 3) amino acid residues of said aforementioned entitites. The mGluR4 used in the above method may also be a functional fragment of any of the aforementioned entitites (including said muteins), i.e. a fragment which retains the mGluR4 activity of the respective aforementioned entity or, in other words, a fragment having essentially the same biological activity (i.e., at least about 60% activitiy, preferably at least about 70% activity, more preferably at least about 80% activity, even more preferably at least about 90% activity) as the respective aforementioned entity. A person skilled in the art is readily in a position to determine whether mGiuR4 activity is retained using techniques known in the art, e.g. knock-out and rescue experiments. Furthermore, the mGluR4 used in the above method may also be a compound comprising any one or more of the aforementioned entitites (including, without limitation, a protein of the accession number NPJ300832, a protein having at least 80% amino acid identity to said protein of the accession number NP_000832, or a functional fragment thereof), wherein the mGluR4 activity is retained. Preferably, the mGluR4 used in the above method is a human form.
The compounds of the general formuia (I) will be described in more detail in the following:
R , R2, R3, and R4 each independently represent a -L-R group.
L represents a bond, CrC10 alkylene, C2-C 0 alkenylene, or C2-C10 alkynyiene. Said alkylene, said alkenylene or said alkynyiene is optionally substituted with one or more (such as, e.g., one, two, three, or four) groups independently selected from halogen, -CF3, -CN, -OH, or -NH2.
Moreover, one or more (such as, e.g., one, two, three, or four), preferably one or two, more preferably one, -CH2- units comprised in said alkylene, said alkenylene or said aikynylene are each optionally replaced by a group independently selected from: -0-, -NR11-, -CO-, -S-, -SO-, or -S02~. This includes the option that the replacing groups -NR11- and -CO- are combined, in any order, to form an amide group. Otherwise, if more than one -CH2- unit is replaced, it is preferred that these -CH2- units are non- adjacent. Preferably, no -CH2- unit, one -CH2- unit or two -CH2- units are each replaced by a group independently selected from -0-, -NR11-, -CO-, or -S-. More preferably, no -CH2- unit is replaced or one -CH2- unit or two -CH2- units are each replaced by a group independently selected from -0-, -NR11-, or -S-. Even more preferably, no -CH2- unit is replaced or one or two -CH2- units are each replaced by -0-,
More preferably, L is a bond, Ci-C6 alkylene, C2-C6 alkenylene, or C2-C6 aikynylene, wherein one or two -CH2- units comprised in said alkylene, said alkenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NR11-, -CO-, or -S-.
Even more preferably, L is a bond, Ci-C3 alkylene, C2-C6 alkenylene, or C2-C6 aikynylene, wherein one or two -CH2~ units comprised in said alkylene, said alkenylene, or said aikynylene are each optionally replaced by -0-.
Most preferably, L is a bond or C C6 alkylene, wherein one or two -CH2- units comprised in said alkylene are each optionally replaced by -0-.
R is selected from hydrogen, C C 0 alkyl, halogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloaikyi, optionally substituted heterocycioalkyi, -NR11R12, -OR11, -SR11, -SOR11, -S02R11, -CF3, or -CN, wherein said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloaikyi, or said optionally substituted heterocycioalkyi may be substituted with one or more (such as, e.g., one, two, three, or four), preferably one or two, substituent groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(C C4 alkyl), -N(C C4 alkyl)(d-C4 alkyi) wherein the two C C4 alkyl moieties of said -N(C C alkyl)(C C4 alkyl) are optionally mutually linked to form a ring (such as, e.g., a pyrrolidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to, or -L1-R13. Said substituent groups are preferably selected from C-1-C4 alkyl, -(C C4 alkylene)-OH, ~{C C alkylene)-0-(Ci-C4 alkyl), -OH, -0{d-C4 alkyl), -0-(CrC4 alkylene)-0-(C C4 alkyl), -NH2, -NH(Ci-C4 alkyl), -N(C C4 alkyl)(C C4 alkyl) wherein the two C C4 alkyl moieties of said -N(C1-C4 alkyl){C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -CF3| or halogen, and more preferably selected from C C4 alkyl, -(C C4 alkylene)-OH, -(C^C^ aikylene)-0-(C C4 alkyl), -0(C -C4 alkyl), -N(C C4 alkyi)(C C4 alky!) wherein the two C C4 alkyl moieties of said -N(C C4 alkyl)(C C4 alky!) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, or halogen, Said halogen may, for example, be selected from fluoro, chloro, bromo, or iodo.
Preferably, R is selected from: hydrogen; optionally substituted aryl; optionally substituted heieroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or and the other ring atoms are carbon atoms; opttonaliy substituted heterocycioalkyl having 3 to 10 ring atoms, wherein one or more (such as, e.g., one, two, or three) ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -NH(C C4 alkyl); -N(CTC4 alkyl)(Ci-C4 alkyl); or -0(C C alkyl). Said aryl may, for example, be a phenyl. Said heteroaryl may, for example, be selected from pyridinyl, thienyi, furyl, pyrrolyl, imidazolyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, isoxazoiyl, or furazanyl. Said heterocycioalkyl may, for example, be selected from tetrahydrofuranyl, piperidinyl, piperazinyl, pyrrolidinyl, imidazolidinyl, morpholinyi, pyrazolidinyl, tetrahydrothienyl, oxazolidinyi, isoxazolidinyl, aziridinyl, azetidinyl, octahydroquinolinyl, octahydroisoquinolinyl, azepanyl, diazepanyl, oxazepanyi or 2-oxa-5-aza- bicyclo[2.2.1]hept-5-yl.
More preferably, R is selected from: hydrogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -0(C C alkyl); or optiona!ly substituted heterocycioalkyl having 3 to 10 ring atoms, wherein one or more ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms.
Even more preferably, R is selected from: hydrogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independentiy selected from O, S, or N and the other ring atoms are carbon atoms; or optionally substituted heterocycloalkyl having 5 to 7 ring atoms, wherein one or two ring atoms are each independentiy selected from N or O and the other ring atoms are carbon atoms.
As is apparent from the above, preferred combinations -L-R are those wherein L is a bond, C-|-C6 alkylene, C2-C6 alkeny!ene, or C2-C6 alkynylene, wherein one or two -CH2- units comprised in said aikylene, said alkenylene, or said aikynyiene are each optionally replaced by -0-, and preferably L is a bond or C C6 alkylene, wherein one or two -CH2- units comprised in said alkylene are each optionally replaced by -0-, and further wherein R is selected from hydrogen, optionally substituted phenyl, optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; or optionally substituted heterocycloalkyl having 5 to 7 ring atoms, wherein one or two ring atoms are each independently selected from N or O and the other ring atoms are carbon atoms. Said optionally substituted phenyl, said optionally substituted heteroaryl having 5 or 6 ring atoms, or said optionally substituted heterocycloalkyl having 5 to 7 ring atoms may be substituted with one or more, preferably one or two, groups independently selected from C C4 alkyl, -(CrC4 aikylene)-OH, -(C C4 aikylene)-0-(C1-C4 alkyl), -OH, -0{CrC4 alkyl), -0-(C C4 alkylene)-0-{C C4 alkyl), -NH2, -NH(CrC4 alkyl), or -N(C C4 a!kyS)(C C4 alkyl) wherein the two C C4 alkyl moieties of said -N(CTC4 alkyl)(C1-C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -CF3, or halogen, and more preferably selected from CrC4 alkyl, ~{C C4 alkylene)-OH, -(C C alky!ene)-0-(d-C4 aikyl), -0{C C4 alkyl), -N(CrC4 alkyl)(C C4 alkyl) wherein the two C-rC4 alkyl moieties of said -N(C -C4 alkyl)(C C alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, or halogen.
Exemplary preferred -L-R groups encompassed by the above preferred definition are independently selected from: hydrogen; (VC4 alkyl; -(C C3 alkyIene)-0~((VC2 alkyl); -(C2-C4 alkyny!ene)-phenyl; -OH; -0(d-C4 alkyl); -0(C C2 alkylene)-0-(C C2 alkyl); -(C C4 alky!ene)-morpholinyl; -0(C C4 alkylene)-phenyl; -0(C CA alkylene)-imidazolyl; -0(C C4 alkylene)-pyrrolidinyl; -0(CVC4 alkylene)-piperidinyl; -0(C C4 alkylene)-morpholinyl; -0(C C4 alkylene)-pyridinyl; -0(C^-C4 a!kylene)-oxazepanyl; -0{C-|-C4 aikylene)-(2-oxa-5-aza-bicydo[2.2.1 ]hept-5-yl); -0(C C4 alkylene)-piperaziny1ene-(C C4 alkyl); or -0(CrC4 alkyiene)-diazepanylene-(C-rC4 alkyl); wherein the phenyl, imidazoiyl, pyrrolidinyi, piperidinyl, morpholinyl, pyridinyl, oxazepanyl, 2-oxa-5-aza-bicyclo[2.2.1]hept-5-yl, piperazinylene, and diazepanyiene moieties are each optionally substituted with one or more, preferably one or two, groups independently selected from halogen, -CF3, C C alkyl, -(d-d alky!ene)-OH, -(C1-C4 alky!ene)-0-(C C4 alkyl), -OH, -0(C C4 alkyl), -0-(d-d alkylene)-0-(d-d alkyl), -NH2, -NH(d-d alkyl), or -N(d-d aikyl)(d-d alkyl) wherein the two d-d alkyl moieties of said -N(d-d alkyl)(d-d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, more preferably selected from halogen, d- alkyl, -(d-d a!kylene)-OH, -{d-d alkylene)-0-(C d alkyl), -0(d-d alkyl). or -N(Crd alkyI){Crd alkyl) wherein the two d-d alkyl moieties of said -N(d-d alkyl)(d-d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
Moreover, in particularly preferred embodiments, either R to R4 are all hydrogen, or one of R1 to R4 is a -L-R group other than hydrogen as defined above and the other ones of R1 to R4 are hydrogen. In this latter preferred embodiment, the -L-R group other than hydrogen is in particular one of R2 and R3, and more preferably R2.
Accordingly, in one preferred embodiment R1 and R4 are each hydrogen, one of R2 and R3 (preferably R3) is hydrogen, and the other one of R2 and R3 (preferably R2) is selected from: hydrogen; d-d alkyl; ~(d~d a!kylene)-0-(d-C2 alkyl); -(d-d alkynylene)-phenyl; -OH; -0(C d alkyl); -0(C C2 alky!ene)-0-(d-C2 alkyl); -(d-d alkylene)-morpholinyl; -0(C d alkylene)-phenyl; ~0{d-d alkylene)-imidazolyl; -0(C d alkylene)-pyrrolidinyl; -0(d-d alkylene)-piperidinyl; -0(d-d alkylene)-morphoiinyl; -0(d-d alkylene)-pyridinyl; -0(C d alkylene)-oxazepanyl; -0(d-d aikylene)-(2-oxa-5-aza-bicyc!o[2.2.1 jhept-5-yl); -0(CrC4 aikylene)-piperazinylene-(d-d alkyl); or -0(C d alkylene)-diazepanylene-(C C4 alkyl); wherein the phenyl, imidazoiyl, pyrrolidinyi, piperidinyl, morpholinyl, pyridinyl, oxazepanyl, 2-oxa-5-aza-bicyclo[2.2.1]hept-5-yl, piperazinylene, and diazepanylene moieties are each optionally substituted with one or more, preferably one or two, groups independently selected from halogen, -CF3, d-d alkyl, -(d-d aikylene)-OH, -(d-d aikylene)-0-(d-d alkyl), -OH, -0(Crd alkyl), -0-(Crd alkylene)-0-(C d alkyl), -NH2, -NH(C d alkyl), or -N(d-C4 alkyl)(C d alkyl) wherein the two CrC4 alkyl moieties of said -N(d-d alkyl)(d-d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, more preferably selected from halogen, C C4 alkyl, -(CrC4 alkylene)-OH, -(C-rC4 aikylene)-0-(C -C4 alkyl), -0(C C4 alkyl), or -N(CrC4 alkyl)(d-C alkyl) wherein the two C C4 alkyl moieties of said -N{C C4 alkyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to. in one aspect of this embodiment, one of R2 and R3 (preferably R3) is hydrogen, and the other one of R2 and R3 (preferably R2) is not hydrogen.
R11 and R12 are each independently selected from hydrogen, optionally substituted C C10 alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionaliy substituted heterocycloalkyi, or -CF3, wherein said optionally substituted alkyl, said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalkyi may be substituted with one or more, preferably one or two, more preferably one, groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 a!kyl), -NH2, -NH(C C4 alkyi), or -N(CrC4 alkyl)(C C4 alkyl) wherein the two C C4 alkyl moieties of said -Ν((_ θ4 alkylXd-C* alkyl) are optionally mutually linked to form a ring (such as, e.g., a pyrrolidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to. It is preferred that the aforementioned groups are unsubstituted. Preferably, R11 and R 2 are each independently selected from hydrogen, or CrC4 alkyl.
L1 is selected from a bond, C C 0 alkylene, C2-C10 aikenylene, or C2-C10 aikynylene, wherein one or two -CH2- units comprised in said alkylene, said aikenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NH-, -N(C C4 alkyl)-, -CO-, -S-, -SO- or -S02-. This includes the option that the replacing group -NH- or the replacing group -N(C C4 alkyl)- is combined with the replacing group -CO-, in any order, to form an amide group. Otherwise, if more than one -CH2- unit is replaced, it is preferred that these -CH2- units are non-adjacent.
Preferably, L1 is selected from a bond, C C6 alkylene, C2-C6 aikenylene, or C2-C6 aikynylene, wherein one or two -CH2- units comprised in said alkylene, said aikenylene or said aikynylene are each optionally replaced by a group independently selected from -0-, -NH-, -CO-, or -S-.
More preferably, L1 is selected from a bond, C C5 alkylene, C2-C6 aikenylene, or C2-C5 alkynylene, wherein one or two -CH2- units comprised in said aikylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -0-, -NH-, or -S-.
Even more preferably, L1 is a bond, C C6 aikylene, C2-C6 alkenylene, or C2-C6 alkynylene, wherein one or two -CH2- units comprised in said aikylene, said alkenylene or said alkynylene are each optionally replaced by -0-.
Most preferably, L is a bond or C Cs aikylene, wherein one or two -CH2- units comprised in said aikylene are each optionally replaced by -0-.
R13 is selected from hydrogen, d-C alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycloa!kyl, optionally substituted heterocycloalkyl, -NH2, -NH(C C4 alkyl), -N(C C4 alkyl)(C C4 alkyl), -OH, -0(C C4 alkyl), -SH, -S(C C4 alkyl), -CF3, or -CN, wherein said optionally substituted phenyl, said optionally substituted heteroaryl, said optionally substituted cycloalkyi, or said optionally substituted heterocycloalkyl may be substituted with one or more, preferably one or two, more preferably one, groups independently selected from CrC4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(C C4 alkyl), or -N(d-C4 a!kyl)(C C4 alkyl) wherein the two CrC4 alkyl moieties of said -N(C C4 alkyl)(C C4 alkyl) are optionally mutually linked to form a ring (such as, e.g., a pyrrolidinyl ring or a piperidinyi ring) together with the nitrogen atom which they are attached to.
Preferably, R13 is selected from hydrogen, C C alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycloalkyi, optionally substituted heterocycloalkyl, -NH(C C4 alkyl), -N(C C4 alkyl)(C C4 alkyl), -0{C C4 alkyl), -S(C-,-C4 alkyl), -CF3, or -CN.
More preferably, R13 is selected from: hydrogen; Ci-C4 alkyl; halogen; optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; cycloalkyi having 3 to 7 ring atoms; optionally substituted heterocycloalkyl having 3 to 10 ring atoms, wherein one or more (such as, e.g., one, two, or three) ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -NH(CrC4 alkyl); -N(CrC4 aIkyl)(C1-C4 alkyl); -0(C C4 alkyl); -S(C C4 alkyl); -CF3; or -CN.
Even more preferably, R13 is selected from: hydrogen; -0(CrC4 alkyl); optionally substituted phenyl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; or optionally substituted heterocycloaikyl having 3 to 10 ring atoms, wherein one or more ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms.
R6 is selected from hydrogen, C C4 alky!, halogen, -CF3, -CN, -OH, -0(C C4 alky!), -COOH, -000(0,-04 alkyl), -CONH2, -CONH(C C4 alkyi), -CON(Ci-C4 alkyl)(C1-C4 alkyl) wherein the two C C4 alkyl moieties of said -CON(C-i-C4 aikyt)(CrC4 alkyl) are optionally mutually linked to form a ring (such as, e.g., a pyrroiidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to, -NH2, -NH{Ci-C4 alkyl), or -N(CrC4 alkyl)(CfC4 alkyl) wherein the two Ci-C4 alkyl moieties of said -N(Ci-C4 alky!)(Ct-C4 aikyl) are optionally mutually linked to form a ring (such as, e.g., a pyrroiidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to. Said halogen may, for example, be selected from fluoro, chloro, bromo, or iodo. Preferably, R5 is selected from hydrogen, Ci-C4 alkyl, halogen, -CF3, -CN, -OH, or -0(C C4 alkyl). More preferably, R5 is selected from hydrogen or C C4 alkyl. Even more preferably, Rs is hydrogen.
A is a bicyclic moiety corresponding to formula (II):
Figure imgf000020_0001
which may be saturated or unsaturated, i.e. one or more double bonds may be present in one or both rings formed by X1 to X9. This includes the option that one or both rings formed by X1 to X9 are aromatic.
n is 0 or 1 .
X1 to Xs are each independently selected from N, N(Rx1), C(Rx2), C(Rx2)(Rx3), O, S, S(0), S(0)2, or C(O). Preferably, X1 to X6 are each independently selected from N, N(R*1), C(R*2); C(Rx2)(Rx3), S or O.
X7 is N or N(Rx1). Included is furthermore the option that any of groups X1 to X7 containing a nitrogen atom can form an N-oxide group.
X8 and X9 are each independently selected from N, C, or C(R*2).
Each Rx1 is independently selected from hydrogen, C C4 alkyi, -OH, -0{CrC4 alkyi), or -(C1-C4 alkyiene)-phenyl. Preferably, each Rx1 is independently selected from hydrogen, or d-d alkyi.
Each Rx2 and each R 3 is independently selected from hydrogen, d-d aikyl, halogen, -CF3, -CN, -OH, -0(CrC4 alkyi), -COOH, -COO(C C4 alkyi), -CONH2, -CONH(C C4 alkyi), -CON(C C4 alkyl)(d-C4 alkyi), -NH2, -NH(C C4 aikyl), or -N(C C alkyl)(d-C4 alkyi), wherein the two C C4 aikyl moieties of said -N(CTC4 alkyl)(C1-C4 alkyi) or of said -CON(C d aikyl)(C C4 aikyl) are optionally mutually linked to form a ring (such as, e.g. , a pyrrolidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to. Preferably, each RxZ and each Rx3 is independently selected from hydrogen, d-C4 aikyl, halogen, -CF3, -CN, -OH, -0(C d alkyi), -NH2, -NH(d-C4 aikyl), or -N(d-d alkyl)(Crd alkyi) wherein the two d-d alkyi moieties of said -N(d-C4 alkyl)(d-d aikyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to. More preferably, each Rx2 and each Rx3 is independently selected from hydrogen, - alkyi, halogen, -CF3, -OH, or -0(C C4 alkyi).
As will be understood, the bicyciic moiety corresponding to formula (II) is linked to the remainder of the compound of general formula (I) by the interrupted bond shown in formula (II), i.e. As will further be understood, the absence or the presence of one or two substituents Rx1, Rx2 and Rx3 in the groups X1 to X9 will depend on the presence and the position of double bonds in the bicyciic ring system of formula (I I).
In preferred embodiments, one or two of the groups X1 to X6, Xs and X9 are each independently selected from N, N(R ), O, S, S(O), or S(0)2 (preferably, selected from N, N(Rx1), S or O) in the case of X1 to Xs and from N in the case of X8 and X9, respectively, and the remaining groups X1 to X6, Xs and X9 are each independently selected from C(Rx2) or C(Rx2)(R 3) in the case of X1 to X6 and from C or C(RxZ) in the case of X8 and X9, respectively. Accordingly, in one embodiment one or two of the groups X1 to X6 are each independently selected from N, N(R ), O, or S, the remaining groups X1 to X6 are each independently selected from C(R 2) or C(R 2)(R 3), and X8 and X9 are each independently selected from C or C(R*2); alternatively, in another embodiment one or none of the groups X1 to X6 is selected from N, N(R ), O, or S, the remaining groups X1 to X6 are each independently selected from C(Rx2) or C(Rx2)(R 3), one of X8 and X9 is N, and the other one of X8 and X9 is C or C(R"2).
In further preferred embodiments, one or two of the groups X1 to X6, Xs and X9 are each independently selected from N, N(Rx1) or S in the case of X1 to X6 and from N in the case of Xs and X9, respectively, and the remaining groups X1 to X6, Xs and X9 are each independently selected from C{Rx2) or C(Rx2)(R 3) in the case of X1 to X6 and from C or C(R 2) in the case of X8 and Xs, respectively. Accordingly, in one embodiment one or two of X1 to X6 are each independently selected from N, N(R 1) or S, the remaining groups X1 to X6 are each independently selected from C(Rx2) or C(Rx2)(Rx3), and Xs and X9 are each independently selected from C or C(Rx2); alternatively, in another embodiment one or none of the groups X1 to X6 is selected from N, N(Rx1) or S, the remaining groups X1 to Xs are each independently selected from C(Rx2) or C(Rx2)(Rx3), one of X8 and X9 is N, and the other one of X8 and X9 is C or C(Rx2).
It is preferred that A is a bicyclic moiety corresponding to formula (II) as described and defined herein above, wherein the first ring of the bicyclic moiety (i.e. the ring which is linked to the remainder of the compound of general formula (I)) is aromatic, and it is more preferred that A is a bicyclic moiety corresponding to formula (II) as described and defined herein above, wherein the two rings of the bicyclic moiety form an aromatic ring system.
It is particularly preferred that A is one of the following groups:
Figure imgf000022_0001
Figure imgf000023_0001
each of which may optionally be substituted on ring carbon atoms with one or more groups independently selected from C^-C4 aikyl, halogen, -CF3, -CN, -OH, -0(Ci-C aikyl), -NH2, -NH(C C4 alkyl), or -N(C C4 alkyl)(C C4 alkyl) wherein the two d-C4 alkyl moieties of said -N(d-C4 aikyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from C C4 alkyl, halogen, -OH, or -0(C C aikyl). Furthermore, each Rx1 (if present) is independently selected from hydrogen, C C4 aikyl, -OH, or -0(C C4 alkyl), and preferably selected from hydrogen or d-d alkyl. Each Rx4 (if present) is independently selected from hydrogen or Cj-C4 aikyl, or the two groups R 4 (if present) are each independently C1-C4 alkyl and are mutually linked to form a ring (such as, e.g., a pyrrolidinyl ring or a piperidinyl ring) together with the nitrogen atom which they are attached to. It is preferred that the above depicted optionaiiy substituted groups are unsubstituted and R 1 (if present) is hydrogen. .
More preferably, A is one of the following groups:
Figure imgf000023_0002
Figure imgf000024_0001
each of which may optional!y be substituted on ring carbon atoms with one or more groups independently selected from C1-C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(d-d alkyl), or -N(d-C4 aikyi)(d-C4 alkyl) wherein the two C C4 alky! moieties of said -N(d-d alky!)(d-C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from d-d alkyl, halogen, -OH, or -0{C-|-C4 alkyl). It is preferred that the above depicted optionally substituted groups are unsubstituted.
Even more preferably, A is one of the following groups:
Figure imgf000024_0002
each of which may optionally be substituted on ring carbon atoms with one or more groups independently selected from d- alkyl, halogen, -CF3l -CN, -OH, -0(d-d alkyl), -NH2, -NH(C alkyl), or -N(d-C4 alkyl)(C C4 alkyl) wherein the two - alkyl moieties of said -N(d~d alkyl)(C d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, and preferably selected from d-d alkyl, halogen, -OH, or -0(C C4 aikyf). It is preferred that the above depicted optionally substituted groups are unsubstituted.
It is particularly preferred that the compound of the general formula (I) is selected from:
2-isoquinolin-3-yl-chromen-4-one oxime;
7~bromo-2-isoquinolin-3-yl-chromen-4-one oxime;
7-bromo-2-isoquinolin-3-yl-6-methyl-chromen-4-one oxime;
6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime; -isoquinolin-3-yl-6-methyl-chromen-4-one oxime;
- fluoro-2-isoquinolin-3-y[-chromen-4-one oxime;
,8-difluoro-2-isoquinoiin-3-yl-chromen-4-one oxime;
-ch!oro-2-isoquinolin-3-yl-chromen-4-orie oxime;
-fluoro-2-isoquinolin-3-yl-chromen-4-one-(Z)- oxime;
-isoquinoiin-3-yi-6-trifluoromethoxy-chromen-4-one oxime;
-isoquinolin-3-yt-6-trifluoromethyl-chromen-4-one oxime;
-(7-fluoro-isoquinolin-3-yl)-chromen-4-one oxime;
-(7-methoxy-isoquino[in-3-yl)-chromen-4-one oxime;
-(6.7-dimethoxy-isoquinolin-3-yl)-chromen-4-one oxime;
-(6-methyl-isoquinolin-3-yl)-chromen-4-one oxime;
-(7-chloro-isoquinolin-3-yl)-chromen-4-one oxime;
-(5-bromo-isoquinoiin-3-yl)-chromen-4-one oxime;
-(5-hydroxy-isoquinoSin-3-yl)-chromen-4-one oxime;
-{5-meihoxy-isoqutno!in-3-yl)-chromen-4-orie oxime;
-isoquinoiin-3-yl-7-phenylethynyi-chromen-4-one oxime;
-isoquinoiin-3-yl-7-((E)-styryl)-chromen-4-one oxime;
-isoquinolin-3-yl-7-phenet yl-chromen-4-one oxime;
- ethynyl-2-isoquinoIin-3-yS-chromen-4-one oxime:
-isoquino[in-3-yl-7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime;
-isoquinofin-3-yl-7-(pyridin-2-yl~ethynyl)-chromen-4-one oxime;
-!soquinolin-3-yi-7-(pyridin-4-y!)ethyny!-chromen-4-one oxime;
-{4-dimethyiaminophenyl)ethynyi-2-isoquino!in-3-yl-chromen-4-one oxime;-isoquinoiiri-3-y!~7-(3-methoxyphenyl)ethynyl-chromen-4-one oxime;
-(3-aminopheny!)ethynyl-2-isoquinoiin-3-yl~chromen-4-one oxime;
-(3-hydroxyphenyi)ethynyl-2-isoquinolin-3-yl-chromen-4-one oxime;
-isoquinolin-3-yl-7-(4-methoxyphenyl)ethynyl-chromen-4-one oxime; 7-(2-chlorophenyl)ethynyl-2-isoqutrioiin-3-yl-chromen-4-one oxime;
7-(3-dimethylamino-prop-1-ynyi)-2-isoquinolin-3-yl-chromen-4-one oxime;
6-cycSopropyi-2-isoquinoiin-3-yi-chromen-4-one oxime;
2-isoquinolin-3-yl-6-(pyrrolidin-1 -yl)-chromen-4-one oxime;
2-isoquinolin-3-yi-6-(vinyI)-chromen-4-one oxime;
6-ethyl-2-isoquinolin-3-yl-chromen-4-one oxime;
6-cyano-2-isoquinolin-3-yl-chromen-4-one oxime;
6-dimethyiamino-2-isoquinolin-3~yl-chromen-4-one oxime;
2-isoquinolin-3-yf-6-(morpholin-4-yi-methyl)-chromen-4-orie oxime;
6-hydroxy-2-isoquinolin-3-yl-chromen-4-one oxime;
2-isoquinolin-3-yl-6-methoxy-chromen-4-one oxime;
2-isoquinofin-3-yl-6-(2-meihoxy-ethoxy)-c romen-4-one oxime;
6- (2-dimethylamino-ethoxy)-2-isoquinoiin-3-yl-chromen-4-one oxime;
2-isoquinolin-3-yl-6-[3-(4-methyi-piperazin-1-yl)-propy!amino]-chromen-4-one oxime; 2-isoquinoltn-3-yl-6-[2-(4-methy!-piperazin-1-yl)-eihoxy]-chromen-4-one oxime;
2-isoquinolin-3-yl-7-phenyl-chromen-4-one oxime;
7- (4-biphenyi)-2-isoquinolin-3-yI-chromen-4-one oxime;
2-isoquinoiin-3-yl-7-[3-(4-methy]-piperazin-1-y[)-propylamino]-chromen-4-one oxime;
2-isoquinolin-3-yi-7-{3-[2-(4-methyl-piperazin-1-yl)-ethoxy]-phenylethynyl}-chromen-4- one oxime;
2-lsoquinolin-3-yl-7-{3-methylaminophenylethynyl}-chromen-4-one oxime;
7-(4-Hydroxy-but-1-ynyI)-2-isoquinoiin-3-yi-chromen-4-one oxime;
2-lsoquinolin-3-yl- 7-f3-(2-methoxy-e†hoxy)-phenylethynyl3-chromen-4-one oxime; 2-lsoquinolin-3-yl-7-[3-(2-methoxy-ethoxy)-prop-1 -ynyl]-chromen~4~one oxime;
7-but-3-en-1 -ynyl-2-!soquinoiin-3-yi-chromen-4-one oxime;
2-isoqusnoiin-3-yl-7-methoxy-chromen-4-one oxime;
2-isoquinoiin-3-yl-7-(2-methoxy-ethoxy)-chromen-4-one oxime; -cyano-2-isoquinolin-3-yl-chromen-4-one oxime;
~lsoquinoiin-3-yl-7-[3-(4-meihy!-piperazin-1 -yl)-prop-1 -ynyi]-chromen-4-one oxime;-(7-hydroxy-isoquinolin-3-yl)-chromen-4-one oxime;
-[2,6]Naphthyridin-3-yl-chromen-4-one oxime;
-[1 ,6]Naphthyridin-3-yl-chromen-4-one oxime;
-Pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
-(5!7-dimethyl-Pyrrolo[1 ,2-c]pyrimidin-3-yl)-chromen-4-one oxime;
-(6-bromo-Pyrrolo[1 ,2-c]pyrtmidin-3-yl)-chromen-4-one oxime;
-bromo-2-Pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
-methoxyethoxy-2-Pyrro!o[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
-(1-8enzy!-1H-imidazo[4,5-c]pyridin-6-yl)-chromen-4-one oxime;
-Thieno[2,3-c]pyridin-5-yl-chromeri-4-one oxime;
-Thieno[3,2-c]pyridin-6-y[-chromen~4-one oxime;
- lsoquinolin-3-yl-3-methyl-chromen-4-one oxime;
- {4-[(E)-Hydroxyimino]-4H-chromen-2-yl}-2H-isoquinolin-1 -one;
-lmidazo[1 ,2-c]pyrimidin-7-y!-chromen-4-one oxime;
-{1 H-Pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxime;
-(1-Hydroxy-1 H-pyrro!o[2,3-c]pyridin-5-yl)-chromen-4-one oxime;
-(1- ethyl-1 H-pyrrolo[2,3-c]pyridin-5-yi)-chromen-4-one oxime;
-(1-Methoxy-iH-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxime;
-Hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
-Thiazolot5,4-c)pyridin-6-yl-chromen-4-one oxime;
-{3-Methoxy-propyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
-(3-Dimethylamino-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3~y!~chromen-4-one oxime;-(3- orphoiin-4-yi-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;-(2I3-Dihydroxy-propoxy)-2-pyrrolo[1 I2-c]pyrimidin-3-yl-chromen-4-one oxime;-(2-Pyrro!idin-1 -yl-ethoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime; 6-(2-Piperidin-1-yl-eihoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4-one oxime;
6-(2-dimethyiamino-ethoxy)-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime; 6-(2-d!ethyiamino-ethoxy)-2-pyrroio[1 l2-c]pynmidin-3-yl-chromen-4-one oxime;
2-Pyrroio[1 ,2-a]pyrazin-3-yl-chromen-4-one oxime;
6-[2-(2-methyl-pyrrolidin-1-y!)-ethoxy3-2-pyrrolo[1 ,2-c]pyrimidin-3-y!-chromen~4-one oxime;
6-[2-(4-methyl-piperazin-1 -yi)-ethoxy]-2-pyrrolo[1 ,2-c] pyrimidiri-3-yi-chromen-4-one oxime;
6-[2-(4-methyl-[1 ,4]diazepan-1 -yl)-ethoxy]-2-pyrro!o[1 ,2-c] pyrimidsn-3-y!-chromen-4- one oxime;
6-[2-((S)-2-hydroxymeihy!pyrro!idin-1-yi)-ethoxy]-2-pyrroio[1 ,2-c] pyrimidin-3-yl- chromen-4-one oxime;
6-[2-({S)-2-methoxymethylpyrrolidin-1-yi)-ethoxy]-2-pyrrolo[1 !2-c]pyrimidin-3-yl- chromen-4-one oxime;
6-[2-({R)-2-meihoxymethylpyrrolidin-1-yl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime;
6-[2-(3-hydroxy-pyrroiidin-1 -yi)-ethoxy]-2-pyrro!o[1 ,2-c] pyrimidin-3-yl-chromei 4-one oxime;
6-[2-(4-dimeihylamino-piperidin-1-yl)-ethoxy]-2-pyrroio[1 ,2-c] pyrimidin-3-yl-chromen-
4- o ne oxime;
6-(2-cyclopentylamino-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-y[-chromen-4-one oxime;
6-(3-morpholin-4-yl-propoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(4-morpholin-4-yi-butyl)-2-pyrrolo[1 !2-c]pyrimidin-3-yi-chromen-4-one oxime;
6-(4.4-difluoro-piperidin-1-y)-2-pyrrolo[1.2-c] pyrimidin-3-yl-chromen-4-one oxime;
6-{2-[bis-(2-methoxy-ethyi)-amino]-ethoxy}-2-pyrroio[1 ,2-c] pyrimidin-3-yi-chromen-4- one oxime;
6-(2-imidazoi-1-yl-ethoxy)-2-pyrrolo[1 ,2-c] pyrimidin-3-yl-chromen-4-one oxime;
5- iS-morpholin^-yl-propyl^-pyrroloil ^-clpyrimidin-S-yl-chromen^-one oxime;
6- ((Z)-3-Morpholin-4-y!-propenyl))-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[2-(3-methoxy-piperidin-1 -yl)-ethox^
oxime;
6-[2-(4-fluoro-phenyl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yS-chromen-4-one oxime; 2-quinazolin-2~yl-chromen-4-one oxime;
6-[(1S,4S)-2-(2-oxa-5-aza-bicyc]o[2.2.1]hept-5-yi)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one oxime;
6-[2-(cis-2,6-dimethyi-morphoiin-4-yi)-ethoxy]-2-pyrrolo[1 ,2-c3pyrimidin-3-yl-c romen^ 4-one oxime;
2-pyrrolo[1 ,2-c]pyrimidin-3-yl-6-[2-(4 rifluoromethyl-piperidin-1-yl)-ethoxy]-chrorrien--4- one oxime;
6-[2-(3,3-difluoro-pipendin-1-yl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
4-(hydroxyimino)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromene-6-carboxylic acid (2- morpholin-4-yi-ethyl)-amide;
6-(2-[1 ,4']bipiperidinyl-1 '-yl-ethoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-y!-chromen-4-one oxime;
6-(2-[1 ,4]oxazepan-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromerh4-one oxime;
6-[2-(4-pyrrolidin-1-y[-piperidin-1-yl)-efo^
one oxime;
6- [2-(3I3-difluoro-pyrroridin-1~yl)-ethoxy]-2-pyrro!o[1 t2-c]pyrimidin-3-yl~chromen-4-one oxime;
7- [3-(3-dimethylamino-propoxy)-phenyiethynyl]-2-pyrrolo[1 ,2-c]pyrimidin-3-yi- chromen-4-one oxime;
6-[2-(4-eihyI-piperazin-1 -yl)-ethoxy]-2-pyrroio[1 ,2~c]pyrimidin-3-yl-chromen-4-one oxime;
6-(2-amino-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(2-MorphoIin-4-yl-ethoxy)-2-(8aH-pyrrolo[1 ,2-a]pyrazin~3-yf)-chromen-4-one oxime;
6-[2-(4,4-Dsfluoro-piperidin-1-yl)-ethoxy]-2-pyrroSo[1 ,2-a]pyrazin-3-yl-chromen-4-one oxime; 6-(2-lmidazol-1 -yi-ethoxy)-2-pyrrolo[1 ,2-a]pyrazirv3-yI-chromen-4-one oxime;
6-[2-(4-Fluoro-phenyl)-ethoxy]-2-pyrrolo[1 I2-a]pyrazin-3-yi-chromen-4-one oxime;
6-(2-Morpholin-4-yl-ethoxy)-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(4,4-Difluoro-piperidin-1-yi}~e†hoxy]- 2-thieno[3,2-c]pyridin-6-y!-chromen-4-one oxime;
6-(2-imidazol-1 -yl-ethoxy) 2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(4-Fiuoro-phenyi)-ethoxy]-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(3 -Difluoro-piperidin-1 -yl)-ethoxy]-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(2!6-Dimethy!-morpholin-4-y[)-ethoxy]-2-thieno[3I2-c]pyridin-6-yi~chromen-4-one oxime;
6-[2-{3,3-Difiuoro-pyrrolidin-1 -yl)-ethoxy]-2 hieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-(3-Pyridin-4-yi-propoxy)-2-.thieno[3,2-c]pyridin-6-yi-chromen-4-one oxime;
6-(3-Pyridin-3-yl-propoxy)-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-(2-Pyridin-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
2-Pyrrolo[1 ,2-c]pynmidin-3-yi-6-[2-(4-trifSuoromethyl-phenyl)-ethoxy]-chromen-4-one, oxime;
6-I2-(3-Fluoro-phenyl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one, oxime;
5- Methoxy-2-pyrroio[1.2-c]pyrimidin-3-yl-chromen-4-one oxime;
6- [2~(4-Chioro-phenyl)-ethoxy3-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
2-Pyrrolo[1 ,2-c]pyrimidin-3-yl-6-[2-(3-trifluoromethyl-phenyl)-ethoxy]-chromen-4-one oxime;
7- (2- orphoiin-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-y!-chromeri-4-one oxime;
7-(3-Morpholin-4-yl-propoxy)-2-pyrro!o[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[(4-Fluoro-benzylamino)-methyi]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-{[2-(4-F(uoro-phenyl}-ethylamino]-methyl}-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4- one oxime; 6-Phenethyloxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[2-(Pyridin-4-yloxy)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[2-(Pyridin-3-yioxy)-ethoxy]-2-pyrroio[1 ,2-c]pyrirnidin-3-yl-chromen-4-one oxime;
6-[3-(Pyridin-3-y!oxy)-propoxy]-2-pyrrolo[1 ,2-G]pyrimidin-3-yl-chromen-4-one oxime;
N-(4'Fiuoro-phenyI)-2-{4-hydroxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromen-6- y!oxy}-acetamide;
N-(4-FIuoro-phenyI)-2 4-hydroxyimino-2-pyrrolo[1 !2-c]pyrimidin-3-yl-4H-chromen-6- yioxy}-N-meihyl-acetamide;
N-(5-Fluoro-pyridin-2-yI)-2-{4-hydroxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromen
6- y I o xy}-a ceta m i de ;
2-Pyrrolo[1 ,2-c]pyrimidin-3-yl-5-trifluoromethyl-chromen-4-one oxime;
2-(7-tert-Butyl-pyrrolo[1 ,2-c]pyrimidin-3-yi)-6-(2-morpholin-4-yi-ethoxy)-chromen-4-one oxime;
7- (2-Morpho[in-4-yi-eihoxy)-2-ihieno[2,3-c]pyridin-5-yl-chromen-4-one oxime;
5-(2- orpholin-4-yf-eihoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
5- {3-Morphoiin-4-yi-propoxy)-2-pyrro!o[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6- (2-Morphoiin-4-yi-ethoxy)-2-thieno[2I3-c]pyridin-5-yl-chromen-4-one oxime;
6-(3-Pyridsn-4-yi-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(2-Phenoxy-eihoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6~[3-(4-Fluoro-phenoxy)-propoxy]-2-pyrrolo[1.2-c]pyrimidin-3-yl-chromen~4-one oxime;
6-(3-Phenoxy-propoxy)-2-pyrro]o[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
e-tS^S-Fluoro-phenoxyj-propoxyl^-pyrroloEI ^-clpyrimidin-S-yl-chromen^-one oxime;
6-[3-(3,4-Difluoro-phenoxy)-propoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3~yl-chromen-4-one oxime;
6-(2-[1 ,4]Oxazepan-4-yl-ethoxy)-2-ihieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(3,4-Dihydro-1 H-isoquinolin~2-yi)-ethoxy]-2-thieno[3,2-c]pyridin-6-yl-chromen-4- orie oxime;
6-[2-(4-fIuoro-piperidin-1-yl)-eihoxy]-2~thieno[3,2-c]pyridin-6-yi-chromen~4~one oxime; 6-[2-(1 ,1-Dioxo-1-†hiomorpholin-4-yI)-ethox
one oxime;
6-{1-Pyrimidin-2-yl-piperidin-4-ylo^ oxime; e-fS^.S.e-Tetrahydro^H-EI ^'jbip ridinyl^-yloxyJ^-thieno S^-cjpyridin-e-yl-chromen- 4-one oxime; and
pharmaceutically acceptable salts, solvates and prodrugs thereof.
Compounds of the present invention are characterized in that they provide mGluR4 activity due to two specific features: 1) the presence of a bicyciic ring on position 2 of the chromone ring (i.e. as group A in formula (I)), and 2) the presence of a nitrogen atom on the first ring of that bicyciic ring and at the position adjacent to the carbon linked to the chromone moiety but not adjacent to any of the two ring atoms which form part of both rings condensed to said bicyciic ring, as shown in the scheme below. It will be understood that this is a schematic representation, and that the N atom in the bicyciic system can be bound via a double bond in the ring structure or can carry a substituent as defined above:
Figure imgf000032_0001
This can be demonstrated, e.g. from the comparison of activities of the compounds depicted in the following scheme:
Figure imgf000033_0001
Example 1
Example 74 {reference) Exam le 7S (reference)
mGluR4 PAM
Inactive on mGI-F!4 up to 100μΜ Inactive on mGluR4 up to 100μΜ
Figure imgf000033_0002
Exam le 63 Exam le 80 (reference)
mGluR4 PAW! Inactive on mGluR4 up to 100μΜ
ΕΟ^Ο.δμΜ
Example 1 is a positive allosteric modulator (PAM) of mGluR4 having an EC50 lower than 1 μΜ. Example 74 (reference), which has no PAM activity on mGluR4 up to 100 μΜ, is the isoster of example 1 without the nitrogen atom on the required position. Example 79 (reference), which has no PAM activity on mGluR4 up to 100 μΜ, is the isomer of example 1 with a nitrogen atom on the other position adjacent to the carbon linked to the chromone moiety. Comparison of activities of these three molecules demonstrates that a nitrogen atom on the position X7 of Formula (II) is required for the mGluR4 PAM activity.
Similarly, example 63 is an mGluR4 PAM having an EC50 lower than 1 μΜ. Example 80 (reference), that has no PAM activity on mGluR4 up to 100 μΜ, is an analog of example 63 with a monocyclic heteroaryl. Comparison of activities of these two molecules demonstrates that a bicyclsc ring is required as moiety A in formula (!) for the mGluR4 PAM activity.
Experimental details of the syntheses and evaluations of examples 1 , 63, 74 (reference), 79 (reference) and 80 (reference) are provided in the Examples section of this application.
All isomers, including configuration isomers and possible stereoisomers of the compounds according to the invention are contemplated as part of the present invention, either in admixture or in pure or substantially pure form, in particular, the compounds and compositions of the invention may have the E- and the Z-configuration of the oxime group (=N(OH)) shown in formula (I), and the inventio comprises compounds and compositions only showing the E-configuration, those showing only the Z-configuration, and mixtures of the E- and the Z-configuration.
As for stereoisomers, it embraces the racemic forms and the isolated optical isomers. The racemic forms can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates using conventional methods, such as, e.g., salt formation with an optically active counterpart followed by crystallization.
As used herein, "a!kyl" represents a straight or branched chain saturated hydrocarbon residue which does not comprise any carbon-to-carbon double bonds or carbon-to- carbon triple bonds.
As used herein, "alkenyl" represents a straight or branched chain unsaturated hydrocarbon residue comprising at least one carbon-to-carbon double bond.
As used herein, "alkynyl" represents a straight or branched chain unsaturated hydrocarbon residue comprising at least one carbon-to-carbon triple bond.
As used herein, "alkylene" represents a straight or branched chain alkanediyl group which does not comprise any carbon-to-carbon double bonds or carbon-to-carbon triple bonds.
As used herein, "alkenylene" represents a straight or branched chain alkenediyl group comprising at least one carbon-to-carbon double bond.
As used herein, "alkyny!ene" represents a straight or branched chain alkynediyl group comprising at least one carbon-to-carbon triple bond.
As used herein, "aryl" represents a 6-10 membered aromatic hydrocarbon ring, including bridged ring or fused ring systems containing at least one aromatic ring. "Aryl" may, for example, refer to phenyl or naphthyl.
As used herein, "heteroaryl" represents a 5-14 membered aromatic ring, including bridged ring or fused ring systems containing at least one aromatic ring, comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein one or more of said S ring atoms (if present) and/or one or more of said N ring atoms (if present) may optionally be oxidized. A "heteroaryl", as defined herein above, preferably represents a 5-14 membered aromatic ring, including bridged ring or fused ring systems containing at least one aromatic ring, comprising one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N. "Heteroaryl" may, for example, refer to thienyl (thiophenyi), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (furany!), isobenzofuranyl, chromenyl, xanthenyl, phenoxathiiny!, pyrrolyl (including, without limitation, 2H-pyrroiyl), imidazolyl, pyrazoiyl, pyridyl (pyridinyl; including, without limitation, 2-pyridyl, 3-pyridyl, and 4-pyridyl), pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, indolyl (including, without limitation, 3H-indolyl), indazolyl, purinyi, isoquinolyl, quino!y!, phthalazinyl, naphthyridinyl, quinoxaiinyl, cinnoiinyl, pteridinyl, carbazoly!, beta-carboSinyl, phenanthridinyl, acridinyl, perimidiny!, phenanthrolinyl (including, without limitation, [1 ,10]phenanthrolinyl, [1 ,7]phenanthro- linyl, and [4,7]phenanthroiinyl), phenazinyl, isothiazolyl, phenothiazinyl, oxazo!y!, isoxazolyl, furazanyl, phenoxazinyl, pyrazolo[1 ,5-a]pyrimidinyl (including, without limitation, pyrazolo[1 ,5-a)pyrimidin-3-yl), 1 ,2-benzoisoxazol-3-yi, or benzimidazolyl.
As used herein, "cyc!oalkyi" represents a 3-10 membered saturated hydrocarbon ring, including bridged ring or fused ring systems. "Cycloalkyl" may, for example, refer to cyclopropy!, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl.
As used herein, "heterocycloalky!" represents a 3-10 membered saturated ring, including bridged ring, spiro ring or fused ring systems, containing one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N, wherein one or more of said S ring atoms (if present) and/or one or more of said N ring atoms (if present) may optionally be oxidized. A "heterocycloalkyl", as defined herein above, preferably represents a 3-10 membered saturated ring, including bridged ring or fused ring systems, containing one or more (such as, e.g., one, two, or three) ring heteroatoms independently selected from O, S, or N. "Heterocycloalkyl" may, for example, refer to tetrahydrofuranyi, piperidinyi, piperazinyl, aziridinyl, azetidinyl, pyrrolidinyl, imidazoiidinyl, morpholinyl, pyrazolidinyi, tetrahydrothienyl, octahydroquinolinyl, octahydroisoquinolinyl, oxazolidiny!, isoxazolidinyl, azepanyl, diazepanyl, oxazepanyl or 2-oxa-5-aza-bicyclo[2.2, 1]hept-5-yl.
As used herein, "halogen" represents fiuoro, chloro, bromo. or iodo, and in particular fiuoro, chloro, or bromo.
Various groups are referred to as being "optionally substituted" in the context of this description. Generally, these groups may carry one or more, such as e.g. one, two, three or four substiiuents. Unless defined otherwise in the specific context, these groups carry preferably not more than two substiiuents.
The scope of the invention embraces ail pharmaceutically acceptable salt forms of the compounds of the general formula (I) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of a carboxylic acid group with a physiologically acceptable cation as they are well known in the art. Exemplary base addition salts comprise, for example, alkaii metal salts such as sodium or potassium salts; alkaline-earth metai salts such as calcium or magnesium salts; ammonium salts; aliphatic amine salts such as trimethyiamine, triethylamine, dicyc!ohexylamine, ethanolamine, diethanolamine, triethano!amine, procaine salts, meglumine salts, diethanol amine salts or ethylenediamine salts; aralkyl amine salts such as Ν,Ν-dibenzylethylenediamine salts, benetamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoHne salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethy!ammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylammonium salts, benzyltributylammonium salts, methyltrioctylammonium salts or tetrabutylammonium salts; and basic amino acid salts such as arginine salts or lysine salts. Exemplary acid addition salts comprise, for example, mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts, nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts or perchlorate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, undecanoate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, nicotinate, benzoate, salicylate or ascorbate salts; sulfonate salts such as methanesulfonate, ethanesulfonate, 2-hydroxyethanesulfonate, benzenesulfonate, p-toluenesuifonate (tosylate), 2-naphthalenesulfonate, 3-phenylsulfonate, or camphorsulfonate salts; and acidic amino acid salts such as aspartate or giutamate salts.
Moreover, the scope of the invention embraces solid forms of the compounds of the general formula (I) in any solvated form, including e.g. solvates with water, for example hydrates, or with organic solvents such as, e.g., methanol, ethano! or acetonitri!e, i.e. as a methanolate, ethanolate or acetonitrilate, respectively; or in the form of any polymorph.
Pharmaceutically acceptable prodrugs of compounds that can be used in the present invention, in particular the compounds of the general formula (I), are derivatives which have chemically or metabolically cieavable groups and become, by solvo!ysis or under physiological conditions, the compounds used in the present invention which are pharmaceuticaily active in vivo. Prodrugs of compounds that can be used in the present invention may be formed in a conventional manner with a functional group of the compounds such as with an amino, hydroxy or carboxy group. The prodrug derivative form often offers advantages of solubility, tissue compatibility or delayed release in a mamma!ian organism (see, Bundgaard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well known to the person skilled in the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine. When a compound employed in the present invention, in particular a compound of the general formula (I), has a carboxyl group, an ester derivative prepared by reacting the carboxyl group with a suitable alcohol or an amide derivative prepared by reacting the carboxyl group with a suitable amine is exemplified as a prodrug. An especially preferred ester derivative as a prodrug is methylester, ethyiester, n-propylester, isopropylester, n-butylester, isobutylester, tert-butylester, morpholinoethylester or Ν,Ν-diethylg!ycoSamidoester. When a compound employed in the present invention has a hydroxy group, an acyloxy derivative prepared by reacting, the hydroxy! group with a suitable acylhalide or a suitable acid anhydride is exemplified as a prodrug. An especially preferred acyloxy derivative as a prodrug is -OC(=0)-CH3, -OC(=0)-C2H5, -OC(=0)-C3H7, -OC(=0)-(tert-butyl), -OC(=0)-C15H31, -OC(=0)-CH2CH2COONa, -0(C=0)- CH(NH2)CH3 or -OC(=0)-CH2-N(CH3)2. When a compound employed in the present invention has an amino group, an amide derivative prepared by reacting the amino group with a suitable acid halide or a suitable mixed anhydride is exemplified as a prodrug. An especially preferred amide derivative as a prodrug is -NHC(=0)- (CH2)2OCH3 or -NHC(=0)-CH(NH2)CH3. Accordingly, the oxime -OH group of the compounds of the general formula (I) may be in the form of an O-acyl-oxime (or acyloxy derivative), such as, e.g., -OC(=0)-CH3, -OC(=0)-C2H5, -OC(=0)-C3H7, -OC(=0)-(tert-butyi), -OC(=0)-C15H31, -OC(=0)-CH2CH2COONa, -0{C=0)-CH{NH2)CH3 or -OC(=0)-CH2-N(CH3)2. The oxime -OH group of the compounds of the general formula (I) may also be in the form of O-alkyl-oxime such as, e.g., -0-CH3, -0-C2H5, -O-C3H7 or -O-(tert-butyl). The oxime -OH group of the compounds of the general formula (I) may also be in the form of O-dialkySphosphinyloxy such as -0-P(=0)-[0-(CH3)2] , -0-P(=OHO-{C2-C5)23 , -O- Ρ(=Ο)-[Ο-(03-07)2] or -0-P(=0)-[0-(tert-butyl)2] or in the form of O-phosphoric acid -O- P(=0)-(OH)2 or in the form of -O-sulfuric acid -0-S02-OH.
The compounds described herein, in particular the compounds of general formula (I), may be administered as compounds per se or may be formulated as medicaments. Within the scope of the present invention are pharmaceutical compositions comprising as an active ingredient one ore more compounds of the general formula (I), as defined herein above. The pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, or antioxidants.
The pharmaceutical compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in Remington's Pharmaceutical Sciences, 20th Edition. The pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, rectal, nasal, topical, aerosol or vaginal administration. Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersibie powders and granules, medicated gums, chewing tablets and effervescent tablets. Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration. Dosage forms for recta! and vaginal administration include suppositories and ovula. Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler. Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems. The compounds according to the invention, in particular the compounds of the general formula (I), or the above described pharmaceutical compositions comprising one ore more compounds of the general formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripheraily or at the site of desired action, including but not limited to one or more of: oral (e.g. as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e. g., using injection techniques or infusion techniques, and including, for example, by injection, e.g. subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously or intramuscularly), pulmonary (e.g., by inhalation or insufflation therapy using, e.g., an aerosol, e.g. through mouth or nose), gastrointestinal, intrauterine, intraocular, subcutaneous, ophthalmic (including intravitreal or intracameral), rectal, and vaginal.
If said compounds or pharmaceutical compositions are administered parenterally, then examples of such administration include one or more of: intravenously, intraarterially, intraperitoneal^, intrathecal^, intraventricularly, intraurethra!ly, intrasternally, intracranial^, intramuscularly or subcutaneously administering the compounds pharmaceutical compositions, and/or by using infusion techniques. For parenteral administration, the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications. The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmeilose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropy!methyicellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diiuents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
Alternatively, said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or it may be appiied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The compounds of the present invention may also be dermaily or transdermaily administered, for example, by the use of a skin patch.
Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route. For ophthalmic use, they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
For topical application to the skin, said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water
Typically, a physician will determine the actual dosage which will be most suitable for an individual subject. The specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
A proposed, yet non-limiting dose of the compounds of the general formula (1) for administration to a human (of approximately 70 kg body weight) may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose. The unit dose may be administered, for example, 1 to 4 times per day. The dose will depend on the route of administration. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
The subject or patient, such as the subject in need of treatment or prophylaxis, may be a eukaryote, an animal, a vertebrate animal, a mammal, a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), a murine (e.g. a mouse), a canine (e.g. a dog), a feline (e.g. a cat), an equine (e.g. a horse), a primate, a simian (e.g. a monkey or ape), a monkey (e.g. a marmoset, a baboon), an ape (e. g. gorilla, chimpanzee, orangutang, gibbon), or a human. The meaning of the terms "eukaryote", "animal", "mammal", etc. is well known in the art and can, for example, be deduced from Wehner und Gehring (1995; Thieme Verlag). In the context of this invention, it is particularly envisaged that animals are to be treated which are economically, agronomica!ly or scientifically important. Scientifically important organisms include, but are not limited to, mice, rats, rabbits, fruit flies like Drosophiia melagonaster and nematodes like Caenorhabditis elegans. Non-limiting examples of agronomica!ly important animals are sheep, cattle and pig, while, for example, cats and dogs may be considered as economically important animals. Preferably, the subject/patient is a mammal; more preferably, the subject/patient is a human.
The term "treatment of a disorder or disease" as used herein is well known in the art. 'Treatment of a disorder or disease" implies that a disorder or disease is suspected or has been diagnosed in a patient/subject. A patient/subject suspected of suffering from a disorder or disease typically shows specific clinical and/or pathological symptoms which a skilled person can easily attribute to a specific pathological condition (i.e. diagnose a disorder or disease).
"Treatment of a disorder or disease" may, for example, lead to a halt in the progression of the disorder or disease (e.g. no deterioration of symptoms) or a delay in the progression of the disorder or disease (in case the halt in progression is of a transient nature only). "Treatment of a disorder or disease" may also lead to a partial response (e.g. amelioration of symptoms) or complete response (e.g. disappearance of symptoms) of the subject/patient suffering from the disorder or disease. "Amelioration" of a disorder or disease may, for example, lead to a halt in the progression of the disorder or disease or a delay in the progression of the disorder or disease. Such a partial or complete response may be followed by a relapse. It is to be understood that a subject/patient may experience a broad range of responses to a treatment (e.g. the exemplary responses as described herein above).
Treatment of a disorder or disease may, inter alia, comprise curative treatment (preferably leading to a complete response and eventually to healing of the disorder or disease) and palliative treatment (including symptomatic relief).
Also the term "prophylaxis of a disorder or disease" as used herein is well known in the art. For example, a patient/subject suspected of being prone to suffer , from a disorder or disease as defined herein may, in particular, benefit from a prophylaxis of the disorder or disease. Said subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition. Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators. It is to be understood that a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in said patient/subject (for example, said patient/subject does not show any clinical or pathological symptoms). Thus, the term "prophylaxis" comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician. The terms "prophylaxis" and "prevention" are used herein interchangeably.
in the method for identifying an agent that binds to metabotropic glutamate receptor 4 (mG!uR4) described herein above, the test agent may, for example, be selected from nucleic acids, DNA, RNA, PNA, oligonucleotides, aptamers (Gold, Ann. Rev. Biochem. 64 (1995), 763-797)), aptazymes, RNAzymes, ribozymes (see e.g., EP-B1 0 291 533, EP-A1 0 321 201 , EP-B1 0 360 257), antisense DNA, antisense oligonucleotides, antisense RNA, siRNA, RNAi, shRNA, amino acids, peptides, polypeptides, proteins, glycoproteins, lipoproteins, nucleoproteins, antibodies (Harlow and Lane "Antibodies, A Laboratory Manual", CSH Press, Cold Spring Harbor, 1988), monocloncal antibodies, polyclonal antibodies, immunoglobulins, affibodies (Hansson, Immunotechnology 4 (1999), 237-252; Henning, Hum Gene Ther. 13 (2000), 1427- 1439), immunoreactive fragments, immunoreactive derivatives, antigens, epitopes, " haptens, cell-surface molecules, cofactors, ligands, small organic molecules, lectins or derivatives thereof, lectin fragments, irinectins (Phylos Inc., Lexington, Massachusetts, USA; Xu, Chem. Biol. 9 (2002), 933), anticalins (EP-B-1 1 017 814), hormones, peptide and protein hormones, non-peptide hormones, steroids, interieukins, interferons, cytokines, neurotransmitters, toxins, enzymes, polysaccharides, carbohydrates, lipids, lipopolysaccharides, vitamins, crown ethers, cyclodextrins, cryptands, calixarenes, aldehydes, thiols, amines, drugs, drugs of abuse, therapeutic agents, medicaments, pharmaceuticals, substrates, fragments, portions, components or products of microorganisms, metabolites of or antibodies to any of the above substances and the like. EXPERIMENTAL PROCEDURES
Method A
The 2-heterocycfic substituted chromones of the invention may be prepared using a common synthetic scheme illustrated in examp!e 1A, differing only in the starting phenol and heieroaryl ester (or heterocycloaikyl ester) compounds. (J. Med. Chem, 1999, 42 (1 1), 1881-1893).
Figure imgf000044_0001
Ill iv
Specifically, a substituted 2'-hydroxyphenyl ketone of Formula I, wherein R1, R2, R3, R4, and Rs are as described and defined herein above, is reacted with an appropriate heteroaryl ester or heterocycloaikyl ester of Formula 11, wherein R is methyl or ethyl and A is as described and defined herein above, in a solvent such as pyridine and in presence of a strong base, preferentially sodium hydride, to yield the corresponding diketone III. The diketone III is treated in strong acidic conditions especially with sulfuric acid (H2S04) in acetic acid (AcOH) to give the desired chromone IV.
Method B
2-heterocyclic substituted chromones IV can also be prepared using a typical 3 steps procedure well known in the art for flavone derivatives synthesis (eg. J. Med. Chem. 2004, 47, 6466-6475 ; J. Org. Chem. 1993, 68, 7903-7905 ; US04065467). Substituted 2!-hydroxyacetophenone I is reacted with a heieroaryl carboxylic acid or a heterocycloaikyl carboxylic acid of formula II, wherein R is hydrogen and A is as described and defined herein above, preferentially activated as acid chloride in pyridine to yield the corresponding ester of Formula V. The ester is treated with a strong base, preferentially potassium hydroxide, in a suitable solvent such as pyridine to perform the mixed Claisen rearrangement and to give the corresponding diketone of formula ill. The diketone III is then treated in strong acidic conditions especially with sulfuric acid in acetic acid to give the desired chromone of formula IV.
Figure imgf000045_0001
V
Method C
The chromone oximes of the invention of formula VI, in particular the compounds of the general formula (1), may be prepared by the foliowing procedure illustrated in example 73. The chromone derivative of formula IV, which may be prepared according to method A or method B as described herein above, is treated with hydroxyiamine hydrochloride in pyridine or methanol under microwave irradiation to yield directly the ox'sme compound of formula VI.
Figure imgf000045_0002
IV VI
Method D
The chromone oximes of the invention of formula VI, in particular the compounds of the general formula (I), may also be obtained using the following two steps procedure illustrated in example 1 B and 1 C: The appropriate chromone of formuia IV, which may be prepared according to method A or method B as described herein above, is reacted with ferf-butyi hydroxyiamine hydrochloride in a suitable solvent such as methanol and under microwave irradiation to yield the protected ie/f-buty!oxime product of formula VII (step 1). The protected oxime VII is treated with a Lewis acid such as titanium tetrachloride in an inert solvent, preferentially dich!oromethane, to give the desired free oxime of formuia VI (WO2004/52S69) (step 2).
Figure imgf000045_0003
The compounds of the present invention can be radio-labeled by carrying out their synthesis using precursors comprising at least one atom which is a radioisotope. Preferably, radioisotopes of carbon atoms, hydrogen atoms, sulfur atoms, or iodine atoms are employed, such as, e.g., 4C, 3H, 35S, or 1251. Compounds labeled with 3H (tritium) can also be prepared by subjecting a compound of the invention to a hydrogen exchange reaction such as, e.g., a platinum-catalyzed exchange reaction in tritiated acetic acid (i.e., acetic acid comprising 3H instead of 1H), an acid-cata!yzed exchange reaction in tritiated trifluoroacetic acid, or a heterogeneous-catalyzed exchange reaction with tritium gas. For a person skilled in the field of synthetic chemistry, various further ways for radio-!abeling the compounds of the present invention or preparing radio-labeled derivatives of the compounds are readily apparent. Fluorescent labels can be bound to the compounds according to the invention following methods well established in the art.
In this specification, a number of documents including patent applications and manufacturer's manuals are cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
The invention is also illustrated by the following illustrative figures. The appended figures show:
• Figure 1 : Effect of the compound of Example 63 administered intraperitoneal^ in the haloperidol-induced catalepsy test in the mouse. As also explained in Example 172, the figure shows the mean time of latency spent on the bar in each group of animals. At each time-point, the anticata!eptic effect of the compound of Example 63 was compared to the vehicle-treated group using ANOVA test followed by the Dunnett's test (*=p<0.05).
• Figure 2: Effect of the compound of Example 85 in the marble burying test in the mouse, administered p.o. at 100 or 300 mg/kg (see Example 173). The figure shows the mean number of marbles burying in each group of animals, as also explained in Example 173.
The invention will now be described by reference to the following examples, which are merely illustrative and are not to be construed as a limitation of the scope of the present invention. It will be understood that both possible configurations of the oxime group in the figures of the examples can be obtained.
Example 1 : 2-isoquinolin-3-yl-chromen-4-one oxim
Figure imgf000047_0001
Example 1A: 2-isoquinolin-3-yl-chromen-4-one
Figure imgf000047_0002
To a suspension of sodium hydride (60% in mineral oil, 227 mg, 5.7 mmol) in dry pyridine (4 ml) was added dropwise a solution of methyl isoquinoline-3-carboxylate (390 mg, 2.08 mmol) and 2'-hydroxy-acetophenone (257 mg, 1.89 mmol) in dry pyridine (4 ml). The mixture was heated at 90°C for 15 min., cooled to room temperature and poured into an ice cooled N hydrochloric acid aqueous solution. The product was extracted in dichloromethane. The organic layers were washed with a 1 N hydrochloric acid solution, brine, dried over sodium sulfate and concentrated to dryness. The residue was dissolved in acetic acid (10 ml) and treated with sulfuric acid (40 μΙ). The solution was heated to 100°C for 30 min. and cooled to room temperature. The solvents were removed under vacuum distillation. The solid was triturated in water, filtered, washed with a saturated solution of sodium hydrogenocarbonate and water. The solid was dried under vacuum to yield 2- lsoquinolin-3-yl-chromen-4-one (459 mg, 89 %) as beige solid.
1 H N R : CDCI3 δ (ppm): 9.32 (s, 1 H), 8.49 (s, 1 H), 8.27 (d, J = 7.3 Hz, 1 H), 8.06 (d, J = 7.9 Hz, 1 H), 8.01 (d, J = 7.9 Hz, 1 H), 7.85-7.63 (m, 4H), 7.59 (s, 1 H), 7.44 (t, J = 7.2 Hz, 1 H). Example 1 B; 2-isoquinotin-3-yl-chromen-4-one O-tert-butyl-oxime
Figure imgf000048_0001
To a suspension of 2-isoquinolin-3-yl-chromen-4-one (459 mg, 1.67 mmol) in methanol (11 ml) was added O-terf-buty! hydroxylamine hydrochloride (421 mg, 3.35 mml). The mixture was heated to 130°C under microwave irradiation for 30 min. The volatiles were removed by vacuum distillation and the residue was purified by flash chromatography (cychlohexane/ ethyl acetate 95/5) to yield 2-isoquinolin-3-yl- chromen-4-one O-te/1-butyl-oxime (387 mg, 67 %) as a yellow solid.
1 H NMR : CDCS3 S (ppm): 9.29 (s, 1 H), 8.30 (s, 1 H), 8.10 (dd, J = 7.9 Hz, J = 1.5 Hz, 1 H), 8.01 (d, J = 7.7 Hz, 1 H), 7.95 (d, J = 7.9 Hz, 1 H), 7.80 (s, 1 H), 7.75 (td, J = 7.0 Hz, J = 1.1 Hz, 1 H), 7.65 (td, J = 7.5 Hz, J = 1.1 Hz, 1 H), 7.42 (td, J = 7.7 Hz, J = 1.7 Hz, 1 H), 7.34 (dd, J = 8.3 Hz, J = 1.3 Hz, 1 H), 7.21 (td, J = 7.4 Hz, J = 1.3 Hz, 1 H), 1 .43 (s, 9H).
Example 1C: 2~isoquinolin-3-yl-chromen-4~one oxime
Figure imgf000048_0002
To an ice cooled solution of 2-isoquinolin-3-yl-chromen-4-one O-f erf- butyl - oxime (136 mg, 0.39 mmol) in dichloromethane (10 ml) was cautiously added dropwise a 1 M solution of titanium tetrachloride in dichloromethane (1.2 ml, 1.2 mmol). The mixture was stirred at 0°C 2 hours, then at room temperature 2 more hours and poured onto ice coid water (100 mi). The mixture was basified using a 6N aqueous sodium hydroxide solution until pH 10 and the yellow precipitate was collected by filtration. The solid was washed with water, dried and purified by flash chromatography (cychlohexane/ethyi acetate/dichloromethane: 80/10/10; 60/30/10 then 0/50/50) to yield 2-isoquinolin-3-yl-chromen-4-one oxime (71 mg, 62 %) as a yellow solid. The product was isolated as a 95/5 mixture of Z/E oxime isomers. Mp: 247-249°C.
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1 % HCOOH): > 95 %; RT = 4.94 min.
MS (ESI+): 289.3 [C18H12N202+H]÷ (m/z).
1 H N R of the major Z isomer : DMSO-d6 δ (ppm): 1 08 (s, 1 H), 9.42 (s, 1 H), 8.50 (s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), 8.15 (d, J = 7.9 Hz, 1 H), 7.93 (dd, J = 8.1 Hz, J = 1 .2 Hz, 1 H), 7.88 (td, J = 7.5 Hz, J = 1.1 Hz, 1 H), 7.78 (td, J = 7.4 Hz, J = 1.1 Hz, 1 H), 7.77 (s, 1 H), 7.60-7.48 (m, 2H), 7.31 (td, J = 7.4 Hz, J = 1.3 Hz, 1 H).
In a similar manner but utilizing the appropriate substituted 2'-hydroxy- acetophenones and/or heteroaryi esters were also prepared:
Example 2: 7-bromo-2~isoquinoiin-3~yl-chromen'4-one oxime
Figure imgf000049_0001
Example 2A: 7-bromo-2-isoquinolin-3-yl~7~bromo-chromen~4-one wasprepared using the method A illustrated in example 1A. 4>-Bromo-2'- hydroxyacetophenone (627 mg, 2.91 mmol) and methy!-2-isoquinoline-3-carboxylate (600 mg, 3.21 mmo!), was treated first with sodium hydride (60% in mineral oil, 350 mg, 8.73 mmol) in dry pyridine (2x6 ml) at 90°C for 15 min. and then with sulfuric acid (73 μί) in acetic acid (15 ml) at 100°C for 45 min. to yield 7-bromo-2-isoquinolin-3-yl- chromen-4-one (684 mg, 67%) as pink solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate : 80/20 then 50/50).
1 H NMR : CDCI3 δ (ppm): 9.32 (s, 1 H), 8.46 (s, 1 H), 8.13 (d, J = 8.5 Hz, 1 H),
8.07 (d, J = 8.1 Hz, 1 H), 8.01 (d, J = 8.1 Hz, 1 H), 7.89 (d, J = 1.7 Hz, 1 H), 7.82 (td, J =
6.8 Hz, J = 1.3 Hz, 1 H), 7.75 (td, J = 6.9 Hz, J = 1.3 Hz, 1 H), 7.59 (s, 1 H), 7.56 (dd, J = 8.5 Hz, J = 1.7 Hz, 1 H).
Example 28: 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared using the method D (step 1) illustrated in example 1 B. 7-bromo- 2-isoquinoiin-3-yl-chromen-4-one (682 mg, 194 mmol) was treated with O-ferf-butyl- hydroxylamine hydrochloride (486 mg, 3.87 mmol) in methanol (9 ml) under microwave irradiation to yield 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-feri-butyl oxime (629 mg, 77%) as a yellow solid after purification by flash chromatography over silica gel (cyc!ohexane/ethyl acetate : 50/50).
1 H NMR : CDCI3 δ (ppm): 9.28 (s, 1 H), 8.26 (s, 1 H), 8.02 (d, J = 8.1 Hz, 1 H), 7.94 (d, J = 8.6 Hz, 1 H), 7.93 (d, J = 8.1 Hz, 1 H), 7.78 (s, 1 H), 7.76 (td, J = 7.6 Hz, J = 1.3 Hz, 1 H), 7.66 (td, J = 7.5 Hz, J = 1.3 Hz, H), 7.53 (d, J = 1.9 Hz, 1 H), 7.31 (dd, J = 8.5 Hz, J = 1 .9 Hz, 1 H) , 1.42 (s, 9H).
Example 2C: 7-bromo-2-isoquinolin-3-yl-chromen-4-one oxime was prepared using the method D, (step 2) illustrated in example 1C. 7-bromo-2- isoquinoiin-3-yl-chromen-4-one O-ferf-butyl oxime (150 mg, 0.35 mmol) was treated with a 1 M solution of titanium tetrachloride (1.1 ml, 1.1 mmol) in dichloromethane (7.5 ml) to yield 7-bromo-2-isoquinoiin-3-y chromen-4-one oxime (56 mg, 37%) as a beige solid after recrystal!ization in hot chioroform. The product was isolated as a 90/10 mixture of Z/E oxime isomers.
p: 279-283°C
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1 % HCOOH): > 95 %; RT = 5.47 min. S (ESI+): 369.3 [deHnBrNzOa+HHm/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.26 (s, 1 H), 9.42 (s, 1 H), 8.52 (s, 1 H), 8.22 (d, J = 8.1 Hz, 1 H), 8.10 (d, J = 7.9 Hz, 1 H), 7.89 (td, J = 7.6 Hz, J = 1.3 Hz, 1 H), 7.86-7.75 (m, 4H), 7.74 (s, 1 H), 7.49 (dd, , J = 8.5 Hz, J = 1 .9 Hz, 1 H).
1 H NMR of the minor E isomer: DMSO-de δ (ppm): 1 1.74 (s, 1 H), 9.38 (s, 1 H), 8.92 (d, J = 8.8 Hz, 1 H), 8.40 (s, 1 H), 8.19 (d, J = 8.1 Hz, 1 H), 8.06 (d, J = 7.9 Hz, 1 H), 7.86 (t, J = 7.1 Hz, 1 H), 7.81 (s, 1 H), 7.75 (t, J = 8.1 Hz, 1 H), 7.46 (dd, , J = 8.8 Hz, J = 2.2 Hz, 1 H), 7.10 (s, 1 H).
Example 3 : 7-bromo-2-isoquinolin-3-yi-6-methyl-chromen-4-one oxime
Figure imgf000051_0001
Example 3A: 7-bromo~2~isoquinolin-3-yl~6~methyl-chromen-4~one was prepared using the method A illustrated in example 1A. 4'-ΒΓοηΊθ-5'-ΓηβΐΓψ1-2'- hydroxyacetophenone (556 mg, 2.42 mmol) and methyi-2-isoquinoiine-3-carboxySate (500 mg, 2.67 mmol), was treated with sodium hydride (60% in mineral oil, 291 mg, 7.28 mmol) in dry pyridine (13 ml) at 90°C for 15 min. and then with sulfuric acid (100 μΙ) in acetic acid (15 ml) at 100°C for 30 min. to yield 7-bromo-2-isoquinolin-3-yl-6- methyl-chromen-4-one (738 mg, 83%) as beige solid.
1 H NMR : CDCI3 δ (ppm): 9.32 (s, 1 H), 8.45 (s, 1 H), 8.09 (s, 1 H), 8.07 (d, J = 8.1 Hz, 1 H), 8.01 (d, J = 8.1 Hz, 1 H), 7.89 (d, J = 1.7 Hz, 1 H), 7.82 (td, J = 6.8 Hz, J = 1 ,3 Hz, 1 H), 7.75 (td, J = 6.9 Hz, J = 1.3 Hz, 1 H), 7.59 (s, 1 H), 7.56 (dd, J = 8.5 Hz, J = 1.7 Hz, 1 H), 2.52 (s, 3H).
Example 3B: 7-bromo-2-isoquinolin-3-yl-6-methyl-chromen-4-one O-tert- butyl oxime was prepared using the method D (step 1 ) illustrated in example 1 B. 7- bromo-2-isoquinoltn-3-yl-6-methyi-chromen-4-one (730 mg, 1.98 mmol) was treated with O-ie f-butyl-hydroxylamine hydrochioride (500 mg, 3.98 mmol) in methanol (13 ml) under microwave irradiation to yieid 7-bromo-2-isoquinolin-3-yl-chromen-4-one O- ferf-butyl oxime (383 mg, 44%) as a yellow solid after purification by flash chromatography over silica gei (cyclohexane/Chloroform/ethyl acetate : 96/4/0; 50/50/0; 0/100/0 0/50/50).
1 H NMR : CDCI3 δ (ppm): 9.27 (s, 1 H), 8.25 (s, 1 H), 8.00 (d, J = 8.1 Hz, 1 H), 7.92 (d, J = 8.5 Hz, 1 H), 7.90 (s, 1 H), 7.77 (s, 1 H), 7.75 (td, J = 8.1 Hz, J = 1.3 Hz, 1 H), 7.65 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.57 (s, 1 H), 2.44 (s, 3H), 1.42 (s, 9H).
Example 3C: 7~bromo-2-isoquinolin~3-yl-6-methyl-chromen-4~one oxime was prepared using the method D, (step 2) illustrated in example 1 C. 7-bromo-2- isoquinolin-3-yl-6-methyl-chromen-4-one O-tert-butyl oxime (100 mg, 0.22 mmol) was treated with a 1 M solution of titanium tetrachloride (0.68 ml, 0.68 mmol) in dfchloromethane (4 ml) to yield 7-bromo-2-isoquinolin-3-yi-6-methyl-chromen-4-one oxime (79 mg, 94%) as a yellow solid after recrystailization in hot chloroform. The compound was isolated as a 88/12 mixture of 2/E oxime isomers.
Mp: 267-270°C
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1 % HCOOH): > 95 %; RT = 5.82 min.
MS (ESI+): 383.3 [Cl9H13BrN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 S (ppm): 1 1.17 (s, 1 H), 9.39 (s, 1 H), 8.47(s, 1 H), 8.20 (d, J = 7.9 Hz, 1 H), 8.08 (d, J = 7.9 Hz, 1 H), 8.00-7.65 (m, 4H), 7.72 (s, 1 H), 2.38 (s, 3H).
Example 4: 6~bromo-2-isoquinolin-3-yl-chromen-4'one oxime
Figure imgf000052_0001
Example 4A: 6-bromo~2-isoquinolin-3-yl-chromen-4-one was prepared using the method A illustrated in example 1A. 5'-Bromo-2'-hydroxyacetophenone (1.56 g, 7.28 mmoi) and methyl-2-isoquinoline-3-carboxyiate (1.50 g, 8.0 mmol), was first treated with sodium hydride (60% in mineral oil, 960 mg, 24.0 mmol) in dry pyridine (40 ml) at 90°C for 15 min. and then with sulfuric acid {440 μΙ) in acetic acid (54 ml) at 100°C for 5 hours to yield 6-bromo-2-isoquinolin-3-yl-chromen-4-one (2.6 g, 90%) as brown needles.
1 H NMR : CDCi3 δ (ppm): 9.31 (s, 1 H), 8.46 (s, 1 H), 8.38 (d, J = 2.4 Hz, 1 H), 8.06 (d, J = 7.7 Hz, 1 H), 8.01 (d, J = 8.1 Hz, 1 H), 7.81 (t, J = 6.3 Hz, 1 H), 7.80 (d, J = 8.6 Hz, 1 H), 7.74 (td, J = 6.9 Hz, J = 1.3 Hz, 1 H), 7.59 (s, 1 H), 7.56 (d, J = 8.8 Hz, H).
Example 4B: 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared using the method D (step 1) illustrated in example B. 6-bromo- 2-isoquinolin-3-yl-chromen-4-one (600 mg, 1.7 mmol) was treated with O-ferf-butyl- hydroxyfamine hydrochloride (427 mg, 3.4 mmol) in methanol (10 mi) under microwave irradiation to yield 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ierf-butyl oxime (546 mg, 76%) as a yellow solid.
1 H NMR : CDCl3 δ (ppm): 9.28 (s, 1 H), 8.27(s, 1 H), 8.19 (d, J = 2.4 Hz, 1 H),
8.02 (d, J = 8.3 Hz, 1 H), 7.94 (d, J = 8.3 Hz, 1 H), 7.78 (s, 1 H), 7.76 (id, J = 8.1 Hz, J =
1.3 Hz, 1 H), 7.67 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.50 (dd, J = 8.6 Hz, J = 2.4 Hz, 1 H), 7.22 (d, J = 8.8 Hz, 1 H ), 1.43 (s, 9H).
Example 4C: 6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime was prepared using the method D, (step 2) illustrated in example 1C. 6-bromo-2- isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (50 mg, 0.12 mmol) was treated with a 1 M solution of titanium tetrachloride (0.35 ml, 0.35 mmol) in dichloromethane (3 ml) to yieid 6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime (37 mg, 85%) as a yellow solid after recrystailization in hot chloroform. The compound was isolated as a 98/2 mixture of Z E oxime isomers.
Mp: 266-269°C
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1 % HCOOH): > 98 %; RT = 5.41 mm.
MS (ESI+): 369.3 [C18H11BrN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.31 (s, 1 H), 9.41 (s, 1 H), 8.49{s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), 8.13 (d, J = 7.9 Hz, 1 H), 7.97 (d, J = 2.4 Hz, 1 H), 7.88 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.78 (td, J = 7.9 Hz, J = 1.3 Hz, 1 H), 7.75 (s, 1 H), 7.72 (dd, J = 8.8 Hz, J = 2.4 Hz, 1 H), 7.50 (d, J = 8.8 Hz, 1 H).
Examp!e 5 : 2-isoquinolin-3-yI-6-methy!~chromen-4~one oxime was isolated in 21 % overall yield as a pale yellow solid and as a 98/2 mixture of Z/E oxime isomers.
Figure imgf000053_0001
Mp: 260-264°C
MS (ESI+):. 303.4 [C19H14N202+H]+ (m/z). 1H NMR of the major Z isomer: DMSO-de δ (ppm): 11.04 (s, 1H), 9.42 (s, 1H), 8.48 (s, 1H), 8.22 (d, J = 8.1 Hz, 1H), 8.15 (d, J = 8.1 Hz, 1H), 7.88 (td, J = 7.8 Hz, J = 1.3 Hz, 1H), 7.78 (td, J = 7.9 Hz, J = 1.3 Hz, 1H), 7.75 (s, 1H), 7.71 (s, 1H), 7.42 (d, J = 8.3 Hz, 1H), 7.36 (dd, J = 8.8 Hz, J = 1.7 Hz, 1H), 2.37 (s, 3H).
Exam pie 6 : 6-fluoro-2-isoquinoHn-3-yl-chromen-4-one oxime was isolated in 36% overall yield as a yellow solid and as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000054_0001
Mp: 278-280°C
MS (ESI+): 307.3 [C SH11FN202+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.27 (s, 1H), 9.42 (s, 1H), 8.50 (s, 1H), 8.22 (d, J = 7.7 Hz, 1H), 8,14 (d, J = 8.1 Hz, 1H), 7.88 (t, J = 6.8 Hz, 1H), 7.78 (td, J = 7.1 Hz, J = 1.3 Hz, 1H), 7.74 (s, 1H), 7.64-7.53 (m, 2H), 7.43 (td, J = 8.6 Hz, J = 3.2 Hz, 1H).
Example 7: 6, 8-difluoro-2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 31% overall yield as a beige solid and as a 96/4 mixture of Z/E oxime isomers.
Figure imgf000054_0002
Mp: 253-256°C
MS (ESR): 325.1 [C1sH10F2N2O2+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11,48 (s, 1H), 9.42 (s, 1H), 8.33 (s, 1H), 8.21 (d, J = 7.5 Hz, 1H), 8.20 (d, J = 7.7 Hz, 1H), 7.87 (td, J = 7.5 Hz, J - 1.2 Hz, 1H), 7.78 (t, J = 7.2 Hz, 1H), 7.76 (s, 1H), 7.63 (ddd, 11.1 Hz, J = 8.7 Hz, 2.8 Hz, 1H), 7.40 (dm, J = 11.1 Hz, 1H). Example 8 : 8~chloro~2-isoquinoHn-3-yl-chromen-4-one oxime was isolated overall yield as a beige powder and as a 95/5 mixture of Z/E oxime isomers.
p: 270-272°C
MS(ESI+): 323.1 [C18H11C1N202+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.34 (s, 1H), 9.44 (s, 1H), 8.38 (s, 1H), 8.23 (d, J = 8.1 Hz, 1H), 8.17 (d, J = 8.1 Hz, 1H), 7.89 (t, J = 7.4 Hz, 1H), 7.87 (d, J = 7.0 Hz, 1H), 7.79 (t, J = 7.4 Hz, 1H), 7.79 (s, 1H), 7.71 (d, J = 7.9 Hz, 1H), 7.31 (t, J = 7.9 Hz, 1H).
Example 9 : 4-fiuoro-2-isoquinolin-3-yl-chromen-4-one-(Z)- oxime was isolated in 54% overall yield as a yellow solid.
Figure imgf000055_0002
Mp: 275-278°C
MS (ESI+): 307.1 [C18H11FN202+H]+ (m/z).
1H NMR: DMSO-d6 δ (ppm): 11.40 (s, 1H), 9.42 (s, 1H), 8.48 (s, 1H), 8.22 (d, J = 7.9 Hz, 1H), ), 8.15 (d, J = 7.9 Hz, 1H), 7.88 (td, J = 7.5 Hz, J = 1.3 Hz, 1H), 7.83 (s, 1H), 7.78 (td, J = 7.5 Hz, J = 1.3 Hz, 1H), 7.54 (td, J = 8.3 Hz, J = 5.6 Hz, 1H), 7.37 (d, J = 8.3 Hz, 1H), 7.16 (ddd, J = 11.1 Hz, J = 8.2 Hz, J = 0.9 Hz, 1H).
Example 10 : 2-isoquinolin-3-yl-6-trifluoromethoxy'Chromen-4-one oxime was isofated in 15% overall yield as a yellow powder and as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000056_0001
Mp: 250-254°C
Figure imgf000056_0002
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.34 (s, 1 H), 9.43 (s, 1 H), 8.51 (s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), ), 8.15 (d, J = 7.9 Hz, 1 H), 7.89 (td, J = 7.6 Hz, J = 1.3 Hz, 1H), 7.79 {td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.76 (m, 1 H), 7.75 (s, 1 H), 7.66 (d, J = 9.0 Hz, 1 H), 7.57 (dd, J = 9.0 Hz, J = 2.5 Hz, 1 H).
Example 11 : 2-isoquinolin-3-yl-6-trifluoromethyl-chromen-4-one oxime was isolated in 10% overall yield starting from 2'-hydroxy-5'-trif!uoromethyl- aceiophenone (example 1 1A) and methyl isoquinoline-3-carboxylate. The yellow solid product was a 95/5 mixture of Z/E oxime isomers.
Figure imgf000056_0003
Mp: 245-247°C
MS (ESI+): 357.1 [CeHnFaNaOz+Hnm/z).
1 H NMR of the major Z isomer: DMSO-de δ (ppm): 11.39 (s, 1 H), 9.43 (s, 1 H), 8.54 (s, 1 H), 8.23 (d, J = 7.9 Hz, 1 H), ), 8.15 {m, 2H), 7.94-7.85 (m, 2H), 7.79 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.78 (s, 1 H), 7.73 (d, J = 8.7 Hz, 1 H).
Example 11 A: 2'-hydroxy-5!-trifluoromethyl-acetophenone was prepared using the following process (JACS, 2004, 126(3), 712-713). To a cooled solution of 2'- methoxy-5'-trifluoromethy!-acetophenone (650 mg, 2.98 mmol) at -78°C in dry dichloromethane (40 ml) was slowly added a 1 M solution of boron trichloride in dichloromethane (7.5 ml, 7.5 mmol) keeping the internal temperature below -70°C. The brown-orange solution was slowly warmed up to room temperature within 2 hours. The solution was ice cooled and cautiously hydrolyzed with a 1 N aqueous hydrochloride solution (40 ml). The organic layer was treated with 1 N HCI, washed with water and dried over sodium sulfate. The filtrated solution was concentrated and the residue purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-10%) to yield 2'~hydroxy-5,-trifluoromethyl-acetophenone (467 mg, 77%) as a light yellow oil.
1 H NMR: CDCI3 δ (ppm): 12.55 (s, 1 H), 7.99 (d, J = 2.0 Hz, 1 H), 7.70 (dd, J = 8.8 Hz, J = 2.0 Hz, 1 H), 7.08 (d, J = 8.9 Hz, 1 H), 2.69 (s, 3H).
Example 11 B: 2'-methoxy-5'-trifluoromethyl-acetophenone was prepared as follow {US2003/0109574): in a water bath at room temperature, to a solution of trifluoromethane sulfonic acid (5 ml, 56.7 mmol) under argon was slowly added a solution of 4-trifiuoromethyl-anisol (2.0 g, 1 1.35 mmol) in acetic anhydride (2.15 ml, 22.7 mmol). The resulting dark mixture was stirred for 3 hours at room temperature then poured onto ice water (26 ml). The product was extracted several times with ether. The ethereal layer was washed with a 10% solution of sodium hydrogenocarbonate, water, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography ove . silica gel (gradient cydohexane/dichloromethane: 0-50%) to yield 2'-methoxy-5'-trifluoromethyi- acetophenone (1.7 g, 67%) as a white solid.
1 H NMR : CDCI3 δ (ppm): 8.00 (d, J = 2.3 Hz, 1 H), 7.71 (dd, J = 8.8 Hz, J = 2.3 Hz, 1 H), 7.05 (d, J = 8.7 Hz, 1 H), 3.98 (s, 3H), 2.63 (s, 3H).
Example 12: 2-(7-fluoro-isoquinolin-3~yl)-chromen-4-one oxime was isolated in 11% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 7-fiuoro-isoquinoline-3-carboxylate (example 12A) as a yellow solid and a 95/5 mixture of Z/E oxime isomers.
Figure imgf000057_0001
p: 233-235°C MS (ESI+): 307.0 [C BH 1FN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.09 (s, 1 H), 9.41 (s, 1 H), 8.53 (s, 1 H), 8.28 (d, J = 9.0 Hz, 1 H), 8.25 (d, J = 9.0 Hz, 1 H), 8.04 (dd, J = 9.2 Hz, J = 2.5 Hz, 1 H), 7.92 (dd, J = 7.9 Hz, J = 1 ,3 Hz, 1 H), 7.81 (td, J = 9.0 Hz, J = 2.6 Hz, 1 H), 7.75 (s, 1 H), 7.60-7.45 (m, 2H), 7.31 (td, J = 8.0 Hz, J = 1.3 Hz, 1 H).
Example 12A: methyl 7-fluoro-isoquinoline-3~carboxylate: A sealed tube was charged with 2-bromo-5-fluorobenzaidehyde (300 mg, 1.47 mmol), dry dimethylformamide (1.5 ml), triethylamine (0.7 ml, 5.02 mmol), methyl 2- acetamidoacryiate (273 mg, 1.91 mmol), fris-o-toiyi-phosphine (89 mg, 0.29 mmol) and palladium acetate (33 mg, 0.14 mmol). The solution was degassed with Argon for 10 min. and then warmed at 110°C for 4.5 hours. The mixture was cooled to room temperature and poured onto an aqueous solution of ammonium chloride. The mixture was extracted with ethyl acetate, washed with brine, dried over sodium sulfate and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/dichloromethane: 0-80%) to yield methyl 7-fluoro-isoqusnoline- 3-carboxylate (120 mg, 39%) as beige solid.
1 H NMR: CDCl3 δ (ppm): 9.29 (s, 1 H), 8.60 (s, 1 H), 8.01 (dd, J = 9.0 Hz , J = 5.2 Hz, 1 H), 7.68 (dd, J = 8.4 Hz, J = 2.4 Hz, 1 H), 7.57 (td, J = 8.4 Hz, J = 2.4 Hz, 1 H), 4.06 (s, 3H).
Example 13: 2-(7-methoxy-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 3% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 7-fluoro-isoquinoline-3-carboxyiate (example 12A). The brown solid product was a 80/20 mixture of Z/E oxime isomers.
Figure imgf000058_0001
Mp: 239-241 °C
S (ES!+): 319.1 [C19H1 N203+H]+ (m/z).
1H N R of the major Z isomer: DMSO-d6 δ (ppm): 11.02 (s, 1H), 9.31 (s, 1H), 8.42 (s, 1H), 8.08 (d, J = 9.0 Hz, 1H) 7.92 (dd, J = 7.9 Hz, J = 1.3 Hz, 1H), 7.70 (s, 1H), 7.63 (d, J = 2.5 Hz, 1H), 7.60-7.44 (m, 3H), 7.30 (td, J = 7.3 Hz, J = 1.5 Hz, 1H), 3.96 (s, 3H).
Example 14: 2-(6.7-dimethoxy-isoquinolin-3-yl)'Chromen-4-one oxime was isolated in 8% overai! yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 6,7-dimethoxy-isoquinoline-3-carboxylate (US054777252) as a yellow solid and a 95/5 mixture of ZJE oxime isomers.
Figure imgf000059_0001
Mp: 266-269°C
MS (ESI+): 349.0 [C20H16N2O4+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 10.99 (s, 1H), 9.16 (s, 1H), 8.33 (s, 1H), 7.92 (dd, J = 7.9 Hz, J = 1.3 Hz, 1H), 7.69 (s, 1H), 7.60 (s, 1H), 7.55 (s, 1H), 7.55 (td, J = 7.7 Hz, J = 1.5 Hz, 1H), 7.45 (d, J = 7.4 Hz, 1H), 7.30 (t, J = 7.4 Hz, 1H), 3.97 (s, 3H), 3.96 (s, 3H).
Example 15: 2-(6-methyl-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 35% overall yield using method A and D starting from 2!-hydroxy- acetophenone and methyl 6-methyl-isoquinoline-3-carboxyiate (example 15A) as a yellow solid and a 95/5 mixture of ZJE oxime isomers.
Figure imgf000060_0001
Mp: 245-247°C
MS (ESI+): 349.0 [C19H14N202+H]+ (m/z).
1 H N R of the major Z isomer: DMSO-d6 δ (ppm): 11.06 (s, 1 H), 9.33 (s, H), 8.37 (s, 1 H), 8.10 (d, J = 8.3 Hz, 1 H), 7.92 (dd, J = 7.9 Hz, J = 1.3 Hz, 1 H), 7.91 (s, 1 H), 7.75 (s, 1 H), 7.6-7.45 (m, 3H), 7.30 (td, J = 7.4 Hz, J = 1.3 Hz, 1 H), 2.55 (s, 3H).
Example 15A: methyl 6-methyl~isoqumolm" e~3~carboxylate was prepared using the procedure described in example 12A: 2-bromo-4-methytbenzaldehyde was reacted with methyl 2-acetamidoacrylate at 1 10°C for 18 hours to yieid methyl 6- methyi-isoquinoline-3-carboxylate (50%) as brown solid.
1 H NMR : CDCI3 δ (ppm): 9.25 (s, 1 H), 8.51 (s, 1 H), 7.94 (d, J = 8.3 Hz, 1 H), 7.73 (s, 1 H), 7.57 (dd, J = 8.4 Hz, J = 1.5 Hz, 1 H), 4.05 (s, 3H), 2.58 (s, 3H).
Example 16: 2-(7-chloro-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 20% overall yield using method A and D starting from 2'-hydroxy- acetophenone and methyl 7-chloro-isoquinoline-3-carboxylate (example 16A) as pale a yellow solid and a 90/10 mixture of Z/E oxime isomers.
Figure imgf000060_0002
Mp: 260-261 °C
MS (ESI+): 323.1 [C18H11CIN202+H]+ (m/z). 1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.13 (s, 1 H), 9.39 (s, 1 H), 8.51 (s, 1 H), 8.34 (d, J = 2.1 Hz, 1 H), 8.19 (d, J = 8.8 Hz, 1 H), 7.90 (td, J = 8.6 Hz, J = 2.1 Hz, 1 H), 7.90 (s, 1 H), 7.75 (s, 1 H), 7.60-7.45 (m, 2H), 7.30 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H).
Example 16A: methyl 7-chloro-isoquinoline-3-carboxy!ate was prepared using the procedure described in example 12A: 2-bromo-5-chlorobenzaldehyde was reacted with methyl 2-acetamidoacrylate at 110°C for 24 hours to yield methyl 7- chloro-isoquinoiine-3-carboxylate (25%) as brown solid.
1 H NMR : CDC!3 δ (ppm): 9.27 (s, 1 H), 8.58 (s, 1 H), 8.05 (d, J = 1.7 Hz, 1 H), 7.93 (d, J = 8.8 Hz, 1 H), 7.73 (dd, J = 8.7 Hz, J = 1.9 Hz, 1 H), 4.06 (s, 3H).
Example 17: 2-(5-bromO"isoquinolin-3~yl)~chromen-4-one oxime was isolated in 15% overall yield using method A and D, starting from 2'-hydroxy- acetophenone and methyl 5-bromo-isoquinoiine-3-carboxylate (example 17A) as a yellow solid and a 90/10 mixture of Z/E oxime isomers.
Mp: 268-270°C
MS (ESI+): 366.9 [C18HnBrN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.17 (br. s, 1 H), 9.47 (s, 1 H), 8.48 (s, 1 H), 8.28 (d, J = 8.1 Hz, 1 H), 8.24 (d, J = 8.1 Hz, 1 H), 7.93 (d, J = 7.2 Hz, 1 H), 7.82 (s, 1 H), 7.71 (t, J = 7.8 Hz, 1 H), 7.60-7.48 (m, 2H), 7.32 (td, J = 7.8 Hz, J = 1.9 Hz, 1 H).
Example 17A: methy-5-bromo-isoquinoline-3-carboxylate was prepared using the foilowing procedure describe in US054777252 for methyl-6,7-dimethoxy- isoquinoiine-3-carboxylate: 5-bromo-1 ,2,3,4-tetrahydroisoquinoline-3-carboxylic acid methyl ester (880 mg, 3.26 mmo!) was treated with 2,3-Dichloro-5,6- dicyanobenzoquinone (1.62 mg, 7.19 mmol) in dry tetrahydrofuran (19 ml) at reflux temperature for 18 hours. The cooled dark mixture was filtered and the solid washed with dichloromethane. The filtrate was treated with a 1 M aqueous sodium hydroxide solution and the aqueous layer was extracted twice with dichloromethane. The organic layer was washed with water, brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-100%) to yield methyl 5-bromo- isoquinoiine-3-carboxylaie (793 mg, 91 %) as beige solid.
1 H N R : CDCI3 δ (ppm): 9.31 (s, 1 H), 8.90 (s, 1 H), 8.05 (t, J = 9.4 Hz, 2H), 7.61 (t, J = 7.5 Hz, 1 H), 4.09 (s, 3H).
Example 18: 2-(5-hydroxy-isoquinotin-3-yl)-chromen-4-one oxime was isolated in 14% yield using method D (step 2), starting from 2-(5-hydroxy-isoquinolin- 3-yi)-chromen-4-one O-tert-butyl oxime (example 8A) as orange solid and as a 80/20 mixture of 27 E oxime isomers.
Figure imgf000062_0001
MS (ESI+): 305.1 [C18H12N203+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-de δ (ppm): 1 104 (s 1 H), 10.82 (br. s, 1 H), 9.32 (s, 1 H), 8.55 (s, 1 H), 7.92 (d, J = 7.7 Hz, 1 H), 7.75 (s, 1 H), 7.70-7.50 (m, 4H), 7.35-7.25 (m, 1 H), 7.20 (d, J = 7.0 Hz, 1 H).
Example 18A: 2-(5-hydroxy-isoquinolin-3-yl)-chromen-4-one O-tert-butyl oxime was prepared using a palladium coupling procedure describes by K. W. Anderson (JACS, 2006, 128, 10694-10695):
A solution of 2-(5-bromo-isoquinolin-3-yl)-chromen-4-one O-ferf-butyl oxime (370 mg, 0.87 mmol), potassium hydroxide (300 mg, 5.24 mmol), tris(dibenzyIideneacetone)dipalladium (40 mg, 0.043 mmol) and 2-di-fert- butyiphosphino-2\4\6'-triisopropylbiphenyi (37 mg, 0.087 mmo!) in dioxane (1 .9 ml) and water (1 .9 mi) was degassed with argon for 5 min. and then warmed to 100°C for 8 hours.
The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-40%) to yield 2-(5-hydroxy-isoquinolin-3-yl)- chromen-4-one O-ierf-butyi oxime (230 mg, 73%) as a yellow solid.
MS (ESI+): 361 .1 .1 [C22H20N2O3+H]+ (m/z).
1 H NMR: acetone-d6 δ (ppm): 9.30 (s, 1 H), 8.69 (s, 1 H), 8.08 (d, J = 7.1 Hz, 1 H), 7.86 (s, 1 H), 7.67 (d, J = 7.9 Hz, 1 H), 7.63-7.48 (m, 3H), 7.35-7.20 (m, 2H), 1 .43 (s, 9H).
Example 19: 2-(5-methoxy-isoquinolin-3-yl)-chromen-4-one oxime was isolated in 94% yield using method D (step 2), starting from 2-(5-methoxy-isoquinolin- 3-yl)-chromen-4-one O-ferf-butyS oxime (example 19A) as a yellow solid and as a 90/ 0 mixture of Z/E oxime isomers.
Figure imgf000063_0001
Mp: 245-247
MS (ES!+): 319.1 [C19H14N203÷H (m/z).
1 H NMR of the major 2 isomer: DMSO-d6 δ (ppm): 1 1 .07 (s, 1 H), 9.37 (s, 1 H), 8.52 (s, 1 H), 7.92 (d, J = 7.7 Hz, 1 H), 7.76 (s, 1 H), 7.75 (d, J = 7.7 Hz, 1 H), 7.70 (t, J = 8.0 Hz, 1 H), 7.55-7.50 (m, 2H), 7.36-7.26 (m, 2H), 4.07 (s, 3H).
Example 19A: 2-(5-methoxy-isoquinolin-3-yl)-chromen-4-one O-tert-butyl oxime:
To a ice-water bath cooled solution of 2-(5-hydroxy-isoquinolin-3-yl)~chromen- 4-one O-fert-buty! oxime (80 mg, 0.22 mmol) in dimethylformamide (2.5 ml) was added sodium hydride (60% in mineral oil, 13 mg, 0.33 mmoi) under argon and the mixture was stirred at room temperature for 1 hour. The solution was cooled to 0°C and iodomethane ( 5 μΙ, 0.24 mmol) was added. The resulting solution was stirred at room temperature for 20 hours and then hydroiyzed with brine. The mixture was extracted with ethyl acetate, washed with brine, dried over sodium sulfate and then purified by flash chromatography over silica gel (cyclohexane/ethyl acetate: 80/20) to yield 2-(5-methoxy-isoquinolin-3-yl)-chromen-4~one O-rerf-butyl oxime (65mg, 79%) as a yellow solid.
MS (ESI+): 375.1 [C23H22N203+H]+ (m/z).
1 H NMR: CDCI35 (ppm): 9.23 (d, J = 0.9 Hz, 1 H), 8.63 (s, 1 H), 8.09 (d, J = 8.5 Hz, 1 H), 7.78 (s, 1 H), 7.43-7.38 (m, 2H), 7.24-7.17 (m, 1 H), 7.08-7.03 (m, 1 H), 4.08 (s, 3H), 1.43 (s, 9H).
Example 20: 2-isoquinolin-3-yl-7-phenylethynyl-chromen-4-one oxime
Figure imgf000064_0001
A solution of 7~bromo-2-isoquinolin-3-yl-chromen-4-one oxime (52 mg, 0.142 mmol), trans-dichiorobis(triphenylphosphine)pa!ladium (10 mg, 0.014 mmoi), copper iodide (5.3 mg, 0.028 mmol) in dimethyiformamide (3 ml) was degassed with argon for 10 min and then treated with triethylamine (30μΙ, 0.213 mmol) and phenyl acetylene (19μ1, 0.170 mmol). The solution was warmed to 90°C for 18 hours and then poured onto a 0.5N aqueous solution of hydrogen chloride, extracted with ethyl acetate and purified by preparative HPLC (gradient 25-10% Water/acetonitriSe+0.05% trifSuoroacetic acid) to yield 2-isoquinolin-3-yl-7-phenylethynyl-chromen-4-one oxime (7.3 mg, 13%) as a yellow solid and a 80/20 mixture of Z7E oxime isomers.
MS (ESI+): 389.2 [C26H16N202+H]+ (m/z).
Example 21: 2~isoquinolin-3-yl~7-((E)-styryl)~chromen~4"One oxime was isolated in 55% yield using method D (step 2), starting from 2-!soquinolin-3-yl-7-((E)- styryl)-chromen-4-one O-ferf-butyl oxime (example 21 A) as a pale yellow solid and as a 90/10 mixture of Z/E oxime isomers
Figure imgf000065_0001
p: 273-276°C
MS (ESI+): 391.5 [C2BH1 SN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.15 (s, 1 H), 9.44 (s, 1 H), 8.51 (s, 1 H), 8.24 (d, J = 8.1 Hz, 1 H), 8.15 (d, J = 8.2 Hz, 1 H), 7.90 (d, J = 8.2 Hz, 1 H), 7.90 (t, J = 7.0 Hz, 1 H), 7.79 (t, J = 7.4 Hz, 2H), 7.76 (s, 1 H), 7.67 (d, J = 7.3 Hz, 2H), 7.56 (d, J = 7.3 Hz, 1 H), 7.5-7.28 (m, 5H).
Example 21A: 2-isoquinolin-3-yl-7-((E)styryl)~chromen~4-one O-tert-butyl oxime:
In a microwave vial, a solution of 7-bromo-2-isoquinoiin-3-yl-chrornen-4-one O- terf-butyl oxime (200 mg, 0.47 mmol), cesium fluoride (213 mg, 1.41 mmol) and trans- styryl boronic acid (160 mg, 1.07 mmol) in 1.2-dimethoxyeihane (3.2 ml) and methanol (Ϊ .6 ml) was degassed with argon for 20 min and then tetrakis(triphenylphosphine)pailadium (30 mg, 0.026 mmoi) was added. The vial was sealed and the mixture was heated under microwave irradiation to 150°C for 5 min. After cooling, the solution was diluted with dichioromethane and absorbed over silica gel for purification by flash chromatography (gradient cyclohexane/dichloromethane: 0-80 to yield 2-lsoquinolin-3-yl-7-((E)-styryl)-chromen-4-one O-ferf-butyl oxime (160 mg, 76%) as a yellow solid.
MS (ESI+): 447.6 [C30H26N2O2+H]+ (m/z).
1 H NMR: CDCI3 δ (ppm): 9.29 (s, 1 H), 8.33 (s, 1 H), 8.06 (d, J = 8.1 Hz, 1 H),
8.02 (d, J = 7.7 Hz, 1 H), 7.96 (d, J = 7.9 Hz, 1 H), 7.80 (s, 2H), 7.76 (td, J = 7.5 Hz, J =
1 .3 Hz, 1 H), 7.66 (id, J = 7.4 Hz, J = 1.3 Hz, 1 H), 7.56 (d, J = 7.3 Hz, 2H), 7.49 (d, J = 1.7 Hz, 1 H), 7.44-7.26 (m, 4 H), 7.23 (d, J = 16 Hz, 1 H), 7.23 (d, J = 16 Hz, 1 H), Example 22: 2-isoquinolin-3-yl-7-phenethyl~chromen-4-one oxime was isolated using method D (step 2), starting from 2-lsoquinolin-3-yl-7-phenethyl- chromen-4-one O-ferf-butyl oxime (example 22A) as a yellow solid and as a 95/5 mixture of Z/E oxime isomers
Figure imgf000066_0001
MS (ESI+): 393.1 [C26H20N2O2+Hl+ (m/z).
H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.04 (s, 1 H), 9.50 (s, 1 H), 8.56 (s, 1 H), 8.30 (d, J = 8.0 Hz, 1 H), 8.21 (d, J = 8.0 Hz, 1 H), 7.97 (t, J = 7.2 Hz, 1 H), 7.90-7.80 (m, 3H), 7.49 (s, 1 H), 7.40-7.35 (m, 4H), 7.35-7.20 (m, 2H), 3.06 (s, 4H).
Example 22A: 2-isoquinolin -3-yl-7-phenethyl-chromen-4-one O-tert-butyl oxime:
A solution of 2-isoquinolin-3-yl-7-phenylethynyl-chromen-4-one O-ferf-butyl oxime (60 mg, 0.13 mmol) (prepared using the procedure described in example 20 starting from example 2B) and Lindlar's catalyst (22.5 mg) in tetrahydrofuran (9.2 ml) and methanol (1.8 ml) was stirred at room temperature under 1 atmosphere of hydrogen for 18 h. The catalyst was removed by filtration and the solvent evaporated under reduce pressure to yield the title compound in 60% yield.
Example 23: 7-ethynyl~2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 72% yield using method D (step 2), starting from 7-ethynyl-2-isoquinoiin-3- yl-chrornen-4-one O-re f-butyl oxime (example 23A) as a yellow solid and as a 90/10 mixture of Z E oxime isomers. Example 23 was purified by preparative HPLC (gradient 35-20% Water/acetonitrile+0.05% trifluoroacetic acid)
Figure imgf000066_0002
MS (ESI+): 313.4 [C20H12N2O2+H]+ (m/z). 1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.29 (s, 1 H), 9.42 (s, 1 H), 8.52 (s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), 8.11 (d, J = 8.0 Hz, 1Ή), 7.90 (d, J = 8.0 Hz, 1 H), 7.89 (t, J = 8.1 Hz, 1 H), 7.79 (t, J = 8.0 Hz, 1 H), 7.74 (s, 1 H), 7.64 (d, J = 1.3 Hz, 1 H), 7.38 (dd, J = 8.3 Hz, J = 1.5 Hz, 1 H), 4.44 (s, 1 H).
Example 23A; 7-ethynyl-2-isoquinoIin-3-yl-chromen-4-one O-tert-butyl oxime:
A solution of 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (120 mg, 0.28 mmol), trans-dichlorobis(triphenylphosphine)palladium (19 mg, 0.028 mmol), copper iodide (10 mg, 0.056 mmol), triphenylphosphine (14 mg, 0.056 mmol) in a mixture of dimethylformamide (3 ml) and acetonitrile (3 ml) was degassed with argon for 10 min and then treated with diethylamine (42 μΙ, 0.40 mmol) and trimethylsiiylaceiylene (46 μΐ, 0.326 mmoi). The solution was warmed to 90°C for 20 hours and then poured onto a 0.5N aqueous solution of hydrogen chloride, extracted with ethy! acetate and purified by flash chromatography over silica gel (gradient cyclohexane/ethy! acetate: 20-50%) to yield 2-isoquinolin-3-yl-7-trimethylsilylethynyi- chromen-4-one O-ferf-butyl oxime (63 mg, 50%) as a yellow solid. The solid was dissolved in methanol (10 ml) and treated with potassium carbonate (39 mg, 0.28 mmol) and stirred at room temperature for 18 hours. Methanol was evaporated and the residue was extracted with ethyl acetate washed with water and brine, dried over sodium sulfate and the solution concentrated to dryness to yield 7-ethynyl-2- isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (49 mg, 89%) as beige solid.
MS (ESI+): 369.3 [C24H20N2O2+H]+ (m/z).
Example 24: 2'isoquinolin-3-yl'7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime was isolated in 42% yield using method D (step 2), starting from 2-isoquinolin- 3-yl-7-(pyridin-2-yl-ethynyl)-chromen-4-one O-ferf-butyl oxime (example 24A) as a yel!ow solid after recrysta!lization in hot chloroform.
Figure imgf000067_0001
Mp: 248-251 °C
MS (ESI+): 390.5 [C25H1 SN302+H]+ (m/z).
1 H NMR: DMSO-d6 δ (ppm): 1 1.33 (s, 1 H), 9.43 (s, 1 H), 8.65 (d, J = 4.1 Hz, 1 H), 8.54 (s, 1 H), 8.23 (d, J = 8.0 Hz, 1 H), 8.12 {d, J = 7.9 Hz, 1 H), 7.96 (d, J = 8.1 Hz, 1 H), 7.90 (t, J = 7.7 Hz, 2H), 7.83-7.75. (m, 3H), 7.71 (d, J = 7.7 Hz, 1 H), 7.51 (dd, J = 8.1 Hz, J = 1.5 Hz, 1 H), 7.46 (ddd, J = 8.1 Hz , J = 5.0 Hz, J * 1.1 Hz, 1 H).
Example 24 : 2-iso quinolin-3-yi- 7-( pyridm" -2-yl-ethynyl)-chromen-4-one O-tert-butyl oxime:
A solution of 7-bromo-2-isoquinolin-3-y!-chromen-4-one O-iert-butyl oxime (150 mg, 0.35 mmoi), trans-dichlorobis(triphenylphosphine)palIadium (22 mg, 0.035 mmol), copper iodide (13 mg, 0.071 mmol), triphenylphosphine (19 mg, 0.071 mmoi) in dimethylformamide (3 rn!) was degassed with argon for 10 min and then treated with triethyiamine (74 μΙ, 0.53 mmol) and 2-ethynyipyridine (45 μΙ, 0.45 mmof). The soiution was warmed to 120X under microwave irradiation (power max = 80 W) for 1 hour and then poured onto a saturated aqueous solution of sodium hydrogenocarbonate, extracted with ethyl acetate, washed with water and brine, dried over sodium sulfate and purified by flash chromatography over silica gei (cyclohexane/ethyi acetate: 20-40%) to yield 2-isoquinolin-3-yl-7-(pyridin-2-yi-ethynyi)- chromen-4-one O-ferf-butyl oxime (100 mg, 63%) as a yellow solid.
1 H NMR: CDCI38 (ppm): 9.29 (s, 1 H), 8.66 (Br. s, 1 H), 8.29 (s, 1 H), 8.08 (d, J = 8.0 Hz, 1 H), 8.02 (d, J = 7.9 Hz, 1 H), 7.96 (d, J = 8.1 Hz, 1 H), 7.80 (s, 1 H), 7.76 (t, J = 8.3 Hz, 2H), 7.66 (t, J = 7.6 Hz, 1 H), 7.63-7.56 (m, 2H), 7.43 (dd, J = 8.2 Hz, J = 1.5 Hz, 1 H), 7.31 (br. t, J = 5.9 Hz , 1 H), 1.43 (s, 9H).
Example 25: 2-isoquinolin-3-y!-7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime was prepared in 18% overall yield using the method described in example 23, starting from 7-bromo-2-isoquinoiin-3-y[-6-methyl-chromen-4-one O-ferf-butyl oxime (example 3B) and phenyiacetylene. The titie compound was isolated as a yellow solid and as a 95/5 mixture of Z E oxime isomers.
Figure imgf000069_0001
Mp: 248-250°C
MS (ESI+): 403.5 [C27H18N202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1123 (s, 1 H), 9.42 (s, 1 H), 8.52 (d, J = 4.1 Hz, 1 H), 8.23 (d, J = 8.1 Hz, 1 H), 8.1 1 (d, J = 7.9 Hz, 1 H), 7.89 (t, J = 7.0 Hz, 1 H), 7.85-7.75 (m, 1 H), 7.81 (s, 1 H), 7.75 (s, 1 H), 7.68 (s, 1 H), 7.66-7.59 (m, 2H), 7.52-7.45 (m, 3H), 2.50 (s, 3H).
Example 26: 2-isoquinolin-3-yl-7-(pyridin~4-yl)ethynyl~chromen-4-one oxime was prepared in 6% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (exampie 2B) and 4-eihynylpyridine. The title compound was isolated as a yellow solid and as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000069_0002
Mp: 265-270°C
MS (ESi+): 390.4 [C25H15N302+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.34 (s, 1 H), 9.43 (s, 1 H), 8.68 (d, J - 5.6 Hz, 2H), 8.52 (s, 1 H), 8.23 (d, J = 8.1 Hz, 1 H), 8.12 (d, J = 7.9 Hz, 1 H), 7.96 (d, J = 8.2 Hz, 1 H), 7.89 (t, J = 7.1 Hz, 1 H), 7.84-7.74 (m, 3H), 7.58 (m, 2H), 7.52 (d, J = 8.1 Hz, H).
Example 27: 7~(4~dimethylamin ophenyl) ethynyl- 2-isoquinoliti-3-yl- chromen-4-one oxime was prepared in 8% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (example 2B) and 4-dimethylaminophenylacetyiene. The title compound was isolated as an orange solid and as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000070_0001
Mp: 235-240°C
MS (ESI+): 432.5 [C28H21N302+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.21 (s, 1 H), 9.43 (s, 1 H), 8.53 (s, 1 H), 8.23 (d, J = 8.0 Hz, 1 H), 8.12 (d, J = 8.0 Hz, 1 H), 7,89 (d, J = 7.8 Hz, 2H), 7.79 (t, J = 7.6 Hz, 1 H), 7.75 (s, 1 H),7.63 (s, 1 H), 7.45-7.25 (m, 3H), 6.74 (d, J = 8.6 Hz, 2H), 2.97 (s, 6H).
Example 28: 2-iso quin olin-3-yl- 7-( 3-methoxyphenyl) ethynyl-chrom en-4- one oxime was prepared in 22% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyi oxime (example 2B) and 3-methoxyphenyiacetyiene. The title compound was isolated as an orange solid and as a 93/7 mixture of Z/E oxime isomers.
Figure imgf000070_0002
Mp: 248-251 °C
MS (ESI+): 419.5 [C27H18N203+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 6 (ppm): 1 1.28 (s, 1 H), 9.43 (s, 1 H), 8.53 (s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), 8.12 (d, J = 8.1 Hz, 1 H), 7.93 (d, J = 8.3 Hz, 2H), 7.89 (t, J = 7.1 Hz, 1 H), 7.79 (t, J = 7.2 Hz, 1 H), 7.76 (s, 1 H),7.72 (d, J = 1.3 Hz, 1 H), 7.46 (dd, J = 8.1 Hz, J = 1 .3 Hz„ 1 H), 7.38 (t, J = 7.9 Hz, 1 H), 7.22-7.14 (m, 2H), 7.04 (dd, J = 8,5 Hz, J = 1.2 Hz, 1 H), 3.82 (s, 3H). Example 29: 7-( 3-aminoph enyl) ethynyl-2-iso quin olin-3-yl-chrom en-4-one oxime was prepared in 17% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyi oxime (example 2B) and 3-aminophenylacetylene. The title compound was isolated as a ye!low solid and as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000071_0001
Mp: 272-275°C
MS (ESI+): 404.4 [C26H17N302+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.26 (s, 1 H), 9.43 (s, 1 H), 8.54 (s, 1 H), 8.23 (d, J = 7.9 Hz, 1 H), 8.12 (d, J = 8.0 Hz, 1 H), 7.95-7.86 (m, 2H), 7.79 (t, J = 7.7 Hz, 1 H), 7.76 (s, 1 H),7.68 (d, J = 1.3 Hz, 1 H), 7.42 (dd, J = 8.1 Hz, J = 1 .3 Hz, 1 H), 7.09 (t, J = 7.7 Hz, 1 H), 6.77 (s, 1 H), 6.73 (d, J = 7.5 Hz, 1 H), 6.65 (d, J = 8.1 Hz, 1 H), 5.30 (s, 2H).
Example 30: 7-(3-hydroxyphenyl)ethynyl-2-isoquinolin-3-yl-chromen-4- one oxime was prepared in 28% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one Oferf-buty! oxime (example 2B) and 3-hydroxyphenylacetyiene. The title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers.
Figure imgf000071_0002
Mp: 255-259X
MS (ESI+): 405.4 [C26H16N203+Hf (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.30 (s, 1 H), 9.79 (s, 1 H), 9.43 (s, 1 H), 8.54 (s, 1 H), 8.23 (d, J = 8.1 Hz, 1 H), 8.12 (d, J = 8.2 Hz, 1 H), 7.92 (d, J = 8.3 Hz, 1 H), 7.90 (t, J = 7.1 Hz, 1 H, 1 H), 7.79 (t, J = 8.1 Hz, 1 H), 7.75 (s, 1 H),7.72 (d, J = 1.4 Hz, 1 H), 7.45 (dd, J = 8.1 Hz, J = 1.3 Hz, 1 H), 7.26 (t, J = 7.9 Hz, 1 H), 6.03 (d, J = 7.5 Hz, 1 H), 6.96 (s, 1 H), 6.85 (dd, J = 7.3 Hz, J = 2.5 Hz, 1 H).
Example 31: 2-isoquinolin-3-yl-7-(4-methoxyphenyl)ethynyl-chromen~4~ one oxime was prepared in 9% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-fert-butyl oxime (example 2B) and 4-methoxyphenyiacetylene. The title compound was purified by preparative HPLC (gradient 35-20% Water/acetonitrile+0.05% trifluoroacetic acid) and isolated as a yellow solid and as a 95/05 mixture of Z/E oxime isomers.
Figure imgf000072_0001
MS (ESI+): 419.1 [C27H18N203+H3+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.31 (s, 1 H), 9.49 (s, 1 H), 8.60 (s, 1 H), 8.29 (d, J = 8.1 Hz, 1 H), 8.18 (d, J = 8.0 Hz, 1 H), 8.00-7.90 (m, 2H), 7.90-7.80 (m, 1 H), 7.81 (s, 1 H), 7.75 (s, 1 H), 7.61 (d, J = 8.6 Hz, 2H), 7.48 (d, J = 7.5 Hz, 1 H), 7.09 (d, J = 8.6 Hz, 2H), 3.88 (s, 3H).
Example 32: 7-( 2-chlorophenyl) ethynyi-2-isoquin olin-3-yl-chrom en-4-one oxime was prepared in 10% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3~yl-chromen-4-one O-iert-butyl oxime (example 2B) and 2-chiorophenylacetylene. The title compound was isolated as an orange solid and as a 80/20 mixture of Z/E oxime isomers.
Figure imgf000072_0002
p: 292-297°C MS (ES!+): 423.4 [C26H15CiN202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.31 (s, 1 H), 9.43 (s, 1 H), 8.56 (s, 1 H), 8.22 (d, J = 7.8 Hz, 1 H), 8.12 (d, J = 7.8 Hz, 1 H), 7.95 (d, J = 8.1 Hz, 1 H), 7.89 (t, J = 7.7 Hz, 1 H), 7.80-7.65 (m, 4H), 7.64 (d, J = 7.5 Hz, 1 H), 7.55-7.30 (m, 3H).
Example 33: 7-( 3- im ethylamino-prop- 1-ynyl) -2-iso quin olin-3-yl-chrom en- 4-one oxime was prepared in 20% overaii yield using the method described in example 24, starting from 7-bromo-2-isoquino!in-3-yl-chromen-4-one O-ierf-butyl oxime (example 2B) and N,N-dimethylamino-2-propyne. The titie compound was purified by preparative HPLC (gradient 70 - 55% Water/acetonitrile + 0.05% trifluoroacetic acid) and isolated as an orange solid and as a 85/15 mixture of Z/E oxime isomers.
Figure imgf000073_0001
MS (ESI+): 370.1 [C23H19N302+H]+ (m/z).
Example 34: 6-cyclopropyl-2-isoquinolin-3-yl-chromen-4-one oxime was isolated in 60% yield using method D (step 2), starting from 6-cyclopropyl-2- isoquinolin-3-yl-chromen -4-one O-tert-butyl oxime (example 34A) as a yellow solid after purification by preparative HPLC (gradient 50 - 35% Water/acetonitrile + 0.05% trifluoroacetic acid). The title compound was isolated as a 85/15 mixture of Z/E oxime isomers.
Figure imgf000073_0002
MS (ESI+): 329.2 [C21H16N202+Hf (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.48 (s, 1 H), 9.38 (s, 1 H), 8.36 (s, 1 H), 8.19 (d, J = 8, 1 Hz, 1 H), 8.12 (d, J = 8.0 Hz, 1 H), 7.90-7.65 (m, 2H), 7.41 (d, J = 8.4 Hz, 2H), 7.29 (d, J = 7.7 Hz, 1 H), 7.08 (s, 1 H), 2.00 (m, 1H), 0.98 (m, 2H), 0.67 (m, 2H).
Example 34 A: 6'Cyclopropy!-2-isoquinolin-3-yl-chromen-4-one O-tert- butyl oxime
A solution of 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-fert-butyl oxime (100 mg, 0.236 mmol), palladium acetate (3 mg, 0.014 mmol), potassium phosphate (175 mg, 0.826 mmol), dicycfohexylbiphenylphosphine (8 mg, 0.024 mmol), Cyclopropylboronic acid pinacol ester (99 mg, 0.59 mmol) in toluene (3 ml) was degassed with argon for 10 min. The reactor was sealed and heated at 120°C for 18 hours. The mixture was poured onto a saturated aqueous solution of ammonium chloride, extracted with ethyl acetate, washed with water and brine, dried over sodium sulfate and purified by flash chromatography over silica gel (cyclohexane/dichloromethane: 0-80%) to yield 2-isoquinolin-3-yl-7-(pyridin-2-yl- ethynyi)-chromen-4-one O-tert-butyl oxime (18 mg, 20%).
MS (ESI+): 385.1 [C25H24N202+H]+ (m/z).
Example 35: 2-isoquinolin-3-yl-6-(pyrroiidin-1-yl)'Chromen-4-one oxime was isolated in 63% yield using method D (step 2), starting from 2-isoquinolin-3-yl-6- (pyrrolidin-1-y!)-chromen-4-one O-ferf-butyl oxime (example 35A) as an orange solid after trituraiing in hot chloroform. The title compound was isolated as a 97/3 mixture of Z/E oxime isomers.
Figure imgf000074_0001
MS (ESI+): 358.1 [C22H19N302+H]+ (m/z).
1 H N R of the major 2 isomer: DMSOd6 δ (ppm): 10.85 (s, 1 H), 9.41 (s, 1 H), 8.45 (s, 1H), 8.21 (d, J = 8.1 Hz, 1H), 8.14 (d, J = 7.9 Hz, 1 H), 7.86 (t, J = 7.0 Hz, 1H), 7.76 (t, J = 7.1 Hz, 1 H), 7.71 (s, 1 H), 7.38 (d, J = 9.0 Hz, 1 H), 6.90 (d, J = 2.6 Hz, 1 H), 6.82 (dd, J = 9.0 Hz, J = 2.6 Hz, 1 H), 3.27 (br. s, 4H), 1.98 (br. s, 4H). Example 35A: 2-isoquinoUn-3-yl-6-(pyrrolidin-1-yl)-chromen-4-one O-tert- butyl oxime
A vial was charged with 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-f erf-butyl oxime (100 mg, 0.236 mmol), Pd-PEPPSI® (3 mg, 0.005 mmol), potassium tert- butoxide (40 mg, 0.354 mmol) and purged with argon for 15 min., Pyrrolidine (24 μΙ, 0.283 mmol) and dry 1 ,2-dimethoxyethane (1 ml) were added and the solution was heated at 80°C for 18 hours. The mixture was diluted with ethyl acetate washed with a saturated aqueous solution of ammonium chloride, water and brine, dried over sodium sulfate and purified by flash chromatography over silica gei (cyclohexane/dichloromethane: 0-80%) to yield 2-isoquinolin-3-yl-6-(pyrrolidin-1-yl)- chromen-4-one O-ferf-butyl oxime (64 mg, 65%).
MS (ESI+): 414.5 [C26H27N302+H]+ (m/z).
Example 36; 2~isoquinolin-3-yl~6-(vinyl)-chromen~4-one oxime was isolated in 40% yield using method D (step 2), starting from 2-isoquinolin-3-yl-6- (vinyl)-chromen-4-one O-terf-butyl oxime (example 36A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate : 0- 40%)
Figure imgf000075_0001
Mp: 198-200°C
MS (ES!+): 315.3 [C20H14N2O2+H]+ (m/z).
1 H NMR of the major Z isomer: D SO-ds δ (ppm): 11.10 (s, 1 H), 9.42 (s, 1 H), 8.50 (s, 1 H), 8.23 (d, J = 7.9 Hz, 1 H), 8.15 (d, J = 7.9 Hz, 1 H), 7.92 (d, J = 2.0 Hz, 1 H), 7.88 (t, J = 7.0 Hz, 1 H), 7.78 (t, J = 7.1 Hz, 1 H), 7.77 (s, 1 H), 7.72 (dd, J = 8.9 Hz, J = 2.2 Hz , 1 H), 7.51 (dd, J = 8.6 Hz, J = 9.0 Hz, 1 H), 6.82 (dd, J - 17.7 Hz, J = 11 .1 Hz, 1 H), 5.85 (d, J = 17.7 Hz, 1 H), 5.32 (d, J = 11 .1 Hz, 1 H).
Example 36A: 2-isoquinoiin-3-yl-6-(vinyl)-chromen-4-one O-tert-butyl In a sealed tube was degassed with argon for 15 min. a solution of 6-bromo-2~ isoquinoiin-3-yl-chromen-4-one O-ferf-butyi oxime (100 mg, 0.236 mmol) in toluene. Tributyi(vinyl)tin (76 μΙ, 0.26 mmol) and tetrakis(t phenylphosphine)palladium (13 mg, 0.012 mmol) were added degassing was continued for 5 min. The tube was sealed and heated at 120°C for 8 hours. The mixture was filtered, concentrated and purified by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-10%) to yield 2- isoquinoiin-3-yl-6-(vinyl)-chromen-4-one O-fert-butyl oxime (84 mg, 96%).
MS (ESI+): 371.0 [C24H22N202+H]+ (m/z).
Example 37: 6-ethyl~2~isoquinolin-3-yl-chromen-4-one oxime was isolated in 50% yield using method D (step 2), starting from 6-ethyl-2-isoquino!in-3-yi-chromen- 4-one O-ferf-butyl oxime (example 37 A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate : 0-20%)
Figure imgf000076_0001
Mp: 210-212°C
MS (ESI+): 317.0 [C2oH16N202+H]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.00 (s, 1 H), 9.42 (s, 1 H), 8.48 (s, 1 H), 8.22 (d, J = 8.0 Hz, 1 H), 8.15 (d, J = 7.9 Hz, 1 H), 7.88 (t, J = 7.0 Hz, 1 H), 7.82-7.72 (m, 2H), 7.77 (s, 1 H), 7.46-7.35 (m, 2H), 2.68 (q, J = 7.5 Hz, 2H), 1.22 (t, J = 7.5 Hz, 3H).
Example 37 A: 6-ethyl~2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime
A solution of 2-isoquinoiirv3~yl-6-(viny1)-chromen-4-one O-ieri-butyl oxime (50 mg, 0.135 mmol) in mixture of tetrahydrofuran (4m!) and methanol (1 ml) was hydrogenated over 10% palladium Pd/C (10 mg) under I atmosphere at room temperature for 6 to 8 hours. The catalyst was removed by filtration and the filtrate was concentrated to dryness to yield 6-ethyi-2-isoquinolin-3-yl-chromen-4-one O-tert- butyl oxime (46 mg, 90%) that was used without further purification. 1 H NMR: CDCI3 δ (ppm): 9.28 (s, 1 H), 8.30 (s, 1 H), 8.01 (d, J = 7.7 Hz, 1 H), 7.94 (d, J = 7.5 Hz, 1 H), 7.89 (m, 1 H), 7.79 (s, 1 H), 7.75 (td, J = 8.2 Hz, J = 1.5 Hz, 1 H), 7.65 (td, J = 7.5 Hz, J = 1.5 Hz, 1 H), 2.71 (q, J = 7.5 Hz, 2H), 1.29 (t, J = 7.5 Hz, 3H).
Example 38: 6~cyano-2-isoquinoIin-3-yI~chromen-4-one oxime was prepared in 63% yield using method D (step 2), starting from 6-cyano-2-isoquinolin-3- yl-chromen-4-one O-ferf-butyl oxime (example 38A) as a paie yellow solid after recrystailization in hot chloroform. The title compound was isolated as a 90/10 mixture of Z/E oxime isomers.
Figure imgf000077_0001
Mp: 280°C dec.
MS (ESI+): 314.3 [CieH aOa+H]* (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 147 (s, 1 H), 9.41 (s, 1 H), 8.49 (s, 1 H), 8.25-8.18 (m, 2H), 8.12 (d, J = 8.1 Hz, 1 H), 7.98 (dd, J = 8.6 Hz, J = 2.0 Hz, 1 H), 7.88 (t, J = 7.0 Hz, 1 H), 7.78 (t, J = 7.0 Hz, 1 H), 7.76 (s, 1 H), 7.68 (d, J = 8.6 Hz, 1 H).
Example 38 A: 6-cyano-2-isoquinolin-3-yl-chromen~4-one O-tert-butyl oxime
In a microwave vial was degassed with argon for 15 min. a mixture of 6-bromo- 2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (50 mg, 0.1 18 mmol), zinc cyanide (14 mg, 0.118 mmol) in dimethylformamide (1.5 ml). Tetrakis(triphenylphosphine)palladium (4 mg, 0.003 mmol) was added and the vial was sealed and heated at 120°C under microwave irradiation for 10 min.. The mixture was diiuted with ethyl acetate, washed with a 5% aqueous solution of ammonia, brine and dried over sodium sulfate. The solution was concentrated and purified by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-20%) to yield 6-cyano-2- isoquinoiin-3-yl-chromen-4-one O-fert-butyl oxime (33 mg, 76%). MS (ESI+): 370.4 [C23H19N302+H]+ (m/z).
Example 39: 6-dimethylamino-2-isoquinolin-3-yl-chromen-4-one oxime was prepared in 51 % overall yield following the method described in example 35, starting from 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime and dimethy!amine (2.0 M in tetrahydrofuran). The title compound was isolated as a pale yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-60%).
Figure imgf000078_0001
Mp: 223-225°C.
MS (ESI+): 332.1 [C20H17N3O2+H]+ (m/z).
1 H NM : DMSO-d6 δ (ppm): 10.91 (s, 1 H), 9.41 (s, 1 H), 8.46 (s, 1 H), 8.21 (d, J = 8.1 Hz, 1 H), 8.14 (d, J = 8.1 Hz, 1 H), 7.87 (t, J = 8.3 Hz, 1 H), 7.76 (t, J = 7.0 Hz, 1 H), 7.71 (s, 1 H), 7.39 (d, J = 9.0 Hz, 1 H), 7.10-7.00 (m, 2 H), 2.94 (s, 6H).
Example 40: 2-iso quin olin-3-yl-6-( morph olin-4-yl-m ethyl)-chrom en-4-one oxime was prepared in 30% yield using method D (step 2), starting from 2-isoquinolin- 3-yl-6-(morpholin-4-yl-methyl)-chromen-4-one O-ferf-butyi oxime (example 40A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-100%). The title compound was isolated as a 70/30 mixture of Z/E oxime isomers.
Figure imgf000078_0002
MS (ESI+): 388.5 [C23H21N303+H]+ (m/z). .
1 H NMR of the major Z isomer: D SO-d5 δ (ppm): 1 1.04 (s, H), 9.42 (s, 8.49 (s, 1 H), 8.22 (d, J = 8.1 Hz, 1 H), 8.19 (d, J = 8.1 Hz, 1 H), 7.90-7.85 (m, 2H), 7.85-7.72 (m, 1 H), 7.76 (s, 1 H), 7.55-7.42 (m, 2H), 3.59 (br. s, 4H), 3.52 (s, 2H), 2.39 (br. S, 4H).
Example 40A: 2-isoquinolin-3-yt-6-(morpholin~4-yl~methyl)~chromen-4- one O-tert-butyl oxime
In a sealed tube was degassed with argon for 15 min. a mixture of 6-bromo-2- isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (100 mg, 0.236 mmol), cesium carbonate (230 mg, 0.708 mmol), dichioro[1 ,1- bis(diphenylphosphino)ferrocene3palladium(ll) dichloromethane adduct (10 mg, 0.012 mmol) and potassium influoroboratomethylmorphoiine in toiuene (7.5 ml) and water (2.5 ml). The tube was sealed and heated at 100°C for 72 hours. The mixture was diluted with ethyl acetate, washed with water, a saturated aqueous solution of ammonium chloride, and extracted three times with a 1 N solution of hydrochloric acid. The aqueous layers were neutralized with a 6N solution of sodium hydroxide and extracted with ethyl acetate. The organic layers were dried over sodium sulfate and concentrated to dryness to yield 2-isoquinolin-3-yl-6-(morpholin-4-yl-methyl)-chromen- 4-one O-ferf-butyl oxime (10 mg, 10%) as a brown residue that was used without further purification.
MS (ES!+): 444.2 [027Η29 3θ3+Η; (m/z).
Example 41: 6-hydroxy-2-isoquinolin-3-yl~chromen-4-one oxime was prepared in 56% yield using method D (step 2), starting from 6-hydroxy-2-isoquinolin- 3-yl-chromen-4-one O-ferf-butyl oxime (example 41 A) as a yellow solid after purification by flash chromatography over silica gel (cyc!ohexane/ethyl acetate: 0- 80%). The title compound was isolated as a 95/5 mixture of Z/E oxime isomers.
Figure imgf000079_0001
Mp: 255-260°C.
MS (ESI+): 305.0 [C18H12N203+H]+ (m/z). 1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 10.96 (s, 1H), 9.66 (s, 1H), 9.41 (s, 1H), 8.45 (s, 1H), 8.21 (d, J = 7.9 Hz, 1H), 8.14 (d, J = 8.2 Hz, 1H), 7.87 (t, J = 6.9 Hz, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.70 (s, 1H), 7.36 (d, J = 8.8 Hz, 1H), 7.27 (s, 1H), 6.96 (d, J = 8.5 Hz, 1H).
Example 41A: 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime was prepared in 89% yieid following the method describe in example 18A, starting from 6-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0- 20%). The title compound was isolated as a yellow solid.
1H NMR: CDCi3 δ (ppm): 9.28 (s, 1H), 8.29 (s, 1H), 8.01 (d, J = 7.7 Hz, 1H), 7.94 (d, J = 7.9 Hz, 1H), 7.75 (t, J = 7.4 Hz, 1H), 7.75 (s, 1H), 7.65 (t, J = 7.5 Hz, 1H), 7.50 (d, J = 3.0 Hz, 1H), 7.28-7.22 (m, 1H), 6.90 (dd, J = 8.8 Hz, J = 2.5 Hz, 1H), 5.08 (s, 1H), 1.42 (s, 9H).
Example 42: 2-isoqum" oHn-3-yl-6-methoxy-chromen-4~one oxime was prepared in 62% yield using method D (step 2), starting from 2-isoqutnoiin-3-yl-6- methoxy-chromen-4-one O-ferf-butyl oxime (example 42A) as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0- 80%, then dichioromethane/methanol: 0-10%). The title compound was isolated as a 90/10 mixture of Z/E oxime isomers.
Figure imgf000080_0001
Mp: 250-255°C.
MS (ESI+): 319.0 [C19H14N203+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d8 δ (ppm): 11.04 (s, 1H), 9.41 (s, 1H), 8.47 (s, 1H), 8.21 (d, J = 8,1 Hz, 1H), 8.14 (d, J = 8.0 Hz, 1H), 7.87 (t, J = 7.5 Hz, 1H), 7.77 (t, J = 7.9 Hz, 1H), 7.75 (s, 1H), 7.47 (d, J = 9.0 Hz, 1H), 7.34 (d, J = 3.0 Hz, 1H), 7.15 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 3.81 (s, 3H). Example 42A: 2-isoquinolin-3-yl-6-methoxy-chromen-4-one O-tert-buty! oxime was prepared in 67% yield following the method described in example 19A, starting from 6-hydroxy~2-isoquinolin-3-yl-chromen-4-one O-ierf-butyl oxime (example 41 A). The title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-20%):
1 H NMR: CDCI3 S (ppm): 9.28 (s, 1 H), 8.29 (s, 1 H), 8.01 (d, J = 7.9 Hz, 1 H), 7.94 (d, J = 7.9 Hz, 1 H), 7.77 (s, 1 H), 7.74 (t, J = 7.5 Hz, 1 H), 7.65 (t, J = 7.5 Hz, 1 H), 7.54 (d, J = 3.0 Hz, 1 H), 7.29 (d, J = 9.4 Hz, 1 H), 7.10 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 3.88 (s, 3H), 1.43 (s, 9H).
Example 43: 2-isoquinolin-3-yl- 6-( 2-methoxy-ethoxy)-chrom en-4-one oxime was prepared in 67% overall yield using the method described in example 42 and 42A, starting from 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyl oxime (example 41A) and bromoethylmethylether instead of iodomethane. The title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (cyclohexane/ethyl acetate: 0-80%), and as a 85/15 mixture of Z E oxime isomers.
Figure imgf000081_0001
Mp: 198-204X.
MS (ESH): 363.2 [C21H18N204+H]+ (m/z).
1 H NMR of the major Z isomer: D SO-d6 δ (ppm): 1 1.04 (s, 1 H), 9.41 (s, 1 H), 8.47 (s, 1 H), 8.21 (d, J = 8.1 Hz, 1 H), 8.14 (d, J = 8.0 Hz, 1 H), 7.87 (t, J = 7.7 Hz, 1 H), 7.77 (t, J = 7.9 Hz, 1 H), 7.73 (s, 1 H), 7.46 (d, J = 8.9 Hz, 1 H), 7.34 (d, J = 3.0 Hz, 1 H), 7.16 (dd, J = 9.0 Hz, J ~ 3.0 Hz, 1 H), 4.14 (m, 2H), 3.39 (m, 2H) 3.32 (s, 3H).
Example 44: 6-(2-dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4- one oxime was prepared in 66% yield using method D (step 2), starting from 6-(2- dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4-one O-iert-butyl oxime (example 44A) as an orange solid and as a 90/10 mixture of Z/E oxime isomers.
Figure imgf000082_0001
Mp: 203-205°C.
MS (ESI+): 376.5 [C22H21N303+H]+ (m/z).
1 H N R of the major Z isomer: DMSO-de S (ppm): 1 1.04 (s, 1 H), 9.41 (s, 1 H), 8.47 (s, 1 H), 8.21 (d, J = 8.1 Hz, 1 H), 8.14 (d, J = 8.0 Hz, 1 H), 7.87 (t, J = 7.0 Hz, 1 H), 7.77 (t, J = 7.7 Hz, 1 H), 7.73 (s, - 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.33 (d, J = 3.0 Hz, 1 H), 7.15 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.09 (m, 2H), 2.64 (m, 2H) 2.23 (s, 6H).
Example 44A: 6-(2-dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4- one O-tert-butyl oxime
To a cold solution of triphenylphosphine ( 10 mg, 0.41 mmoi) in tetrahydrofuran (3 ml) under argon were successively added 2-dimethylamino-ethanol (30 μΙ, 0.27 mmol), a solution of diethylazodicarboxylate (40% in toluene, 190 μΐ, 0.41 mmol) and 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O-tert-butyl oxime (100 mg, 0.27 mmol). The solution was stirred at room temperature for 18 hours. The mixture was poured onto a saturated solution of sodium hydrogenocarbonate and extracted three times with dichloromethane. The organic layers were washed with brine, dried over sodium sulfate and concentrated to dryness to yield 2-isoquinolin-3-yl-6- (morpholin-4-yl-methyl)-chromen-4-one O-f erf-butyl oxime (30 mg, 22%) as a yellow solid after purification by flash chromatography over silica gel (dichloromethane/methanol: 0-20%)
MS (ESi+): 432.3 [0Ζ6Η29Ν303+ΗΓ (m/z).
Example 45: 2-isoquinolin-3-yl- 6-[3-(4-methyl-piperazin- 1-yl)- propylamino]-chromen-4~one oxime was prepared in 85% overall yield using the method described in examples 35 and 35A, starting from 6-bromo-2-isoquinolsn-3-yl- chromen-4-one O-ferf-butyl oxime (example 4B) and 3-(4-methyl-piperazin-1 -yl)- propytamine. The title compound was isolated as a yellow solid and as a 95/5 mixture of Z E oxime isomers.
Figure imgf000083_0001
Mp: 208-211°C.
MS (ESl÷): 444.5 [C25H29N602+Hf (m/z).
1H N R of the major Z isomer: DMSO-d6 δ (ppm): 10.83 (s, 1H), 9.40 (s, 1H), 8.43 (s, H), 8.20 (d, J = 8.1 Hz, 1H), 8.13 (d, J = 8.1 Hz, 1H), 7.85 (t, J = 7.2 Hz, 1H), 7.76 (t, J = 7.5 Hz, 1H), 7.69 (s, 1H), 7.28 (d, J = 8.8 Hz, 1H), 6.94 (d, J = 3.0 Hz, 1H), 6.82 (dd, J = 8.9 Hz, j = 3.0 Hz, 1H), 5.88 (m, 1H), 3.04 (m, 2H) 2,45-2.20 (m, 10H), 2.14 (s, 3H), 1.70 (m, 2H).
Example 46: 2-isoquinolin-3-yl-6-[2-(4-methyl-piperazin-1-yl)-ethoxyJ- chromen-4-one oxime was prepared in 49% overall yield using the method described in examples 44 and 44A, starting from 6-hydroxy-2-isoquinolin-3-yl-chromen-4-one O- fe/ -butyl oxime (example 41 A) and 2-(4-methyl-piperaztn-1-yl)-ethanoi. The title compound was isolated as a yellow solid and as a 90/10 mixture of ZE oxime isomers.
Figure imgf000083_0002
Mp: 233-236X.
MS (ES1+): 431.3 [C25H26N403+H]+ (m/z).
1H NMR of the major Z isomer: DMSO-d6 δ (ppm): 11.03 (s, 1H), 9.41 (s, 1H), 8.47 (s, 1H), 8.21 (d, J = 8.1 Hz, 1H), 8.14 (d, J = 8.1 Hz, 1H), 7.87 (t, J = 7.4 Hz, 1H), 7.77 (t, J = 7.5 Hz, 1H), 7.73 (s, 1H), 7.46 (d, J = 9.2 Hz, 1H), 7.33 (d, J = 2.5 Hz, 1H), 7.15 (dd, J = 9.0 Hz, J = 2.5 Hz, 1H), 4.11 (t, J = 5.6 Hz, 2H), 2.70 (t, J = 5.6 Hz, 2H) 2.60-2.40 {m, 4H), 2.40-2.20 (m, 4H), 2.14 (s, 3H).
Example 47: 2-isoquinolin-3-yl-7-phenyl-chromen-4-one oxime was isolated in 30% yield using method D (step 2), starting from 2-lsoquinolin-3-yl-7- phenyl-chromen-4-one O-iert-butyl oxime (example 47A) as a pale yellow solid after recrystallization in hot chloroform.
Figure imgf000084_0001
Mp: 275-278°C
MS (ESI+): 365.4 [C24H16N202+H]+ (m/z).
1 H N R: DMSOd6 δ (ppm): 11.13 (s, 1 H), 9.44 (s, 1 H), 8.57 (s, 1 H), 8.23 (d, J = 7.9 Hz, 1 H), 8.13 (d, J = 8.1 Hz, 1 H), 7.99 (d, J = 8.2 Hz, 1 H), 7.90 (t, J = 7.0 Hz, 1 H), 7.86-7.75 (m, 4H), 7.79 (s, 1 H), 7.65 (dd, J = 8.3 Hz, J = 1.9 Hz, 1 H), 7.54 (t, J = 7.7 Hz, 2H), 7.44 (t, J = 7.3 Hz, 1 H).
Example 47A: 2-isoquinolin-3-yl-7-phenyI-chromen-4-one O-tert-butyl oxime
In a sealed tube, a solution of 7-bromo-2-isoquinoiin-3-yl-chromen-4-one O- fe/t-butyl oxime (150 mg, 0,35 mmol) phenylboronic acid (60 mg, 0.49 mmol) a 2M aqueous solution of sodium carbonate (350 pi, 0.70 mmol) in toluene (2.4 ml) and ethanol (0.5 ml) was degassed with argon for 10 min and then tetrakis(triphenylphosphine)palladium (20 mg, 0.018 mmol) was added. The tube was sealed and the mixture was heated under argon at 120°C for 4 hours. After cooling, the reaction mixture was poured onto a saturated solution of ammonium chloride and extracted with ethyl acetate. The extracts were washed with brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography {gradient cydohexane/dichloromethane: 0-80%) to yield 2-lsoquinolin-3-yl-7-phenyl-chromen-4- one O-ferf-butyl oxime (135 mg, 91 %) as a yellow solid.
MS (ESI+): 447.6 [C30H26N2O2+H]+ (m/z).
1 H N R: CDCI3 δ (ppm): 9.30 (s, 1 H), 8.34 (s, 1 H), 8.15 (d, J = 8.3 Hz, 1 H),
8.02 (d, J = 7.7 Hz, 1 H), 7.96 (d, J = 7.9 Hz, 1 H), 7.82 (s, 1 H), 7.76 (td, J = 6.8 Hz, J =
1.3 Hz, 1 H), 7.72-7.63 (m, 3H), 7.59 (d, J = 1.7 Hz, 1 H), 7.53-7.44 (m, 3H), 7.44-7.36 (m, 1 H), 1.45 (s, 9H). Example 48: 7-(4-biphenyl)~2-i$oquinoiin~3-yl-chromen-4-one oxime was isolated in 26% overall yield using the method described in examples 47 and 47A, starting from 7-bromo-2-lsoquinolin-3-yl-chromen-4-one O-terf-butyl oxime and 4- biphenylboronic acid. The title compound was isolated as a beige solid after triturating in hot chloroform and as a 80/20 mixture of Z/E oxime isomers.
Figure imgf000085_0001
Mp: 283-285°C
MS (ESI+): 441.0 [C30H20N2O2+H]+ (m/z).
1 H NMR: DMSO-d6 δ (ppm): 1 1.20 (s, 1 H), 9.45 (s, 1 H), 8.59 (s, H), 8.24 (d, J = 8.3 Hz, 1 H), 8.15 (d, J = 8.1 Hz, 1 H), 8.01 (d, J = 8.3 Hz, 1 H), 7.96-7.68 (m, 1 1 H), 7.51 (t, J = 7.7 Hz, 2H), 7.41 (t, J = 7.3 Hz, 1 H).
Example 49: 2-isoquinoiin-3-yl-7-[3-(4-methyl-piperazin-1-yl)- propylaminoJ'Chromen-4-one oxime was prepared in 19% overall yield using the method described in examples 45, starting from 7-bromo-2-isoquinolin-3-yl-chromen- 4-one O-terf-butyl oxime (example 2B) and 3-(4-methyl-piperazin-1-yl)-propylamine. The title compound was isolated as a yellow solid.
Figure imgf000085_0002
Mp: 228-233°C.
MS (ESI+): 444.4 [C26H29N502+H]+ (m/z).
1 H NMR: DMSO-d6 δ (ppm): 10.45 (s, 1 H), 9.40 (s, 1 H), 8.45 (s, 1 H), 8.20 (d, J = 8.1 Hz, 1 H), 8.15 (d, J = 8.1 Hz, 1 H), 7.87 (t, J = 7.0 Hz, 1 H), 7.76 (t, J = 7.0 Hz, 1 H), 7.70 (s, 1 H), 7.58 (d, J = 8.7 Hz, H), 6.57 (d, J = 8.7 Hz, 1 H), 6.52 (s, 1 H), 6.31 (m, 1 H), 3.13 (m, 2H) 2.45-2.20 (m, 10H), 2.18 (s, 3H), 1.75 (m, 2H). Example 50: 2-lsoquinolin-3-yl-7-{3-[2-(4~methyl-piperazin-1-yl)-ethoxy]- phenylethynyl}-chromen-4-one oxime was prepared in 9% overall yield Using the method described in examples 44 and 44A, starting from 7-(3-hydroxyphenyl)ethynyl- 2-isoquinoiin-3-yl-chromen-4-one O-terf-butyl oxime (te/f-buiyl protected oxime of example 30) and 2-(4-methyi-piperazin-1-yl)-ethanol. The title compound was isolated as a yellow solid and as a 90/10 mixture of 2 E oxime isomers after Preparative HPLC purification (gradient 65-50% Water/aceionitrile+0.05% trifluoroacetic acid).
Figure imgf000086_0001
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1 % HCOOH): > 95 %; RT = 4.94 min.
MS (ESI+): 531.2 [¾3Η3οΝ4θ3+Η]+ (m/z).
1 H NMR of the major Z isomer: DMSO-d6 δ (ppm): 1 1.30 (s, 1 H), 9.44 (s, 1 H),
8.54 (s, 1 H), 8.30 (d, J = 8.1 Hz, 1 H), 8.18 (d, J = 8.1 Hz, 1 H), 8.01 (d, J = 8.1 Hz, 1 H), 7.96 (t, J = 7.4 Hz, 1 H), 7.86 (t, J = 7.5 Hz, 1 H), 7.82 (s, 1 H), 7.79 (s, 1 H), 7.45-
7.55 (m, 2H), 7.32 (s, 1 H), 7.21 -7.18 (m, 1 H), 4.1 1 (t, J = 5.6 Hz, 2H), 2.70 (t, J = 5.6 Hz, 2H) 2.60-2.40 (m, 4H), 2.40-2.20 (m, 4H), 2.14 (s, 3H).
Example 51: 2-lsoquinolin-3-yl-7-{3-methylaminophenylethynyl}- chromen-4-one oxime was prepared using method D (step 2) starting from 2- isoquinolin-3-yl-7-{3-methylaminophenyiethynyl}-chromen-4-one O-tert-butyi oxime (example 51 A). The title compound was isolated as a yellow solid and as a 90/10 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 50-35% Wa†er/aceionitrile÷0.05% trifluoroacetic acid).
Figure imgf000087_0001
HPLC (gradient 95 % - 40 % H20/ACN + 0.05% TFA (in 12 min.): > 90%; RT = 12.25 min.
MS (ESI+): 418.1 [C27H19N302÷Hf (m/z).
1 H NMR of the major Z isomer: (400 MHz) DMSO-de δ (ppm): 1 1.26 (s, 1 H), 9.43 (s, 1 H), 8.54 (s, 1 H), 8.23 (d, J = 8.0 Hz, 1 H), 8.12 (d, J = 8.1 Hz, 1 H), 7.95-7.85 (m, 2H), 7.82-7.75 (m, 1 H), 7.76 (s, 1 H), 7.70 (s, 1 H), 7.43 (d, J = 8.0 Hz, 1 H), 7.15 (t, J = 8.0 Hz, 1 H), 6.77 (d, J = 7.6 Hz, 1 H), 6.72 (s, 1 H), 6.63 (d, J = 8.0 Hz, 1 H), 5.88 (m, 1 H), 2.71 (d, J = 5.2 Hz, 3H).
Example 51 A: 2-!s oquinolin-3-yl-7-{3-methylamin oph enylethynyl}- chromen-4-one O-tert-butyl oxime. A solution of 7-(3-aminophenylethynyl)-2- (lsoquinoiin-3-yl)-chromen-4-one O-fert-butyl oxime (iert-butyi protected oxime of example 29) (127 mg, 0.286 mmol) in dimethy!formamide (3 ml) was added to a cold suspension of sodium hydride (60% in mineral oii, 24 mg, 0.608 mmol) in DMF (1 ml). The reaction mixture was stirred for 1 hour at room temperature and cooled to 0°C. lodomethane (43 μ!, 0.690 mmol) was added and the solution was stirred at room temperature for 18 hours. The mixture was poured onto water and extracted with ethyl acetate. The organic extracts were washed with water, brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography (gradient cyclohexane / dichloromethane: 0-45%) to yield 2-lsoquinoiin-3-yl-7-{3- methylaminophenylethynyl}-chromen-4-one O-terf-butyS oxime (65 mg, 50%) as a beige solid.
1 H NMR: (300 MHz) CDCI33 (ppm): 9.29 (s, 1 H), 8.29 (s, 1 H), 8.05 (d, J = 8.3 Hz, 1 H), 8.02 (d, J = 7.9 Hz, 1 H), 7.94 (d, J = 7.9 Hz, 1 H), 7.79 (s, 1 H), 7.76 (td, J = 7.4 Hz, J = 1.2 Hz, 1 H), 7.66 (id, J = 7.5 Hz, J = 1.2 Hz, 1 H), 7.52 (d, J = 1.5 Hz, 1 H), 7.35 (dd, J = 8.3 Hz, J = 1.5 Hz, 1 H), 7.18 (t, J = 7.9 Hz, 1 H), 6.93 (d, J = 8.4Hz, 1 H), 6.80 (m, 1 H), 6.62 (dd, J = 8.3 Hz, J = 1.7 Hz, 1 H), 3.78 (br. s, 1 H), 2.87 (s, 3H), 1.43 (s, 9H). Example 52: 7-(4-Hydroxy-b ut-1-ynyl) -2-isoquinolin-3-yl-chrom en-4-on e oxime was prepared in 10% overall yield using the method described in example 24, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ferf-butyS oxime (example 2B) and 3-butyn-1 -ol. The title compound was isolated as a yellow solid and as a 70/30 mixture of Z/E oxime isomers.
Figure imgf000088_0001
Mp: 238-240°C.
MS (ESI+): 357.1 [C22H16N203+Hl+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.20 (s, 1 H), 9.42 (s, 1 H), 8.51 (s, 1 H), 8.22 (d, J = 7.9 Hz, 1 H), 8.1 1 (d, J = 8.0 Hz, 1 H), 7.92-7.72 (m, 3H), 7.73 (s, 1 H), 7.55 (s, 1 H), 7.29 (dd, J = 8.1 Hz, J = 1.5 Hz, 1 H), 4.95 (t, J = 6.7 Hz, 1 H), 3.63 (d, J = 6.7 Hz, 2H), 2.61 (d, J = 6.7 Hz, 2H).
Example 53: 2-!soquinolin-3-yl- 7-[3-(2-methoxy-ethoxy)'phenylethynyl]~ chromen-4-one oxime was prepared in 48% overall yield using the methods described in examples 42 and 42A, starting from 7-(3-hydroxyphenyl)ethynyl-2- isoquinolin-3-yl-chromerv4-one O-fert-butyl oxime (ferf-butyl protected oxime of example 30) and bromoethylmethylether instead of iodomethane. The title compound was isolated as a yeiiow solid and as a 90/10 mixture of Z/E oxime isomers after purification by flash chromatography over silica gel (gradient cyciohexane/ethyl acetate: 0-50%).
Figure imgf000088_0002
Mp: 155-160°C. MS (ESI+): 463.1 [C29H22N204+Hf (m/z).
Η NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.28 (s, 1 H), 9.43 (s, 1 H), 8.53 (s, 1 H), 8.23 (d, J = 8.1 Hz, 1 H), 8.1 1 (d, J = 8.1 Hz, 1 H), 7.96-7.86 (m, 2H), 7.82-7.70 (m, 3H), 7.46 (dd, J = 8.2 Hz, J = 1.5 Hz, 1 H), 7.37 (t, J = 8.2 Hz, 1 H), 7.21-7.16 (m, 2H), 7.05 (d, J = 9.2 Hz, 1 H), 4.16 (m, 2H), 3.68 (m, 2H), 3.32 (s, 3H).
Example 54: 2-ls oquinolin-3-yl- 7-[3-(2-meth oxy-ethoxy)-prop- 1-ynylJ- chromen-4-one oxime was prepared in 7% overall yield using the methods described in examples 42 and 42A, starting from 7-(3-hydroxy-prop-1 -ynyl)-2-lsoquinolin-3-yl- chromen-4-one O-ierf-buty! oxime (example 54A) and bromoethylmethyiether. The title compound was isolated as a beige solid and as a 90/10 mixture of Z E oxime isomers after triturating in diethyl ether.
Figure imgf000089_0001
Mp: 158-160°C.
MS (ESI+): 401.1 [C24H2o 204+H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.26 (s, 1 H), 9.43 (s, 1 H), 8.52 (s, 1 H), 8.22 (d, J = 8.3 Hz, 1 H), 8.1 1 (d, J = 7.9 Hz, 1 H), 7.94-7.86 (m, 2H), 7.79 (m, 2H), 7.74 (s, 1 H), 7.63 (s, 1 H), 7.36 (d, J = 8.2 Hz, 1 H), 4.45 (s, 1 H), 3.68 (m, 2H), 3.52 (m, 2H), 3.31 (s, 3H).
Example 54 A: 7-(3-hydroxy-prop-1-ynyl)-2-isoquinolin-3-yl-chromen-4- one O-tert-butyl-oxime was prepared in 58% overall yield using the method described in example 23A, starting from 7-bromo-2-isoquinoiin-3-yl-chromen-4-one O- ferf-butyl oxime (example 2B) and 2-propyn-1 -o!.
Figure imgf000089_0002
1 H NMR: (300 MHz) CDCI3 δ (ppm): 9.28 (s, 1 H), 8.26 (s, 1 H), 8.02 (2d, J = 7.9 Hz, 2H), 7.94 (d, J = 7.9 Hz, 1 H), 7.78 (s, 1 H), 7.76 (td, J = 7.4 Hz, J = 1.2 Hz, 1 H), 7.66 (td, J = 7.5 Hz, J = 1.2 Hz, H), 7.42 (d, J = 1.5 Hz, 1 H), 7.30-7.23 (m, 1 H), 4.54 (d, J = 6.2 Hz, 2H), 1.76 (i, J = 6.2 Hz, 1 H), 1.42 (s, 9H).
Example 55: 7-but-3-en-1-ynyl-2-lsoquinolin-3-yl-chromen-4-one oxime was prepared using method D (step 2) starting from 7-(but-3-en-1-ynyl)-2-lsoquinolin- 3-yi-chromen-4-one O-ferf-butyl oxime (example 55A). The title compound was isolated in 93% yield as a yellow solid and as a 90/10 mixture of Z/E oxime isomers.
Figure imgf000090_0001
p: > 210°C dec.
MS (ESI+): 339.1 [C22H14N202+H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.27 (s, 1 H), 9.42 (s, 1 H), 8.51 (s, 1 H), 8.22 (d, J = 8.0 Hz, 1 H), 8.1 1 (d, J = 8.0 Hz, 1 H), 7.92-7.86 (m, 2H), 7.76 (t, J = 7.0 Hz, 1 H), 7.74 (s, 1 H), 7.62 (s, 1 H), 7.36 (dd, J = 8.2 Hz, J = 1.5 Hz, 1 H), 6.2 (dd, J = 17.5 Hz, J = 11.2 Hz , 1 H), 5.82 (d, J = 17.5 Hz, 1 H), 5.73 (d, J = 11.2 Hz, 1 H).
Example 55A: 7-(but-3-en-1-ynyl)-2-lsoquinolin-3-yl-chromen-4-one O- tert-butyl oxime. To a cold solution of 7-(4-Hydroxy-but-1 -ynyl)-2-isoquinolin-3-yl- chromen-4-one O-ferf-butyl oxime (f erf-butyl protected oxime of example 52) (200 mg, 0.485 mmol) and triethylamine (0.15 ml, 1 .067 mmol) in dichloromethane (3 ml) was added dropwise methanesulfony! chloride (75 μί, 0.970 mmol). The reaction mixture was stirred for 1.5 hours at 0°C and poured onto a saturated solution of sodium hydrogenocarbonate. The product was extracted with dichloromethane, washed with brine, dried over sodium sulfate and concentrated to dryness to yield methanesulfonic acid 4-{4-[(EHert-buioxyimino]-2-isoquinolin-3-yl-4H-chromen-7-yi}-but-3-ynyi ester. The methansulfonic ester was dissolved in dimethyiformamtde (4 ml) and treated with potassium carbonate (201 mg, 146 mmol) and methyl piperazine (73 mg, 0.728 mmol) and the mixture was stirred at 40°C for 20 hours. The mixture was poured onto a saturated solution of sodium hydrogenocarbonate. The reaction mixture was extracted with dichloromethane, washed with water, brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography (gradient cyclohexane/dichioromethane; 0-45%) to yield 7-(but-3-en-1 -yny!)-2-lsoquinolin-3-yi~ chromen-4-one O-rert-butyl oxime (156 mg, 82%) as a yellow solid.
1 H NMR: (300 MHz) CDCI3 δ (ppm): 9.29 (s, 1 H), 8.27 (s, 1 H), 8.03 (d, J = 8.3 Hz, 1 H), 8.02 (d, J = 7.9 Hz, 1 H), 7.94 (d, J = 7.9 Hz, 1 H), 7.79 (s, 1 H), 7.76 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.66 (td, J = 7.5 Hz, J = 1.3 Hz, 1 H), 7.44 (d, J = 1 .5 Hz, 1 H), 7.27 (m, 1 H), 6.05 (dd, J = 17.5 Hz, J = 11.1 Hz , 1 H), 5.79 (dd, J = 17.5 Hz, J = 2.1 Hz, 1 H), 5.60 (d, J = 11.2 Hz, J = 2.1 Hz, 1H), 1.43 (s, 9H).
Example 56: 2"isoquinolin~3-yl-7-methoxy-chromen-4-one oxime was prepared in 18% overall yield using the methods described in examples 41 and 42, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-rerf-butyl oxime (example 2B). The title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers.
Figure imgf000091_0001
Mp: 246-248°C.
MS (ESI+): 319.0 [C19H14N203+H]+ (m/z).
1 H NMR of the major Z isomer. (300 MHz) DMSO-d6 δ (ppm): 10.85 (s, 1 H), 9.42 (s, 1 H), 8.48 (s, 1 H), 8.22 (d, J = 8.0 Hz, 1 H), 8.12 (d, J = 8.0 Hz, 1 H), 7.88 (t, J = 6.9 Hz, 1 H), 7.85-7.75 (m, 2H), 7.75 (s, 1 H), 7.09 (d, J = 2.5 Hz, 1 H), 6.91 (dd, J = 8.8 Hz, J = 2.9 Hz, 1 H), 3.87 (s, 3H).
Example 57: 2-isoquinolin-3-yl-7-(2-methoxy-ethoxy)~chromen-4~one oxime was prepared in 22% overall yield using the methods described in examples 41 and 43, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-ie/ -butyl oxime (example 2B). The title compound was isolated as a yellow solid and as a 80/20 mixture of Z/E oxime isomers after purification by flash chromatography over silica ge! (gradient cyclohexane/ethyl acetate: 20-50%).
Figure imgf000092_0001
p: 223-225°C.
MS (ES1+): 363.2 [021Η18Ν204+Η]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 10.84 (s, 1 H), 9.42 (s, 1 H), 8.50 (s, 1 H), 8.22 (d; J = 8.0 Hz, 1 H), 8.13 (d, J = 8.0 Hz, 1 H), 7.89 (t, J = 7.0 Hz, 1 H), 7.85-7.75 (m, 2H), 7.75 (s, 1 H), 7.11 (d, J = 2.5 Hz, 1 H), 6.91 (dd, J = 8.8 Hz, J = 2.9 Hz, 1 H), 4.22 (m, 2H), 3.72 (m, 2H), 3.34 (s, 3H).
Example 58: 7-cyano-2-isoquinolin-3-yl-chromen-4-one oxime was prepared in 8% overall yield using the methods described in examples 38 and 38A, starting from 7-bromo-2-isoquinolin-3-yl-chromen-4-one O-iert-butyl oxime (example 2B). The title compound was isolated as a beige solid after purification by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-60%).
Figure imgf000092_0002
Mp; 279-281 °C.
MS (ESI+): 314.3 [C19H11N302+H]* (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.57 (s, 1 H), 9.43 (s, 1 H), 8.49 (s, 1 H), 8.23 (d, J = 8.1 Hz, 1 H), 8.12-8.00 (m, 3H), 7.90 (t, J = 7.6 Hz, 1 H), 7.80-7.68 (m, 2H), 7.75 (s, 1 H).
Example 59: 2-lsoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-yl)-prop-1~ynyl]~ chromen-4-one oxime was prepared in 67% yield using method D (step 2) starting from 2-isoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-y!)-prop-1-ynyl]-chromen-4-one O- ferf-butyl oxime (example 59A). The title compound was isolated as a ye!low solid and as a 95/5 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 75-60% Water/acetonttrile+0.05% trifluoroacetic acid).
Figure imgf000093_0001
HPLC (gradient 95 % - 40 % H20/ACN + 0.05% TFA (in 12 min.): > 95%; RT = 8.45 min.
S (ESl+): 425.2 [C26H24N402+H]+ (m/z).
1 H NMR of the major Z isomer: (400 MHz) DMSO-d6 δ (ppm): 9.48 (s, 1 H), 8.58 (s, 1 H), 8.29 (d, J = 8.0 Hz, 1 H), 8.18 (d, J = 8.0 Hz, 1 H), 8.00-7.90 (m, 2H), 7.86 (t, J = 7.2 Hz, 1 H), 7.81 (s, 1 H), 7.80 (s, 1 H), 7.50 (d, J = 8.4 Hz , 1 H), 4.26 (s, 2H), 3.80-3.20 (br multiplets, 8H), 2.92 (s, 3H).
Example 59A: 2-lsoquinolin-3-yl-7-[3-(4-methyl-piperazin-1-yl)-prop-1- ynyl]-chromen -4-one O-tert-butyl oxime. To a coid solution of 7-(3-Hydroxy-prop-1- ynyl)-2-isoquinolin-3-yl-chromen-4-one O-terf-butyi oxime (example 54A) (230 mg, 0.577 mmoi) and triethylamine (0.18 ml, 1.27 mmol) in dichioromethane (3 m!) was added dropwise methanesuSfonyl chloride (90 μΙ, 1.15 mmo!). The reaction mixture was stirred for 4 hours at 0°C and poured onto a saturated solution of ammonium chloride. The product was extracted with dichioromethane, washed with brine, dried over sodium sulfate and concentrated to dryness. The residue methansulfonic ester was dissolved in acetone (3 ml) and treated with potassium carbonate (239 mg, 1.73 mmoi) and methyl piperazine (87 mg, 0.866 mmol) and the mixture was stirred at 25°C for 20 hours. The mixture was poured onto water and extracted with ethyl acetate, washed with water, brine, dried over sodium sulfate and absorbed over silica gel for purification by flash chromatography (ethyl acetate/methanol : 90/10 then ethyl acetate/7N ammoniac in methanol : 90/10) to yield 2-.lsoquinoltn-3-yi-7-[3-(4-rnethyl- piperazin-1-y[)-prop-1-ynyl]-chromen -4-one O-tert-butyl oxime (49 mg, 18%) as a brown solid.
Example 60: 2-(7-hydroxy-isoquinolin~3-yl)-chromen-4-one oxime was prepared in 51 % yield using the method described in example 18, starting from 2-(7- chloro-isoquinolin-3-yi)~chromen-4-one oxime (example 16) instead of 2-(5-bromo- isoquinolin-3-yl)-chromen-4-one O-fert-butyl oxime. The title compound was isolated as a yellow solid after purification by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-80%).
Figure imgf000094_0001
MP: 248-250°C
MS (ESI+): 305.1 [C18H12N203÷Hf (m/z).
H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.98 (s, 1 H), 10.43 (br. s, 1 H), 9.20 (s, 1 H), 8.36 (s, 1 H), 8.02 (d, J = 8.8 Hz, 1 H), 7.91 (dd, J = 8.1 Hz, J = 2.6 Hz, 1 H), 7.67 (s, 1 H), 7.60-7.45 (m, 2H), 7.44-7.36 (m, 2H), 7.29 (dd, J = 7.3 Hz, J = 1.5 Hz, 1 H).
Example 61: 2-[2,6]Naphthyridin-3-yl-chromen-4-one oxime was isolated using methods A and D starting from 2'-hydroxy-acetophenone and [2,6]Naphthyridine-3-carboxylic acid methyl ester (example 61A) as a yellow solid after preparative HPLC purification (gradient 75-60% Water/acetonitrile+0.05% trifluoroacetic acid).
Figure imgf000094_0002
HPLC (gradient 95 % - 40 % H20/ACN + 0.05% TFA (in 12 min.): > 95%; RT = 9.04 min.
MS (ESI+): 290.1 [C17H11N302÷Hf (m/z).
1 H NJVIR: (400 MHz) DMSO-d6 δ (ppm): 1 1.17 (s, 1 H), 9.61 (s, 1 H), 9.57 (s, 1 H), 8.81 (d, J = 5.6 Hz, 1 H), 8.67 (s, 1 H), 8.13 (d, J - 5.6 Hz, 1 H), 7.93 (dd, J = 8.0 Hz, J = 1.2 Hz, 1 H), 7.79 (s, 1 H), 7.57 (td, J = 8.4 Hz, J = 1.2 Hz, 1 H), 7.52 (d, J = 7.6 Hz, 1 H), 7.32 (td, J = 8.0 Hz, J = 1.2 Hz, 1 H). Example 61 A: methyl 6-methyl-isoquinoline-3-carboxylate
Figure imgf000095_0001
To a cold solution of 4-Dimethoxymethyl-pyrtdine-3-carbaldehyde (tetrahedron letters 2004 (45), 553-556) (400 mg, 1.91 mmol) in dich!oromethane (10 ml) was slowly added a solution of Acetylamino-(dimethoxy-phosphoryI)-acetic acid methyl ester (synthesis 1984, 53-60) (503 mg, 2.1 mmoi) and 1.8-diazabicyclo[5.4.0]undec-7- ene (0.31 ml, 2.1 mmol). The solution was stirred at 0°C for 1 hour then at room temperature for 18 hours. The mixture was poured onto a cold saturated solution of sodium hydrogenocarbonate, extracted with dichloromethane, dried over sodium sulfate and concentrated to dryness. The residue was dissolved in toluene (49 ml) and treated with p-toleunesuifontc acid (315 mg, 1.66 mmol). The mixture was heated at reflux temperature for 18 hours and then concentrated under vacuum. The brown residue was dissolved in ethyl acetate, washed with a saturated solution of sodium hydrogenocarbonate, brine, dried over sodium sulfate and concentrated to yield methyl 6-methyi-isoquinoiine-3-carboxylate (241 mg, 62%) as a brown solid.
1 H NMR: (300 MHz) CDCl3 S (ppm): 9.49 (s, 1 H), 9.43 (s, 1 H), 8.87 (d, J = 5.6 Hz, 1 H), 8.72 (s, 1 H), 7.88 (d, J = 5.6 Hz, 1 H), 4.09 (s, 3H).
Example 62: 2-[1,6]Naphthyridin-3-yl-chromen-4-one oxime was isolated in 7% overall yield using methods A and D starting from 2'-hydroxy-ace†ophenone and [1 ,6]Naphthyridine-3-carboxylic acid methyl ester (example 62A) as a yellow solid and a 80/20 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 45-30% Water/MethanoS+0.05% trifluoroacetic acid).
Figure imgf000095_0002
HPLC (gradient 85 % - 25 % H20/MeOH + 0.05% TFA (in 12 min.): > 95%; RT = 1 1.72 min. (Z isomer); 12.09 min. (E isomer).
MS (ESi+): 290.1 [C17H11N302+H]t (m/z).
1 H NMR of the major Z isomer: (400 MHz) DMSO-d6 δ (ppm): 1 1.16 (s, 1 H), 9.52 (s, 1 H), 9.25-9.15 (m, 1 H), 8.70-8.60 (m, 1 H), 8.50 (s, 1 H), 7.93 (dd, J = 7.6 Hz, J = 1.2 Hz, 1 H), 7.83 (s, 1 H), 7.82-7.72 (m, 1 H), 7.65-7.50 (m, 2H), 7.35-7.25 (m, 1 H).
Example 62A: methyl 6-methyl-isoquinoline-3-carboxylate was prepared in 43 % yield according to the method described in example 61 A, starting from 3- Dimethoxymethyl-pyridine-2-carbaldehyde instead of 4-Dime†hoxymethy!-pyridfne-3- carbaidehyde.
Figure imgf000096_0001
1 H NMR: (300 MHz) CDCI3 δ (ppm): 9.39 (dd, J = 2.4 Hz, J = 0.75 Hz, 1 H), 9.21 (dd, J = 4.3 Hz, J = 1.9 Hz, 1 H), 8.81 (s, 1 H), 8.39 (dm, J = 8.4 Hz, 1 H), 7.67 (ddd, J = 8.3 Hz, J = 4.1 Hz, J = 2.0 Hz, 1 H), 4.09 (s, 3H).
Example 63: 2-Pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime was isolated in 43% overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Pyrrolo[1 ,2-c]pyrimidine-3-carboxyIic acid methyl ester as a green- yellow solid after recrystallization in 1-butanol.
Figure imgf000096_0002
MP: 260-263°C
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): > 95%; RT = 5.38 min.
MS (ESI+): 278.1 [C16Hn 302+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-de δ (ppm): 10.97 (s, 1 H), 9.22 (s; 1 H), 8.05 (s, 1 H), 7.88 (dd, J = 7.9 Hz, J = 1.4 Hz, 1 H), 7,82 (d, J = 2.4 Hz, 1 H), 7.54-7.42 (m, 2H), 7.46 (s, 1 H), 7.27 (t, J = 6.8 Hz, 1 H), 6.97 (t, J = 2.9 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H).
Example 64: 2-(5, 7-dimethyl-Pyrrolo[ 1,2-c]pyrimidin-3-yl)-chromen-4~one oxime was isolated as a yeliow solid in 19% overall yield using method A and D starting from 2'-hydroxy-acetophenone and 5,7-dimethyl-Pyrrolo[1,2-cjpyrimidine-3- carboxylic acid methyl ester.
Figure imgf000097_0001
MP; >250°C dec.
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): > 95%; RT = 5.63 mm.
MS(ESI+); 306.1 [C18H15N302+H]+ (m/z).
1H N R; (300 MHz) D SO-d6 δ (ppm); 10.89 (s, 1H), 8.89 (s, 1H), 7.94 (d, J = 1.3 Hz, 1H), 7.88 (d, J = 7.9 Hz, 1H), 7.54-7.45 (m, 2H), 7.41 (s, 1H), 7.26 (m, 1H), 6.61 (s, 1H), 2.54 (s, 3H), 2.35 (s, 3H).
Example 65: 2-(6-bromo-Pyrro!o[1,2-c]pyrimidin-3-yI)-chromen-4-one oxime was isolated as a yellow solid using method A and D starting from 2'-hydroxy- acetophenone and 6-bromo-PyrroIo[1,2-c]pyrimidine-3-carboxylic acid methyl ester.
Figure imgf000097_0002
MP; 244-245°C.
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): > 95%; RT = 5.69 mm.
MS(ESI+): 356.1 [C16H10BrN3O2+H]+ (m/z).
1H NMR: (300 Mhz) DMSO-d6 δ (ppm): 11.03 (s, 1H), 9.15 (s, 1H), 8.01 (s, 1H), 7.98 (d, J = 0.75 Hz, 1H), 7.88 (dd, J = 8.0 Hz, J = 1.3 Hz, 1H), 7.52 (td, J = 7.7 Hz, J = 1.5 Hz, 1H), 7.48 (s, 1H), 7.43 (dd, J = 8.4 Hz, J = 1.1 Hz, 1H), 7.27 (td, J = 7.5 Hz, J = 1.3 Hz, 1H), 6.86 (s, 1H). Example 66: 6-bromo-2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4-one oxime was prepared in 34% overall yield using methods A and D, starting from 5'- bromo-2!-hydroxy-acetopbenone and pyrrolo[1 ,2-c]pyrimidine-3-carboxylic acid methyl ester. The title compound was isolated as a yellow solid and a 80/20 mixture of Z/E oxime isomers after Preparative HPLC purification (gradient 45-30% Water/acetonitrsle÷0.05% trifluoroacetic acid).
Figure imgf000098_0001
HPLC (gradient 5 % - 60 % ACN / H20 + 0.1 % TFA): > 95%; RT = 4.52 min. (Z isomer); 3.95 min. (E isomer).
MS (ESI+): 357.9 [Ο16Η10ΒΓΝ3Ο2+Η]+ (m/z).
1 H NMR of the major Z isomer: (400Mz) DMSO-d6 δ (ppm): 1 1.23 (s, 1 H), 9.27 (s, 1 H), 8.12 (s, 1 H), 8.00 (d, J = 2.4 Hz, 1 H), 7.88 (dT J = 2.4 Hz, 1 H), 7,75 (dd, J = 9.2 Hz, J = 2.4 Hz, 1 H), 7.54-7.48 (m, 2H), 7.07-7.00 (m, 1 H), 6.80 (d, J = 3.6 Hz, 1 H).
Example 67: 6-methoxyethoxy-2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4- one oxime was prepared in 23% overall yield using the methods described in examples 41 and 43, starting from 6-bromo-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4- one O-ferf-butyl oxime (tert-butyl protected oxime of example 66). The title compound was isolated as an orange solid.
Figure imgf000098_0002
p: 212-215°C.
MS (ESI+): 352.1 [C19H17N304+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 S (ppm): 10.93 (s, 1 H), 9.21 (s, 1 H), 8.03 (s, 1 H), 7.81 (d, J = 2.5 Hz, 1 H), 7.43 (s, 1 H), 7.40 (d, J = 9.0 Hz, 1 H), 7.30 (d, J = 3.0 Hz, 1 H), 7.13 {dd, , J = 9.2 Hz, , J = 3.2 Hz, 1 H), 6.98 (dd, J = 3.8 Hz, , J = 2.8 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.13 (m, 2H), 3.67 (m, 2H), 3.39 (s, 3H). Example 68: 2-(1-B enzyl- 1 H-imidazo[ 4, 5-c]pyridin-6-yl)-chrom en-4-one oxime was prepared in 20% overall yield using methods A and D, starting from 2'- hydroxy-acetophenone and 1-Benzyl-1 H-imidazo[4,5-c]pyridine-6-carboxylic acid methyl ester (example 68A). The title compound was isolated as a ye!low solid and a 90/10 mixture of Z E oxime isomers.
Figure imgf000099_0001
p: 148-150°C.
MS (ESI+): 369.2 [C22HieN402+H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) D SO-d6 δ (ppm): 10.94 (s, 1 H), 9.03 (s, 1 H), 8.74 (s, 1 H), 8.34 (s, 1 H), 7.89 (d, J = 7.4 Hz, 1 H), 7.59 (s, 1 H), 7.55- 7.50 (m, 2H), 7.45-7.27 (m, 6H), 5.66 (s, 2H).
Example 68A: 1-Benzyl-1H-imidazo[4,5-c]pyridin-6-carboxylic acid methyl ester
Figure imgf000099_0002
A solution of H-His-(Bzl)-OH (2.0 g, 8.16 mmol) and formaldehyde (37% in water, 0.92 ml, 12.24 mmol) in water (16 ml) was heated to reflux for 4 hours and concentrated to dryness. The residue was dissolved in methanol (4.6 ml) cooled to 0°C and treated with thionyl chloride (0.6 ml, 8.12 mmol). The reaction mixture was reflux for 2.5 hours and concentrated. The solid was dissolved in dichloromethane, washed with a saturated solution of sodium hydrogenocarbonate, brine, dried over sodium sulfate and concentrated to dryness . to yield 1 -Benzyl-4,5,6,7-tetrahydro-1 H- imidazo[4,5-c]pyridine-6-carboxyfic acid methyl ester (1.58 g, 71%). The ester was dissolved in tetrahydrofuran (26 mi), cooled to 0°C and treated with 2,3-Dichloro~5,6- dicyanobenzoquinone (2.9 g, 12.81 mmol) in tetrahydrofuran (26 ml) and the reaction mixture was reflux for 4 hours. The solvent was removed and the black residue was dissolved in dichioromethane and treated with a 1 M aqueous solution of sodium hydroxide. The aqueous solution was extracted several times with dichioromethane. The combined organic extracts were washed with brine, dried over sodium sulfate, concentrated and purified by flash chromatography over silica gel (gradient ethyl acetate/methanoi: 0-10%) to yield 1 -Benzyl-1 H-imidazo[4,5-c]pyridin-6-carboxy!ic acid methyl ester (525 mg, 34%) as a beige solid.
1 H N R: (300 MHz) CHCIs-di δ (ppm): 8.77 (s, 1 H), 8.62 (s, 1 H), 8.15 (s, 1 H), 7.42-7.35 (m, 3H), 7.28-7.21 (m, 2H), 5.48 (s, 2H), 4,02 (s, 3H).
Example 69: 2-Thieno[2,3-c]pyridin-5-yl-chromen-4-one ox/me was prepared in 21 % overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Thieno[2,3-c]pyridin-5-carboxylic acid methyl ester (J. Med. Chem. 2006, 49, 4425-4436). The title compound was isolated as a beige solid and a 95/5 mixture of Z/E oxime isomers.
Figure imgf000100_0001
Mp: 243-245X.
MS (ESI+): 295.0 [C15H10N2O2S÷H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.04 (s, 1 H), 9.38 (s, 1 H), 8.52 (s, 1 H), 8.24 (d, J = 5.3 Hz, 1 H), 7.92 (dd, J = 7.9 Hz, J = 1.4 Hz, 1 H), 7.72 (d, J = 4.9 Hz, ' l H), 7.71 (s, 1 H), 7.58-7.45 (m, 2H), 7.30 (t, J = 7.4 Hz, 1 H).
Example 70: 2-Thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime was prepared in 43% overall yield using methods A and D, starting from 2'-hydroxy- acetophenone and Thieno[3,2-c]pyridin-6-carboxylic acid methyl ester (J. Med. Chem. 2006, 49, 4425-4436). The title compound was isolated as a yellow solid.
Figure imgf000100_0002
Mp: 272-275°C.
MS (ESI+): 295.0 [C16H10N2O2S+Hf (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.05 (s, 1 H), 9.25 (s, 1 H), 8.76 (s, 1 H), 8.03 (d, J = 5.3 Hz, 1 H), 7.91 (dd, J = 7.9 Hz, J = 1.3 Hz, 1 H), 7.72 (d, J = 4.8 Hz, 1 H), 7.71 (s, 1 H), 7.58-7.46 (m, 2H), 7.30 (t, J = 7.5 Hz, 1 H).
Example 71: 2-lsoquinolin-3-yl-3-methyl-chromen-4-one oxime
Figure imgf000101_0001
To a suspension of 2-lsoquinolin-3-yl-3-methyt-chromen-4-one (example 71 A) (200 mg, 0.69 mmol) in anhydrous pyridine (5 mi) was added hydroxyiamine hydrochloride (96 mg, 1.39 mmol). The mixture was heated to 130°C under microwave irradiation for 30 min. The reaction mixture was poured into a 1 N aqueous solution of hydrochloric acid and extracted with ethyl acetate. The organic layer was washed successively with a 1 N aqueous solution of hydrochloric acid, brine and dried over sodium sulfate and concentrated. The title compound was isolated as a white solid after Preparative HPLC purification (gradient 45-30% Water/Methanol +0.05% trifluoroacetic acid) (9 mg, 4%).
HPLC (gradient 85 % - 25 % H20/MeOH + 0.05% TFA (in 12 min.): > 95%; RT = 12.00 min.
MS (ESI+): 303.1 [C19H14N202+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.13 (s, 1 H), 9.49 (s, 1 H), 8.44 (s, 1 H), 8.22 (d, J = 8.4 Hz, 1 H), 8.16 (d, J = 8.4 Hz, 1 H), 7.88 (t, J = 7.2 Hz, 1 H), 7.79 (t, J = 7.2 Hz, 1 H), 7.42 (d, J = 7.6 Hz, 1 H), 7.38 (d, J = 7.6 Hz, 1 H), 7.05 (d, J = 8.0 Hz, 1 H), 6.98 (t, J = 7.6 Hz, 1 H), 2.27 (s, 3H).
Example 71A: 2-lso quin olin-3-yl-3-m ethyl-chromen-4-on e was prepared in 76% yield using method A, starting from 2!-hydroxy-propiophenone and methyl isoquino!ine-3-carboxylate.
Figure imgf000102_0001
1 H N R: (300 MHz) CHC -d, δ (ppm): 9.40 (s, 1 H), 8.29 (dd, J = 7.9 Hz, J = 1.3 Hz, 1 H), 8.18 (s, 1 H), 8.09 (d, J = 8.3 Hz, 1 H), 7.98 (d, J = 8.2 Hz, 1 H), 7.84-7.65 (multipieis, 3H), 7.55 (dd, J = 8.5 Hz, J = 0.6 Hz, 1 H), 7.42 (td, J = 7.6 Hz, J = 1 .1 Hz, 1 H), 2.37 (s, 3H).
Example 72: 3-{4-[(E)-Hydroxyimino]-4H-chromen-2-yl}-2H-isoquinolin-1- one was prepared in 9% overall yield using methods A and D, starting from 2'- hydroxy-acetophenone and 1-oxo-1 ,2-dihydro-isoquinotine-3-carboxylic acid methyl ester. The title compound was isolated as a pale yellow solid and a 90/10 mixture of Z/E oxime isomers.
Figure imgf000102_0002
Mp: 310-3 5°C dec.
MS (ESI+): 305.3 [CieH12N203+H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 1 1.66 (br. s, 1 H), 1 1.84 (s, H), 2.23 (d, J = 7.9 Hz, 1 H), 7.90-7.74 (m, 3H), 7.60-7.45 (m, 3H), 7.46 (S, 1 H), 7.39 (s, 1 H), 7.29 (t, J = 7.3 Hz, 1 H).
Example 73: 2-lmidazo[1,2-c]pyrimidin-7-yl-chromen-4-one oxime was prepared in 0.3% overall yield using methods A and C, starting from 2'-hydroxy- acetophenone and !midazo 1 ,2-c]pynrnsdine-7-carboxyi!c acid ethyl ester (US2003/236264). The title compound was isolated as a yellow solid and a 60/40 mixture of Z E oxime isomers after purification by silica gel thin layer chromatography (dichloromethane/methanol : 96/4).
Figure imgf000103_0001
MS (ES1+): 279,1 [C^H^C H]* (m/z).
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): > 95%; RT = 4.44 min. (E isomer) and 4.59 min. (Z isomer).
Example 74 (Reference): 2-Naphthalen-2-yl-chromen-4-one oxime was isolated in 69% yield using method D starting from 2-Naphthalen-2-yl-chromen-4-one (compound 74A) as a yellow solid and a 95/5 mixture of Z/E oxime isomers.
Figure imgf000103_0002
Mp: 224-226°C
MS (ESI+): 288.0 [C19H13N02+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.06 (s, 1 H), 8.56 (s, 1 H), 8.14-8 06 (m, 1 H), 8.04-7.95 (m, 3H), 7.92 (dd, J = 8.1 Hz, J = 1.3 Hz, 1 H), 7.64-7.56 (m, 2H), 7.55-7.47 (m, 2H), 7.34-7.26 (m, 1 H), 7.28 (s, 1 H).
Compound 74A: 2-Naphthalen-2-yl-cbromen-4-one.
Figure imgf000103_0003
Method B : To a cold suspension of 2-naphtoic acid (2.0 g, 1 1.6 mmol) in dtchloromethane (60 ml) were added oxalyl chloride (1.12 ml, 12.7 mmol) and then dimethySformamide (10 drops). The mixture was stirred at room temperature for 2 hours and concentrated to dryness to give the crude 2-naphtoic acid chloride (2.5 g). The crude product was dissolved in dry pyridine (50 ml) under argon, cooled to 0°C and treated with 2'-hydroxy-acetophenone (1.43 g, 10.5 mmol). The reaction mixture was heated to 60°C for 2 hours and poured into ice-cold water (150 ml). The solution was acidified to pH 1 with concentrated hydrochloric acid and the precipitate isolated by filtration, washed with water to yield Naphthalene-2-carboxylic acid 2'-acetyl-phenyl ester (2.7 g, 80%). The ester was dissolved in dimeihylsulfoxide (30 ml) under argon and treated with freshly crushed potassium hydroxide (1 .4 g, 25.8 mmol) and stirred at room temperature for 14 hours. The mixture was poured into ice-cold water and acidified to pH 3-4 with a 6N aqueous hydrochloric acid solution. The solid was filtrated, washed with water, dried under vacuum to yield 1 -(2-Hydroxy-phenyl)-3- naphthalen-2-y!-propane-1 ,3-dione (1 .44 g, 86%). The diketone was dissolved in dimethylsulfoxide (25 ml), treated with para-toiuenesulfontc acid monohydrate (660 mg, 3.47 mmol) and heated to 90°C for 4 hours. The reaction mixture was poured into ice-cold water and filtrated. The solid was dissolved in dich!oromethane, dried over sodium sulfate and concentrated. The residue was purified by silica gel flash chromatography (cyclohexane/ethyl acetate: 80/20) to yield 2-Naphthalen-2-yl- chromen-4-one (956 mg, 70%) as a brown solid.
1 H NMR: (300 MHz) DMSO-d5 δ (ppm): 8.50 (s, 1 H), 8.26 (dd, J = 8.1 Hz, J = 1 .3 Hz, 1 H), 8.02-7.87 (m, 4H), 7.74 (td, J = 7.8 Hz, J = 1.7 Hz, 1 H), 7.65 (dd, J = 8.2 Hz, J = 1 .0 Hz, 1 H), 7.62-7.55 (m, 2H), 7.45 (td, J = 8.1 Hz, J = 1 .1 Hz, 1 H), 6.99 (s, 1 H).
Example 75: 2-(1H-Pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxime
Figure imgf000104_0001
Mp: 270-275°C dec.
MS (ESl+): 278.1 [deHnNsOj+Hnm/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1 .89 (br. S, 1 H), 10.83 (s, 1 H), 8.85 (s, 1 H), 8.26 (s, 1 H), 7.90 (d, J = 7.9 Hz, 1 H), 7.70 (s, 1 H), 7.60 (s, 1 H), 7.52-7.46 (m, 2H), 7.28 (t, J = 7.1 Hz, 1 H), 6.67 (s, 1 H).
Example 76: 2-(1-Hydroxy-1H-pyrrolo(2,3-c]pyridin-5-yl)-chromen-4-one oxime
Figure imgf000105_0001
1 H NMR: (400 MHz) DMSO-d@ δ (ppm): 1 1.99 (br. S, 1 H), 10.88 (s, 1 H), 8.89 (s, 1 H), 8.28 (s, 1 H), 7.91 (d, J = 7.9 Hz, 1 H), 7.83 (s, 1 H), 7.60 (s, 1 H), 7.56-7.46 (m, 2H), 7.29 (t, J = 7.4 Ηζ,Ί Η), 6.58 (t, J = 2.4 Hz, 1 H).
Examples 75 and 76 were prepared simultaneously by the following procedure:
To a suspension of 2-(4-Methyl-5-nitro-pyridin-2-yl)-chromen-4-one O-tert- buiyl-oxime (example 75A) (250 mg, 0.70 mmol) in dimethyiformamide (6 ml) under argon was added dimethyfformamide-dimethylacetal (127μΙ, 0.95 mmo!) and the mixture was stirred at 90°C for 2.5 hours. The solvents were removed under vacuum and the residue was dissolved in absolute ethanol. 10% palladium black was added (50 mg) and the mixture was stirred under 1 atmosphere of hydrogen at room temperature for 16 hours. The catalyst was removed by filtration ant the filtrate purified by silica gei chromatography (cyciohexane/ethy! acetate 0-20%) to give a 70/30 mixture of 2-(1 H-Pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one O-tert-butyl-oxsme and 2- (1-Hydroxy-1 H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one 0-tert-butyl-oxime (165 mg, 70%). The former mixture tert-butyl protected oximes (70 mg) was treated with titanium tetrachloride (0.63 mmol) in dichioromethane (6 ml) (method D, step 2) to yield separately after purification by flash chromatography (dich!oromethane/ethyl acetate: gradient 50-100%) and preparative HPLC (gradient 70-55% Water/Methano! +0.05% trifluoroacetic acid) 2-(1 H-Pyrroio[2,3-c]pyridin-5-yi)-chromen-4-one oxime (example 75; 39 mg, 70%) and 2-(1-Hydroxy-1 H-Pyrroio[2,3-c]pyridin~5-yl)-chromen- 4-one oxime (example 77; 5 mg, 8%).
Example 75A: 2~(4~Methyl~5~nitro^yridin-2-yl)~chromen-4-one O-tert- butyl-oxime
Figure imgf000106_0001
A solution of 2'-hydroxyacetophenone (1.68 g, 12.39 mrrtol) in tetrahydrofuran (120 mi) under argon was cooled to -78°C and treated dropwise with lithium hexamethyldisilazane (1 M in tetrahydrofuran, 2.25 ml, 2.25 mmol). The solution was stirred at -78°C for 1 hour and at -10°C for 2 hours then cooled down to -78°C and treated dropwise with a solution of 4- efhy!-5-nitro-pyridine-2-carboxylic acid methyl ester (WO2005/103003) (2.42 g, 12.39 mmo!) in tetrahydrofuran (60 ml). The dark red solution was stirred at -78°C for 1 hour then at room temperature for 18 hours. The mixture was poured into a ice-cold 1 N solution of hydrochloric acid (200 ml) and extracted several times with ethyl acetate. The combined extracts were dried over sodium sulfate and concentrated to dryness. The residue was dissolved in acetic acid (60 ml), treated with sulfuric acid (0.33 ml) and heated to 100X for 30 minutes. After cooling to room temperature, the solution was concentrated and the residue added with water and neutralized with a saturated solution of sodium hydrogenocarbonate. The precipitate was filtrated, washed with water and dried under vacuum to yield 2-(4- Methyl-5-nitro-pyridin-2-yl)-chromen-4-one (2.33 g, 66%) as a brown solid. The previous chromen-4-none (770 mg, 2.72 mmol) was treated with tert-buty!- hydroxylamine hydrochloride (685 mg, 5.45 mmol) in methanol (20 ml) at 130°C for 30 minutes under microwave irradiation (method D, step 1) to yield the title compound (394 mg, 41 %) after purification by silica ge! flash chromatography (gradient cyciohexane/dichloromethane 0-80%) as a gold solid.
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 9.23 (s, 1 H), 8,08 (dd, J = 7.9 Hz, J = 1.7 Hz, 1 H), 7.89 (s, 1 H), 7.78 (s, 1 H), 7.42 (td, J = 7.6 Hz, J = 1.7 Hz, 1 H), 7.32-7.19 (m, 2H), 2.76 (s, 3H), 1.42 (s, 9H).
Example 77: 2-(1-Methyl-1H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxime
Figure imgf000107_0001
Mp: 270-275X dec.
MS (ESI+): 292.1 [C17H13N302+H]+ (m/z).
H NMR: (400 MHz) DMSO-d6 δ (ppm): 1 1.00 (br. s, 1 H), 9.05 (s, 1 H), 8.39 (s, 1 H), 7.90 (d, J = 8.0 Hz, 1 H), 7.83 (s, 1 H), 7.61 (s, 1 H), 7.57-7.47 (m, 2H), 7.30 (t, J = 7.4 Hz, 1 H), 6.76 (t, J = 2.8 Hz, 1 H), 4.02 (s, 3H).
Example 78: 2-(1-Methoxy-1H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxtme
Figure imgf000107_0002
MS (ESI+): 308.0 [C17H13N303+Hf (m/z).
1 H NMR: (400 MHz) DMSOd6 δ (ppm): 10.92 (s, 1 H), 9.00 (s, 1 H), 8.30 (s, 1 H), 8.05 (s, 1 H), 7.91 (d, J = 7.9 Hz, 1 H), 7.62 (s, 1 H), 7.56-7.46 (m, 2H), 7.29 (td, J = 7.4 Hz, J = 1.2 Hz , 1 H), 6.58 (d, J = 3.2 Hz, 1 H), 4.21 (s, 3H).
Examples 77 and 78 were prepared simultaneously by the following procedure:
A 70/30 mixture of 2-(1 H-Pyrroio[2,3-cjpyridin-5-yl)-chrornen-4-one O-tert- butyl-oxime and 2-(1-Hydroxy-1 H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one O-tert- butyl-oxime (77 mg) in dimethyiformamide (5 ml) was cooled to 0°C and treated with sodium hydride (60% in mineral oil, 10 mg, 0.25 mmol) and stirred at room temperature for 1 hour. The solution was cooled down to 0°C and treated with iodomethane (16 μΙ, 0.25 mmol). The solution was slowly warmed up to room temperature during 1 hour and poured into brine, extracted with ethyl acetate, washed with brine, dried over sodium sulfate and concentrated to dryness. The crude mixture of 2-(1 -methyl-1 H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one 0 ert-butyi-oxime and 2- (1 -methoxy-1 H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one O-tert-butyl-oxime was treated with titanium tetrachloride (0.70 mmoi) in dichioromethane (7.5 ml) (method D) to yield separately after purification by preparative HPLC (gradient 65-50% Water/ ethanoi +0.05% trifluoroacetic acid), 2-(1-methyl-1 H-pyrrolo[2,3-c]pyridin-5- yl)-chromen-4-one oxime (example 77, 17 mg) and 2-(1-methoxy-1 H-pyrrolo[2,3- c]pyridin-5-yl)-chromen-4-one oxime (example 78, 12 mg) as yellow solids.
Example 79 (Reference): 2-Quinolin~2~yl~chromen-4~one oxime was prepared in 24% overall yield using method B and D as described in compound A, starting from quinaldyl chloride. The title compound was isolated as a yellow solid,
Figure imgf000108_0001
Mp: 232-235
MS (ESI+): 289.0 [C1SH12N202+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-de δ (ppm): 11.20 (s, 1 H), 8.59 (d, J = 8.5, 1 H), 8.19 (d, J = 8.7, 1 H), 8.14 (d, J = 8.5, 1 H), 8.07 (d, J = 8.5, 1 H), 7.93 (d, J = 8.3, 1 H), 7.85 (td, J = 7.6 Hz, J = 1.3 Hz, 1 H), 7.80 (s, 1 H), 7.68 (td, J = 7.9 Hz, J = 1.2 Hz, 1 H), 7.60-7.49 (m, 2H), 7.31 (td, J = 7.1 Hz, J = 1.4 Hz, 1 H).
Example 80 (Reference): 2-Pyrimidin-4-yl-chromen-4-one oxime was prepared in 61 % overall yield using method A and D, starting from 2'- hydroxyacetophenone and pyrimidine-4-carboxylic acid methy! ester. The title compound was isolated as a yellow solid.
Figure imgf000109_0001
Mp: 208-211 °C
S(ESI+): 240.1 [C13H9N302+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.37 (br. s, 1H), 9.31 (d, J = 1.3, H), 9.04 (d, J = 5.3, 1H), 8,05 (dd, J = 5.3, J = 8.7, 1H), 7.90 (dd, J = 8.1, J = 1.5, 1H), 7.75 (s, 1H), 7.54 (td, J = 7.7 Hz, J = 1.3 Hz, 1H), 7.47 (dd, J = 8.3 Hz, J = 1.2 Hz, 1H), 7.31 (td, J = 7.4 Hz, J = 1.3 Hz, 1H).
Example 81: 6-Hydroxy-2~pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4-one oxime was prepared using method D (step 2) starting from 6-Hydroxy-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 81A) and isolated as a yellow powder.
Figure imgf000109_0002
" Mp: 263-235°C.
Figure imgf000109_0003
1H NMR: (300 MHz) DMSO-d65 (ppm): 9.51 (s, 1H), 9.20 (s, 1H), 8.00 (s, 1H), 7.80 (d, J = 2.5 Hz, 1H), 7.40 (s, 1H), 7.29 (d, J = 8.9 Hz, 1H), 7.23 (d, J = 2.9 Hz, 1H), 6.97 (dd, , J = 3.7 Hz, , J = 2.8 Hz, 1H), 6.90 (dd, J = 8.9 Hz, , J = 3.0 Hz, 1H), 6.72 (d, J = 3.8 Hz, 1H).
Example 81 A: 6-Hydroxy-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen'4-one O- tert-butyl-oxime was prepared in 59% yield using the procedure described in example 18A, starting from 6-bromo-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one O- ferf-butyl oxime (tert-butyl protected oxime of example 66). The title compound was isolated as a yellow solid.
Figure imgf000110_0001
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 8.79 (s, 1 H), 7.90 (s, 1 H), 7.52 (d, J = 3.0 Hz, 1 H), 7.49 (s, 1 H), 7.44 (d, J = 2.9 Hz, 1 H), 7.17 (d, J = 8.8 Hz, 1 H), 6.95-6.88 (m, 2H), 6.63 (d, J = 3.8 Hz, 1 H), 5.56 (br s., 1 H), 1 .41 (s, 9H).
Example 82: 2-Thiazolo[5,4-c]pyridin-6-yl-chromen-4-one oxime was prepared in 0.2% overall yield using the method described in example 75A, starting from Thiazolo[5,4-c]pyridine-6-carboxylic acid ethyl ester (US6342606).
Figure imgf000110_0002
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH); > 95%; RT = 5.1 1 mm.
MS (ESI+): 296.1 [C15H9N302S÷H]+ (m/z).
Example 83: 6-(3-Methoxy-propyl)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime was prepared in 32% yield using method D (step 2) starting from 6-(3-Methoxy-propyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyl- oxime (example 83A) and isolated as a ye!low solid and a 95/5 mixture of Z/E isomers.
Figure imgf000110_0003
Mp: 172-173°C.
MS (ESI+): 350.2 [Ο20ΗΝ3θ3+Η]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 10.93 (s, 1 H), 9.22 (s, 1 H), 8.03 (s, 1 H), 7.81 (d, J = 2.4 Hz, 1 H), 7.68 (s, 1 H), 7.45 (s, 1 H), 7.36 (s, 2H), 6.98 (m, 1 H), 6.74 (d, J = 3.8 Hz, 1 H), 3.35-3.25 (m, 2H), 3.23 (s, 3H), 2.66 (m, 2H), 1.80 (m, 2H).
Example 83 A: 6-( 3-Methoxy-propyl)-2-pyrrolo[1, 2-c]py midin- chromen-4-one O-tert-butyl-oxime
N(O-tButyt)
Figure imgf000111_0001
A solution of 6-(3-Methoxy-prop-1 -ynyi)-2-pyrroio[1 ,2-c3pyrimidin-3-yl-chrornen~ 4-one O-tert-butyi-oxime (example 83B) (30 mg, 0.075 mmol) and 10 % platinum black (10 mg) in ethyi acetate (10 ml) was hydrogenated at room temperature under 1 atmosphere of hydrogen during 48 hours. The catalyst was removed by fiitration over celite® and the filtrates concentrated to dryness to yield the title compound (18 mg, 60%) as a yellow solid that was used for the next step without further purification.
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): > 80%; RT = 6.47 min.
MS (ESI+): 405.5 [C24H27N3O3+H]* (m/z).
Example 83 B : 6~( 3-Meth oxy-prop- 1 ~ynyl)-2-pyrrolo[ 1, 2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 39% yield using the procedure described in example 23A, starting from 6-bromo-2-pyrrolo[1 ,2-cjpyrimidin-3-yl- chromen-4-one O-ferf-butyl oxime (tert-butyl protected oxime of example 66) and 3- Methoxy-propyne. The title compound was isolated as a yellow solid.
Figure imgf000111_0002
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 8.84 (s, 1 H), 8.30 (d, J
7.93 (s, 1 H), 7.55 (s, 1 H), 7.56-7.46 (m, 2H), 7.24 (m, 1 H), 6.96 (t, J
6.68 (d, J = 3.8 Hz, 1 H), 4.35 (s, 2H), 3.48 (s, 3H), 1.43 (s, 9H). Example 84: 6-(3-Dimethylamino-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime hydrochloride
6-(3-Dsmethyiamino-propoxy)-2-pyrroto[
oxime was prepared in 22% overall yield using the procedure described in example 42A, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-fert-butyl oxime (example 81A) and (3-Chioro-propyl)-dimethy!-amine hydrochloride. The compound was treated with a 1 .25 N solution of hydrogen chloride in isopropanol to yield 6-(3-Dimethylamino-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime hydrochloride as an orange solid.
Figure imgf000112_0001
Mp: > 270°C dec.
MS (ES1+): 793.2 [C21H22N403+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.93 (br. s, 1 H), 10.38 (br. s, 1 H), 9.22 (s, 1 H), 8.03 (s, 1 H), 7.82 (s, 1 H), 7.43 (s, 1 H), 7.42 (d, J = 8.3 Hz, 1 H), 7.33 (d, J = 3.0 Hz, 1 H), 7.14 (d, J = 3.0 Hz, 1 H), 7.1 1 (d, J = 3.0 Hz, 1 H), 6.98 (d, J = 3.7 Hz, 1 H), 4.10 (t, J = 5.9 Hz, 2H), 3.18 (m, 2H), 2.79, 2.77 (2s, 6H), 2.27 (m, 2H).
Example 85: 6-(3-Morpholin-4-yt-ethoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime hydrochloride was prepared in 65% overall yield using method D (step 2), starting from 6-(3-Morpholin-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin- 3-yl-chromen~4-one O-terf-butyl oxime (example 85A). The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanol.
Figure imgf000112_0002
Mp: > 270°C dec.
MS (ESI+): 407.3 [C22H22 404+H]+ (m/z). 1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.99 (br. s, 1 H), 10.67 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (d, J = 2.6 Hz, 1 H), 7.46 (d, J = 9.2 Hz, 1 H), 7.44 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.98 (dd, J = 3.6 Hz, J = 2,8 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.45 (m, 2H), 3.98 (br. d, J = 1 1 .1 Hz, 2H),3.90-3.70 (m, 2H), 3.65-345 (m, 4H), 3.30-3.12 (m, 2H).
Example 85 A: 6-(3-Morpholin-4-yl-ethoxy)-2-pyrrolo[1, 2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime
Figure imgf000113_0001
A mixture of 6-Hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert- butyl-oxime (example 81 A) (100 mg, 0.286 mmol) ,4-(2-chioroethyl)morpholine hydrochloride (80 mg, 0.430 mmol) and potassium carbonate (1 19 mg, 0.858 mmol) in dry acetone (2.5 ml) was stirred at 60°C for 20 hours. The solvent was removed under vacuum. The residue was treated with water and extracted with ethyi acetate. The combined organic layers was washed with brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by silica gel flash chromatography (gradient cyclohexane/ethyl acetate: 20-100%) to yield the title compound (89 mg, 67%) as a yellow solid.
1 H NMR: (300 MHz) CHCl3-d1 δ (ppm): 8.79 (m, 1 H), 7.90 (s, H), 7.51 (d, J = 3.0 Hz, 1 H), 7.49 (s, 1 H), 7.45 (d, J = 2.5 Hz, 1 H), 7.20 (d, J = 9.1 Hz, 1 H), 6.99 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.92 (dd, J = 3.8 Hz, J = 2.8 Hz, 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 4.21 (br. m, 2H), 3.79 (br. m, 4H), 2.89 (br. m, 2H), 2.63 (br. m, 4H), 1.41 (s, 9H).
Example 86: 6-(2,3-Dihydroxy-propoxy}-2-pyrrolo[ 1,2-c]pyrimidin~3~yl- chromen-4-οηβ oxime
A solution of 6-(2,3-Dihydroxy-propoxy)-2-pyrrolot1 ,2-c]pyrimidin-3-yl-chromen- 4-one O-ferf-butyl oxime (example 86A) (100 mg, 0.236 mmol) in toluene (8 ml) was treated at 0°C with trifluoro acetic acid (2 ml) and heated at 60°C for 5.5 hours. The soiution was concentrated under vacuum, the residue dissolved in ethyi acetate and washed successively with a saturated solution of sodium hydrogenocarbonate, brine, dried over sodium sulfate and concentrated. The residue was purified by silica gel flash chromatography (gradient cyclohexane/ethyl acetate: 0-100% then ethyi acetate/methanol: 90/10) to yield the title compound (38 mg, 44%) as a brown-yellow solid.
Figure imgf000114_0001
MS (ESI+): 368.1 [C1SH17N305+H]+ (m/z).
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 10.92 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (d, J = 2.4 Hz, 1 H), 7.44 (s, 1 H), 7.41 (d, J = 9.2 Hz, 1 H), 7.34 (s, 1 H), 7.13 (dd, J = 9.2 Hz, J = 2.3 Hz, 1 H), 6.99 (dd, J = 3.6 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 4.4 Hz, 1 H), 4.05 (m, 2H), 3.89 (m, 2H), 3.81 (m, 1 H).
Example 86 A: 6-(2,3-Dihydroxy-propoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime was prepared in 74% yield using the method described in example 85A, starting from 6-Hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (example 81 A) and 3-chloro-1 ,2-propandiol. The title compound was isolated as a yellow solid.
Figure imgf000114_0002
1 H NMR: (300 MHz) CHCI^ δ (ppm): 8.81 (s, 1 H), 7.96 (s, 1 H), 7.57 (m, 1 H), 7.47 (s, 1 H), 7.29-7.15 (m, 2H), 7.06 (m, 1 H), 6.94 (m, 1 H), 6.67 (d, J = 3.6 Hz, 1 H), 4.30-4.00 (m, 3H), 3.95-3.60 (m, 2H), 1.45 (s, 9H).
Example 87: 6-(2-Pyrrolidin-1-yl-ethoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 37% yield using method D (step 2), starting from 6-(2-Pyrrolidin-1 -yl-ethoxy)-2-pyrroio[1 ,2-c]pyrimidtn-3-yl- chromen-4-one O-tert-butyi oxime (example 87A). The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1 .25 M solution of hydrogen chloride in isopropanol.
Figure imgf000115_0001
Mp: > 260°C dec.
MS (ESI+): 391.2 [C22H22N403+H]+ (m/z).
1H NMR: (300 MHz) DMSO-ds δ (ppm): 10.94 (br. s, 1 H), 10.16 (br. s, 1H), 9.22 (s, 1H), 8.04 (s, 1H), 7.82 (s, 1 H), 7.46 (d, J = 9.2 Hz, 1H), 7.44 (s, 1 H), 7.38 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.6 Hz, J = 2.8 Hz, 1H), 6.74 (d, J = 3.6 Hz, 1 H), 4.37 (m, 2H), 3.50 (m, 4H), 3.15 (m, 2H), 2.03 (m, 2H), 1.90 (m, 2H).
Example 87 A : 6-(2~PyrroUdin- 1 -yl-ethoxy)-2~pyrrolo[ 1, 2-cJpyrimidin-3-yl- chromen-4-one O-tert-butyl oxime
N(0 Butyl)
Figure imgf000115_0002
A mixture of 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2-c]pyrim!din-3-yl-chromen-4-one O-tert-butyi oxime (example 87B) (64 mg, 0.16 mmol), potassium carbonate (64 mg, 0.46 mmol) and pyrrolidine (19μί, 0.24 mmol), in acetonitri!e (1.5 ml) was heated in a sealed tube at 100°C for 18 hours. The yellow suspension was filtered, the soiid washed with ethyl acetate and the combined filtrates concentrated to dryness. The residue was purified by silica gel flash chromatography (gradient ethyl acetate/methanol 0-2 %) to yield the title compound (49 mg, 71%) as a yellow solid.
1 H NMR: (300 MHz) CHC^ δ (ppm): 8.79 (s, 1 H), 7.90 (s, 1H), 7.51 (d, J = 3.0 Hz, 1 H), 7.49 (s, 1 H), 7.45 (d, J = 2.8 Hz, 1 H), 7.21 (d, J = 9.0 Hz, 1 H), 7.00 (d, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.92 (d, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.63 (d, J = 3.7 Hz, 1 H), 4.40-4.25 (br. m, 2H), 3.24-3.09 (m, 2H), 3.08-2.60 (m, 4H), 2.03-1.90 (m, 4H), 1.41 (s, 9H).
Example 87B: 6-(2-chloro-ethoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen- 4-one O-tert-butyl oxime was prepared in 60% yield using the method described in example 85A, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O- tert-butyl-oxime (example 81 A) and 1 ,2-dichforoethane in dimethyiformamide. The title compound was isolated as a yeflow solid.
Figure imgf000116_0001
1 H NMR: (300 MHz) CHCIs-di δ (ppm): 8.79 (s, 1 H), 7.89 (s, 1 H), 7.54 (d, J = 3.0 Hz, 1 H), 7.50 (s, 1 H), 7.44 (s, 1 H), 7.22 (d, J = 9.0 Hz, 1 H), 7.01 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.62 (d, J = 3.7 Hz, 1 H), 4.30 (t, J = 5.7 Hz, 2H), 3.85 (t, J = 5.7 Hz, 2H), 1.42 (s, 9H).
Example 88: 6~(2~Piperidin~1-yl'ethoxy)-2~pyrrolo[ 1 ,2~c]pyrimidm-3-yl- chromen-4-one oxime, hydrochloride was prepared in 27% overalf yield using the method described in example 85, starting from 6-hydroxy-2-pyrroio[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyl oxime (example 81A) and 1-(2-chloroethyl)-piperidine hydrochloride .
Figure imgf000116_0002
Mp: > 255°C dec.
MS (ESI+): 405.2 [C23H24N403+H]+ (m/z).
1 H NMR: (300 MHz) D SO-de δ (ppm): 11.00 (br. s, 1 H), 10.26 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.47 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.45 (m, 2H), 3.49 (m, 4H), 3.02 (m, 2H), 1.79 (m, 4H), 1.72 (m, 1 H), 1.41 (m, 1 H).
Example 89: 6-(2-dimethylamino-ethoxy)-2~pyrrolo[1,2-c]pyrimidin-3~yl- chromen-4-one oxime, hydrochloride was prepared in 39% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyl oxime (example 81 A) and 1-(2-chloroethyl)- dimethyiamine hydrochlonde.
Figure imgf000117_0001
Mp: > 275°C dec.
MS (ESI+): 365.2 [C2oH2oN403+H]+ (m/z).
1 H NMR: (300 MHz) D SO-d6 δ (ppm): 11.00 (br. s, 1 H), 10.40 (br. s, 1 H), 9.23 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.48 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.42 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J - 3.7 Hz, 1 H), 4.40 (m, 2H), 3.52 (m, 2H), 2.86 (s, 3H), 2.84 (s, 3H).
Example 90: 6-(2-diethylamino-ethoxy)-2-pyrrolo[ 1, 2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen- 4-one 0-£er£-buiyl oxime (example 87B) and 1-(2-chloroethyl)-diethylamine hydrochloride.
Figure imgf000117_0002
MS (ESI+): 393.2 [C22H24N403+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.94 (br. s, 1 H), 8.80 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.83 (s, 1 H), 7.47 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.7 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.40 (m, 2H), 3.50 (m, 2H), 3.23 (m, 4H),- 1.26 {m, 6H).
Example 91: 2-Pyrrolo[1,2-a]pyrazin-3-yl-chromen-4-one oxime was prepared in 41 % yield using methods A and D, starting from 2'-hydroxyacetophenone and Pyrrolo[1 ,2-a]pyrazine-3-carboxyiic acid methyl ester (example 91 A). The title compound was isolated as a pale yellow solid.
Figure imgf000118_0001
Mp: > 276-277°C.
MS (ESI+): 278.0 [C16HnN302+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.96 (s, 1 H), 8.97 (s, 1 H), 8.93 (s, 1 H), 7.92 (s, 1 H), 7.90 (dd, J = 7.9 Hz, J = 1.5 Hz, 1 H), 7.53 (td, J = 7.8 Hz, J = 1.3 Hz, 1 H), 7.48 (s, 1 H), 7.40 (d, J = 8.3 Hz, 1 H), 7.28 (t, J = 7.5 Hz, 1 H), 7.00 (dd, J = 3.9 Hz, J = 2.4 Hz, 1 H), 6.96 (d, J = 3.9 Hz, 1 H).
Example 91 A: Pyrrolo[1,2-a]pyrazine-3-carboxyiic acid methyl ester
Figure imgf000118_0002
A solution of di-(2-tert-Butoxycarbonyl)-amino-3-{2-formyl-pyrrol-1-yl)-propionic acid methyl ester (J. Chem. Soc, Perkin Trans 1 , 2000, 3317-3324) (500 mg, 1.26 mmoi) in trifiuoroacetic acid (4 mi) was stirred at room temperature for 1 hour. The reaction mixture was cautiously poured into a cold saturated aqueous solution of sodium hydrogenocarbonate and extracted with ethyl acetate. The extracts were washed with brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by silica gel flash chromatography (gradient cyclohexane/ethy! acetate: 0-100%) to yield 3,4-Dihydro-pyrrolo[1 ,2-a]pyrazine-3-carboxylic acid methyl ester (169 mg, 75%) as an orange oii. This oil was dissolved in dichloromethane and treated with manganese dioxide (800 mg, 9.2 mmol) at 40°C for 1 hour. The black solids were removed by filtration over celite® and washed abundantly with dichloromethane. The filtrate was concentrated to dryness to yield Pyrrolo[1 ,2- a]pyrazine-3-carboxylic acid methyl ester ( 30 mg, 80%) as a yellow solid.
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 8.85 (s, 1 H), 8.78 (s, 1 H), 7.56 (d, J = 2.5 Hz, 1 H), 7.02 (dd, J = 4.1 Hz, J = 2.6 Hz, 1 H), 6.91 (d, J = 3.9 Hz, 1 H), 4.00 (s, 3H).
Example 92: 6~[2-(2-methyl-pyrrolidin-1-yl)-ethoxy]-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 22 % overall yield using the method described in example 87, starting from 6-(2-chioro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 2-methylpyrrolidine.
Figure imgf000119_0001
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.98 (br. s, 1 H), 10.19 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.47 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.40 (m, 2H), 3.80-3.60 (m, 2H), 3.60-3.30 (m, 2H), 3.30-3.10 (m, 1 H), 2.30-2.15 (m, 1 H), 2.10-1.85 (m, 2H), 1.70-1.55 (m, 1 H), 1.43 (d, J = 8.9 Hz, 3H).
Example 93: 6-[2-(4-methyI-piperazin-1-yl)-ethoxy]-2-pyrroIo[ 1,2-c] pyrimidin-3-yl-chromen-4-one oxime, dihydrochlo de was prepared in 26 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrro!o[1 ,2-c]pyrimldin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 4-methyl-piperazine.
Figure imgf000119_0002
p: > 230°C dec.
MS (ESI+); 420.2 [023Η25Ν6θ3+Η; (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.56 (br. s, 1 H), 10.98 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.44 (m, 2H), 4.00-3.20 (multiplets, 10H), 2.84 (s, 3H).
Example 94: 6-[2-(4-methyl-[ 1,4Jdiazepan-1-yl)-ethoxy]-2-pyrrolo[ 1,2-c] pyrimidin-3-yl-chromen"4-one oxime, dihydrochloride was prepared in 13 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrroio[ ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 1-methy!-[1 ,4]diazepane.
Figure imgf000120_0001
Mp: > 210°C dec.
MS (ESI+): 434.3 [C24H27N603+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm); 10.90 (br. s, 1 H), 9.20 (s, 1 H), 8.03 (s, 1 H), 7.81 (s, 1 H), 7.43 (s, 1 H), 7.40 (d, J = 9.1 Hz, 1 H), 7.31 (d, J = 3.0 Hz, 1 H), 7.12 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.98 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.73 (d, J = 2.9 Hz, 1 H), 4.10 (m, 2H), 3.33-3.27 (m, 2H), 3.10-2.70 (multiplets, 11 H), 1.84 (m, 2H).
Example 95: 6-[2-((S)-2~hydroxym ethylpyrrolidin- 1-yl)-eth oxy]-2- pyrrolo[1,2-c] pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 8 % overall yieid using the method described in example 87, starting from 6-(2- chioro-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4-one O-rert-butyl oxime (example 87B) and 2-((S)-2-hydroxymethylpyrrolidine.
Figure imgf000120_0002
Mp: > 220°C dec.
MS (ESI+): 421.3 [C23H24N404+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-de δ (ppm): 10.94 (br. s, 1 H), 9.61 (br. s, 1 H), 9.22 (s, 1 H), 8.04 (s, 1 H), 7.82 (s, 1 H), 7.46 (d, J = 9.1 Hz, 1 H), 7.44 (s, 1 H), 7.38 (d, J = 3.0 Hz, 1 H), 7.19 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 2.9 Hz, 1 H), 4.40 (m, 2H), 3.85-3.60 (muitiplets, 3H), 3.60-3.40 (muitiplets, 3H), 3.34-3.19 (m, 1 H), 2.17-1.95 (m, 2H), 1.95-1.82 (m, 1 H), 1.72-1.67 (m, 1 H).
Example 96: 6-[2-((S)-2-methoxymethylpyrrolidin- 1-yl)-ethoxy]-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 40 % overall yieid using the method described in example 87, starting from 6-(2- chloro-ethoxy)-2-pyrroio[1 T2-c]pyrimidin-3-yl-chromen-4-one O-ferf-buty! oxime (example 87B) and 2-((S)-2-methoxymethylpyrrolidine.
Figure imgf000121_0001
Mp: > 225°C dec.
MS (ESI+): 435.3 [CZ4 26H404+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.99 (br. s, 1 H), 10.39 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.83 (s, 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.38 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.8 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J = 2.9 Hz, 1 H), 4.41 (m, 2H), 3.85-3.45 (muitiplets, 6H), 3.35-3.15 (m, 1 H), 3.30 (s, 3H), 2.20-1.80 (muitiplets, 3H), 1.75-1.55 (m, 1 H).
Example 97: 6-[2-((R)-2-methoxymethyipyrroiidin-1-yi)-ethoxy]-2- pyrrolof 1, 2-c]pyrimidin-3-yl"Chromen-4-one oxime, hydrochloride was prepared in 33 % overall yield using the method described in example 87, starting from 6-(2- chioro-ethoxy)-2-pyrrolo[ ,2-c]pyrimidin-3-yi-chromen-4-one O-terf-butyl oxime (example 87B) and 2-((R)-2-meihoxymethylpyrrolidine.
Figure imgf000122_0001
Mp: 2 0°Cciec.
MS (ESI+): 435.3 [C24H26N404+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.00 (br. s, 1H), 10.39 (br. s, 1H), 9.23 (s, 1H), 8.06 (s, 1H), 7.83 (s, 1H), 7.46 (d, J = 9.0 Hz, 1H), 7.45 (s, 1H), 7.40 (d, J = 3.0 Hz, 1H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.8 Hz, J = 2.9 Hz, 1H), 6.74 (d, J = 2.9 Hz, 1H), 4.41 (m, 2H), 3.85-3.45 (multiplets, 6H), 3.35-3.15 (m,. 1H), 3.30 (s, 3H), 2.22-1.80 (multiplets, 3H), 1.78-1.55 (m, 1H).
Example 98: 6-[2-(3-hydroxy-pyrrolidin- 1-yl)-ethoxy]-2-pyrroIo[ 1, 2-c] pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 23 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and 3- pyrrolidinoi.
Figure imgf000122_0002
Mp: > 230°C dec.
MS (ESI+): 407.2 [C22H22N404+H]+ (m/z).
1H NMR: (300 MHz) DMSO-ds δ (ppm): 10.96 (br. s, 1H), 10.62 (br. s, 1H), 10.39 (br. s, 1H), 9.22 (s, 1H), 8.04 (s, 1H), 7.82 (s, 1H), 7.46 (d, J = 9.1 Hz, 1H), 7.45 (s, 1H), 7.38 (d, J = 3.0 Hz, 1H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.7 Hz, J = 2.8 Hz, 1H), 6.74 (d, J = 2.9 Hz, 1H), 4.46 (m, 1H), 4.38 (m, 1H), 3.80- 3.05 (multiplets, 6H), 3.35-3.15 (m, 1H), 2.40-2.15 (m, 0.5 H), 2.10-1.75 (multiplets, 1.5H). Example 99: 6-[2-(4-dimethylammo-piperidin-1-yl)-ethoxyJ-2-pyrrolo[1,2-c] pyrimidin-3-yl-chromen-4-one oxime, dihydrochloride was prepared in 30 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-buty! oxime (example 87B) and 4-dsmethylarninopiperidine,
Figure imgf000123_0001
Mp: > 255°C dec.
MS (ESI+): 448.3
Figure imgf000123_0002
(rn/z).
H N R: (300 MHz) DMSO-d6 δ (ppm): 1.30-10.80 (br. s, 3H), 9.23 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J - 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 2.9 Hz, 1 H), 4.46 (m, 1 H), 3.73 (br. d, J = 1 1.4 Hz, 2H), 3.54 (br. s, 2H), 3.38 (br. m, 1 H), 3.22-3.05 (m, 2H), 2.80-2.60 (m, 1 H), 2.73, 2.72 (2s, 6H) , 2.35-2.00 (multiplets, 4H).
Example 100: 6~(2~cyclopentylamino-ethoxy)-2-pyrrolo[ 1,2~c]pyrimidin-3- yl-chromen-4-one oxime, hydrochloride was prepared in 17 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2- c]pyrimsdin-3-yl-chromen-4-one O-fert-butyl oxime (example 87 B) and cyclopentylamine.
Figure imgf000123_0003
Mp: > 230X dec.
Figure imgf000123_0004
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.98 (br. s, 1 H), 9.22 (s, 1 H), 9.01 (br. s, 2H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.48 (d, J = 9.4 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.0 Hz, 1 H), 7.20 (dd, J = 9.2 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.4 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.0 Hz, 1 H), 4.30 (m, 2H), 3.56 (m, 1 H), 3.36 (m, 2H), 1.99 (m, 2H), 1.80-1.45 (multiplets, 6H).
Example 101: 6-(3-morpholin-4-yl-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one ox/me, hydrochloride was prepared in 29 % overall yield using the method described in example 87, starting from 6-(3-chloro-propoxy)-2-pyrrolo[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-iert-butyl oxime (example 101A) and morphoiine.
Figure imgf000124_0001
p: > 220°C dec.
MS (ES1+): 421.3 [C^H^ Hf (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.05 (br. s, 2H), 9.22 (s, 1 H), 8.06 (s, 1 H), 7.82 (s, 1 H), 7.46 (d, J = 8.9 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 2.8 Hz, 1 H), 7.15 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.4 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.0 Hz, 1 H), 4.12 (m, 2H), 3.97 (cl, J = 11.9 Hz, 2H), 3.82 (t, J = 1 1.8 Hz, 2H), 3.46 (d, J = 1 .8 Hz, 2H), 3.27 (m, 2H), 3.20-3.00 (m, 2H), 2.22 (m, 2H).
Example 101 A: 6-( 3-Chloro-propoxy}-2-pyrrolo[1 ', 2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime was prepared in 27% yield using the method described in example 85A, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (example 81 A) and 1-bromo-3-chioropropane in dimethylformamide. The title compound was isolated as a yellow solid.
Figure imgf000124_0002
1 H NMR: (300 MHz) CHCIa- δ (ppm): 8.79 (s, 1 H), 7.81 (s, 1 H), 7.56 (d, J = 2.6 Hz, 1 H), 7.50 (s, 1 H), 7.45 (d, J = 2.2 Hz, 1 H), 7.21 (d, J = 9.0 Hz, 1 H), 6.98 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.92 (dd, J = 3.8 Hz, J = 3.0 Hz, 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 4.19 (t, J = 5.6 Hz, 2H), 3.38 (t, J = 6.4 Hz, 2H), 2.27 (quint, J = 6.0 Hz, 2H), 1.43 (s, 9H). Example 102: 6-(4-morpholin-4-yl-butyl)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 45 % overall yield using the method described in example 87, starting from methanesulfonic acid 4-{4-[(E)-tert- butoxyimino]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromen-6-yl}-butyl ester (example 102A) and morphoiine.
Figure imgf000125_0001
Mp: 180-185X.
MS (ESI+): 419.3 [024Η26Ν.,03+Η]+ (m/z).
1 H NMR: (300 MHz) DMSO-ds δ (ppm): 10.97 (br. s, 1 H), 10.70 (br. s, 1 H), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.73 (s, 1 H), 7.47 (s, 1 H), 7.39 (s, 2H), 6.99 (dd, J = 3.2 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.0 Hz, 1 H), 3.92 (d, J = 1 1.7 Hz, 2H), 3.76 (t, J = 11.5 Hz, 2H), 3.37 (d, J = 1 1.8 Hz, 2H), 3.20-2.90 (m, 4H), 2.75-2.60 (m, 2H), 1.80-1.55 (m, 4H).
Example 102A: methanesulfonic acid 4-{4-[(E)-tert-butoxyimino]~2~ pyrrol o[ 1, 2-c]pyrimidin-3-yf-4H-chromen-6-y}}-butyl ester.
A ice-cooled solution of 6-(4-Hydroxy-butyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (1 15 mg,- 0.28 mmol) (example 102B) in dichioromethane (5 ml) was treated with triethyiamine (100μΙ, 0.7 mmol) and dropwise with methanesulfonyl chloride (26 μΙ, 0.34 mmol). The mixture was stirred at room temperature for 3 hours and treated with a saturated solution of ammonium chloride, extracted with dichioromethane, washed with brine, dried over sodium sulfate and concentrated to dryness. The crude mesylate was used for the next step without further purification.
Example 102B: 6-(4-hydroxy-butyl)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 17% overall yield using the methods described in examples 83A and 83B, starting from 6-bromo-2-pyrroio[1 ,2- c]pyrimidin-3-yl~chromen-4-one O-tert-butyl-oxime (tert-butyl protected oxime of example 66) and But-3-yn-1 -oi. The title compound was isolated as a yellow solid.
N(O-tButyl)
Figure imgf000126_0001
1 H NMR: (300 MHz) CHC^-d! δ (ppm): 8.80 (s, 1 H), 8.20 (d, J = 1.9 Hz, 1 H), 7.90 (s, 1 H), 7.52 (s, 1 H), 7.45 (d, J = 3.7 Hz, 1 H), 7.43 (dd, J = 8.7 Hz, J = 2.1 Hz, 1 H), 7.20 (d, J = 8.5 Hz, 1 H), 6.93 (dd, J = 3.8 Hz, J = 3.3 Hz, 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 3.85 (t, J = 6.2 Hz, 2H), 2.72 (t, J = 6.2 Hz, 2H), 1.43 (s, 9H).
Example 103: 6-(4,4-difluoro-piperidin-1-y)-2-pyrrolo[1,2-c] pyrimidin-3-yl- chromen-4-one ox/me, hydrochloride was prepared in 48 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrroio[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-fert-butyl oxime (example 87B) and 4,4- difluoropipersdine.
Figure imgf000126_0002
MS (ES!+): 441.1 [C23H22F2N403+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 .27 (br. s, 1 H), 10.99 (br. s, 1 H), 9.23 (S, 1 H), 8.06 (s, 1 H), 7.82 (s, 1 H), 7.47 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.41 (d, J = 3.0 Hz, 1 H), 7.22 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 2.9 Hz, 1 H), 4.49 (m, 2H), 3.90-3.70 (m, 2H), 3.64 (m, 2H), 3.31 (m, 2H), 2.60-2.30 (m, 4H).
Example 104: 6-{2-[bis-(2-methoxy-ethyl)-amino]-ethoxy}-2-pyrrolo[ 1,2-c] pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 40 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2- pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 87B) and bis- (2-methoxy-ethy!)-amine.
Figure imgf000127_0001
Mp: 155-160°C.
MS (ESi+): 453.3 [C24H2BN405+Hf (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.00 (br. s, 1Ή), 10.23 (br. s, 1 H), 9.23 (s, 1 H), 8.06 (s, 1 H), 7.82 (s.' l H), 7.47 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.0 Hz, 1 H), 7.19 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.3 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.0 Hz, 1 H), 4.43 (m, 2H), 3.75 (m, 4H), 3.65 (m, 2H), 3.48 (m, 4H), 3.31 (s, 6H).
Example 105: 6-(2-lmidazol-1-yl-ethoxy)-2-pyrrolo[1,2-c] pyrimidin-3-yl- chromen-4-one oxime was prepared in 57 % overall yield using the method described in example 87, starting from 6-{2-ch!oro-ethoxy)-2-pyrro!o[1 ,2-c]pyrimidin-3- yi-chromen-4-one O-fert-butyl oxime (example 87B) and sodium imidazolate (prepared in situ from imidazole and sodium hydride in dimethylformamide).
Figure imgf000127_0002
Mp: > 250°C dec.
MS (ESI+): 388.2 [C21H17N503-t-H]+ (m/z). H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.86 (br. s, 1 H), 9.21 (s, 1 H), 8.02 (s, 1 H), 7.80 (s, 1 H), 7,69 (s, 1 H), 7.42 (s, 1 H), 7.39 (d, J = 9.0 Hz, 1 H), 7.29 (d, J = 3.0 Hz, 1 H), 7.26 (s, 1 H), 7.11 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.97 (dd, J = 3.3 Hz, J = 3.0 Hz, 1 H), 6.89 (s, 1 H), 6.73 (d, J = 3.0 Hz, 1 H), 4.36 (m, 2H), 4.30 (m, 2H).
Example 106: 6-(3~morpholin~4-yl-propyl)~2~pyrro!o[ 1,2-cjpyrimidin~3-yl- chromen-4-one oxime, hydrochloride was prepared in 4 % overall yield using the methods described in examples 102, 102A and 102B, starting from 6-bromo-2- pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyi-oxime (tert-butyl protected oxime of example 66) and prop-2-yn-1 -oi.
Figure imgf000128_0001
MS (ESI+): 405.2 [C23H24N403+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.91 (br. s, 1 H), 10.20 (br. s, 1Ή), 9.22 (s, 1 H), 8.05 (s, 1 H), 7.82 (s, 1 H), 7.75 (s, 1 H), 7.47 (s, 1 H), 7.41 (s, 2H), 6.99 (dd, J = 3.2 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.0 Hz, 1 H), 3.95 (d, J = 12.0 Hz, 2H), 3.71 (t, J = 12.0 Hz, 2H), 3.20-2.95 (m, 4H), 2.75-2.65 (m, 2H), 2.60-2.45 (m, 2H), 2.10-1.95 (m, 2H).
Example 107: 6-((Z)-3-Morpholin-4-yl-propenyl))-2-pyrrolo[1,2-c]pyrimidin- 3-yl-chromen-4-one oxime was isolated as byproduct during the synthesis of example 106.
Figure imgf000128_0002
MS (ESI+): 403.2 [C23H22 403+H]+ (m/z).
Example 108 : 6-[2-(3-methoxy-piperidin-1-yl)-ethoxy]-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 37 % overall yiefd using the method described in example 87, starting from 6-(2-chioro- ethoxy)-2-pyrro!o[1 ,2-c]pyrim!din-3-yl-chromen-4-one O-terf-butyl oxime (example 87B) and 3-methoxy-piperidine.
Figure imgf000128_0003
Mp: 178-184°C.
MS (ESI+): 435,3 [C24H26N404+H]+ (m/z),
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.07 (br. s, 1 H), 9.55 (br. sT 1 H), 9.24 (s, 1 H), 8.07 (s, 1 H), 7.84 (s, 1 H), 7.50 (d, J = 9.2 Hz, 1 H), 7.47 (s, 1 H), 7.42 (t, J = 2.8 Hz, 1 H), 7.21 (dt, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (t, J = 3.2 Hz, 1 H), 6.75 (d, J = 3.0 Hz, 1 H), 4.55-4.35 (m, 2H), 3.80-2.75 (muitiplets, 7H), 3.31 (s, 3H), 2.20-1.15 (muStiplets, 4H).
Example 109: 6-[2-(4-fluoro-pheny!)-ethoxy]-2-pyrrolo[ 1,2-c]pyrimidin-3- yl-chromen-4-one oxime, hydrochloride was prepared in 9% overail yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyl oxime (example 81 A) and 1 -(2-Chloroethy!)-4-fiuoro- benzene.
Figure imgf000129_0001
Mp: 234-236°C.
MS (ESI+): 416.3 [C24Hi eN303+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-ds δ (ppm): 10.91 (s, 1 H), 9.21 (s, 1 H), 8.03 (s, 1 H), 7.81 (s, 1 H), 7.45-7.30 (m, 5H), 7.20-7.05 (m, 3H), 6.98 (m, 1 H), 6.73 (d, J = 3.0 Hz, 1 H), 4.22 (t, J = 6.4 Hz, 2H), 3.05 (t, J = 6.4 Hz, 2H).
Example 110 : 2-quinazolin-2-yl-chromen-4-one oxime was prepared in 23% overall yield using methods A and D, starting from 2'-hydroxyacetophenone and ethyl quinazoline-2-carboxyiate.
Figure imgf000129_0002
Mp: 239-241 °C. MS (ESI+): 290.1 [C^HnNaOa+H]* (m/z).
1 H WR tSOO MHz) DMSO-d@ δ (ppm): 1 1.28 (s, 1 H), 9.74 (s, 1 H), 8.25 (d, J - 8.3 Hz, 1 H), 8.17 {d, J = 7.9 Hz, 1 H), 8.1 1 (td, J = 7.6 Hz, J = 1.3 Hz, 1 H), 7.99 (s, 1 H), 7.93 (td, J = 7.9 Hz, J = 1.5 Hz, 1 H), 7.84 (td, J = 7.4 Hz, J = 1.3 Hz, 1 H), 7.55 (td, J = 8.4 Hz, J = 1.5 Hz, 1 H), 7.50 (td, J = 8.3 Hz, J = 1.5 Hz, 1 H), 7.32 (td, J = 7.3 Hz, J = 1.3 Hz, 1 H).
Example 111: 6-[(1S,4S)-2-(2-oxa-5-aza-bicyclo[2.2.1]hept-5-yl)-ethoxy]-2- pyrrololl^-cJpyrimidinS-yl-chromen-A-one oxime, hydrochloride was prepared in 37 % overail yield using the method described in example 87, starting from 6-(2- chloro-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ierf-butyl oxime (example 87B) and (1 S,4S)-2-oxa-5-aza-bicyclo[2.2.1]heptane,
Figure imgf000130_0001
MS (ESI+): 419.1 [C23H22N404+H] (m/z).
1 H NMR: (400 MHz) DMSO-de δ (ppm): 10.94 (br. s, 1 H), 10.41 (br. s, 1 H), 9.23 (s, 1 H), 8.05 (s, 1 H), 7.82 (d, J = 2.4 Hz, 1 H), ), 7.47 (d, J = 9.2 Hz, 1 H), 7.45 (s, 1 H), 7.39 (d, J = 3.8 Hz, 1 H), 7.21 (dd, J = 9.2 Hz, J = 4.0 Hz, 1 H), 6.99 (dd, J = 4.0 Hz, J = 3.2 Hz, 1 H), 6.74 (d, J = 4.0 Hz, 1 H), 4.70-4.50 (m , 2H), 4.50-4.30 (m, 2H), 4.30-4.10 (m, 1 H), 3.80-3.50 (m, 4H), 3.35-3.05 (m, 1 H), 2.10-1.95 (m, 2H).
Example 112; 6-[2-(cis-2,6~dimethyl~morphoIin*4-yl)~ethoxy]*2~pyrrolo[ 1,2- c]pyrimidin-3-yl-chrom en-4-one oxime, hydrochloride was prepared in 17 % overail yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-rerf-butyl oxime (example 87B) and (c/s)-2,6-dirnethylmorpholine.
Figure imgf000131_0001
MS (ESI+): 435.1 [C24H26N404+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 11.01 (br. s, 2H), 9.23 (s, 1 H), 8.06 (s, 1 H), 7.83 (s, 1 H), 7.47 (d, J = 10.0 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.2 Hz, 1 H), 7.22 (dd, J = 9.2 Hz, J = 3.2 Hz, 1 H), 6.99 (dd, J = 3.4 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 3.6 Hz, H), 4.47 (m, 2H), 3.98 (m, 2H), 3.54 (m, 4H), 2.78 (m, 2H), 1.16 (s, 3H), 1.14 (s, 3H)
Example 113: 2-pyrrolo[ 1,2-c]pyrimidin-3-yI-6-[2-(4-trifluoromethyl- piperidin~1-yl)'ethoxy]'Chromen-4-one oxime, hydrochloride was prepared in 34 % overall yield using the method described in example 87, starting from 6~(2-chloro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-terf-butyl oxime (example 87B) and 4-trifluoromethyl-piperidine.
Figure imgf000131_0002
MS (ESI+): 473.1 ^Η^Ν^+Η] (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.96 (br. s, 1 H), 10.19 (br. s, 1 H), 9.23 (s, 1 H), 8.05 (s, 1 H), 7.83 (s, 1 H), 7.46 (d, J = 9.2 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.2 Hz, 1 H), 7.21 (dd, J = 9.2 Hz, J = 3.2 Hz, 1 H), 6.99 (dd, J = 3.4 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 3.2 Hz, 1 H), 4.44 (m, 2H), 3.70 (d, J = 1.2 Hz, 2H), 3.20-3.00 (m, 3H), 2.75-2.50 (m, 2H), 2.10-2.00 (m, 2H), 2.00-1.90 (m, 2H)
Example 114: 6-[2-(3,3-difluoro-piperidin-1-yl)-ethoxy]-2-pyrro!o[ 1,2- c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 72 % overall yield using the method described in example 87, starting from 6-(2-chioro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-teri-butyl oxime (example 87B) and 3,3-difluoro-piperidine.
Figure imgf000132_0001
Mp : 190-195°C.
MS (ESI+): 441.3 [C23H22F2N403+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d@ δ (ppm): 11.27 (br. s, 2H), 9.23 (s, 1H), 8.07 (s, 1H), 7.82 (s, 1H), 7.47 (d, J = 9.0 Hz, 1H), 7.46 (s, 1H), 7.43 (d, J = 3.0 Hz, 1H), 7.23 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.2 Hz, J = 2.8 Hz, 1H), 6.74 (d, J = 3.2 Hz, 1H), 4.52 (m, 2H), 4.15-3.10 (br. muttiplets, 4H), 3.62 (m, 2H), 2.35-1.80 (br. mu!tiplets, 4H).
Example 115: 4-(hydroxyimino)-2-pyrrolo[1,2-c]pyrimidin-3-yl~4H- chromene-6-carboxyIic acid (2-morpholin-4-yl-ethyl)-amide, hydrochloride was prepared in 54% yield using method D (step 2) starting from 4-(tert-Butoxyimino)-2- pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromene-6-carboxyiic acid (2-morpholin-4-yl-ethy!)- amide (example 115A). The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1.25 solution of hydrogen chloride in isopropanoi.
Figure imgf000132_0002
Mp : 230-235°C.
MS (ESI+): 434.3 [C23H23N60 +H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.11 (br. s, 1H), 10.75 (br. s, 1H), 9.23 (s, 1H), 9.01 (m, 1H), 8.42 (s, 1H), 8.10-8.04 (m, 2H), 7.83 (s, 1H), 7.53 (d, J = 8.7 Hz, 1H), 7.49 (s, 1H), 6.99 (dd, J = 3.5 Hz, J = 3.0 Hz, 1H), 6.76 (d, J = 3.8 Hz, 11-0,4.99 (d, J = 13.5 Hz, 2H), 3.80 (t, J = 12.0 Hz, 2H), 3.69 (m, 2H), 3.55 (d, J = 11.8 Hz,2H), 3.32 (m, 2H), 3.13 (m, 2H). Example 115A: 4~( tert-Butoxyimino)~2~pyrrolo[ 1, 2-c]pyrimidin-3-yl-4H- chromene-6-carboxylic acid (2-morpholin-4-yl-ethyl)-amide.
Figure imgf000133_0001
A suspension of 4-{tert-Butoxyimino)-2-pyrrolo[1 !2-c]pyrimidin-3-yi-4H- chromene-6-carboxylic acid methyl ester (example 115B) (170 mg, 0.43 mmol) in a mixture of tetrahydrofuran/methano!/water (2.0 ml/0.5 ml/0.5 ml) was treated with lithium hydroxide monohydrate (55 mg, 1 .29 mmol) and heated at 60°C for 2 hours. The solvents were removed and the residue was diluted with water, neutralized to pH 5-6 with a 1 N solution of hydrochloric acid and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate and concentrated to dryness to give 4-(tert-Butoxyimino)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-4H- chromene-6-carboxylic acid (135 mg, 84%) as a green solid. A solution of the previous carboxylic acid, (3-Dimeihyiamino-propyl)-ethyl-carbodiimide, hydrochloride (90 mg, 0.47 mmol), 1-Hydroxybenzotriazo!e hydrate (68 mg, 0.50 mmol) and triethylamine (250 μΙ, 1.80 mmol) in dimethylformamide (6.0 ml) was stirred at 0°C for 1 hour then 4- (2-amtnoethyl)-morpholine (52 μΙ, 0.40 mmol) was added. The mixture was stirred at room temperature for 18 hours. The volatiles were removed under vacuum and the residue taken into ethyl acetate, washed several times with a saturated solution of sodium hydrogenocarbonate, brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by silica gel flash chromatography (gradient ethyl acetate/methanol 0-8%) to yield the title compound as a yellow solid (137 mg, 78%).
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.79 (s, 1H), 8.41 (s, 1 H), 7.97 (dd, J = 8.7 Hz, J = 2.0 Hz, 1 H), 7.92 (s, 1 H), 7.53 (s, 1 H), 7.46 (d, J = 2.6 Hz, 1 H), 7.32 (d, J = 8.6 Hz, 1 H), 6.94 (dd, J = 4.0 Hz, J = 3.0 Hz, 1 H), 6.65 (d, J = 3.8 Hz, 1 H), 3.82 (m, 4H), 3.63 (m, 2H), 2.73 (m, 2H), 2.64 (m, 4H), 1.43 (s, 9H).
Example 115B: 4-(tert-8utoxyimino)-2-pyrrolo[1,2-c]pyrimidin-3-y}-4H- chromene-6-carboxylic acid methyl ester.
Figure imgf000134_0001
!n a 75 mi sealed tube were charged 6-bromo-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (tert-butyi protected oxime of example 66) (250 mg, 0.60 mmol), palladium acetate (14 mg, 0.06 mmoi), 1 , 1 '- bis(diphenylphosphino)ferrocene (68 mg, 0.12 mmol), triethyiamine (170 μΙ, 1.22 mmol), methanol (125 μΙ, 3.03 mmol) and dimethylformamide (5 ml). The suspension was degazed with carbone monoxide for 5 minutes. The tube was sealed and heated to 100X for 18 hours. The mixture was degazed with argon to remove the excess of carbone monoxide and then diluted with ethyl acetate, washed with a saturated solution of ammonium chloride, water, brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by silica gel flash chromatography (gradient cyclohexane/ethyl acetate 0-80%) to yield the title compound as a yellow solid (172 mg, 72%).
1 H NMR: (300 MHz) CHCIs-d, δ (ppm): 8.80 (s, 1 H), 8.71 (d, J = 2.1 Hz, 1 H), 8.05 (dd, J = 8,7 Hz, J = 2.0 Hz, 1 H), 7.90 (s, 1 H), 7.54 (s, 1 H), 7.45 (d, J = 2.1 Hz, 1 H), 7.28 (d, J = 8.7 Hz, 1 H), 6.93 (dd, J = 4.0 Hz, J = 3.0 Hz, 1 H), 6.65 (d, J = 3.8 Hz, 1 H), 3.95 (s, 3H), 1.43 (s, 9H).
Example 116: 6-(2~[ 1,4J" bipiperidinyl~1'-yl~ethoxy)-2~pyrrolo[ 1,2· c]pyrimidin-3-yl-chromen-4-one oxime, dihydrochloride was prepared in 34 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrrolo[ ,2-cjpyrimidin-3-yl-chromen-4-one O-terf-buty! oxime (example 87B) and /V-(4-piperidino)piperidine.
Figure imgf000134_0002
Mp : > 235°C dec.
MS (ESI+): 488.4 [C28H33N503+H]+ (m/z). 1 H NMR: (300 MHz) DMSO-da δ (ppm): 10.99 (br. s, 1 H), 10.74 (br. s, 1 H), 9.23 (s, 1 H), 8.05 (s, 1 H), 7.82 (d, J = 2.4 Hz, 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H)„ 7.39 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.2 Hz, J = 2.8 Ηζ, Η), 6.74 (d, J = 3.7 Hz, 1 H), 4.46 (m, 2H), 3.8-3.45 (multipiets, 4H), 3.45-3.25 (m, 3H), 3.14 (m, 2H), 2.92 (m, 2H), 2.45-2.10 (multipiets, 4H), 2.00-1.60 (multipiets, 5H), 1.50-1.30 (m, 1 H),
Exampl e 117; 6-( 2-[ 1, 4]oxazepan-4-yl-ethoxy)-2-pyrrolo[ 1, 2-c]pyrimidin-3- yl-chromen-4-one oxime, hydrochloride was prepared in 45 % overall yield using the method described in example 87, starting from 6-(2-chloro-ethoxy)-2-pyrrolo[1 ,2- c]pyrimidiri-3-yl-chromen-4-one O-fe/ -butyl oxime (example 87B) and /■"/, /oxazepane.
Figure imgf000135_0001
p : 240°C dec.
MS (ESI+): 421.3 [C^H^C H] (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.87 (br. s, 2H), 9.23 (s, 1 H), 8.06 (s, 1 H), 7.82 (d, J = 2.0 Hz, 1 H), 7.46 (d, J = 9.0 Hz, 1 H), 7.45 (s, 1 H), 7.40 (d, J = 3.0 Hz, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.2 Hz, J = 2.8 Hz, 1 H), 6.74 (d, J = 3.8 Hz, 1 H), 4.47 (m, 2H), 3.88 (m, 2H), 3.85-3.68 (m, 2H), 3.65-3.47 (m, 4H), 3.45-3.25 (m, 2H), 2.37-2.17 (m, 1 H), 2.12-1.95 (m, 1 H).
Example 118: 6-[2-(4-pyrrolidin-1-yl-piperidin~1-yl)-ethoxy]-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one oxime, dihydrochlonde was prepared in 90 % overall yield using the method described in example 87, starting from 6-(2-chloro- ethoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-ierf-butyl oxime (example 87B) and 4-(1-pyrrolidinyl)piperidine.
Figure imgf000136_0001
Mp : > 210°C dec.
MS (ESI+): 474.3 [027Η31 503+Η]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.25-10:75 (br. multip!ets, 2H), 9.23 (s, 1H), 8.05 (s, 1H), 7.82 (d, J = 2.3 Hz, 1H), 7.47 (d, J = 9.0 Hz, 1H), 7.45 (s, 1H), 7.40 (d, J = 3.0 Hz, H), 7.21 (dd, J = 9.0 Hz, J = 3.0 Hz, 1H), 6.99 (dd, J = 3.3 Hz, J = 2.8 Hz, 1H), 6.74 (d, J = 3.7 Hz, 1H), 4.46 (m, 2H), 3.72 (m, 2H), 3.65-3.42 (multipiets, 4H), 3.40-3.22 (m, 1H), 3.20-2.95 (m, 4H), 2.40-2.05 (muitiplets, 4H), 2.05-1.80 (multiplets, 4H).
Example 119: 6-[2-(3,3-difluoro-pyrrolidin-1-yl)-ethoxy]-2-pyrrolo[ 1,2- c]pyrimidin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 42 % overali yield using the method described in example 87, starting from 6-(2-ch!oro- ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yt-chromen-4-one O-ferf-butyl oxime (example 87B) and 3,3-difluoro-pyrroiidine.
Figure imgf000136_0002
Mp : > 230°C dec.
MS (ES1+): 427.3 [C22H20F2N4O3+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.96 (br. s, 1H), 11.02 (br. s, 1H), 9.23 (s, 1H), 8.06 (s, 1H), 7.82 (s, 1H), 7.47 (d, J = 9.0 Hz, 1H), 7.45 (s, 1H), 7.40 (d, J = 3.0 Hz, 1H), 7.24 (dd, J = 9.0 Hz, J = 2.8 Hz, 1H), 6.99 (dd, J = 3.2 Hz, J = 3.0 Hz, 1H), 6.74 <d, J = 3.4 Hz, 1H), 4.43 (m, 2H), 4.30-3.40 (m, 4H), 3.72 (m, 2H), 2.80-2.40 (m, 2H). Example 120: 7-[3-(3-dimethylamino-propoxy)-phenylethynyl]-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4~one oxi e was prepared in 12% overall yieid using the methods described in method D (step 1 ) and in example 86, starting from 7-[3-(3-dimethyiamino-propoxy)-phenyiethynyl]-2-pyrrolo[1 ,2-c]pyrimidin-3-y!- chromen-4-one (example 120A), The title compound was isolated as a yellow
Figure imgf000137_0001
MS (ESI+): 479.1 [C29H26N403+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 9.23 (s, 1 H), 8.08 (s, 1 H), 7.90 (d, J = 8.0 Hz, 1 H), 7.84 (s, 1 H), 7.66 (d, J = 1.6 Hz, 1 H), 7.47 (s, 1 H), 7.43 (dd, J = 10.4 Hz, J = 1.6 Hz, 1 H), 7.36 (t, J = 7.6 Hz, 1 H), 7.18-7.12 (m, 2H), 7.05-7.00 (m, 2H), 6.75 (d, J = 4 Hz, 1 H), 4.06 (t, J = 6.4 Hz, 2H), 2.36 (t, J - 6.4 Hz, 2H), 2.17 (s, 6H), 1.88 (m, 2H).
Example 120 : 7-[3-(3-dimethylamino-propoxy)-phenylethynyl]-2- pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one was prepared in 52% yieid using the method described in example 42, starting from 7-(3-hydroxy-phenylethynyl)-2- pyrrolo[1 ,2-c3pyrimidin-3-yl-chromen-4-one (example 120B) and (3-Chloro-propyl)- dimethyi-amine hydrochloride.
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.84 (t, J = 1.3 Hz, 1 H), 8.20 (d, J = 8.5 Hz, 1 H), 8.13 (d, J = 1.2 Hz, 1 H), 7.73 (d, J = 0.9 Hz, 1 H), 7.55-7.50 (m, 2H), 7.33 (s, 1 H), 7.28 (d, J = 12.2 Hz, 1 H), 7.17 (td, J = 7.7 Hz, J = 1.1 Hz, 1 H), 7.10 (dd, J = 2.4 Hz, J = 1.3 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.6 Hz, 1 H), 6.94 (ddd, J = 8.3 Hz, J = 2.6 Hz, J = 1.0 Hz, 1 H ), 6.77 (d, J = 3.8 Hz, 1 H), 4.09 (t, J = 6.2 Hz, 2H), 2.72 (t, J = 6.9 Hz, 2H), 2.46 (s, 6H), 2.13 (quint, J = 6.6 Hz, 2H).
Example 120B: 7-(3-hydroxy-phenylethynyl)-2-pyrrolo[ 1, 2-c]pyrimidin-3- yl-chromen-4-one was prepared in 71 % yield using the method described in example 23A, starting from 7-bromo-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one (example 120C) and 3-hydroxyphenylacetylene. The title compound was isolated as a brown powder.
1 H NMR: (300 MHz) DMSO-de δ (ppm): 9.87 (s, 1 H), 9.28 (s, 1 H), 8.40 (s, 1 H), 8.04 (d, J = 8.3 Hz, 1 H), 8.00 (d, J = 1.3 Hz, 1 H), 7.90 (d, J = 2.5 Hz, 1 H), 7.60 (dd, J = 8.3 Hz, J = 1.5 Hz, 1 H), 7.27 (t, J = 7.9 Hz, 1 H), 7.10-6.98 (m, 4H), 6.93-6.85 (m, 2H
)■
Example 120C: 7-bromo-2-pyrrolo[ 1,2-c]pyrimidin-3-yl-chromen-4-one was prepared in 89% yield using method A, starting from 4'-bromo-2'- hydroxyacetophenone and pyrrolo[1 ,2-c]pyrimidin-2-carboxylic acid ethyl ester. The title compound was isolated as a green powder.
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 9.28 (s, 1 H), 8.40 (s, 1 H), 8.16 (d, J = 1.9 Hz, 1 H), 7.96 (d, J = 8.5 Hz, 1 H), 7.91 (d, J = 2.4 Hz, 1 H), 7.68 (dd, J = 8.4 Hz, J = 1.7 Hz, 1 H), 7.08-7.02 (m, 2H), 6.88 (d, J = 3.8 Hz, 1 H).
Example 121: 6-[2-(4-ethyl-piperazin-1-yl)-ethoxy]-2-pyrrolo[ 1,2- c]pyrimidin-3-yl-chromen-4-one oxime, dihydrochforide was prepared in 12 % overall yield using the method described in example 87, starting from 6-(2-chSoro- ethoxy)-2-pyrrolo[1 ,2-cjpyrimidin-3-yi-chromen-4-one O-f erf-butyl oxime (example 87B) and 4-ethy!-piperazine.
Figure imgf000138_0001
Mp: > 240°C dec.
MS (ESI+): 434.2 [C24H27N503+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.95 (br. s, 1 H), 9.22 (s, 1 H), 8.04 (s, 1 H), 7.81 (s, 1 H), 7.46 (d, J = 8.7 Hz, 1 H), 7.44 (s, 1 H), 7.38 (d, J = 2.8 Hz, 1 H), 7.20 (d, J = 9.0 Hz, 1 H), 6.99 (dd, J = 3.2 Hz, J = 3.0 Hz, 1 H), 6.74 (d, J = 3.7 Hz, 1 H), 4.41 (m, 2H), 4.00-3.00 (muitiplets, 12H), 1.26 (t, J = 6.9 Hz, 3H).
Example 122: 6-(2-amino-ethoxy)-2-pyrrolo[ 1f2-c]pyrimidin-3~yl~chromen- 4-one oxime, hydrochloride was prepared in 26% yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-cjpyrimidin-3-yl-chromen-4-one O-ferf-butyl oxitne (example 81 A) and 2-(Boc-amino)eihyl bromide.
Figure imgf000139_0001
Mp: > 270°C dec.
MS (ESI+); 337.2 [C18H16N403+H]+ (m/z).
1 H N R: (300 MHz) DMSO-d6 δ (ppm): 10.99 (br. s, 1 H), 9.22 (s, 1 H), 8.15 (br. s, 3H), 8.05 (s, 1 H), 7.82 (d, J = 2.6 Hz, 1 H), 7.46 (d, J - 8.7 Hz, 1 H), 7,45 (s, 1 H), 7.39 (d, J = 3.0 Hz, 1 H), 7.18 (dd, J = 9.0 Hz, d, J = 3.0 Hz, 1 H), 6.99 (dd, J = 3.7 Hz, J = 2.9 Hz, 1 H), 6.74 (d, J = 3.7 Hz, H), 4.22 (m, 2H), 3.24 (m, 2H).
Example 123: 6-(2-Morpholin-4-yl-ethoxy)-2-(8aH-pyrrolo[ 1,2-ajpyrazin~3- yl)-chromen-4~one oxime, hydrochloride was prepared in 66% yield using method D (step 2), starting from 6-(2-Morpholin-4-yS-ethoxy)-2-pyrrolo[1 ,2-a]pyrazin-3-yl- chromen-4-one O-tert-butyl-oxime (example 123A). The hydrochloride salt of the title compound was isolated as a yellow solid after treatment with a 1 .25 M solution of hydrogen chloride in isopropanoi.
Figure imgf000139_0002
Mp: > 250°C dec.
MS (ESI+): 407.2 [C22H22N404+H3+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 6 ' (ppm): 11.33 (br. s, 1 H), 10.95 (s, 1 H), 8.95 (d, J = 7.1 Hz, 2H), 7.92 (s, 1 H), 7.41 (m, 3H), 7.21 (dd, J = 9.0 Hz, J = 3.0, 1Ή), 6.97 (m, 2H), 4.48 (br. s, 2H), 3.89 (m, 4H), 3.52 (m, 4H), 3.19 (br. s, 2H).
Example 123 A: 6-(2-Morpholin-4-yl-ethoxy}-2-pyrrolo[ 1,2-a]pyrazin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 68% yield using the procedure described in exampie 85A, starting from 6-Hydroxy-2-pyrrolo[1 ,2-a]pyrazin-3-yl- chromen-4-one O-tert-butyl-oxime (example 123B) and 4-(2-chloroethyl) morpholine hydrochloride. The title compound was isolated as a beige solid.
Figure imgf000140_0001
1 H NMR: (300 MHz) CHCIa-d, δ (ppm): 8.82 (s, 1 H), 8.45 (s, 1 H), 7.51 (m, 3H), 7.16 (d, J = 9.0 Hz, 1 H), 6.98 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.91 {dd, J = 4.1 Hz, J = 2.6 Hz, 1 H), 6.83 (d, J = 4.1 Hz, 1 H), 4.17 (t, J = 5.6 Hz, 2H), 3.75 (t, J = 4.7 Hz, 4H), 2.83 (t, J = 5.6 Hz, 2H), 2.60 (t, J = 4.7 Hz, 4H), 1.41 (s, 9H).
Example 123B: 6-Hydroxy-2-pyrrolo[1r2-a]pyrazin-3-yl-chromen-4-one O- tert-butyl-oxime was prepared in 85% yield using the procedure described in example 18A, starting from 6-Bromo-2-pyrroio[1 ,2-a]pyrazin-3-yl-chromen-4-one O- tert-butyl-oxime (example 123C). The title compound was isolated as a brown solid.
N{0-tButyl)
Figure imgf000140_0002
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 8.99 (d, J = 2.2 Hz, 1 H), 7.97 (s, 1 H), 7.33 (s, 1 H), 7.32 (d, J = 3.0 Hz, 1 H), 7.26 (d, J = 8.8 Hz, 1 H), 7.03 (m, 2H), 6.96 (dd, J = 9.0 Hz, J = 3.0, 1 H), 5.03 (br s, 1 H), 1.36 (s, 9H).
Example 123C: 6-Bromo-2-pyrrolo[1,2-a]pyrazin-3-yl-chromen-4-one O- tert-butyl-oxime was prepared in 65% overall yield using methods A and D (stepi), starting from 5'-bromo-2'-hydroxy-acetophenone and Pyrrolo[1 ,2-a]pyrazine-3- carboxylic acid methyl ester (example 91A).
N(O-tButyl)
Figure imgf000140_0003
1 H NMR: (300 MHz) CHCIs-dt δ (ppm): 8.83 (s, 1 H), 8.44 (s, 1 H), 8.16 (d, J = 2.4 Hz, 1 H), 7.53-7.52 (m, 2H), 7.47 (dd, J = 8.8 Hz, J = 2.4, 1 H), 7.21 (dd, J = 9.0 Hz, J = 3.0, 1 H), 7.12 (d, J = 8,6 Hz, 1 H), 6.93 (dd, J = 4.1 Hz, J = 2.6, 1 H), 6.85 (dt, J = 3.9 Hz, J = 1.1 Hz, 1 H), 1.41 (s, 9H). Example 124: 6-[2-(4,4-Difluoro-piperidin-1-y!)-ethoxy]-2~pyrrolo[ 1,2- a]pyrazin-3-yl-chromen-4-one oxime, hydrochloride was prepared in 50% overall yield using the method described in example 123, starting from 6-Hydroxy-2- pyrrolo[1 ,2-a]pyrazin-3-yl-chromen-4-one O-tert-butyl-oxime (exampie 123B) and 1-(2- Chloro-ethyl)-4,4-difluoro-piperidine hydrochloride.
Figure imgf000141_0001
Mp: 225°C - 230°C.
MS (ESI+): 441.3 [C23H22F2N403+H3+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1 .21 (br. s, 1 H), 10.98 (br. s, 1 H), 8.97 (d, J = 7.1 Hz, 2H), 7.94 (s, 1 H), 7.46 (s, 1 H), 7.41 (m, 2H), 7.23 (dd, J = 9.0 Hz, J = 3.0, 1 H), 7.00 (m, 2H), 4.48 (t, J = 4.35Hz, 2H), 3.67 (m, 4H), 3.31 (br. s, 2H), 2.39 (m, 4H).
Example 125: 6-(2-lmidazol-1-yl-ethoxy)-2-pyrrolo[ 1,2-a]pyrazin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 13% overall yield using the method described in example 123, starting from 6-Hydroxy-2-pyrrolo[1 ,2-a]pyrazin-3- y[-chromen-4-one O-tert-butyi-oxime (exampie 123B) and 1-(2-Ch!oro-ethyi)-1 H- - imidazole hydrochloride.
Figure imgf000141_0002
Mp: 238°C - 240°C.
MS (ES1+): 388.2 [C21H 7N503+Hf (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 14.68 (br. s, 1 H), 10.94 (br. s, 1 H), 9.25 (s, 1 H), 8.95 (d, J = 7.1 Hz, 2H), 7.93 (s, 1 H), 7.87 (s, 1 H), 7.71 (s, 1 H), 7.44 (s, 1 H), 7.36 (m, 2H), 7.14 (dd, J = 9.0 Hz, J = 3.0, 1 H), 7.00 (m, 2H), 4.65 (br. s, 2H), 4.45 (s, 2H).
Example 126: 6-[2-(4-Fluoro~phenyf)~ethoxy]~2-pyrrolo[ 1,2-a]pyrazin~3-yt~ chromen-4-one oxime was prepared in 27% overall yield using the method described in example 123, starting from 6-Hydroxy-2-pyrrolo[1 ,2-a]pyrazin-3-yl-chromen-4-one O-tert-butyl-oxime (example 123B) and 1-(2-Chloro-ethyi)-4-fluorophenyl.
Figure imgf000142_0001
Mp: 248°C - 250°C.
MS (ESI+): 416.3 [C24H18FN303+H]+ (m/z).
1 H N R: (300 MHz) D SO-da δ (ppm): 10.94 (br. s, 1 H), 8.97 (d, J = 7.1 Hz, 2H), 7.94 (s, 1 H), 7.44 (s, 1 H), 7.36 (m, 4H), 7.14 (m, 3H), 7.00 (m, 2H), 4.22 (t, J = 6.7 Hz, 2H), 3.05 (t, J = 6.7 Hz, 2H).
Example 127: 6-(2-MorphoIin-4^l~ethoxy)-2-thieno[3,2-c]pyridin-6-yf- chromen-4-one oxime, hydrochloride was prepared in 39% yield using method D (step 2), starting from 6-(2-Morpholin-4-yl-ethoxy)-2-thseno[3,2-c]pyridin-6-yl-chromen- 4-one O-tert-butyl-oxime (exampie 127A). The hydrochloride salt of the title compound was isolated as a yellow solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanol.
Figure imgf000142_0002
Mp: >230°C dec.
MS (ESI+): 424.3 [C22H21N304S+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.50 (br. s, 1 H), 11.13 (br. s, 1 H), 9.27 (s, 1 H), 8.79 (s, 1 H), 8.06 (d, J = 5.3 Hz, 1 H), 7.74-7.69 (m, 2H), 7.51 (d, J = 9.0 Hz, 1 H), 7.43 (s, 1 H), 7.23 (d, J = 9.0 Hz, 1 H), 4.52-4.49 (m, 2H), 3.99-3.86 (m, 4H), 3.58-3.49 (m, 4H), 3.23-3.20 (m, 2H).
Example 127 A: 6-(2~Morpholin-4-yl'ethoxy)'2-thieno[3,2-c]pyridin-6-yl- chromen-4-one O-tert-butyl-oxime was prepared in 62% yield using the procedure described in example 85A, starting from 6-Hydroxy-2-thieno[3,2-c]pyridin-6-yi- chromen-4-one O-tert-butyl-oxime (example 127B) and 4-(2-chloroethyl)morpho!ine hydrochloride. The title compound was isolated as a yellow solid.
Figure imgf000143_0001
1 H NMR: (300 MHz) CHCfe-d, δ (ppm): 9.15 (s, 1 H), 8.45 (s, 1 H), 7.71 (s, 1 H), 7.58 (d, J = 5.4 Hz, 1 H), 7.54-7.48 (m, 2H), 7.23 (m, 1 H), 7.01 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.18 (t, J = 5^6 Hz, 2H), 3.76 (t, J = 4.5 Hz, 4H), 2.84 (t, J = 5.6 Hz, 2H), 2.61 (t, J = 4.7 Hz, 4H), 1.42 (s, 9H).
Example 127B: 6-Hydroxy-2-thieno(3,2-c]pyridin-6-yl-chromen-4-one O- tert-butyl-oxime was prepared in 99% yield using the procedure described in example 18A, starting from 6-Bromo-2-thieno[3,2-c]pyridin-6-y!-chromen-4-one O-tert- butyl-oxime (example 127C). The title compound was isolated as a yellow solid.
Figure imgf000143_0002
1 H NMR: (300 MHz) CHCis-d! δ (ppm): 9.15 (s, 1 H), 8.45 (s, 1 H), 7.69 (s, 1 H), 7.58 (d, J = 5.4 Hz, 1 H), 7.50-7.48 (m, 2H), 7.20 (d, J = 9.0 Hz, 1H), 6.92 (dd, J = 9.0 Hz, J = 3.2 Hz, H), 5.42 (s, 1 H), 1.40 (s, 9H).
Example 127C: 6-Bromo-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one O- tert-butyl-oxime was prepared in 64% overall yield using methods A and D (stepl), starting from 5'-bromo-2'-hydroxy-acetophenone and Thieno[3,2-c]pyridin-6-carboxylic acid methyl ester.
Figure imgf000144_0001
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 9.16 (s, 1 H), 8.43 (s, 1 H), 8.17 (d, J = 2.4 Hz, 1 H), 7.73 (s, 1 H), 7.60 (d, J = 5.4 Hz, 1 H), 7.51-7.46 (m, 2H), 7.18 (d, J = 8.6 Hz, 1 H), 1.42 (s, 9H).
Example 128: 6-[2-(4,4-Difluoro-piperidin-1-yl)-ethoxy]- 2-thieno[3,2- c]pyridin-6-yl-chromen-4-one oxime, hydrochloride was prepared in 28% overall yield using the method described in example 127, starting from 6-Hydroxy-2- thieno[3,2-c]pyridin-6-yl-chromen-4-one O-tert-butyl-oxime (example 127B) and 1-(2- Chioro-ethyi)-4,4-difluoro-piperidine hydrochloride.
Figure imgf000144_0002
Mp: 225°C - 230°C.
MS (ES1+): 458.3 [C23H21F2N303S÷H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.34 (br. s, 1 H), 1 1.06 (br, s, 1 H), 9.26 (s, 1 H), 8.77 (s, 1 H), 8.04 (d, J = 5.3 Hz, 1 H), 7.72 (d, J = 5.4, 1 H), 7.69 (s, 1 H), 7.51 (d, J = 9.0 Hz, 1 H)77.42 (d, J = 2.9 Hz, 1 H), 7.23 (dd, J = 9.0 Hz, J = 2.9 Hz, 1 H), 4.50 (s, 2H), 3.70-3.64 (m, 4H), 3.31 (s, 2H), 2.43 (m, 4H).
Example 129: 6-(2-imidazol-1~yl-ethoxy) 2-thieno[3,2-c]pyridin-6-yl- chromen-4-one oxime, hydrochloride was prepared in 25% overall yield using the method described in example 127, starting from 6-Hydroxy-2-thieno[3,2-c]pyridin-6-yi- chromen-4-one O-tert-butyi-oxime (example 127B) and 1-(2-Chloro-ethyl)-1 H- imidazole hydrochloride.
Figure imgf000145_0001
Mp: 218°C - 220°C.
MS (ESI+): 405.2 [C21H16N403S-t-H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.10 (br. s, 1 H), 9.26 (s, 2H), 8.78 (s, 1 H), 8.06 (d, J = 5.3 Hz, 1 H), 7.87 (s, 1 H), 7.74-7.68 (m, 3H), 7.48 (d, J = 9.0 Hz, 1 H), 7.38 (d, J = 2.9 Hz, 1 H), 7.18 (dd, J = 9.0 Hz, J = 2.9 Hz, 1 H), 4.65 (t, J = 4.6 Hz, 2H), 4.46 (t, J = 4.6 Hz, 2H).
Example 130: 6-[2-(4-Fluoro-phenyl)-ethoxy]~2-thieno[3,2-c]pyridin~6-yl- chromen-4-one oxime was prepared in 18% overall yield using the method described in example 127, starting from 6-Hydroxy-2-thieno 3,2-c]pyridin-6-yl-chromen-4-one O- tert-bui -oxime (example 127B) and 1~(2-Chloro-ethyl)-4-fluorophenyl hydrochloride.
Figure imgf000145_0002
Mp: 228°C - 234°C.
Figure imgf000145_0003
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.10 (br. s, 1 H), 9.28 (s, 1 H), 8.78 (s, 1 H), 8.05 (d, J = 5.3 Hz, 1 H), 7.72 (d, J = 5.4 Hz, 1 H), 7.68 (s, 1 H), 7.46-7.34 (m, 4H), 7.17-7.1 1 (m, 3H), 4.23 (t, J = 6.6 Hz, 2H), 3.05 (t, J = 6.6 Hz, 2H).
Example 131: 6-[2-( 3, 3-Dtfluoro-piperidin- 1 -yI)-eihoxy]-2-thieno[ 3,2- c]pyridin-6-yl-chromen-4-one oxime, hydrochloride was prepared in 67% overall yield using the method described in example 127, starting from 6-Hydroxy-2- thieno[3,2-c]pyridin-6-yl~chromen-4-one O-tert-butyl-oxime (example 127B) and 1-(2- Chloro-ethyl)-3,3-difluoro-piperidine hydrochloride.
Figure imgf000146_0001
Mp: >250°C dec.
MS (ESI+): 548.2 [C23H21F2N303S+H]+ (m/z).
1 H-NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.04 (br. s, 1 H), 9.25 (s, 1 H), 8.76 (s, 1 H), 8.03 (d, J = 5.4 Hz, 1 H), 7.72 (d, J = 5.4 Hz, 1 H), 7.68 (s, 1 H), 7.50 (d, J = 9.1 Hz, 1 H), 7.40 (d, J = 3.0 Hz, 1 H), 7.22 (dd, J = 9.1 Hz, J = 3.1 Hz, 1 H), 4.49 (br. s, 2H), 3.61 (m, 6H), 2.16-1.99 (m, 4H).
Example 132: 6~[2-(2, 6-Dimethyl-morpholin-4~yf)-ethoxy]"2-thieno[3l 2- c]pyridin-6-yl-chromen-4-one oxime, hydrochloride was prepared in 48% overall yield using the method described in example 127, starting from 6-Hydroxy-2- thieno[3,2-c]pyridin~6-yl-chromen-4-one O-tert-butyl-oxime (example 127B) and 4-(2- Chioro-ethyi)-2,6-dimethyl-morpholine hydrochloride (JACS 1949, 500-505; US2006/14948).
HCi
Figure imgf000146_0002
. 2.2.2 ~~ 225 ^ »
Figure imgf000146_0003
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.33 (br. s, 1 H), 1 1.08 (br. s, 1 H), 9.27 (s, 1 H), 8.78 (s, 1 H), 8.06 (d, J = 5.4 Hz, 1 H), 7.73 (d, J = 5.4 Hz, 1 H), 7.70 (s, 1 H), 7.51 (d, J = 9.1 Hz, 1 H), 7.42 (d, J = 3.0 Hz, 1 H), 7.24 (dd, J = 9.1 Hz, J = 3.1 Hz, 1 H), 4.02 (t, 6.4 Hz, 2H), 3.56-3.52 (m, 4H), 2.82-2.73 (m, 2H).
Example 133: 6-[2-(3, 3-Difluoro-pyrrolidin- 1-yl)-ethoxy]-2-thieno[3, 2- cJpyridin-6-yl-chromen-4-one oxime, hydrochloride was prepared in 62% overall yield using the method described in example 127, starting from 6-Hydroxy-2- thieno[3,2-c]pyridin-6-yl-chromen-4-one O-tert-butyl-oxime (example 127B) and 1-(2- Chloro-ethyl)-3,3-difiuoro-pyrrolidine hydrochloride (W02008/86404).
Figure imgf000147_0001
Mp: 226°C - 229°C.
MS (ESI+): 444.2 [C22H19F2N303S+H]+ (m/z),
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.06 (br. s, 1 H), 9.27 (s, 1 H), 8.78 (s, 1 H), 8.06 (d, J = 5.4 Hz, 1 H), 7.73 (d, J = 5.4 Hz, 1 H), 7.70 (s, 1 H), 7.51 (d, J = 9.1 Hz, 1 H), 7.42 (d, J = 3.0 Hz, 1 H), 7.26 (dd, J = 9.1 Hz, J = 3.1 Hz, 1 H), 4.45-3.73 (m, 10H).
Example 134: 6-(3-Pyridin-4-yl-propoxy)-2-thieno[3,2-c]pyridin-6-yl- chromen-4-one oxime, hydrochloride was prepared in 7% overall yield using the method described in example 127, starting from 6-hydroxy-2-thieno[3,2-c]pyrtdin-6-yl- chromen-4-one O-ierf-buiyl oxime (example 127A) and 4-(3-Chloro-propyl)-pyridine hydrochloride, prepared from 4-pyridine propanol (US 6,362,336).
HCI
Figure imgf000147_0002
Mp: 236°C - 238°C.
Figure imgf000147_0003
1 H NMR: (300 MHz) DMSO-d65 (ppm): 11.02 (br. s, 1 H), 9.25 (s, 1 H), 8.84 (d, J = 6.6 Hz, 2H), 8.76 (s, 1 H), 8.03 (dd, J = 8.8 Hz, J = 5.3 Hz, 3H), 7.72 (d, J = 5.4 Hz, 1 H), 7.67 (s, 1 H), 7.45 (d, J = 9.0 Hz, 1 H), 7.30 (d, J = 3.0 Hz, 1 H), 7.11 (dd, J = 9.2 Hz, J = 3.0 Hz, 1 H), 4.07 (t, J = 6.0 Hz, 2H), 3.09 (t, J = 7.3 Hz, 2H), 2.18 (q, J = 7.7 Hz, 2H). Example 135: 6-(3-Pyridin-3-yl-propoxy)-2-thieno[3,2-c]pyridin~6~yl- chromen-4-one oxime, hydrochloride was prepared in 10% overail yield using the method described in example 127, starting from 6-hydroxy-2-thieno[3,2-c]pyridin-6~y!- chromen-4-one O-terf-butyl oxime (example 127A) and 3-(3-Chloro-propyl)-pyridine hydrochloride, prepared from 3-pyridine propanoi (US 6,362,336).
Figure imgf000148_0001
Mp: 252°C - 254°C.
MS (ES1+): 430.2 [024Η19Ν3033+Η]+ (m/z).
1 H NMR: {300 MHz) DMSO-d6 δ (ppm): 11.04 (br. s, 1 H), 9.25 (s, 1 H), 8.89 (s, 1 H), 8.80 (d, J = 5.4 Hz, 1 H), 8.77 (s, 1 H), 8.54 (d, J = 7.9 Hz, 1 H), 8.06-8.00 (m, 2H), 7.73 (d, J = 5.0 Hz, 1 H), 7.68 (s, 1 H), 7.45 (d, J = 9.0 Hz, 1 H), 7.31 (d, J = 3.0 Hz, 1 H), 7.1 1 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.07 (t, J = 6.0 Hz, 2H), 3.01 (t, J = 7.3 Hz, 2H), 2.16 (q, J = 7.3 Hz, 2H).
Example 136: 6-(2'Pyridin-4-yl-ethoxy)'2~pyrrolo[ 1,2-cJpyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 30% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolof 1 ,2-c]pyrimidin-3- yl-chromen-4-one O-terf-butyl oxime (example 81A) and 4-(2-Chioro-ethyi)-pyridine.
Figure imgf000148_0002
Mp: >270°C dec.
MS (ESI+): 399.2 [C24H1SN403+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 5 (ppm): 10.86 (br. s, 1 H), 9.60 (br.s, 1 H), 9.19 (s, 1 H), 8.84 (d, J = 6.57, 2H), 8.00 (m, 3H), 7.41 (s, 1 H), 7.27 (m, 2H), 6.93 (dd, J = 8.9 Hz, J = 3.0 Hz, 1 H), 6.76 (d, J = 3.8 Hz, 1 H), 6.67 (d, J = 3.8 Hz, 1 H), 3.46 (m, 2H), 3.39 (m, 2H).
Example 137: 2-Pyrrolo[1, 2-c]pyrimidin-3-yl- 6-[2-(4-trifluoromethyl- phenyl)-ethoxy]-chromen-4-one, oxime was prepared in 7% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-iert-butyl oxime (example 81 A) and 1-(2-Bromo-ethyl)-4- trifiuaromethyl-benzene.
Figure imgf000149_0001
Mp: 214DC - 215°C.
MS (ESi+): 466.2 [CMHI BFSNSOS+H]* (m/z).
1 H N R: (300 MHz) DMSO-de δ (ppm): 10.90 (s, 1 H), 9.20 (s, 1 H), 8.02 (s, 1 H), 7.81 (s, 1 H), 7.68 (d, J = 8.1 Hz, 2H), 7.57 (d, J = 8.1 Hz, 2H), 7.42 (s, 1 H), 7.38 (d, J = 9.1 Hz, 1 H), 7.31 (d, J = 3.0 Hz, 1 H), 7.10 (dd, J = 9.1 Hz, J = 3.0 Hz, 1 H), 6.97 (dd, J = 8.9 Hz, J = 3.0 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.29 (t, J = 6.5 Hz, 2H), 3.16 (t, J = 6.4 Hz, 2H).
Example 138: 6-[2-(3~Fluoro-phenyl)-ethoxy]'2-pyrrolo[ 1 ,2-c}pynmidin-3- yl-chromen-4-one, oxime was prepared in 44% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrroio[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-f erf-butyl oxime (example 81 A) and 1 -(2-Bromo~ethyl)-3-fluoro~ benzene.
Figure imgf000149_0002
Mp: 205°C - 207°C.
MS (ESI+): 416.2 [C24H18FN303+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-de 5 (ppm): 10.90 (s, 1 H), 9.20 (s, 1 H), 8.02 (s, 1H), 7.81 (s, 1H), 7.42-7.30 (m, 4H), 7.21 -6.97 (m, 5H), 6.73 (d, J = 3.8 Hz, 1H), 4.25 (t, J = 6.6 Hz, 2H), 3.08 (t, J = 6.5 Hz, 2H).
Example 139: 5-Met oxy~2-pyrrolo[1,2-c]pyrimidin-3~yl-c romen-4-one oxime was prepared in 8% overali yield using methods A and C, starting from 6'- methoxy-2'-hydroxy-acetophenone and pyrrolo[1,2-c]pyrimidine~3-carboxylic acid methyl ester.
Figure imgf000150_0001
Mp; 240°C - 243°C.
MS (ESI+): 308.1 [C17H13N303+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.11 (s, 1H), 9.19 (s, 1H), 7.99 (s, 1H), 7.79 (s, 1H), 7.54 (s, 1H), 7.41 (t, J = 8.3 Hz, 1H), 7.21-6.97 (m, 5H), 7.01 (d, J = 8.1 Hz, 1H), 6.96 (t, J = 3 Hz, 1H), 6.90 (d, J = 8.3 Hz, 1H), 6.70 (d, J = 3.4 Hz, 1H), 3.80 (s, 3H).
Example 140: 6~[2-(4~Chloro^henyl)-ethoxy]-2^yrrolo[1,2~c]pyrimidin~3- yl-chromen-4-one oxime was prepared in 28% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1,2-c]pyrimidin-3-yl- chromen-4-one O-ferf-buty! oxime (example 81 A) and 1-(2-Bromo-ethyl)-4-chSoro- benzene.
Figure imgf000150_0002
Mp: 247°C - 249°C.
MS (ESI+): 432.2 [C24H,8C!N303+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.90 (s, 1H), 9.20 (s, 1H), 8.02 (s, 1 H), 7.81 (s, 1 H), 7.42 (s, 1 H), 7.38 (m, 5H), 7.30 (d, J = 3,0 Hz, 1 H), 7.10 (dd, J = 9.1 Hz, J = 3.0 Hz, 1 H), 6.97 (dd, J = 8.9 Hz, J = 3.0 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.23 (t, J = 6.6 Hz, 2H), 3.05 (t, J = 6.5 Hz, 2H).
Example 141: 2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-6-[2-(3-trifluoromethyl- phenyl)-ethoxy]-chromen-4-one oxime was prepared in 22% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yl-chromen-4-one O-iert-butyi oxime (example 81 A) and 1 -(2-Bromo-ethy!)-3- trifluoromethyl-benzene.
Figure imgf000151_0001
p: 217°C - 218°C.
MS (ESi+): 466.2 [C25H18F3N303+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.90 (s, 1 H), 9.20 (s, 1 H), 8.02 (s, 1 H), 7.81 (s, 1 H), 7.72 (s, 1 H), 7.67 (d, J = 7.0 Hz, 1 H), 7.61 -7.55 (m, 2H), 7.42 (s, 1 H), 7.39 (d, J = 9.1 Hz, 1 H), 7.31 (d, J = 3.0 Hz, 1 H), 7.10 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.97 (dd, J = 3.8 Hz, J = 2.9 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.28 (t, J - 6.6 Hz, 2H), 3.16 (t, J = 6.5 Hz, 2H).
Example 142: 7-(2-Morpholin-4-yl-ethoxy)-2-pyrrolo[1,2-c]pyrimidin-3-yl' chromen-4-one oxime, hydrochloride was prepared in 75% yield using method D (step 2), starting from 7-(2-Morpholin-4-yi-ethoxy)-2-pyrrolo[1 ,2-c]pyrimsdsn-3-yl- chromen-4-one O-tert-butyl-oxime (example 142A). The hydrochloride salt of the title compound was isolated as a yellow solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanol.
Figure imgf000151_0002
MS (ESI+): 407.0 [C22H22N404+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-de δ (ppm): 1 1.37 (br. s, 1 H), 10.94 (br. s, 1 H), 9.24 (s, 1 H), 8.08 (s, 1 H), 7.87 (d, J = 8.8 Hz, 2H), 7.84 (s, 1 H), 7.47 (s, 1 H), 7.18 (s, 1 H), 7.01 -6.96 (m, 2H), 6.75 (d, J = 1.6 Hz, 1 H), 4.56 (t, J = 4.8 Hz, 1 H), 3.99 (m, 2H), 3.87 (m, 2H), 3.60 (m, 2H), 3.53 (m, 2H), 3.25 (m, 2H).
Example 142 A : 7-(2"Morpholin~4-y!-ethoxy)-2-pyrrolo[1f2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 57% yield using the procedure described in example 85A, starting from 7-Hydroxy- pyrrolo[1 ,2-c]pyrimidin-3-yi- chromen-4-one O-tert-butyl-oxime O-tert-butyl-oxime (example 142B) and 4-(2- chloroethyl) morphoiine hydrochloride. The title compound was isolated as a yellow solid.
Figure imgf000152_0001
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.79 (s, 1 H), 7.96 (d, J = 8.9 Hz, 1 H), 7.88 (s, 1 H), 7^50 (s, 1 H), 7.44 (d, J = 2.6 Hz, 1 H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.80-6.75 (m, 2H), 6.62 (d, J = 3.9 Hz, 1 H), 4.17 (t, J = 5.6 Hz, 2H), 3.75 (t, J = 4.7 Hz, 4H), 2.84 (t, J = 5.6 Hz, 2H), 2.60 (t, J = 4.5 Hz, 4H), 1.40 (s, 9H).
Exampl 142B 7-Hydroxy-2-pyrrolo[ 1, 2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime was prepared in 78% yield using the procedure described in example 18A, starting from 7-Bromo-2-pyrroio[1 !2-c]pyrimidin-3-yl-chromen-4-one O- tert-butyl-oxime (example 142C). The title compound was isolated as an orange solid.
Figure imgf000152_0002
1 H NMR: (300 MHz) CHCl3-d1 δ (ppm): 8.78 (s, 1 H), 7.92 (d, J = 8.4 Hz, 1 H), 7.49 (s, 1 H), 7.44 (s, 1 H), 7.43 (s, 1 H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.71 -6.61 (m, 3H), 5.53 (br s, 1 H), 1.39 (s, 9H).
Example 142C: 7-Bromo-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one O- tert-butyl-oxime was prepared in 64% overall yield using methods A and D (step1), starting from 4'-bromo-2'-hydroxy-acetophenone and pyrrolo[1 ,2-c]pyrimidine-3- carboxylic acid methyl ester.
Figure imgf000153_0001
1 H NMR: (300 MHz) CHCl3-d1 δ (ppm): 8.79 (s, 1 H), 7.91 (d, J = 6.4 Hz, 1 H), 7.50 (s, 1 H), 7.45 (dd, J = 3.9 Hz, J = 2.8 Hz, 2H), 7.29 (dd, J = 6.4 Hz, J = 1.3 Hz, 1 H), 6.94-6.92 (m, 2H), 6.64 (d, J = 3.0 Hz, 1 H), 1.40 (s, 9H).
Example 143: 7-(3-Morpholin-4-yl-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yf- chromen-4-one oxime hydrochloride was prepared in 71 % yield using method D (step 2), starting from 7-(3-Morpholin-4-y!-propoxy)-2-pyrrolo[1 ,2~c]pyrimidin-3-yi- chromen-4-one O-tert-butyl-oxime (example 143A). The hydrochloride salt of the title compound was isolated as a yellow solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanoi.
Figure imgf000153_0002
WIS (ESI+): 421.0 [C^H^C^+H]* (m/z).
1 H NMR: (400 MHz) DMSO-de δ (ppm): 10.99 (br. s, 1 H), 10.85 (br. s, 1 H), 9.24 (s, 1 H), 8.06 (s, 1 H), 7.85-7.83 (m, 2H), 7.46 (s, 1 H), 7.10 (s, 1 H), 7.01 -6.99 (m, 1 H), 6.91 (dd, J - 8.8 Hz, J = 2.8 Hz, 1 H), 6.75 (s, 1 H), 3.99 (d, J = 12.8 Hz, 2H), 3.82 (t, J = 1 1.2 Hz, 2H), 3.48 (d, J = 12.0 Hz, 2H), 3.30-3.25 (m, 2H), 3.14-3.06 (m, 2H), 2.29-2.22 (m, 2H).
Example 143A: 7-(3-Morpholin-4-yl-propoxy)-2-pyrrolo[ 1, 2-c]pyrimidin-3- yl-chromen-4-one O-tert-butyi-oxime was prepared in 55 % yield using the method described in example 87A, starting from 7-(3-Chioro-propoxy)-2-pyrrolo[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-tert-butyl oxime (example 143B) and morpholine.
Figure imgf000154_0001
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 8.79 (s, 1 H), 7.95 (d, J = 9.2 Hz, 1 H), 7.88 (s, 1 H), 7.50 (s, 1 H), 7.44 (d, J = 2.6 Hz, 1 H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.79-6.76 (m, 2H), 6.61 (d, J = 3.7 Hz, 1 H), 4.08 (t, J = 6.4 Hz, 2H), 3.74 (t, J = 4.7 Hz, 4H), 2.54 (t, J = 6.9 Hz, 2H), 2.48 (t, J = 4.5 Hz, 4H), 2.00 (quint, J = 6.4 Hz, 2H), 1.40 (s, 9H).
Example 143B: 7-(3-Chloro-propoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime was prepared in 100% yield using the method described in example 85A, starting from 7-Hydroxy- pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl-oxime (example 142B) and 1 -bromo-3-chloropropane in dimethylformamide. The title compound was isolated as a brown solid.
Figure imgf000154_0002
1 H NMR: (300 MHz) CHCh-d^ δ (ppm): 8.79 (s, 1 H), 7.96 (d, J = 9.4 Hz, H), 7.89 (s, 1 H), 7.50 (s, 1H), 7.44 (d, J = 2.4 Hz, 1 H), 7.46 (s, 1 H), 7.10 (s, 1 H), 7.01 - 6.99 (m, 1 H), 6.91 (dd, J = 8.8 Hz, J = 2.8 Hz, 1 H), 4.18 (t, J = 5.8 Hz, 2H), 3.77 (t, J = 6.4 Hz, 2H), 2.28 (quint, J = 6.0 Hz, 2H), 1.40 (s, 9H).
Example 144: 6-[(4-Fluoro-benzylamm' o)-methyl]-2'pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one oxime hydrochloride was prepared in 54% yield using method D (step 2), starting from 6-[(4- uoro-benzytamino)-methyl]-2- pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 44B). The hydrochloride salt of the title compound was isolated as a red solid after treatment with a 1.25 M solution of hydrogen chloride in Isopropanol.
Figure imgf000155_0001
p: >210°C dec.
MS (ESI+): 415.2 [C24H1SFN402+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d65 (ppm): 11.04 (br. s, 1 H), 9.58 (br. s, 2H), 9.22 (s, 1 H), 8.04 (s, 2H), 7.83 (s, 1 H), 7.66-7.60 (m, 3H), 7.50 (d, J = 9.6 Hz, 1 H), 7.48 (s, 1 H), 7.28 (t, J = 8.8 Hz, 2H), 6.99 (t, J = 3.6 Hz, 1 H), 6.74 (d, J = 3.8 Hz, 1 H), 4.20 (s, 4H).
Example 144A: 6-[(4-Fluoro-benzylamino)-methyI]-2-pyrrolo[1,2- c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime
Figure imgf000155_0002
To a solution of 4-tert-Butoxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-4H-chromene- 6-carbaldehyde (example 144B) (50 mg; 0.14 mmol) in dichloromethane (5m!), was added molecular sieves and 4-Fluoro-benzylamine (19 mg; 0.15 mmol). After 2 hours at room temperature, sodium triacetoxyborohydride (29 mg, 0.14 mmol) and acetique acid (2ml), were added. After stirring at room temperature for 18 hours, the reaction mixture was quenched with a saturated solution of sodium hydrogenocarbonate and extracted with ethyl acetate. The organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-100%) to afford the title compound (20 mg, 30%) as a green solid.
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 8.79 (s, 1 H), 7.96 (s, 1 H), 7.90 (s, 1 H), 7.50 (s, 1 H), 7.45-7.33 (m, 4H), 7.23 (d, J = 8.4 Hz, 1 H), 7.05-7.00 (m, 2H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.63 (d, J = 3.7 Hz, 1 H), 3.83 (s, 2H), 3.81 (s, 2H), 2.30 (br s, 1 H), 1.42 (s, 9H).
Example 144B: 4~tert~Butoxyimmo-2-pyrrolo[1,2-c]pyrimidin-3-yl-4H- chrom ene-6- carbaldehyde
Figure imgf000156_0001
To a solution of 6-Hydroxymethyl-2-pyrrolo[1 ,2-cjpyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 144C) (575 mg, 1.58 mmol) in dichloromethane (60 ml), was added a Dess Martin solution (15 wt.% in DCM) (6.7 ml; 2.37 mmol). After 1 hour stirring at room temperature, an additionnel amount of Dess Martin solution (6 ml) was added to complete the reaction. The reaction mixture was quenched with water and added to a 1 :1 solution of saturated sodium hydrogenocarbonate and 5% aq solution of Na2S203. After extraction with ethyl acetate, the organic layers were washed with water, brine, dried over sodium sulfate and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-40%) to afford the title compound (355 mg, 62%) as a yellow solid.
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 10.03 (s, 1 H), 8.81 (s, 1 H), 8.56 (d, J = 2.0 Hz, 1 H), 8.04 (s, 1H), 7.94 (dd, J = 8.6 Hz, J = 1 ,9 Hz, 1 H), 7.55 (s, 1H), 7.47 (dd, J = 2.4 Hz, 1 H), 7.37 (d, J = 8.4 Hz, 1 H), 6.95 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.67 (d, J = 3.7 Hz, 1 H), 1.43 (s, 9H).
Example 144C: 6-Hydroxym ethyl-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl-chromen- 4-one O-tert-butyl-oxime
Figure imgf000156_0002
To a solution of 4-(tert-Butoxyimino)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H- chromene-6-carboxy!ic acid methyl ester (example 1 15B) (574 mg, 1.46 mmol) in dichloromethane (9 ml) at 0°C was added a 1 M solution of DIBAL-H in THF (12.8 ml). After 2 hours at 0°C an additional amount of 1 M solution of DIBAL-H in THF (6 mi) was added to complete the reaction. After hydrolysis with a 1 aqueous solution of hydrogen chloride, the reaction mixture was extracted with ethyl acetate. The organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated. The product was used without further purification.
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.79 (s, 1 H), 8.05 (d, J = 1.9 Hz, 1 H), 7.91 (s, 1 H), 7.52 (s, 1 H), 7.45-7.41 (m, 2H), 7.27-7.24 (m, 2H), 6.92 (dd, J = 3.7 Hz, J = 2.8 Hz, 1 H), 6.64 (d, J = 3.7 Hz, 1 H), 4.73 (s, 2H), 1.42 (s, 9H).
Example 145: 6-{[2-(4-Fluoro-phenyl)-ethylamino]-methyl}-2-pyrrolo[ 1,2- cJpyrimidin-3-yl-chromen-4-one oxime hydrochloride was prepared in 24% overai! yield using the method described in example 144, starting from 4-tert-Butoxyimino-2- pyrrolo[1 ,2-c]pyrim!din-3-yl-4H-chromene-6-carbaldehyde (example 144B) and 4- fluorophenethylamine.
Figure imgf000157_0001
HPLC (gradient 15 % - 75 % MeOH / H20 + 0.05% TFA): > 95%; RT = 10.51 min.
MS (ESI+): 429.0 [C25H2iFN4Oz+H]+ (m/z).
Example 146: 6-Phenethyloxy-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl-chromen-4- one oxime was prepared in 21 % overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yI-chromen-4-one O-ferf-buty! oxime (example 81 A) and (2-Bromo-ethyJ)-benzene.
Figure imgf000157_0002
Mp: 210°C - 212°C.
MS (ES!+): 398.2 [024Η19Ν303+Η]+ (m/z),
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.90 (s, 1 H), 9.21 (s, 1 H), 8.02 (s, 1 H), 7.81 (s, 1 H), 7.42-7.23 (m, 8H), 7.10 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.98 (s, 1 H), 6.74 (d, J = 3.8 Hz, 1 H), 4.23 (t, J = 6.7 Hz, 2H) 3.05 (t, J = 6.5 Hz, 2H). Example 147; 6-[2-(Pyridin~4-yloxy)-ethoxy]~2~pyrrolo[ 1,2-c]pyrimidin~3- yl-chromen-4-one oxime was prepared in 17% yield using method D (step 2), starting from 6-[2-(Pyridin-4-yloxy)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-y!-chromen-4- one O-f erf-butyl oxime (example 147 A).
Figure imgf000158_0001
Mp: 198DC - 200°C.
MS (ESI+): 415.2 [C23H18N404+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d65 (ppm): 10.97 (br. s, 1 H), 9.22 (s, 1 H), 8.79 (d, J = 7.32 Hz, 2H), 8.05 (s, 1 H), 7.82 (d, J = 2.5 Hz, 1 H), 7.67 (d, J = 7.4 Hz, 2H), 7.45- 7.42 (m, 2H), 7.36 (d, J = 3.0 Hz, 1 H), 7.16 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.98 (dd, J = 3.8 Hz, J = 2.9 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.74 (s, 2H), 4.46 (s, 2H).
Example 147 A: 6-[2-(Pyridin-4-yloxy)-ethoxy]-2-pyrrolo[ 1r2-c]pyrimidin-3- yl-chromen-4-one O-ferf-butyi oxime ^^i N(O-tButyl)
Figure imgf000158_0002
To a mixture of sodium hydride 60% in oil (1 1.5 mg, 0.28 mmol) in NMP (0.5 ml) was added at 0°C a solution of 6-hydroxy-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen- 4-one O-tert-butyl-oxime (example 81 A) {100 mg, 0.28 mmol) in NMP (0.5 ml). After 1 hour at room temperature, a solution of 4-(2-Chloro-ethoxy)-pyridine (103 mg, 042 mmol), tetrabutyl ammonium iodide (53 mg, 0.14 mmol), 15-crown-5 (63 mg, 0.28 mmol) in NMP (0.5 ml) was added at 0°C. After stirring at room temperature for 24 hours, the reaction mixture was diluted with a saturated solution of ammonium chloride and extracted with ethyl acetate. The organic layers were washed with a saturated solution of ammonium chloride, water, brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-70%) to yield the title compound (54 mg, 42%) as a yellow solid. 1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.79 (s, 1 H), 8.41 (d, J = 2.8 Hz, 1 H), 8.27 (dd, J = 4.5 Hz, J = 1.3 Hz, 1 H), 7.90 (s, 1 H), 7.57 (d, J = 3.0 Hz, 1 H), 7.50 (s, 1 H), 7.44 (d, J = 2.4 Hz, 1 H), 7.36-7.28 (m, 2H), 7.22 (d, J = 9.0 Hz, 1 H), 7.02 (dd, J = 9.0 Hz, J - 3.0 Hz, 1 H), 6.92 (dd, J = 3.8 Hz, J = 2.8 Hz, 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 4.42 (s, 4H), 1.41 (s, 9H).
Example 148: 6-[2-(Pyridin-3-yloxy)-ethoxy]-2-pyrrolo[ 1,2-c]pyrimidin-3- yI-chromen-4-one oxime was prepared in 16% overall yield using the method described in example 147, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-teri-buty!-oxime (example 81A) and 3-(2-Chloro-ethoxy)-pyridine.
Figure imgf000159_0001
Mp: 170°C - 175°C.
MS (ESI+): 415.2 [C23H18N404+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.97 (br. s, 1 H), 9.22 (s, 1 H), 8.64 (s, 1 H), 8.42 (d, J = 5.13 Hz, 1 H), 8.05 (s, 1 H), 7.99 (d, J = 6.63 Hz, 1 H), 7.82-7.74 (m, 2H), 7.45-7.42 (m, 2H), 7.36 (d, J = 3.0 Hz, 1 H), 7.16 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 6.98 (m, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.54 (m, 2H), 4.74 (m, 2H).
Example 149: 6-[3-(Pyridin-3-yloxy)-propoxy]-2-pyrrolo[ 1,2-c]pynmidin-3- yl~chromen~4~one oxime was prepared in 9% overall yield using the method described in example 147, starting from 6-hydroxy-2-pyrroio[1 ,2-c]pyrjrnidin-3-yi- chromen-4-one O-tert-butyl-oxime (example 81 A) and 3-(3-Chioro-propoxy)-pyridine.
Figure imgf000159_0002
Mp: 202°C - 205°C.
MS (ESI+): 429.2 [C24H20N4CVH]+ (m/z). 1 H NMR: (300 MHz) DMSO-de 8 (ppm): 10.91 (s, 1 H), 9.21 (s, 1H), 8.32 (d, J = 2.8 Hz, 1.H), 8.17 (dd, J = 4.6 Hz, J = 1.2 Hz, 1 H), 8.03 (s, 1 H), 7.89 (s, 1 H), 7.43-7.18 (m, 5H), 7.20 (dd, J = 9.1 Hz, J = 3.0 Hz, 1 H), 6.97 (t, J - 3.0 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.25-4.18 (m, 4H), 2.21 (t, J = 6.3 Hz, 2H).
Example 150: N'(4-Fluoro-phenyl)-2-{4-hydroxyimino-2-pyrrolo[ 1,2- c]pyrimidin-3-yl-4H-chromen-6-yloxy}-acetamide was prepared in 41 % overall yield using the method described in example 147, starting from 6-hydroxy-2- pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-buiyl-oxime (example 81 A) and 2- Bromo-N-(4-fluoro-phenyl)-acetamide (Bioorg. Med. Chem. 1 1 (2003) 2769-2782).
Figure imgf000160_0001
Mp: 278°C - 270°C.
MS (ES!+): 445.2 [C2 Hi7F 404+H]+ (m/z).
1H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.97 (s, 1 H), 10.19 (s, 1 H), 9.22 (s, 1 H), 8.03 (s, 1 H), 7.81 (d, J = 2.6 Hz, 1 H), 7.69-7.64 (m, 2H), 7.46-7.38 (m, 3H), 7.23- 7.14 (m, 3H), 6.98 (dd, J = 3.8 Hz, J = 2.9 Hz, 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.75 (s, 2H).
Example 151: N-(4-Fluoro~phenyl)-2-{4-hydroxyimino-2-pyrrolo[ 1,2- c]pyrimidin-3-yl-4H-chromen-6-yloxy}-N-methyI-acetamide. was prepared in 90% yield using method D (step 2), starting from 2-{4-tert-Butoxyimino-2-pyrro!o[1 ,2- c]pyrimidin-3-yl-4H-chromen-6-yloxy}-N-(4-fluoro-phenyl)-N-methyl-acetamide
(example 151A).
Figure imgf000160_0002
Mp: 268°C - 272°C.
MS (ES1+): 459.2 [C25H19FN404+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.95 (s, 1 H), 9.21 (s, 1 H), 8.02 (s, 1 H), 7.81 (d, 2.6 Hz, 1 H), 7.54 (br. s, 2H), 7.42 (s, 1 H), 7.38-7.33 (m, 3H), 7.1 (br. s, 1 H), 6.97 (t, J = 3.0 Hz, 2H), 6.73 (d, J = 3.8 Hz, 1 H), 4.48 (s, 2H), 3.18 (s, 3H).
Example 151 A: 2-{4-tert-Butoxyimino-2-pyrrolo[ 1,2-c]pyrimidin-3-yl-4H- chromen-6-yloxy}~N-(4-fluoro-phenyl)-N-methyl-acetamide To a solution of 2-{4- tert~Butoxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yl~4H-chrorrten-6-yloxy}-N-(4--fluoro- phenyl)-acetamide (tert-butyl protected oxime of exampie 150) (220 mg; 0.44 mmoi) in dimethylformamide (4 mi), was added at 0°C NaH 60% in oil (21 mg; 0.53 mmoi). After 1 hour at room temperature, lodomethane (41 μΙ; 0.66 mmoi) was added at 0°C. After stirring at room temperature for 18 hours, water was added at 0°C and the resulting precipitate was fi!tred and purified by flash chromatography over silica gel (gradient dichloromethane/ethyl acetate: 0-4%) to yield the title compound (152 mg, 67%) as a yellow solid.
Figure imgf000161_0001
1 H NMR: (300 MHz) CHC^-d, δ (ppm): 8.78 (s, 1 H), 7.89 (s, 1 H), 7.47 (s, 1 H), 7.44 (d, 2.8 Hz, 1 H), 7.33-7.29 (m, 3H), 7.18-7.12 (m, 3H), 6.97 (dd, J = 8.8 Hz, J = 2.8 Hz, 1 H), 6.92 (t, J = 3.8 Hz, 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 4.44 (s, 2H), 3.32 (s, 3H), 1.41 (s, 9H).
Example 152: N-(5-Fl oro-pyridin~2-y})-2-{4~hydroxyimino-2'pyrrolo[ 1,2- c]pyrimidin-3-yI-4H-chromen-6-yloxy}-acetamide was prepared in 2% overall yield using the method described in example 147, starting from 6-hydroxy-2-pyrrolo[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 81A) and 2-Bromo-N-(5- fluoro-pyridin-2-yl)-acetamide (Bioorg. Med. Chem. 1 1 (2003) 2769-2782).
Figure imgf000162_0001
MS (ESI+); 446.0 [C23H,6F 504+H]+ (m/z).
1H NMR: (400 MHz) DMSO-d65 (ppm): 11.00 (br. S, 1H), 10.80 (s, 1H), 9.27 (s, 1H), 8.42 (s, 1H), 8.19 {dd, 7.6 Hz, 1H), 8.10 (s, 1H), 7.87-7.80 (m, 2H), 7.50 (d, J = 7.2 Hz, 1H), 7.48 (s, 1H), 7.39 (d, J = 3.2 Hz, 1H), 7.25 (dd, J - 8.8 Hz, J = 2.8 Hz, 1H), 7.04 (d, J = 3.6 Hz, 1H), 6.80 (s, 1H), 4.91 (s, 2H).
Example 153: 2-Pyrrolo[ 1,2-c]pyrimidin-3-yl-5-trifluoromethyl-chromen-4- one oxime was prepared in 3% overall yield using methods A and C, starting from 6'- trif!uoromethyl-2'-hydroxy-acetophenone and pyrrolo[1 ,2-c]pyrimidine-3-carboxylic acid methyl ester.
Figure imgf000162_0002
Mp: 255°C - 257°C.
MS (ESI+): 346.1 [C17H10F3N3O2+H]+ (m/z).
1H NMR; (300 MHz) DMSO-d6 δ (ppm): 11.34 (s, 1H), 10.19 (s, 1H), 9.23 (s, 1H), 8.15 (s, 1H), 8.07 (d, J = 8.3 Hz, 1H), 7.90 (s, 1H), 7.83 (s, 1H), 7.60 (d, J = 8.3 Hz, 1H), 7.46 (s, 1H), 7.00 (t, J = 3.2 Hz, 1H), 6.73 (d, J - 3.8 Hz, 1H).
Example 154: 2-(7-tert-Butyl-pyrrolo[ 1,2-c]pyrimidin-3-yl)-6-(2-morpholin- 4-yl-ethoxy)-chromen-4-one oxime, hydrochloride is isolated as a by product from the reaction described in example 85.
Figure imgf000163_0001
MS (ESI+): 463.0 [C26H3o 404+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-ds δ (ppm): 1 1.10 (br. s, 1 H), 10.95 (br. s, 1 H), 9.36 (s, 1 H), 8.05 (s, 1 H), 7.48-7.41 (m, 3H), 7.22 (dd, J = 9.2 Hz, J = 3.2 Hz, 1 H), 6.78 (s, 1 H), 6.72 (s, 1 H), 4,48 (t, J = 4.4 Hz, 2H), 3.81 (t, J = 7.2 Hz, 2H), 3.59-3.50 (m, 4H), 3.25-3.17 (m, 4H), 1.44 (s, 9H).
Example 155: 7-( 2-Morpholin-4-yl-ethoxy)-2-thieno[2, 3-c]pyridin-5-yl- chromen-4-one oxime, hydrochloride was prepared in 40% overali yield using the methode described in example 142, starting from 4'-bromo-2'-hydroxy-acetophenone and Thieno[2,3-c]pyridine-5-carboxyiic acid methyl ester.
Figure imgf000163_0002
MS (ESI+): 424.0 [C22H21IM304S+H]+ (m/z).
1 H NMR: (400 MHz) DMSOd6 δ (ppm): 10.99 (br. s, 1 H), 10.88 (br. s, 1 H), 9.36 (s, 1 H), 8.51 (s, 1 H), 8.20 (d, J = 5.2 Hz, 1 H), 7.73 (d, J = 8.8 Hz, 1 H), 7.64 (d, J = 5.6 Hz, 1 H), 7.28 (s, 1 H), 6.65 (s, 1 H), 6.59 (dd, J = 8.8 Hz, J = 2.4 Hz, 1 H), 4.39 (m, 2H), 3.90 (d, J = 11.6 Hz, 2H), 3.73 (t, J = 12.0 Hz, 2H), 3.50 (s, 2H), 3.41 (d, J = 12.4 Hz, 2H), 3.16-3.11 (m, 2H).
Example 156: 5-(2-Morpholin'4-yf~ethoxy)-2-pyrrolo[1,2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 12% overall yield using methods A and C, starting from 2,-hydroxy-6'-(2-rnorpholino-4-yl-ethoxy)- acetophenone (example 156A) and pyrroio[1 ,2-c]pyrim!dine-3-carboxyiic acid methyl ester. The hydrochloride salt of the title compound was isolated as a yellow solid after treatment with a 1.25 M solution of hydrogen chloride in isopropanol.
Figure imgf000164_0001
Mp: 200°C - 205°C.
MS (ESI+): 407.2 [C22H22N404+H (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 10.09 (s, 1 H), 9.24 (s, 1 H), 8.1 1 (s, 1 H), 7.85 <d, J = 2.5 Hz, 1 H), 7,63 (s, 1 H), 7.55 (t, J = 7.28 Hz, 1 H), 7.20 (d, J = 8.28 Hz, 1 H), 7.05 (d, J = 8.3 Hz, 1 H), 7.00 (t, J = 3.8 Hz, 1 H), 6.78 (d, J = 3.8 Hz, 1 H), 4.56 (s, 2H), 3.91 (s, 4H), 3.61 (s, 2H), 3.47 (br. s, 4H).
Example 156 A : 2'-hydroxy-6'-(2-morpholino-4-yl-ethoxy)-acetophenone. in a sealed tube, to a solution of 6'-hydroxy-2'-methoxy-acetophenone (620 mg, 4.075 mmol) in butanone (20 mi) were added potassium carbonate (850 mg, 6.11 mmol) and 2-chioroethyimorpholine hydrochloride (800 mg, 4.28 mmol) and the mixture was stirred at 130°C for 18 h. Water was added and the organics were extracted with ethylacetate, washed with brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography over silica gel (gradient ethyl acetate/methanol: 0-2%) to yield 2'-methoxy-6!-(2-morpholino-4-yl-ethoxy)- acetophenone (1.0 g, 96%) as colorless oil. (1 H NMR (300 MHz) CHCI3-di δ (ppm): 7.24 (t, J = 8.5 Hz, 1 H), 6.56 (d, J = 8.5 Hz, 1 H), 6.53 (d, J = 8.5 Hz, 1 H), 4.1 1 (t, J = 5.7 Hz, 2H), 3.79 (s, 3H), 3.70 (m, 4H), 2.75 (t, J = 5.8 Hz, 2H), 2.54 (m, 4H), 2.48 (s, 3H). To a solution of 2'-methoxy-6'-(2-morpholino-4-yl-ethoxy)-acetophenone (1 .0 g, 3.58 mmol) in dichloromethane (50 ml) at 0°C was added dropwise a 1 M solution of boron tribromide in dichloromethane (14.0 mi, 14.0 mmol). The solution was stirred for 1 hour at this temperature and hydrolyzed with water. After 0.5 hour at room temperature the reaction mixture was cautiously neutralized with a saturated solution of sodium hydrogenocarbonate, and extraxted with dichloromethane. The organic layer was washed with brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography over silica gel (ethyl acetate/ methanol: 0-4%) to yield 2!-hydroxy-6 '-(2-morphoiino-4-yl-ethoxy)-acetophenone (495 mg, 52%) as light yellow oil. .
Figure imgf000165_0001
1 H NMR (300 MHz) CHCI3-di δ (ppm): 13.21 (s, 1 H), 7.32 (t, J = 8.5 Hz, 1 H), 6.56 (d, J = 8.5 Hz, 1 H), 6.36 (d, J = 8.5 Hz, 1 H), 4.16 (t, J = 5.7 Hz, 2H), 3.71 (m, 4H), 2.84 (t, J = 5.8 Hz, 2H), 2.73 (s, 3H), 2.54 (m, 4H).
Example 157: 5-(3-Morpholin-4-yl-propoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one oxime, hydrochloride was prepared in 2% overall yield using methods A and C, starting from 2'-hydroxy-6'-(3-rriorphoiino-4-yl-propoxy)- acetophenone (example 157A) and pyrro!o[1 ,2-c]pyrimidine-3-carboxylic acid methyi ester.
Figure imgf000165_0002
MS (ESI+): 421.0 [C23H24N404+H (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.09 (s, 1 H), 9.24 (s, 1 H), 8.11 (s, 1 H), 7.85 (d, J = 2.5 Hz, 1 H), 7,63 (s, 1 H), 7.55 (t, J = 7.28 Hz, 1 H), 7.20 (d, J = 8.28 Hz, 1 H), 7.05 (d,_ J = 8.3 Hz, 1 H), 7.00 (t, J = 3.8 Hz, 1 H), 6.78 (d, J = 3.8 Hz, 1 H), 4.19-3.93 (m, 8H), 3.25-3.04 (m, 4H), 2.24-2.20 (m, 2H).
Example 157A: 2'-hydroxy-6'-(3-morpholino-4'yl-propoxy)- acetophenone
To a solution of 2',6'-dihydroxy-acetophenone (500 mg, 3.28 mmol) in dimethylformamide (20 ml) were added potassium carbonate (680 mg, 4.93 mmol) and 1-bromo-3-chloropropane (0.34 mi, 3.45 mmo!) and the mixture was stirred at 70°C for 4 h. Water was added and the organics were extracted with ethylacetate, washed with brine, dried over sodium sulfaie and concentrated to dryness. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyi acetate: 0-15%) to yield 6'-(3-ch!oropropoxy)-2'-hydroxy- acetophenone (348 mg, 46%) as a pale yellow solid. To a solution of 6'-(3- chloropropoxy)-2'-hydroxy-acetophenone (345 mg, 1.50 mmol) in acetonitrsle (15 ml) was added potassium carbonate (416 mg, 3.0 mmol) and morpho!ine (0.396 ml, 4.52 mmol). The mixture was stirred for 18 hours at room temperature then 60°C for 3 days. Water was added and the organics were extracted with ethylacetate, washed with brine, dried over sodium sulfate and concentrated to dryness. The residue was purified by flash chromatography over silica gei (gradient ethyl acetate/methanol: 0- 10%) to yield 2'-hydroxy-6'-(3-morpholino-4-y]-propoxy)-acetophenone (171 mg, 40%) as an orange oil that solidified .
Figure imgf000166_0001
(1 H NMR (300 MHz) CHCfe-di δ (ppm): 13.24 (s, 1 H), 7.32 (t, J = 8.3 Hz, 1 H), 6.56 (d, J = 8.4 Hz, 1 H), 6.38 (d, J = 8.4 Hz, 1 H), 4.1 1 (t, J = 6.4 Hz, 2H), 3.73 (m, 4H), 2.69 (s, 3H), 2.55 (t, J = 6.2 Hz, 2H), 2.48 (m, 4H), 2.07 (quint, J = 6.2 Hz, 2H).
Example 158: 6-(2-Morpholm~4-yi-ethoxy)-2-th}eno[2,3-c]pyridin-5-yl- chromen-4'one oxime, hydrochloride was prepared in 41 % overall yield using the methode described in exampfe 127, starting from 5'-bromo-2'-hydroxy-acetophenone and Thieno[2,3-c]pyridine-5-carboxylic acid methyl ester.
Figure imgf000166_0002
Mp: 258°C - 260°C.
MS (ESI+): 424.2 [C22H21N304S+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 11.24 (br. s, 1 H>, 1 1.05 (br. s, 1 H), 9.39 (s, 1 H), 8.52 (s, 1 H), 8.26 (d, J == 5.34 Hz, 1 H), 7.71 (d, J = 5.46, 1 H), 7,69 (s, 1 H), 7.51 (d, 9.0 Hz, 1 H), 7.43 (d, J = 3.0 Hz, 1 H), 7.23 (dd, J = 9.1 Hz, J = 3.0 Hz, 1 H), 4.50 (m, 2H), 3.97 (d, J = 10,4 Hz, 2H), 3.83 {t, J = 1 1.7 Hz, 2H), 3.59-3.49 (m, 4H), 3.23-3.19 (m, 2H).
Example 159: 6-(3-Pyridin-4-yl-propoxy)~2-pyrrolo[ 1,2-c]pyrimidin-3~yl- chromen-4-one oxime, hydrochloride was prepared in 10% overall yield using the method described in example 85, starting from 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3- yi-chromen-4-one O-ferf-buiyl oxime (example 81A) and 4-(3-Chloro-propyl)-pyridine hydrochloride, prepared from 4-pyridine propanol (US 6,362,336).
Figure imgf000167_0001
Mp: 210°C - 215°C.
MS (ESI+): 413.2 [C24H20H4O3+Hf (m/z).
1 H NMR: (300 MHz) DMSO-d65 (ppm): 10.91 (br. s, 1 H), 9.21 (s, 1 H), 8.82 (d, J = 2.8 Hz, 2H), 8.03-7.98 (m, 3H), 7.81 (d, J = 2.5 Hz, 1 H), 7.43 (s, 1 H), 7.40 <d, J = 9.1 Hz, 1 H), 7.29 (d, J = 3.0 Hz, 1 H), 7.07 (dd, J = 9.1 Hz, J = 3.1 Hz, 1 H), 6.97 (dd, J = 3.7 Hz, J = 2.8 Hz , 1 H), 6.73 (d, J = 3.8 Hz, 1 H), 4.05 (t, J = 6.2 Hz, 2H), 3.08 (t, J = 7.3 Hz, 2H), 2.17 (quint., J = 4.7 Hz, 2H).
Example 160: 6-(2-Phenoxy-ethoxy)-2-pyrrolo[ 1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime was prepared in 12% yield using method D (step 2), starting from 6-(2-Phenoxy-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-y!-chromen-4-one O-te/f-butyl oxime (example 160A).
Figure imgf000167_0002
Mp: 210°C - 215°C.
HPLC (gradient 15 % - 75 % MeOH / H20 + 0.05% TFA): > 95%; RT = 8.54 min.
MS (ESI+): 414.1 [C24H19N302+H]+ (m/z).
Example 160A: 6~(2~Phenoxy-ethoxy)-2-pyrrolo[ 1,2-c]pyrimidin-3-yl- chromen-4-one O-tert-butyl oxime
Figure imgf000168_0001
A mixture of 6-hydroxy-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert- butyl-oxime (example 81 A) (150 mg, 0.43 mmol), cesium carbonate (280 mg, 0.85 mmol) and (2-lodo-ethoxy)~benzene (413 mg, 3.87 mmol), in dimethylformamide (6 ml) was stirring at room temperature for 24 hours. The reaction mixture was diluted with water and extracted with eihy! acetate. The organic layers were washed with a saturated solution of ammonium chloride, water, brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/ethyl acetate: 0-10%) to yield the title compound (77 mg, 38%) as a yellow solid.
1 H NMR: (300 MHz) CHC -d, δ (ppm): 8.80 (s, 1 H), 7.91 (s, 1 H), 7.61 (s, 1 H), 7.50 (s, 1 H), 7.45 (d, J = 2.6 Hz, 1 H), 7.33-7.28 (m, 2H), 7.22 (d, J = 9.0 Hz, 1 H), 7.04 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 7.00-6.95 (m, 3H), 6.92 (dd, J = 3.8 Hz, J = 2.8 Hz 1 H), 6.63 (d, J = 3.8 Hz, 1 H), 4.41 -4.34 (m, 4H), 1.42 (s, 9H).
Example 161: 6-[3-(4-Fluoro-phenoxy)-propoxy]-2~pyrrolo[ 1,2-c]pyrimidin-
3- yl-chromen-4-one oxime was prepared in 74% yield using method D (step 2), starting from 6-[3-(4-Fluoro-phenoxy)-propoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-y!-chromen-
4- one O-tert-but l-oxime (example 161A).
Figure imgf000168_0002
MS (ESI+): 446.0 [C2gH20FN3O4+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d65 (ppm): 10.93 (br. s, 1 H), 9.21 (s, 1 H), 8.04 (s, 1 H), 7.82 (s, 1 H), 7.44 (s, 1 H), 7.41 (d, J = 9.2 Hz, 1 H), 7.33 (s, 1 H), 7.15-7.09 (m, 3H), 7.00-6.96 (m, 3H), 7.74 (d, J = 3.2 HzT 1 H), 4.18-4.1 1 (m, 4H), 2.20-2.17 (m, 2H).
Example 161 A: 6-[3-(4-Fluoro-phenoxy)-propoxy]-2-pyrrolo[ 1, 2- c]pyrimidin-3-yl-chromen~4-one O-tert-butyl-oxime A mixture of 6-(3-chloro- propoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one O-tert-butyl-oxime (example 101A) (125 mg, 0.29 mmol), potassium carbonate (120 mg, 0.87 mmol), potassium iodide (48 mg, 0.29 mmol) and 4-fluorophenol (65 mg, 0.58 mmol), in butanone (3,5 ml) was heated in a sealed tube at 130°C for 18 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic layers were washed with, brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography over silica gel (gradient cyclohexane/dichloromethane: 0-100%) to yield the title compound (100 mg, 69%) as a yellow solid.
Figure imgf000169_0001
1 H NMR: (300 MHz) CHC^-d, δ (ppm): 8.78 (s, 1 H), 7.90 (s, 1 H), 7.55 (d, J = 2.8 Hz, 1 H), 7.50 (s, 1 H), 7.44 (d, J = 2.6 Hz, 1 H), 7.20 (d, J = 8.8 Hz, 1H), 7.00-6.84 (m, 6H), 6.63 (d, J = 3.8 Hz, 1 H), 4.22 (t, J = 6.0 Hz, 2H), 4.15 (t, J = 6.0 Hz, 2H), 2.28 (quint., J = 6.2 Hz, 2H), 1.42 (s, 9H).
Example 162: 6-(3~Phen oxy-prop oxy) -2-pyrrolo[ 1, 2~c]pyrimidin-3~yl- chromen-4-one oxime was prepared in 24% overall yield using the method described in example 161 , starting from 6-(3-chioro-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one O-ferf-butyl oxime (example 101A)-and phenol.
Figure imgf000169_0002
MS (ESI+): 428.1 [C25H21FN304+HJ+ (m/z).
Mixture 80:20 of E~Z isomers
1 H NMR of the major Z isomer: (300 MHz) DMSO-d6 δ (ppm): 10.90 (s, 1 H), 9.21 (s, 1 H), 8.04 (s, 1 H), 7.81 (s, 1 H), 7.43-7.25 (m, 5H), 7.13 (dd, J = 9.1 Hz, J = 3.0 Hz, 1 H), 6.98-6.90 (m, 4H), 6.73 (d, J = 3.8 Hz, 1 H), 4.18-4.11 (m, 4H), 2.20-2.17 (m, 2H).
Example 163: 6-[3-(3-Fluoro-phenoxy)-propoxy]-2-pyrrolo[1,2-c]pyrimidin- 3-yl-chromen-4-one oxime was prepared in 46% overall yieid using the method described in example 161 , starting from 6-(3-chloro-propoxy)-2-pyrrolo[1 ,2- c]pyrimidin-3-yl-chromen-4-one O-ferf-butyl oxime (example 101 A) and 3- fluorophenot.
Figure imgf000170_0001
MS (ESI+): 446.0 [C25H20FN3O4+H]+ (m/z).
1 H N R: (400 MHz) DMSO-d6 δ (ppm): 10.93 (s, 1 H), 9.21 (s, 1 H), 8.04 (s, 1 H), 7.81 (s, 1 H), 7.44 (s, 1 H), 7.41 (d, J = 8.8 Hz, 1 H), 7.34-7.30 (m, 2H), 7.13 (dd, J = 9.2 Hz, J = 3.0 Hz, 1 H), 6.98 (dd, J = 3.6 Hz, J = 2.8 Hz, 1 H), 6.87-6.73 (m, 4H), 4.18-4.1 1 (m, 4H), 2.23-2.17 (m, 2H).
Example 164: 6-[3-(3,4-Di†luoro-phenoxy)-propoxy]-2-pyrrolo[ 1,2- c]pyrimidin-3~yl-chromen-4-one oxime was prepared in 53% overall yield using the method described in example 161 , starting from 6-{3-chloro-propoxy)-2-pyrro!o[1 ,2- c]pyrimidin-3-y!-chromen-4-one O-tert-butyi oxime (example 101 A) and 3,4- dif!uorophenol.
Figure imgf000170_0002
MS (ES1+): 464.1 [C25H19F2N304+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 10.93 (br. s, 1 H), 9.21 (s, 1 H), 8.04 (s, 1 H), 7.81 (s, 1 H), 7.44-7.30 (m, 4H), 7.16-7.07 (m, 2H), 6.98 (dd, J = 3.6 Hz, J = 2.8 Hz, 1 H), 6.82-6.80 (m, 1 H), 6.74 (d, J = 3.6 Hz, 1 H), 4.18-4.1 1 (m, 4H), 2.23-2.17 (m, 2H). Example 165: 6~{2-[1,4]Oxazepan-4-yl-ethoxy)-2-thieno[3,2-c]pyridin-6-yl- chromen-4-one oxime, hydrochloride was prepared in 18 % yield using method D (step 2), and the procedure described in example 87A starting from 6-(2-chloro- ethoxy)-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime O-tert-butyl oxime {example 165A) and /"^4/oxazepane. The hydrochloride salt of the title compound was isolated as an orange solid after treatment with a 1.25 soiution of hydrogen chloride in isopropanol.
Figure imgf000171_0001
Mp: 223-226°C.
MS (ESI+): 438,2 [C23H23N304S+H (m/z).
1 H NMR: (300 MHz) DMSO-d6 δ (ppm): 1 1.1 1 (br. s, 1 H), 10.83 (br. s, 1 H), 9.27(s, 1 H), 8.78 (s, 1 H), 8.05 (d, J = 5.2 Hz, 1 H), 7.73 (d, J = 5,5 Hz, 1 H), 7.69 (s, 1 H), 7.51 (d, J = 9.2 Hz, 1 H), 7.41 (d, J = 3.0 Hz, 1 H), 7.24 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.47 (m, 2H), 3.87 (m, 2H), 3.77 (m, 2H), 3.62 (m, 4H), 3.41 (m, 2H), 2.26 (m, 1 H), 2.05 (m, 1 H).
Example 165A: 6-(2-chioro-ethoxy)-2-thieno[3,2-c]pyridin-6-yl-chromen-4~ one oxime O-tert-butyl oxime was prepared in 41 % yield using the method described in example 85A, starting from 6-hydroxy-2-tfueno[3,2-c]pyridin-6-yl- chromen-4-one oxime O-tert-butyl-oxime (example 1278) in dimethyiformamide.
Figure imgf000171_0002
1 H NMR: (300 MHz) CHCI3-d1 δ (ppm): 9.16 (s, 1 H), 8.46 (s, 1 H), 7.72 (s, 1 H), 7.59 (d, J =5.5 Hz, 1 H), 7.53 (d, J =3.0 Hz, 1 H), 7.50 (d, J =5.5 Hz, 1 H), 7.27 (m, 2H), 7.02 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.31 (t, J = 5.8 Hz, 2H), 3.85 (t, J = 5.8 Hz, 2H), 1.42 (s, 9H). Example 166: 6-[2-(3,4-Dihydro-1H-isoquinolin-2-yl)-ethoxy]-2~thieno[3,2- c]pyridin-6-yl-chromen-4-one oxime, hydrochloride was prepared in 7.5% overall yield using the method described in example 165, starting from 6-(2-chloro-ethoxy)-2- thieno[3,2-c3pyridin-6-yl-chromen-4-one oxime O-tert-butyl oxime (example 165A) and 1 ,2,3,4-tetrahydro-isoquinoiine.
Figure imgf000172_0001
Mp: 234-237 .
Figure imgf000172_0002
1 H NMR: (300 MHz) DMSO-ds δ (ppm): 1 1.03 (br. s, 2H), 9.27 (s, 1 H), 8.80 (s, 1 H), 8.06 (d, J = 5.5 Hz, 1 H), 7.73 (dT J = 5.5 Hz, 1 H), 7.70 (s, 1 H), 7.52 (d, J - 9.0 Hz, 1 H), 7.45 <d, J = 3.0 Hz, 1 H), 7.30-7.20 (m, 5H), 4.40-4.80 (m, 2H), 3.85 (m, 1 H), 3.70 (m, 2H), 3.60-3.20 (m, 2H), 3.10-3.00 (m, 1 H), 1.90 (m, 2H).
Example 167: 6-[2~(4-fluoro-piperidin'1-yl)'ethoxy]-2~thieno[3,2-c]pyridin- 6-yl-chromen-4-one oxime, hydrochloride was prepared in 25% overall yield using the method described in example 165, starting from 6-(2-chloro-ethoxy)-2-thieno[3,2- c]pyridin-6-yi-chromen-4-one oxime O-tert-butyl oxime (example 165A) and 4- fiuoropiperidine hydrochloride.
Figure imgf000172_0003
MS (ESI+): 440.1 [C23H22FN303S+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 1 1.10 (br. s, 1 H), 10.79 (br. s, 1 H), 9.27 (s, 1 H), 8.78 (s, 1 H), 8.06 (d, J = 3.9 Hz, 1 H), 7.73 (d, J = 3.9 Hz, 1 H), 7.70 (s, 1 H), 7.51 (d, J = 6.6 Hz, 1 H), 7.42 (s, 1 H), 7.24 (d, J = 6.9 Hz, 1 H), 5.07, 5.95 (2xm, 1 H), 4.45 (m, 2H), 3.65-3.45 (m, 4H), 3.35-3.10 (m, 2H), 2.30-2.00 (m, 4H).
Example 168: 6-[2-(1,1-Dioxo-1-thiomorpholin-4-yl)-ethoxy]-2-thieno[3,2- c]pyridin-6-yl-chromen-4'one oxime, hydrochloride was prepared in 4% overai! yield using the method described in example 165, starting from 6-(2-chloro-ethoxy)-2- thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime O-tert-butyl oxime (example 165A) and thiomorpholine-1 , 1 -dioxide.
Figure imgf000173_0001
Mp; 195-197°C.
MS (ESI+): 472.2 [C22H21N305S2+H]+ (m/z).
1 H NMR: (300 MHz) DMSO-d6 8 (ppm): 1 1.05 (br. s, 1 H), 9.25 (s, 1 H), 8.76 (s, 1 H), 8.05 (d, J = 5.5 Hz, 1 H), 7.72 (d, J = 5.5 Hz, 1 H), 7.68 (s, 1 H), 7.50 (d, J = 9.0 Hz, 1 H), 7.40 (d, J = 3.0 Hz, J = 2.8 Hz, 1 H), 7.23 (dd, J = 9.0 Hz, J = 3.0 Hz, 1 H), 4.42 (m, 2H), 3.60 (m, 4H), 3.43 (m, 6H).
Example 169: 6-( 1-Pyrimidin-2-yl-piperidin-4-yloxy)-2-thieno[3,2-c]pyridin- 6-yl-chromen-4-one oxime was prepared in 9% yield using method D (step 2), starting from 6-(1-Pyrimidin-2-yl-pfperidin-4-yloxy)-2-thieno[3,2-c]pyndin-6-yl-chromen- 4-one O-tert-but l-oxime (example 169A).
Figure imgf000173_0002
MS (ESI+): 472.1 [C2SH21N503S+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 1 1.20 (br. s, 1 H), 9.28 (d, J = 0.8 Hz, 1 H), 8.82 (s, 1 H), 8.43 (d, J = 4.8 Hz, 2H), 8.06 (d, J = 5.2 Hz, 1 H), 7.74 (dd, J = 5.2 Hz, J = 0.8 Hz, 1 H), 7.69 (s, 1 H), 7.50 (d, J = 9.2 Hz, 1 H), 7.43 (d, J = 3.2 Hz, 1 H), 7.24 (dd, J = 9.2 Hz, J = 3.2 Hz, 1 H), 6.71 (t, J = 5.2 Hz, 1 H), 4.72 (quint., J = 4 Hz, 1 H), 4.22-4.16 (m, 2H), 3.63-3.57 (m, 2H), 2.07-2.02 (m, 2H), 1.68-1.65 (m, 2H).
Example 169A: 6-(1-Pyrimidm" -2-yl-piperidin-4-yloxy)-2-thieno[3,2- c]pyridin-6-yl-chromen-4-one O-tert-butyl-oxime was prepared using the procedure described in example 85A, starting from 6-Hydroxy-2-thieno[3,2-c]pyridin-6- yl-chromen-4-one O-tert-buiyl-oxime (example 127B) and methanesulfonic acid 1- pyrimidin-2-yl-piperidin-4-yl ester (example 169B) in dimethylformamide.
Figure imgf000174_0001
HPLC (gradient 5 % - 95 % ACN / H20 + 0.1% HCOOH): RT MS (ESI+): 528.3 [C29H29N503S+H]+ (m/z).
Example 169B: Methanesulfonic acid 1-pyrimidin-2-yl-piperidin-4-y! ester
Figure imgf000174_0002
To a cooled solution of 1-Pyrimidin-2-yl-piperidin-4-ol (WO2008/8895 A1 ), (100 mg; 0.55 mmoi) and triethylamine (233 μί_, 1 .67 mmol) in dry dichioromethane (1 mL), was added meihanesuifonyl chloride (52 0.67 mmol). The reaction mixture was stirred at room temperature for 5 hours. Water was added. After extraction with dichioromethane, the combined organic extracts were dried over sodium sulfate and concentrated to afford a crude product that was used in the next step without further purification.
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.30 (d, J = 4.7 Hz, 2H), 6.49 (t, J = 4.7 Hz, 1 H), 4.97 (m, 1 H), 4.20 (m, 2H), 3.64 (m, 2H), 3.05 (s, 3H), 2.09 (m, 2H), 1.89 (m, 2H).
Example 170: 6-(3,4, 5, 6- Tetrahydro-2H-[1 , 2 "]bipyridinyi-4~yloxy)-2- thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime was prepared in 6% overall yield using the method described in example 169, starting from 6-Hydroxy-2-thieno[3,2- c3pyridin-6-yl-chromen-4-one O-tert-butyl-oxime (example 127B) and methanesulfonic acid 3,4,5,6-tetrahydro-2H-[1 ,2[]bipyridiny[-4-yl ester (example 170A).
Figure imgf000175_0001
MS (ESI+): 471.1 [C25H22N403S+H]+ (m/z).
1 H NMR: (400 MHz) DMSO-d6 δ (ppm): 1.04 (br. s, 1 H), 9.26 (d, J = 0.8 Hz, 1 H), 8.77 (s, 1 H), 8.06-8.01 (m, 3H), 7.73 (dd, J = 5.2 Hz, J = 0.8 Hz, 1 H), 7.69 (s, 1 H), 7.50-7.42 (m, 3H), 7.25 (dd, J = 8.8 Hz, J = 2.8 Hz, 1 H), 6.94 (t, J = 6.8 Hz, 1 H), 4.78 (quint, J = 4 Hz, 1 H), 3.92-3.91 (m, 2H), 3.71-3.65 (m, 2H), 2.13-2.11 (m, 2H), 1.83-1.81 (m, 2H).
Example 170A: Methanesulfonic acid 3,4,5,6-tetrahydro-2H- [1,2']bipyridinyl-4-yl ester was prepared in 48 % overall yield using the method described in example 1698, starting from 3,4,5,6-Tetrahydro-2H-[1 ,2']bipyridiny!-4-ol (WO2008/62276 A2).
Figure imgf000175_0002
1 H NMR: (300 MHz) CHCI3-di δ (ppm): 8.19-8.16 (m, 1 H), 7.51-7.45 (m, 1 H), 6.66 (dd, J = 13.8 Hz, J = 8.6 Hz, 1 H), 6.63-6.60 (m, 1 H), 4.96-4.92 (m, 1 H), 3.96-3.88 (m, 2H), 3.45-3.36 (m, 2H), 3.1 1-3.07 (m, 2H), 3.04 (s, 3H), 2.12-2.04 (m, 2H), 1.98- 1.89 (m, 2H).
Example 171 : Human mGluR4 Positive Allosteric Modulator evaluation using Ca++ functional assay
Compounds of the present Invention were tested successively for their agonist and positive allosteric modulator activities on human mGluR4 transiently over-expressed in HEK-293 cells. They exert agonist activity if, by themselves in absence of the endogenous glutamate, they are able to activate mGluR4; and they exert positive aliosteric modulator activity if they increase the action of the endogenous glutamate.
Cell Culture and Transfection
HEK-293 cells were maintained in Modified Eagle's Medium supplemented with 0% Foetal Calf Serum, 1% Penicillin/Streptomycin and 1% non essential amino acids at 37°C/5% C02.
Cells were co-transfected by electroporation with two DNA plasmids encoding hmGluR4 and a chimeric G protein allowing redirection of the activation signal to intracellular calcium pathway. Cells were plated after transfection onto polyornithine coated, clear bottom, black-walled, 96-well plates and cultured for 24h.
Calcium Assay ECse determination
Receptor activity was detected by changes in intracellular calcium measured using the fluorescent Ca sensitive dye, Fiuo4AM (Molecular Probes),
The day of the assay, medium was aspirated and replaced during 3 hours by medium without serum supplemented with 1% Glutamax, 1 % Penicillin/Streptomycin and 1% non essential amino acids. Then, cells were washed with freshly prepared buffer B (HBSS 1X (PAA), Hepes 20mM, MgS04-7H20 1mM, Na2C03 3.3m , CaCI2-2H20 1 ,3mM, 0.5% BSA, Probenecid 2.5mM) and loaded at 37°C in 5% C02 for 1.5 hours with buffer B containing μΜ Fluo4AM , 0.1mg/mL Pluronic Acid, 7pg/mL Glutamate Pyruvate Transaminase and 2mM sodium pyruvate. Afterwards cells were washed twice with buffer B and 50pL of this buffer were added to each well. Addition of compounds and intracellular Ca2+ measurements {excitation 485nm, emission 525nm) were performed by the fluorescence microp!ate reader FlexStation (Molecular Devices).
Agonist and positive allosteric modulator activities of compounds were consecutively evaluated on the same cells plate. Agonist activity was first tested during 60s with the addition of compound alone on the cells. Then, the cells were stimulated by an ECt0 gSutamate concentration and fluorescence recorded for additional 60s. EC1D giutamate concentration is the concentration giving 10% of the maximal giutamate response. Agonist and/or positive alSosteric modulator activity(ies) were evaluated in comparison to basal signal evoked by EC10 giutamate alone.
For EC50 determination, a dose-response test was performed using 6 to 9 concentrations of each compound of the invention. Dose-response curves were fitted using the sigmol'dal dose-response (variable slope) analysis in GraphPad Prism program (Graph Pad Inc) and EC50 of agonist / positive altosteric modulator activity was calculated. Dose-response experiments were all performed in duplicate, two times independently.
Compounds of the present invention can either be mix agonists / positive allosteric modulators or pure positive allosteric modulators. Their ECS0 are preferably 5μ or less, more preferably 1 or less.
The following lists represent the mean EC50 obtained for selected compounds of the present invention:
- Examples with an ECS0 < 1 μΜ: 1 , 4, 5, 20, 23, 28, 37, 43, 51 , 53, 61 , 62, 63, 67, 69, 70, 73, 81 , 83, 85, 91 , 94, 96, 97, 98, 99, 101 , 102, 103, 104, 105, 106, 107, 108, 109, 111 , 112, 113, 114, 115, 116, 117, 118, 1 19, 123, 124, 125, 126, 127, 128, 129, 130, 131 , 132, 133, 134, 135, 137, 138, 139, 140, 141 , 142, 143, 146, 147, 148, 149, 150, 152, 156, 158, 159, 160, 161 , 162, 163, 164.
- Examples with an EC50 < 5μΜ: 2, 6, 10, 11 , 15, 21 , 22, 24, 26, 30, 31 , 34, 36, 39, 41 , 42, 44, 52, 54, 55, 56, 57, 58, 65, 66, 71 , 76, 77, 78, 87, 88, 92, 93, 95, 121 , 136, 144, 151 , 153, 157.
Example 172: ln-vivo evaluation in the ha!doperidol-induced catalepsy model in the mouse
Introduction This method, which detects anti-Parkinsonian activity, follows that described by Pires ei a/. (J Med and Biol Res 38, 1867-1872, 2005; Shiozaki et a/., Psychopharmacology 147, 90-95, 1999)
Protocol
Catalepsy was assessed using the bar test in mice submitted to acute intra-peritoneal administration of Haioperidol 1 mg/kg,.Mice (male Rj: NMRI mice, weighing 25 - 30 g at the beginning of the experiment) placed in group of 5 in Plexiglas cages, were injected with Haioperidol (1 mg/kg i.p.). Within 15 min after Haioperidol administration, mice were calm and showed slow spontaneous activity. The catalepsy response of one mouse was measured as the time the animal maintained an imposed posture with both forelimbs placed on a horizontal 0.9cm diameter wire bar suspended 4cm above a platform. The end point of catalepsy was considered to occur when both forelimbs were removed from the bar, the mouse climbed onto the bar or if the animal moved its head in an exploratory manner. A cut-off time of 180 seconds was applied. The degree of catalepsy was scored 45min after Haioperidol administration and continued at 45 minutes intervals for a total of 270 minutes. Between determinations, the animals were returned to their home cages. The compound of Example 63, administered i.p. consecutively with haioperidol, was evaluated at 30 mg/kg and compared with a vehicle control group. The Figure 1 shows the mean time of latency spent on the bar in each group of animals. At each time-point, the anticataieptic effect of compound of Example 63 was compared to vehicle-treated group using ANOVA test followed by the Dunnett's test.
Results
It can be dearly observed in Figure 1 that the compound of Example 63 at the dose of 30 mg/kg administered i.p. consecutively with haioperidol exerts a significant anticataieptic activity.
Example 173: ln-vivo evaluation in the marble burying test in the mouse
Introduction
The method, which detects anxiolytic/tranquillizing activity, follows that described by Broekkamp et at. (Eur. J. Pharmacol,, 126, 223-229, 1986). Mice exposed to novel object (marbles) will bury them in the sawdust floor covering. Anxiolytics decrease the number of marbles buried at non-sedative doses.
Protocol
Mice were individually placed in transparent plastic cages (33 x 21 x 18 cm) with 5 cm of sawdust on the floor and 25 marbles grouped in the centre of the cage. The cage was covered with an inverted plastic cage. Each test cage, together with the marbles, was impregnated with mouse odor before-hand by leaving 10 mice in the cage for 15 minutes. These mice then played no further ro!e in the experiment. The number of marbles covered by sawdust (2/3 or more) was counted at the end of a 30 minute test. 12 mice were studied per group. The test was performed blind (apart from positive control). The compound of Example 85 was evaluated at 2 doses (100 and 300 mg/kg), administered p.o. 30 minutes before the test, and compared with vehicle control groups. Clobazam (8 mg/kg i.p.), administered 30 minutes before the test, was used as reference substance and was compared with the vehicle control group. Data were analyzed by comparing treated groups with control group using unpaired Student's t tests.
Results
The compound of Example 85 (100 and 300 mg/kg), administered p.o. 30 minutes before the test, markedly and dose-dependently decreased the number of marbles covered by sawdust, as compared with vehicle controls (-70% and -83%, respectively, p < 0.001 ). The Figure 2 shows the mean number of marbles burying in each group of animals.

Claims

Claims
1. A compound of the general formula (I)
Figure imgf000180_0001
wherein:
R1, R2, R3, and R4 each independently represent a -L-R group, wherein:
L is selected from a bond, C C10 aikylene, C2-C10 alkenylene, or C2-C10 alkynylene, wherein said aikylene, said alkenylene or said alkynylene is optionally substituted with one or more groups independently selected from halogen, -CF3, -CN, -OH, or -NH2t and further wherein one or more -CH2- units comprised in said aikylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from: -0-, -NR11-, -CO-, -S-, -SO-, or -S02-;
R is selected from hydrogen, d-do alkyl, halogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, -NR 1R12, -OR11, -SR11, -SOR11, -S02R \ -CF3, or -CN, wherein said optionally substituted aryl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloaikyl may be substituted with one or more groups independently selected from C C4 aikyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(Ci-C4 alkyl), -N(CrC4 alkyl)(C C4 alkyl) wherein the two C C alkyl moieties of said -N(CrC4 alkyl)(CrC4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, or -L1-R13; R1 and R12 are each independently selected from hydrogen, optionally substituted C C10 alkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or -CF3, wherein said optionally substituted alkyl, said optionally substituted aryi, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalky! may be substituted with one or more groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(d-C4 alkyl), -NH2, -NH(d-d alkyl), or -N(Ci-C4 alkyl)(C C4 alkyl) wherein the two C C4 alkyl moieties of said -N(d-d alkyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to;
L is selected from a bond, d-do alkylene, C2-C10 alkeny!ene, or d-do alkynyiene, wherein one or two -CH2- units comprised in said alkylene, said alkenyiene or said alkynyiene are each optionally replaced by a group independently selected from -0-, -NH-, -N(d-C4 alkyl)-, -CO-, -S-, -SO-, or -S02-;
R 3 is selected from hydrogen, d-d alkyl, halogen, optionally substituted phenyl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalky!, -NH2, -NH(d-C4 alkyl), -N(C C4 alkyl)(C C4 alkyl), -OH, -0(C d alkyl), -SH, -S(C d alkyl), -CF3, or -CN, wherein said optionally substituted phenyl, said optionally substituted heteroaryl, said optionally substituted cycloalkyl, or said optionally substituted heterocycloalky! may be substituted with one or more groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(C C4 alkyl), or -N(C C4 alkyl)(d-C4 alkyl) wherein the two d-d alkyl moieties of said -N(C C4 alkyl)(C C4 alky!) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to;
R5 is selected from hydrogen, C C alkyl, halogen, -CF3, -CN, -OH, -0(d-C4 alkyl), -COOH, -COO(C C4 alkyl), -C0NH2, -CONH(d-C4 alkyl), -CON(d-C4 alkyl)(d-C4 alkyl), wherein the two d-C4 alkyl moieties of said -CON(d-C alkyl)(C C4 a!kyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, -NH2, -NH(C C alkyl), or -N{d-C aikyl)(C C4 alky!), wherein the two C C4 alkyl moieties of said -N(C C aikyl)(CrC4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to;
A is a bicyclic moiety corresponding to formula (II):
Figure imgf000182_0001
which may be saturated or unsaturated, and wherein:
n is 0 or 1 ;
X1 to X6 are each independently selected from N, N(R 1), C(R 2), C(Rx2)(Rx3), O, S, S(O), S(0)2l or C(O);
X7 is N or N(Rx1);
any of groups X1 to X7 containing a nitrogen atom may form an N-oxide group;
X8 and X9 are each independently selected from N, C, or C(Rx2); each R 1 is independently selected from hydrogen, Ci-C4 alkyl, -OH, -0(C C4 alkyl), or -{C C4 alkylene)-phenyl; and
each R*2 and each Rx3 is independently selected from hydrogen, C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -COOH, -COOfd-d aikyl), -CONH2, -CONH(C C4 alkyl), -CON(C C4 alkyl)(C C4 alkyl), -NH2, -NH(Ci-C4 alkyl), or -N{C C4 alkyl)(C C4 alkyl), wherein the two C1 -C4 alkyl moieties of said -N(Ci-C4 aikyl)(CrC4 alkyl) or of said -CON(C C4 a!kyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
The compound of claim 1 , wherein L is a bond, d-C6 alkylene, C2-C6 alkenyiene, or C2-C6 alkynyiene, wherein one or two -CH2- units comprised in said alkylene, said alkenyiene or said alkynyiene are each optionally replaced by a group independently selected from -0-, -NR1 -, -CO-, or -S-.
The compound of claim 1 or 2, wherein R is selected from: hydrogen: optionally substituted aryl; optionally substituted heteroaryl having 5 or 6 ring atoms, wherein 1 , 2, or 3 ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; optionally substituted heterocycioalkyl having 3 to 10 ring atoms, wherein one or more ring atoms are each independently selected from O, S, or N and the other ring atoms are carbon atoms; -NH(d-C4 alkyl); -N(d-d alkyl)(C C4 alkyl); or -0(d-d alkyl); wherein said optionally substituted aryl, said optionally substituted heteroaryl or said optionally substituted heterocycioalkyl may be substituted with one or more groups independently selected from d-d alkyl, halogen, -CF3, -CN, -OH, -0(d-d alkyl), -NH2, -NH(C C4 alkyl), -N(d-C4 alkyl)(CrC alkyl) wherein the two d-d alkyl moieties of said -N(d-d alkyl)(d-d alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, or -L1-R1S.
4. The compound of claim 1 , wherein R1 and R4 are each hydrogen, one of Rz and R3 is hydrogen, and the other one of R2 and R3 is selected from: hydrogen; d-d alkyl; -(d-d alkylene)-0-(d-d alkyl); -(d-d alkynylene)-phenyl; -OH; -0(C C4 alkyl); -0(d-d alkylene)-0-(C C2 alkyl); -(d-d aikylene)-morpholinyl; -0(C d a!kylene)-phenyl; -0(d-d alkyiene)-imidazolyl; -0(Ci-d alkylene)-pyrrolidinyl, -0(d-d aikylene)-piperidinyl; -0(d-d a!kylene)-morpholinyl; -0(d-d alkylene)-oxazepanyl; -0(d-d alkylene)-(2-oxa-5-aza-bicyclo[2.2.1 ]hept-5-yl); -0(C d alkylene)-piperazinyiene-(d~d alkyl); or -0(d-d alkylene)- diazepanylene-(C d alkyl); wherein the phenyl moiety, the imidazolyl moiety, the pyrrolidinyl moiety, the piperidinyl moiety, the morpholinyl moiety, the oxazepanyl moiety, the 2-oxa-5-aza-bicyclo[2.2.1 ]hept-5-yl moiety, the piperazinylene moiety, and the diazepanylene moiety are each optionally substituted with one or more groups independently selected from halogen, -CF3, - alkyl, -(d-d alkylene)-OH, -(d-d alkylene)-0-(d-C4 alkyl), -OH, -0(C d alkyl), -0-(d-C4 alkylene)-0-(d-d alkyl), -NH2, -NH(d-d alkyl), or -N(d-d alkyl)(C d alkyl) wherein the two C d alkyl moieties of said -N(d-d alkyl)(d-d aikyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to.
5. The compound of any of claims 1 to 4, wherein R5 is hydrogen.
6. The compound of any of claims 1 to 5, wherein A is a bicyclic moiety corresponding to formula (II) as defined in claim 1 , wherein the first ring of the bicyclic moiety, which ring is linked to the remainder of the compound of general formula (I), is aromatic.
7. The compound of any of claims 1 to 5, wherein A is one of the following groups:
Figure imgf000184_0001
each of which may optionally be substituted on ring carbon atoms with one or more groups independently selected from C C4 alkyl, halogen, -CF3, -CN, -OH, -0(C C4 alkyl), -NH2, -NH(C C4 alkyl), or -N(C C4 alky!Xd^ a!kyl) wherein the two C-1-C4 alkyl moieties of said -N(d-C4 aikyl)(C C4 alkyl) are optionally mutually linked to form a ring together with the nitrogen atom which they are attached to, wherein each Rx (if present) is independently selected from hydrogen, C C4 alkyl, -OH, or -0(C,-C4 a!kyl), and further wherein each Rx4 (if present) is independently selected from hydrogen or C1-C4 alkyl, or the two groups Rx4 (if present) are each independently C1-C4 alkyl and are mutually linked to form a ring together with the nitrogen atom which they are attached to.
. The compound of claim 1 , wherein said compound is selected from: 2-isoquinolin-3-yl-chromen-4-one oxime;
7-bromo-2-isoquinolin-3-yi-chromen-4-one oxime;
7- bromo-2-isoquinolin-3-yl-6-methyl-chromen-4-one oxime;
6-bromo-2-isoquinolin-3-yl-chromen-4-one oxime;
2-isoquinolin-3-yl-6-methyl-chrornen-4-one oxime;
6- fluoro-2-isoquinolin-3-yl-chromen~4-one oxime;
6,8-difluoro-2-isoquino!in-3-yl-chromen-4-one oxime;
8- chloro-2-isoquinolin-3-yl-chromen-4-one oxime;
4-fluoro-2-isoquinolin-3-yf-chromen-4-one-(Z)- oxime;
2-isoquinolin-3-yl-6-trifluoromethoxy-chromen-4-one oxime;
2-isoquinolin-3-yl-6-trifluoromethyl-chromen-4-one oxime;
2-(7~fluoroHsoquinolin-3-yl)-chromen-4-one oxime;
2-(7-methoxy-isoquinoiin-3-yl)-chromen-4-one oxime;
2-(6.7-dimethoxy-isoquinolin-3-yl)-chromen-4-one oxime;
2-(6-methy!-isoquinolin-3-yl)-chromen-4-one oxime;
2-(7-chloro-isoquinoiin-3-yl)-chromen-4-one oxime;
2-(5~bromo-isoquinolin-3-yl)-chromen-4-one oxime;
" 2-(5-hydroxy-isoquinolin-3-y!)-chromen-4~one oxime;
2-(5-methoxy-isoquinoiin-3-yl)-chromen-4-one oxime;
2-isoquinoIin-3-yl-7-phenylethynyl-chromen-4-one oxime;
2-isoquinoiin-3-yi-7-((E)-styryi)-chromen-4-one oxime;
2-isoquinoiin-3-y!-7-phenethyi-chromen-4-one oxime;
7- ethynyl-2-isoquinolin-3-yl-chromen-4-one oxime;
2-isoquinolin-3-yl-7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime; 2-isoquinolin-3-y!-7-(pyridin-2-yl-ethynyl)-chromen-4-one oxime; 2-isoquinolin-3-yi-7-(pyridin-4-yl)ethynyl-chromen-4-one oxime; 7-(4-dimethylaminophenyl)ethynyl-2-isoquinoIin-3-yt-chromen-4-one oxime; 2-isoquinolin-3-yi-7-(3-tnethoxyphenyl)ethynyS-chromen-4-one oxime;
7-(3-aminopheny!)ethynyl-2-isoquino[in-3-yl-chromen-4-one oxime;
7-(3-hydroxyphenyi)eihynyI-2~isoquinoiin-3-yl-chromen-4-one oxime;
2-isoquinolin-3-yl-7-(4-methoxyphenyl)ethynyl-chromen-4-one oxime;
7-{2-chlorophenyi)ethynyl-2-isoquinolin-3-yi-chromen-4~one oxime;
7-(3-dimethylamino-prop-1-ynyi)-2-isoquinolin-3-yi-chromen-4-one oxime; 6-cyclopropyi-2-isoquinoiin-3-yi-chromen-4-one oxime;
2-ssoquinoiin-3-yi-6~(pyrroiidin-1 -yl}-chromen-4-one oxime;
2-isoquinolin-3-yi-6-(vinyl)-chromen-4-one oxime;
6-ethyl-2-isoquinoiin-3-yl-chromen-4-one oxime;
6-cyano-2-isoquinolin~3-yl-chromen-4-one oxime;
6-dimethylamino-2-isoquinolin-3-yI-chromen-4-one oxime;
2-isoquinoiin-3-yl-6-(morphoiin-4-yl-methyl)-chromen-4-one oxime;
6-hydroxy-2-isoquinolin-3-yi-chromen-4-one oxime;
2-isoquinolin-3-yt-6-methoxy-chromen-4-one oxime;
2-isoquinoliri-3-yl-6-(2-methoxy-ethoxy)-chromen-4-one oxime;
6- (2-dimethylamino-ethoxy)-2-isoquinolin-3-yl-chromen-4-one oxime;
2-isoqdinolin-3-yl-6-[3-(4-methyl-piperazin-1-yi)-propyiamino]-chromen-4-one oxime;
2-isoquinoiin-3-yl-6-[2-(4-methyi-piperazin-1-yl)-ethoxy]-chromen-4-one oxime; 2-isoquinoiin-3-yl-7-phenyl-chromen-4-one oxime;
7- (4-biphenyl)-2-isoquinoSin-3-yi-chromen-4-one oxime;
2-isoquinolin-3-yl-7-[3-(4-methyi-piperazin-1-yi)-propy[amino]-chromen-4~one oxime;
2-lsoquinolsn-3-yl-7-{3-[2-(4-methyi-piperazin-1-yl)-ethoxy]-phenylethynyl}- chromen~4-one oxime;
2-lsoquinoiin-3-yl-7-{3-me†hylaminophenyiethynyl}-chromen-4-one oxime; 7-(4-Hydroxy-but-1-ynyl)-2-isoquinoiin-3-yl-chromen-4-one oxime;
2-lsoquinoiin-3-yl- 7-[3-{2-methoxy-ethoxy)-pheny!eihynyl]-chromen-4-one oxime; 2-isoquinolin-3-y!-7-[3-(2-methoxy-ethoxy)-prop-1-ynyl]-chromen-4-one oxime; 7-but-3-en-1 -ynyl-2-isoquinolin-3-yi-chromen-4-one oxime;
2-isoquinoiin-3-yl-7-methoxy-chromen-4-one oxime;
2-isoquinolin-3-yl~7-(2-methoxy-ethoxy)~chromen-4-one oxime;
7-cyano-2-isoquinofin-3-yl-chromen-4-one oxime;
2-fsoquino[in-3-yl~7-[3-(4-methy!-piperazin-1 ~yi)~prop-1 -ynyl]-chromen-4-one oxime;
2-(7-hydroxy-isoquinoiin-3-yi)-chromen-4-one oxime;
2-[2,6]Naphthyridin-3-yl-chromen-4-one oxime;
2-[1 ^Naphthyridin-S-yl-chromen^-one oxime;
2~Pyrro!o[1 ,2-c]pyrimidin-3-y!~chromen-4-one oxime;
2-(5,7-dimethyl-Pyrroloi1 ,2-c]pyrimidin-3-yl)-chromen-4-one oxime;
2-(6-bromo-Pyrrolo[1 ,2-c]pyrimidin-3-yl)-chromen- -one oxime;
6-bromo-2-PyrroIo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-methoxyethoxy-2-Pyrro)o[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
2-( -Benzyl- 1 H-imidazo[4,5-c]pyridin-6-yl)-chromen-4-one oxime;
2-Thieno[2,3-cJpyridin-5-yl-chromen-4-one oxime;
2-Thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
2- lsoquinolin-3-yi-3-methyl-chromen-4-one oxime;
3- {4-[(E)-Hydroxyimino]-4H-chromen-2-yl}-2H-isoquinoiin-1-one;
2-lmidazo[1 ,2-c]pyrimidin-7-yl-chromen-4-one oxime;
2-(1 H-Pyrroio[2,3-c]pyridin-5-y[)-chromen-4-one oxime;
2-(1 -Hydroxy- H-pyrrolo[2,3-c]pyridin-5-yl)-chromen-4-one oxime;
2-{1-Methyl-1 H-pyrroio[2,3-c]pyridin-5-yl)-chromen-4-one oxime;
2-(1 -Methoxy-1 H-pyrroio[2,3-c]pyridin-5-yi)-chromen-4-one oxime; 6-Hydroxy-2-pyrrolo[1 ,2-c]pyrimtdin-3-yl-chromen-4-one oxime;
2-Thiazoio[5,4-c]pyridin-6-yl-chromen-4-one oxime;
6-(3-Methoxy-propyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(3-Dimethylamino-propoxy)-2-pyrrofo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(3-Morpholin-4-yI-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidiri-3-yl-chromen-4-one oxime; 6-(2,3-Dihydroxy-propoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime; 6-(2-Pyrrolidin-1 -yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime; 6-(2-Piperidin-1 -yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime; 6-(2-dimeihylamino-eihoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yS-chromen-4-one oxime; 6-(2-diethylamino-ethoxy)-2-pyrrolo[1 ,2~c]pyrimidin-3-yl-chromen-4-one oxime; 2-Pyrrolo[1 ,2-a]pyrazin-3-yi-chromen-4-one oxime;
6-[2-(2-methyl-pyrroIidin-1 -yl)-ethoxy]-2-pyrrofo[1 ^-clpyrimidin-S-yl-chromen^- one oxime;
6-[2-(4-methyl-piperazin-1 -yl)-ethoxy]-2-pyrrolo[1 ,2-c] pyrimidin-3-yl-chromen-4- one oxime;
6-[2-(4-methyi-[1 ,4]diazepart-1 -yl)-ethoxy]-2-pyrrolo[1 ,2-c] pyrimidin-3-yi-chromen- 4-one oxime;
6-[2-((S)-2-hydroxymethyipyrrolidin-1-yl)-ethoxy]-2-pyrrolo[1 ,2-c] pyrimidin-3-yi- chromen-4-one oxime;
6-[2-({S}-2-methoxymethyipyrroiidin-1 -yt)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime;
6-[2-((R)-2-methoxymethylpyrrolidin-1-yl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromeri-4-one oxime;
6-[2-(3-hydroxy-pyrro'idin-1 -y!}-eihoxy]-2-pyrrolo[1 ,2-c] pyrimidin-3-yl-chromen-4- one oxime;
6~[2~(4-dimethylamino-piperidin-1-yi)-ethoxy]-2-pyrrolo[1 ,2-c] pyrimidin-3-yi- chromen-4-one oxime;
6-(2-cyclopentyiamino-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-{3-morpholin-4-y(-propoxy)-2-pyrroio[1 ,2-c]pyrimidin-3-yi-chrornen-4-one oxime;
6-(4-morpholin-4-yi-butyl)-2-pyrrolo[1 ,2-c]pyrimidin-3-yi-chromen-4-one oxime;
6-{4,4-difluoro-piperidin-1-y)-2-pyrrolo[1 ,2-c] pyrimidin-3-yl-chromen-4-one oxime;
6-{2-[bis-(2-methoxy-ethyl)-amino]-ethoxy}-2-pyrrolo[1 ,2-c] pyrimidin-3-yl- chromen-4-one oxime;
6-(2-lmidazol-1 -yl-ethoxy)-2-pyrrolo[1 ,2-c] pyrimidin-3-yl-chromen-4-one oxime;
6-(3-morpholin-4-yl-propyl)-2-pyrroto[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-((Z)-3-Morpholin-4-yl-propenyl))-2-pyrroSo[1 ,2-c]pynmidin-3-yl-chromen-4-one oxime;
6-[2-(3-methoxy-piperidin-1 -yl)-ethoxy]-2-pyrroiot1 ,2-c]pyrimidin-3-yl-chromen-4- one oxime;
6-t2-(4-f!uoro-phenyl)-eihoxy]-2-pyrroio[1 ,2-c]pyrimidin-3-y[-chromen-4-one oxime;
2-quinazoiin-2-yl-chromen-4-one oxime;
6-[(1S,4S)-2-(2-oxa-5-aza-bicycto[2.2.1]hept-5-yl)-ethoxy]-2-pyrrolo[1 ,2- c]pyrimidin-3-y!-chromen-4-one oxime;
6-[2-(cis-2>6-dimeihyl-morpholin-4-yl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime;
2-pyrroloI1 ,2-c]pyrimidin-3-yl-6-[2-(4-trifluoromethyl-piperidiri-1-y[)-ethoxy]- chromen-4-one oxime;
6-[2-(3,3-difluoro-piperidin-1-yl)-ethoxy]-2-pyrroio[1 i2-c]pynmidin-3-yl-chromen-4- one oxime;
4-(hydroxyimino)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromene-6-carboxyiic acid (2- morpholm-4-yi-ethy!)-amide;
6-(2-[1 ;4']bipiperidiny[-1 '-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(2-[1 ,4]oxazepan-4-yi-ethoxy)-2-pyrroio[1 I2-c]pyrimidsn-3-y[-chromen-4-one oxime;
6-[2-(4-pyrrolidin-1 -yl-piperidin-1-yl)-ethoxy]-2-pyrro!o[1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime;
6- [2-(3,3-difluoro-pyrrolidin-1-y!)-ethoxy}-2-pyrrolo[1 ,2-c]pyrimidi
one oxime;
7- [3-{3-dimethylamino-propoxy)-phenyiethynyl]-2-pyrrolo[1 ,2-c]pyrimtdin-3-yi- chromen-4-one oxime;
6-[2-(4-ethyl-piperazin-1-yl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-y[-chromen-4-one oxime;
6-(2-amino-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-(2-Morphoiin-4-yl-ethoxy)-2-(8aH-pyrrolo[1 ,2-a]pyrazin-3-yi)-chromen-4-one oxime;
6-[2-{4,4-Difluoro-piperidin-1 -yl)-ethoxy]-2-pyrroio[1 ,2-a]pyrazin-3-yl-chromen-4- one oxime;
6-(2-lmidazol-1 -yI-ethoxy)-2-pyrrolo[1 ,2-a]pyrazin-3-yi-chromen-4-one oxime;
6-[2-(4-Fiuoro-phenyl)-ethoxy]-2-pyrrolo[1 ,2-a]pyraztn-3-yl-chromen-4-one oxime;
6-(2-Morphoiin-4-yl-ethoxy)-2 hieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(4,4-Drfluoro-piperidin-1-yl)-ethoxy]- 2-thieno[3,2-c]pyridtn-6-yl-chromeri-4- one oxime;
6-(2-imidazoI-1-yl-ethoxy) 2-thieno[3,2-c]pyridin-6-yi-chromen-4-one oxime;
6-[2-(4-Fluoro-phenyl)-ethoxy]-2-ihieno[3!2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(3,3-Diftuoro-pipendin-1-yl)-ethoxy]-2-thienoE3(2-c]pyridin~6-yl-chromen-4-one oxime;
6-[2-(2,6-Dimethyl-morpholin-4-yl)-ethoxy]-2-ihieno[3,2-c]pyridin~6-yl-chromen-4- one oxime;
6-[2-(3,3-Difiuoro-pyrroiidin-1-yi)-ethoxy]-2-thieno[3,2-c]pyridin-6-yl-chromen-4- one oxime;
6-(3-Pyridin-4-yi-propoxy)-2-ihieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-{3-Pyrsdsn-3-y!-propoxy)-2-thieno{3,2-c3pyndin-6~yl-chromen-4-one oxime;
6-(2-Pyridin-4-yl-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime; 2-Pyrroio[1 ,2-c]pynmidin-3-y[-6-[2-(4-trifluoromeihyl-phenyi)-ethoxy]-chromen-4- one, oxime;
6-[2-(3-Fluoro-phenyl)-ethoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one, oxime;
5- Methoxy-2-pyrroio[1 ,2-c]pyrimidin-3-yi-chromen-4-one oxime;
6- [2-(4-Chloro-phenyl)-ethoxy]-2-pyrroio[1 ,2-cjpyrimidin-3-yl-chromen-4-one oxime;
2~Pyrroio[1 ,2-c]pyrimidin-3-yi-6-[2-(3-trifiuoromethyl-pheny!)-ethoxy]-chromen-4- one oxime;
7- (2-Morpholin-4-yl-eihoxy)-2-pyrro!o[1 ,2-c3pyrimidin-3-yl-chromen-4-one oxime;
7-(3-Morpholsn-4-yi-propoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[(4-Fluoro-benzyiamino)-methyl]-2-pyrroio[1 l2-c]pyrimidin-3-y[-chromen-4-one oxime;
6-{f2-(4-F)uoro-phenyl)-ethylamino]-methy(}-2-pyrrolo[1 ,2-c]pyrimidin-3-yl- chromen-4-one oxime;
e-Phenethyloxy^-pyrroioCI ^-cJpyrimidin-S-yi-chromen^-one oxime;
6-[2-(Pyridin-4-yioxy)-ethoxy]-2-pyrrolo[1 l2-c]pyrimid'in-3-yi-chromen-4-one oxime;
6-[2~{Pyridin-3-yioxy)-ethoxy]-2-pyrrolo[1 I2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[3-(Pyridin-3-yloxy)-propoxy]-2-pyrroloi1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
N-(4-Fluoro-phenyl)-2-{4-hydroxyimino-2-pyrrolo[1 2-c]pyrimidin-3-yl-4H-chromen- 6-yloxy}-acetamide;
N-(4-Fluoro-phenyl)-2-{4-hydroxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H-chromen-
6- yloxy}-N-methyl-acetamide;
N-(5-Fluoro-pyndin-2-yl)-2-{4-hydroxyimino-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-4H- chromen-6-yloxy}-acetamide;
2-Pyrrolo[1 ,2-c]pyrimidin-3-yl-5-trifluoromethyl-chromen-4-one oxime;
2-(7-tert-Butyi-pyrroio[1 l2-c]pyrimidin-3-yl)-6-(2-morpholin-4-yi-ethoxy)-chromeri- 4-one oxime;
7- (2-Morpho[in-4-yl-ethoxy)-2-thieno[2,3-c]pyridin-5-yl-chromen-4-one oxime; 5-{2- orpholin-4-y[-ethoxy)-2-pyrrolo[1,2-c]pyrimidin-3-yl-chromen-4-one oxime;
5- (3-Morpholin-4-yi-propoxy)-2-pyrroSo[1 ,2-c]pynmidin-3-yl-chromen-4-one oxime;
6- (2- orpholin-4-yl-ethoxy)-2-thieno[2,3-c]pyridin-5-yi-chromen-4-one oxime;
6-(3-Pyridin-4-yl-propoxy)-2-pyrrolo[1 ,2-c]pyrimidsn-3-yl-chromen-4-one oxime;
6-(2-Phenoxy-ethoxy)-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[3-{4-Fluoro-phenoxy)-propoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
e-iS-Phenoxy-propoxyJ^-pyrrolof ^-clpyrimidin-S-yi-chromen^-one oxime;
6-[3-(3-Fluoro-phenoxy)-propoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4-one oxime;
6-[3-(3,4-Difluoro-phenoxy)-propoxy]-2-pyrrolo[1 ,2-c]pyrimidin-3-yl-chromen-4- one oxime;
6-(2-[1 ,4]Oxa2epan-4-yi-ethoxy)-2-ihieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(3,4-Dihydro-1 H-isoquinolin-2-yl)-ethoxy]-2-thieno[3,2-c]pyridin-6-yi-ch 4-one oxime;
6-[2-{4-fiuoro-piperidin-1-yl)-eihoxy]-2-thieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-[2-(1 , 1 -Dioxo-1 -thiomorpholin-4-yl)-et^
4-one oxime;
6-(1-Pyrimidin-2-y!-piperidin-4-yloxy)~2-ihieno[3,2-c]pyridin-6-yl-chromen-4-one oxime;
6-(3,4,5l6-Tetrahydro-2H-[1 ,2']bipyridinyl-4-yloxy)-2-thieno[3,2-c]pyridsn-6-yi- chromen-4-one oxime; or
a pharmaceutically acceptable salt, solvate or prodrug thereof.
9. A pharmaceuticai composition comprising the compound of any of claims 1 to 8 and a pharmaceutically acceptable excipient.
10. The compound of any of claims 1 to 8 for use as a medicament.
1 1. A method of treating or preventing a disease or disorder, the method comprising the administration of the compound of any of ciaims 1 to 8 or the pharmaceutical composition of claim 9 to a subject in need of such treatment or prevention.
12. The compound of any of claims 1 to 8 or the pharmaceutical composition of claim 9 for use in treating or preventing a condition associated with altered glutamatergic signailing and/or functions, or a condition which can be affected by alteration of glutamate level or signalling.
13. A method of treating or preventing a condition associated with altered glutamatergic signalling and/or functions, or a condition which can be affected by alteration of glutamate level or signalling, the method comprising the administration of the compound of any of claims 1 to 8 or the pharmaceuticai composition of claim 9 to a subject in need of such treatment or prevention.
14. The compound or the pharmaceutical composition of claim 12 or the method of claim 13, wherein said condition associated with altered glutamatergic signalling and/or functions, or said condition which can be affected by alteration of glutamate level or signalling is selected from: dementias, Parkinsonism and movement disorders, multiple sclerosis and demyelinating diseases, acute or chronic pain, anxiety disorders, schizophrenia, diabetes, or cancers.
15. The compound or the pharmaceutical composition or the method of claim 14, wherein said dementias are selected from: dementias of the Alzheimer's type (DAT); Alzheimer's disease; Pick's disease; vascuiar dementias; Lewy-body disease; dementias due to metabolic, toxic and deficiency diseases, including alcoholism, hypothyroidism, and vitamin B12 deficiency; AlDS-dementia complex; Creutzfeld-Jacob disease; or atypical subacute spongiform encephalopathy.
16. The compound or the pharmaceutical composition or the method of claim 14, wherein said Parkinsonism and movement disorders are selected from: Parkinson's disease; multiple system atrophy; progressive supranuclear palsy; corticobasal degeneration; hepatolenticular degeneration; chorea, including Huntington's disease and hemiballismus; athetosis; dystonias, including spasmodic torticollis, occupational movement disorder, and Gilles de la Tourette syndrome; tardive or drug induced dyskinesias; tremor; or myoclonus.
17. The compound or the pharmaceutical composition or the method of claim 14, wherein said anxiety disorders are selected from: panic disorders, phobias, obsessive-compulsive disorders, stress disorders, or generalized anxiety disorders.
18. The pharmaceutical composition of any of claims 9, 12 or 14 to 17 or the compound of any of claims 10, 12 or 14 to 17 or the method of any of claims 11 or 13 to 17, whereby said pharmaceutical composition or said compound is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestina! route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
19. The method of any of claims 11 or 13 to 18, wherein said subject is a human.
20. A method for identifying an agent that binds to metabotropic giutamate receptor 4 (mGluR4), comprising the following steps:
(a) contacting mGluR4 with the compound of any of claims 1 to 8, wherein said compound is radio-labeled or fluorescence-labeled, under conditions that permit binding of the compound to mGiuR4, thereby generating bound, iabeled compound;
(b) detecting a signal that corresponds to the amount of bound, labeled compound in the absence of test agent;
(c) contacting the bound, iabeled compound with a test agent;
(d) detecting a signal that corresponds to the amount of bound labeled compound in the presence of test agent; and
(e) comparing the signal detected in step (d) to the signal detected in step (b) to determine whether the test agent binds to mGluR4.
PCT/EP2010/066537 2009-10-30 2010-10-29 Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors WO2011051478A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
ES10781455.0T ES2522827T3 (en) 2009-10-30 2010-10-29 New oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
CN201080059805.3A CN102812014B (en) 2009-10-30 2010-10-29 The purposes of novel 9 oxime derivate and the allosteric modulators as metabotropic glutamate receptor thereof
DK10781455.0T DK2493871T3 (en) 2009-10-30 2010-10-29 New oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
JP2012535861A JP5806672B2 (en) 2009-10-30 2010-10-29 Use of novel oxime derivatives and metabotropic glutamate receptors as allosteric modulators
CA2779073A CA2779073C (en) 2009-10-30 2010-10-29 Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
US13/505,235 US8962627B2 (en) 2009-10-30 2010-10-29 Oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
AU2010311321A AU2010311321B2 (en) 2009-10-30 2010-10-29 Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
EP10781455.0A EP2493871B1 (en) 2009-10-30 2010-10-29 Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
IL219149A IL219149A (en) 2009-10-30 2012-04-15 Oxime derivatives and thier use as allosteric modulators of metabotropic glutamate receptors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP09360049 2009-10-30
EP09360049.2 2009-10-30

Publications (1)

Publication Number Publication Date
WO2011051478A1 true WO2011051478A1 (en) 2011-05-05

Family

ID=41796035

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/066537 WO2011051478A1 (en) 2009-10-30 2010-10-29 Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors

Country Status (10)

Country Link
US (1) US8962627B2 (en)
EP (1) EP2493871B1 (en)
JP (1) JP5806672B2 (en)
CN (1) CN102812014B (en)
AU (1) AU2010311321B2 (en)
CA (1) CA2779073C (en)
DK (1) DK2493871T3 (en)
ES (1) ES2522827T3 (en)
IL (1) IL219149A (en)
WO (1) WO2011051478A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140135840A (en) * 2012-03-23 2014-11-26 피티씨 테라퓨틱스, 인크. Compounds for treating spinal muscular atrophy
WO2016030444A1 (en) 2014-08-27 2016-03-03 Prexton Therapeutics Sa Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
WO2017032874A1 (en) 2015-08-27 2017-03-02 Prexton Therapeutics Sa Brain-penetrant chromone oxime derivative for the therapy of levodopa-induced dyskinesia
US10350266B2 (en) 2017-01-10 2019-07-16 Nodus Therapeutics, Inc. Method of treating cancer with a multiple integrin binding Fc fusion protein
WO2020020783A1 (en) 2018-07-24 2020-01-30 Biontech Rna Pharmaceuticals Gmbh Il2 agonists
WO2020021064A1 (en) 2018-07-26 2020-01-30 Domain Therapeutics Substituted quinazolinone derivatives and their use as positive allosteric modulators of mglur4
US10603358B2 (en) 2017-01-10 2020-03-31 Nodus Therapeutics Combination tumor treatment with an integrin-binding-Fc fusion protein and immune stimulator
WO2022223617A1 (en) 2021-04-20 2022-10-27 BioNTech SE Virus vaccine
WO2023067193A2 (en) 2021-10-22 2023-04-27 BioNTech SE Compositions for administration of different doses of rna
WO2023066496A1 (en) 2021-10-21 2023-04-27 BioNTech SE Coronavirus vaccine
WO2023083434A1 (en) 2021-11-09 2023-05-19 BioNTech SE Rna encoding peptidoglycan hydrolase and use thereof for treating bacterial infection
WO2023194508A1 (en) 2022-04-05 2023-10-12 BioNTech SE Nucleic acid compositions comprising a multivalent anion, such as an inorganic polyphosphate, and methods for preparing, storing and using the same
WO2024017479A1 (en) 2022-07-21 2024-01-25 BioNTech SE Multifunctional cells transiently expressing an immune receptor and one or more cytokines, their use and methods for their production

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104119244B (en) * 2014-06-27 2016-09-07 上海师范大学 Chiral resolution and the method for on-line checking of DL tyrosine is realized based on functional nano channel array
CN111978203A (en) * 2020-08-27 2020-11-24 浙江工业大学 Microwave synthesis method of benzaldehyde oxime compound
CN115594655B (en) * 2022-09-16 2023-08-15 桂林医学院 Chromone oxime derivative and preparation method and application thereof

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4065467A (en) 1973-12-27 1977-12-27 Carlo Erba, S. P. A. 5:6-Benzo δ-pyrone derivatives and process for their preparation
US4777252A (en) 1987-08-13 1988-10-11 E. R. Squibb & Sons, Inc. 2-oxo-1-[[(substituted sulfonyl)amino]-carbonyl]azetidines
EP0321201A2 (en) 1987-12-15 1989-06-21 Gene Shears Pty. Limited Ribozymes
EP0291533B1 (en) 1986-12-03 1995-10-18 University Patents, Inc. Rna ribozyme restriction endoribonucleases and methods
EP0360257B1 (en) 1988-09-20 1996-11-13 The Board Of Regents For Northern Illinois University RNA catalyst for cleaving specific RNA sequences
EP0787723A1 (en) 1994-10-21 1997-08-06 Suntory Limited Cyclopropachromenecarboxylate derivatives
US6342606B2 (en) 1997-10-23 2002-01-29 Kuraray Co., Ltd. Method for producing pyridine derivatives
US6362336B1 (en) 1995-12-18 2002-03-26 Zeneca Limited Chemical compounds
US20030109574A1 (en) 2001-09-28 2003-06-12 Central Glass Company, Limited Process for producing 4-substituted benzopyran derivatives
EP1101781B1 (en) 1999-11-15 2003-10-01 Air Products And Chemicals, Inc. Thixotropic aqueous polymer emulsions comprising polyurethane-polyurea and polyacrylate
US20030236264A1 (en) 2002-02-19 2003-12-25 Rogers Bruce N. Fused bicyclic-N-bridged-heteroaromatic carboxamides for the treatment of disease
WO2004052869A1 (en) 2002-12-12 2004-06-24 F. Hoffmann-La Roche Ag 5-substituted-pyrazine or pyridine glucokinase activators
US20040198750A1 (en) * 2003-04-03 2004-10-07 Jeremy Green Compositions useful as inhibitors of protein kinases
WO2005103003A2 (en) 2004-04-26 2005-11-03 Pfizer Inc. Pyrrolopyridine derivatives and their use as hiv-integrase inhibitors
US20060014948A1 (en) 1998-12-14 2006-01-19 Fujisawa Pharmaceutical Co. Ltd. Piperazine derivatives
WO2008008895A1 (en) 2006-07-13 2008-01-17 Smithkline Beecham Corporation Gpr119 agonists for the treatment of diabetes and related disorders
WO2008062276A2 (en) 2006-11-20 2008-05-29 Glenmark Pharmaceuticals S.A. Acetylene derivatives as stearoyl coa desaturase inhibitors
WO2008086404A1 (en) 2007-01-10 2008-07-17 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
WO2009010455A2 (en) 2007-07-13 2009-01-22 Addex Pharma S.A. Pyrazole derivatives as modulators of metabotropic glutamate receptors
WO2009010454A2 (en) 2007-07-13 2009-01-22 Addex Pharma S.A. Amido derivatives and their use as positive allosteric modulators of metabotropic glutamate receptors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2281580C (en) * 1997-02-27 2003-04-22 Pfizer Inc. Quinoxalinediones
FR2805538B1 (en) * 2000-02-29 2006-08-04 Hoechst Marion Roussel Inc NOVEL FLAVON DERIVATIVES, PROCESS FOR THE PREPARATION THEREOF, THEIR USE AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND NOVEL USE
HUP0400179A3 (en) * 2001-06-12 2004-10-28 Neurogen Corp Branford 2,5-diarylpyrazines, 2,5-diarylpyridines and 2,5-diarylpyrimidines, their use and pharmaceutical compositions containing them
JO2525B1 (en) * 2004-04-08 2010-03-17 شركة جانسين فارماسوتيكا ان. في Substiuted 4-alkyl-and 4-alkanoyl piperidine derivatives and there use as neurokinin antagonist
TW200639163A (en) * 2005-02-04 2006-11-16 Genentech Inc RAF inhibitor compounds and methods
US7999107B2 (en) * 2007-01-31 2011-08-16 Merck Sharp & Dohme Corp. Substituted pyrano[2,3-B]pyridine derivatives as cannabinoid-1 receptor modulators

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4065467A (en) 1973-12-27 1977-12-27 Carlo Erba, S. P. A. 5:6-Benzo δ-pyrone derivatives and process for their preparation
EP0291533B1 (en) 1986-12-03 1995-10-18 University Patents, Inc. Rna ribozyme restriction endoribonucleases and methods
US4777252A (en) 1987-08-13 1988-10-11 E. R. Squibb & Sons, Inc. 2-oxo-1-[[(substituted sulfonyl)amino]-carbonyl]azetidines
EP0321201A2 (en) 1987-12-15 1989-06-21 Gene Shears Pty. Limited Ribozymes
EP0360257B1 (en) 1988-09-20 1996-11-13 The Board Of Regents For Northern Illinois University RNA catalyst for cleaving specific RNA sequences
EP0787723A1 (en) 1994-10-21 1997-08-06 Suntory Limited Cyclopropachromenecarboxylate derivatives
US6362336B1 (en) 1995-12-18 2002-03-26 Zeneca Limited Chemical compounds
US6342606B2 (en) 1997-10-23 2002-01-29 Kuraray Co., Ltd. Method for producing pyridine derivatives
US20060014948A1 (en) 1998-12-14 2006-01-19 Fujisawa Pharmaceutical Co. Ltd. Piperazine derivatives
EP1101781B1 (en) 1999-11-15 2003-10-01 Air Products And Chemicals, Inc. Thixotropic aqueous polymer emulsions comprising polyurethane-polyurea and polyacrylate
US20030109574A1 (en) 2001-09-28 2003-06-12 Central Glass Company, Limited Process for producing 4-substituted benzopyran derivatives
US20030236264A1 (en) 2002-02-19 2003-12-25 Rogers Bruce N. Fused bicyclic-N-bridged-heteroaromatic carboxamides for the treatment of disease
WO2004052869A1 (en) 2002-12-12 2004-06-24 F. Hoffmann-La Roche Ag 5-substituted-pyrazine or pyridine glucokinase activators
US20040198750A1 (en) * 2003-04-03 2004-10-07 Jeremy Green Compositions useful as inhibitors of protein kinases
WO2004092154A1 (en) 2003-04-03 2004-10-28 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
WO2005103003A2 (en) 2004-04-26 2005-11-03 Pfizer Inc. Pyrrolopyridine derivatives and their use as hiv-integrase inhibitors
WO2008008895A1 (en) 2006-07-13 2008-01-17 Smithkline Beecham Corporation Gpr119 agonists for the treatment of diabetes and related disorders
WO2008062276A2 (en) 2006-11-20 2008-05-29 Glenmark Pharmaceuticals S.A. Acetylene derivatives as stearoyl coa desaturase inhibitors
WO2008086404A1 (en) 2007-01-10 2008-07-17 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
WO2009010455A2 (en) 2007-07-13 2009-01-22 Addex Pharma S.A. Pyrazole derivatives as modulators of metabotropic glutamate receptors
WO2009010454A2 (en) 2007-07-13 2009-01-22 Addex Pharma S.A. Amido derivatives and their use as positive allosteric modulators of metabotropic glutamate receptors

Non-Patent Citations (63)

* Cited by examiner, † Cited by third party
Title
BATTAGLIA G, J. NEUROSCI., vol. 26, no. 27, 2006, pages 7222 - 9
BIOORG. MED. CHEM., vol. 11, 2003, pages 2769 - 2782
BOLDYREV AA; JOHNSON P, J. ALZHEIMERS DIS., vol. 11, no. 2, 2007, pages 219 - 28
BRAUNER-OSBORNE H ET AL., J. MED. CHEM., vol. 43, no. 14, 2000, pages 2609 - 45
BRIDGES TM ET AL., ACS CHEM BIOL, vol. 3, no. 9, 2008, pages 530 - 41
BROEKKAMP ET AL., EUR. J. PHARMACOL, vol. 126, 1986, pages 223 - 229
BRUNO V ET AL., J. CEREB. BLOOD FLOW METAB., vol. 21, no. 9, 2001, pages 1013 - 33
BUNDGAARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
CHANG HJ ET AL., CLI. CANCER RES., vol. 11, no. 9, 2005, pages 3288 - 95
CONN PJ ET AL., NAT. REV. NEUROSCIENCE, vol. 6, no. 10, 2005, pages 787 - 98
CONN PJ ET AL., TRENDS PHARMACOL. SCI., vol. 30, no. 1, 2009, pages 25 - 31
CONN PJ; PIN JP., ANNU. REV. PHARMACOL. TOXICOL., vol. 37, 1997, pages 205 - 37
CRYAN J F ET AL., EUR. J. NEUROSC., vol. 17, no. 11, 2003, pages 2409 - 17
ENGERS DW ET AL., J. MED. CHEM., 27 May 2009 (2009-05-27)
ESCHLE BK., NEUROSCIENCE, vol. 155, no. 2, 2008, pages 522 - 9
FLOR PJ; LUKIC S; RUEGG D; LEONHARDT T; KNBPFEL T; KUHN R., NEUROPHARMACOLOGY, vol. 34, 1995, pages 149 - 155
GOLD, ANN. REV. BIOCHEM., vol. 64, 1995, pages 763 - 797
GOUDET C ET AL., PAIN, vol. 137, no. 1, 2008, pages 112 - 24
HANSSON, LMMUNOTECHNOLOGY, vol. 4, 1999, pages 237 - 252
HARLOW; LANE: "Antibodies, A Laboratory Manual", 1988, CSH PRESS
HENNING, HUM GENE THER., vol. 13, 2000, pages 1427 - 1439
J. CHEM. SOC., PERKIN TRANS, vol. 1, 2000, pages 3317 - 3324
J. MED. CHEM, vol. 42, no. 11, 1999, pages 1881 - 1893
J. MED. CHEM, vol. 47, 2004, pages 6466 - 6475
J. MED. CHEM., vol. 49, 2006, pages 4425 - 4436
J. ORG. CHEM., vol. 68, 1993, pages 7903 - 7905
JAGS, vol. 126, no. 3, 2004, pages 712 - 713
K. W. ANDERSON, JACS, vol. 128, 2006, pages 10694 - 10695
KLAK K ET AL., AMINO ACIDS, vol. 32, no. 2, 2006, pages 169 - 72
KONIECZNY J ET AL., NAUNYN-SCHMIEDERBERGS ARCH. PHARMACOL., vol. 358, no. 4, 1998, pages 500 - 2
LACOVELLI L ET AL., J. NEUROSCI., vol. 26, no. 32, 2006, pages 8388 - 97
LOPEZ S ET AL., NEUROPHARMACOLOGY, vol. 55, no. 4, 2008, pages 483 - 90
MAJ M ET AL., NEUROPHARMACOLOGY, vol. 45, no. 7, 2003, pages 895 - 903
MAKOFF A; LELCHUK R; OXER M; HARRINGTON K; EMSON P., BRAIN RES. MOL. BRAIN RES., vol. 37, 1996, pages 239 - 248
MARINO MJ ET AL., PROC. NATL. ACAD. SCI. USA, vol. 100, no. 23, 2003, pages 13668 - 73
MATHIESEN JM ET AL., BR. J. PHARMACOL., vol. 138, no. 6, 2003, pages 1026 - 30
MATTSON MP., NEUROMOLECULAR MED., vol. 3, no. 2, 2003, pages 65 - 94
MELI E ET AL., PHARMACOL. BIOCHEM. BEHAV., vol. 73, no. 2, 2002, pages 439 - 46
NISWENDER CM ET AL., BIOORG. MED. CHEM. LETT., vol. 18, no. 20, 2008, pages 5626 - 30
NISWENDER CM ET AL., MOL. PHARMACOL., vol. 74, no. 5, 2008, pages 1345 - 58
NISWENDER CM ET AL., MOL. PHARMACOL., vol. 74, no. 5, 2008, pages 1345 - 88
PALUCHA A ET AL., NEUROPHARMACOLOGY, vol. 46, no. 2, 2004, pages 151 - 9
PALUCHA IA, PHARMACOL.THER., vol. 115, no. 1, 2007, pages 116 - 47
PALUCHA-PONIEWIERA A ET AL., NEUROPHARMACOLOGY, vol. 55, no. 4, 2008, pages 517 - 24
PESSIMISSIS N ET AL., ANTICANCER RES., vol. 29, no. 1, 2009, pages 371 - 7
PILC A ET AL., NEUROPHARMACOLOGY, vol. 43, no. 2, 2002, pages 181 - 7
PIN JP; ACHER F., CURR. DRUG TARGETS CNS NEUROL. DISORD., vol. 1, no. 3, 2002, pages 297 - 317
PIRES ET AL., J MED AND BIOL RES, vol. 38, 2005, pages 1867 - 1872
SCHOEPP DD ET AL., NEUROPHARMACOLOGY, vol. 38, no. 10, 1999, pages 1431 - 76
SHIOZAKI ET AL., PSYCHOPHARMACOLOGY, vol. 147, 1999, pages 90 - 95
STACHOWICZ K ET AL., EUR. J. PHARMACOL., vol. 498, no. 1-3, 2004, pages 153 - 6
STEPHANS SE; YAMAMOTO BK, SYNAPSE, vol. 17, no. 3, 1994, pages 203 - 9
SYNTHESIS, 1984, pages 53 - 60
TANABE Y; MASU M; ISHII T; SHIGEMOTO R; NAKANISHI S., NEURON, vol. 8, 1992, pages 169 - 179
TETRAHEDRON LETTERS, 2004, pages 553 - 556
UEHARA S., DIABETES, vol. 53, no. 4, 2004, pages 998 - 1006
VERNON AC ET AL., J. PHARMACOL. EXP. THER., vol. 320, no. 1, 2007, pages 397 - 409
VERNON AC ET AL., NEUROREPORT, vol. 19, no. 4, 2008, pages 475 - 8
WILLIAMS R ET AL., BIOORG. MED. CHEM. LETT., vol. 19, no. 3, 2009, pages 962 - 6
WILLIAMS R ET AL: "Positive allosteric modulators of the metabotropic glutamate receptor subtype 4 (mGluR4). Part II: Challenges in hit-to-lead", 1 February 2009, BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, PAGE(S) 962 - 966, ISSN: 0960-894X, XP025925862 *
WU S; WRIGHT RA; ROCKEY PK; BURGETT SG; ARNOLD JS; ROSTECK PR JR; JOHNSON BG; SCHOEPP DD; BELAGAJE RM., BRAIN RES. MOL. BRAIN RES., vol. 53, 1998, pages 88 - 97
XU, CHEM. BIOL., vol. 9, 2002, pages 933
ZHANG HM ET AL., NEUROSCIENCE, vol. 158, no. 2, 2009, pages 875 - 84

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9914722B2 (en) 2012-03-23 2018-03-13 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
EP2828247A4 (en) * 2012-03-23 2015-12-16 Ptc Therapeutics Inc Compounds for treating spinal muscular atrophy
KR102109992B1 (en) * 2012-03-23 2020-05-13 피티씨 테라퓨틱스, 인크. Compounds for treating spinal muscular atrophy
KR20140135840A (en) * 2012-03-23 2014-11-26 피티씨 테라퓨틱스, 인크. Compounds for treating spinal muscular atrophy
RU2672569C2 (en) * 2014-08-27 2018-11-16 Прекстон Терапьютикс Са Chromon oxime new derivative and its use as allosteric modulator of metabotropic glutamate receptors
JP2017525776A (en) * 2014-08-27 2017-09-07 プレクストン・セラピューティクス・ソシエテ・アノニム A novel chromone oxime derivative and its use as an allosteric modulator of metabotropic glutamate receptors
KR101870589B1 (en) 2014-08-27 2018-06-25 프렉스톤 테라퓨틱스 에스에이 Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
US10017521B2 (en) 2014-08-27 2018-07-10 Prexton Therapeutics Sa Chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
WO2016030444A1 (en) 2014-08-27 2016-03-03 Prexton Therapeutics Sa Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
AU2015308438B2 (en) * 2014-08-27 2018-12-13 Prexton Therapeutics Sa Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
EP3502115A1 (en) 2014-08-27 2019-06-26 Prexton Therapeutics SA Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
EP3696183A1 (en) 2014-08-27 2020-08-19 Prexton Therapeutics SA Novel chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
US10442818B2 (en) 2014-08-27 2019-10-15 Prexton Therapeutics Sa Chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
WO2017032874A1 (en) 2015-08-27 2017-03-02 Prexton Therapeutics Sa Brain-penetrant chromone oxime derivative for the therapy of levodopa-induced dyskinesia
US10532057B2 (en) 2015-08-27 2020-01-14 Prexton Therapeutics Sa Brain-penetrant chromone oxime derivative for the therapy of levodopa-induced dyskinesia
EA035016B1 (en) * 2015-08-27 2020-04-17 Прекстон Терапьютикс Са Brain-penetrant chromone oxime derivative for the therapy of levodopa-induced dyskinesia
EP3696182A1 (en) 2015-08-27 2020-08-19 Prexton Therapeutics SA Brain-penetrant chromone oxime derivative for the therapy of levodopa-induced dyskinesia
US10350266B2 (en) 2017-01-10 2019-07-16 Nodus Therapeutics, Inc. Method of treating cancer with a multiple integrin binding Fc fusion protein
US10603358B2 (en) 2017-01-10 2020-03-31 Nodus Therapeutics Combination tumor treatment with an integrin-binding-Fc fusion protein and immune stimulator
WO2020020783A1 (en) 2018-07-24 2020-01-30 Biontech Rna Pharmaceuticals Gmbh Il2 agonists
WO2020021064A1 (en) 2018-07-26 2020-01-30 Domain Therapeutics Substituted quinazolinone derivatives and their use as positive allosteric modulators of mglur4
WO2022223617A1 (en) 2021-04-20 2022-10-27 BioNTech SE Virus vaccine
WO2023066496A1 (en) 2021-10-21 2023-04-27 BioNTech SE Coronavirus vaccine
WO2023067193A2 (en) 2021-10-22 2023-04-27 BioNTech SE Compositions for administration of different doses of rna
EP4238577A2 (en) 2021-10-22 2023-09-06 BioNTech SE Compositions for administration of different doses of rna
WO2023083434A1 (en) 2021-11-09 2023-05-19 BioNTech SE Rna encoding peptidoglycan hydrolase and use thereof for treating bacterial infection
WO2023194508A1 (en) 2022-04-05 2023-10-12 BioNTech SE Nucleic acid compositions comprising a multivalent anion, such as an inorganic polyphosphate, and methods for preparing, storing and using the same
WO2023193892A1 (en) 2022-04-05 2023-10-12 BioNTech SE Nucleic acid compositions comprising an inorganic polyphosphate and methods for preparing, storing and using the same
WO2024017479A1 (en) 2022-07-21 2024-01-25 BioNTech SE Multifunctional cells transiently expressing an immune receptor and one or more cytokines, their use and methods for their production

Also Published As

Publication number Publication date
ES2522827T3 (en) 2014-11-18
CN102812014A (en) 2012-12-05
CA2779073C (en) 2017-10-24
US20120277212A1 (en) 2012-11-01
AU2010311321A1 (en) 2012-05-10
JP5806672B2 (en) 2015-11-10
CN102812014B (en) 2016-01-20
IL219149A0 (en) 2012-06-28
EP2493871A1 (en) 2012-09-05
IL219149A (en) 2016-06-30
EP2493871B1 (en) 2014-09-03
AU2010311321B2 (en) 2014-11-27
DK2493871T3 (en) 2014-11-10
CA2779073A1 (en) 2011-05-05
JP2013509384A (en) 2013-03-14
US8962627B2 (en) 2015-02-24

Similar Documents

Publication Publication Date Title
EP2493871B1 (en) Novel oxime derivatives and their use as allosteric modulators of metabotropic glutamate receptors
US10562915B2 (en) Bromodomain inhibitors
CA2861609C (en) Compounds for treating spinal muscular atrophy
JP4709849B2 (en) Bicyclic and tricyclic substituted phenylmethanones as glycine transporter I (GLYT-1) inhibitors for the treatment of Alzheimer&#39;s disease
DK2770997T3 (en) SUBSTITUTED 2- (4-HETEROCYCLYLBENSYL) isoindolin-1-ON-ANALOGS AS POSITIVE allosteric modulators OF THE muscarinic acetylcholine receptor M1
JP4719317B2 (en) Fused heterocyclic derivatives and uses thereof
CN107428690A (en) Mutation IDH1 inhibitor available for treating cancer
KR101030629B1 (en) HETEROARYL SUBSTITUTED 2-PYRIDINYL AND 2-PYRIMIDINYL-6,7,8,9-TETRAHYDROPYRIMIDO[1,2-a]PYRIMIDIN-4-ONE DERIVATIVES
CN108834415B (en) Substituted tricyclic 1, 4-benzodiazepine derivatives as allosteric modulators of group II metabotropic glutamate receptors
JP2015523406A (en) 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta [a] naphthalene
TW201319067A (en) Triazolopyridine compounds
EA027082B1 (en) Imidazole derivatives as pde10a enzyme inhibitors
KR20150105319A (en) Novel substituted pyrido-piperazinone derivatives as gamma secretase modulators
CN116546985A (en) Pyridopyrimidine derivative and preparation method and application thereof
KR20210039368A (en) Substituted quinazolinone derivatives and their use as positive allosteric modulators of mGluR4
WO2016172547A1 (en) Substituted imidazopyridine and triazolopyridine analogs as positive allosteric modulators of muscarinic acetylcholine receptor m1
WO2016046404A1 (en) Substituted pyrazoloquinazolinones and pyrroloquinazolinones as allosteric modulators of group iimetabotropic glutamate receptors
WO2017126635A1 (en) Heterocyclic compound and use thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080059805.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10781455

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 219149

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2010311321

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2779073

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012535861

Country of ref document: JP

Ref document number: 3734/DELNP/2012

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2010311321

Country of ref document: AU

Date of ref document: 20101029

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010781455

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13505235

Country of ref document: US