WO2017040993A1 - Small molecule inhibitors of dyrk1a and uses thereof - Google Patents

Small molecule inhibitors of dyrk1a and uses thereof Download PDF

Info

Publication number
WO2017040993A1
WO2017040993A1 PCT/US2016/050198 US2016050198W WO2017040993A1 WO 2017040993 A1 WO2017040993 A1 WO 2017040993A1 US 2016050198 W US2016050198 W US 2016050198W WO 2017040993 A1 WO2017040993 A1 WO 2017040993A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
dyrk1a
nmr
mhz
compound
Prior art date
Application number
PCT/US2016/050198
Other languages
French (fr)
Other versions
WO2017040993A4 (en
Inventor
Christopher Hulme
Original Assignee
The Arizona Board Of Regents On Behalf Of The University Of Arizona
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 The Arizona Board Of Regents On Behalf Of The University Of Arizona filed Critical The Arizona Board Of Regents On Behalf Of The University Of Arizona
Priority to EP16843107.0A priority Critical patent/EP3344039A4/en
Priority to US15/756,917 priority patent/US10730842B2/en
Priority to JP2018531299A priority patent/JP2018526452A/en
Priority to MX2018002631A priority patent/MX2018002631A/en
Priority to AU2016315881A priority patent/AU2016315881B2/en
Priority to CA2997556A priority patent/CA2997556C/en
Publication of WO2017040993A1 publication Critical patent/WO2017040993A1/en
Publication of WO2017040993A4 publication Critical patent/WO2017040993A4/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 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
    • C07D405/02Heterocyclic 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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

Definitions

  • This invention is in the field of medicinal chemistry.
  • the invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRK1A protein, and their use as therapeutics for the treatment of Alzheimer's disease, Down syndrome, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), and other diseases.
  • AD Alzheimer's disease
  • the underlying treatment of learning and/or memory disorders is a huge and significantly unmet medical need and also included learning and memory repair after, for example, incidents of stroke or significant brain damage. As such, an improved understanding of the dementia (and other neuropathology) and related improved treatment methods are needed.
  • NFTs neuroofibrillary tangles
  • DYRKIA phosphorylation regulated kinase-1 A
  • AD Alzheimer's disease
  • DS Down syndrome critical region
  • DYRKIA decreased expression of the DYRKIA gene has been implicated in both the cognitive deficits of Down syndrome (DS) and the early onset of tau and amyloid neuropathologies that are associated with this genetic disorder.
  • DYRKIA levels are increased in transgenic mouse models of DS and develop DS-like phenotypes including hippocampal-dependent spatial learning and memory deficits and developmental delays. Together these data strongly support a central function for DYRKIA in cognitive deficits associated with DS.
  • inhibition of excess DYRKIA activity has been shown to improve these DYRKlA-mediated cognitive deficits after administration of the natural products epigallocatechin-3-gallate (EGCg) and harmine, the standards for DYRKIA inhibition at the on-set of this translational campaign.
  • ECCg epigallocatechin-3-gallate
  • DYRKIA inhibitors described herein can also be considered as potential
  • DYRKIA inhibitors of the present invention have been also implicated as potential therapeutics for the treatment of glioblastomas and further potential utility is highlighted in the oncology arena (see, e.g., Ionescu et al, Mini-reviews in Medicinal Chemistry, 2012, 12, 1315-1329).
  • DYRKIA inhibitors may also have utility as general cognitive enhancers, given the published findings that DYRKIA can phosphorylate sirtuin 1 , a key regulator of learning and memory (see, e.g., Michan et al, J. Neurosci. 2010, 30(29), 9695-9707; Guo et al, J Biol. Chem. 2010, 285 (17), 13223-13232).
  • the potential utility of these DYRKIA compound series is further reinforced by findings that harmine, a potent, but relatively less selective DYRKIA inhibitor, enhances memory performance in wild-type rodents (Mennenga et al, Physiol. Behav. 2015, 138, 260-265).
  • DYRKIA as a physiologically relevant regulator of T reg cell differentiation and suggest a broader role for other DYRK family members in immune homeostasis. As such, new roles may be found in autoimmune diseases such as inflammatory bowel disease and type 1 diabetes (see, e.g., Khor B, et al, eLife 2015;4: e05920).
  • this invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRKIA protein, and their use as therapeutics for the treatment of disorders related to DYRKIA activity (e.g., AD, DS, neuropathology, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation)).
  • disorders related to DYRKIA activity e.g., AD, DS, neuropathology, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation)
  • benzimidazole and imidazo ridine compounds e.g., benzimidazole and imidazo ridine compounds
  • Formula I is not limited to a particular chemical moiety for Rl, R2, R3, R4, X, Y and Z.
  • the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit DYRKIA activity.
  • the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit one or more of: DYRKIA related PI3K/Akt signaling; DYRKIA related tau
  • X-Y-Z is , thereby rendering the resulting compound a benzimidazole compound having the following formula:
  • X-Y-Z is , thereb rendering the compound
  • Rl is selected from hydrogen, aryl and substituted aryl
  • R5 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
  • R6 is selected from hydrogen, C1 -C4 alkyl, heterocyclyl alkyl, heteroaryl alkyl, aryl alkyl, aryl, heterocyclyl, and heteroaryl.
  • R7 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
  • each of R2 and R3 is independently selected from hydrogen, aryl,
  • the compound is one or more of the compounds shown in Examples II and/or III.
  • the invention further provides processes for preparing any of the compounds of the present invention.
  • the invention also provides the use of compounds to not only inhibit DYRKIA activity but also signaling pathways dependent upon DYRKIA phosphorylation (e.g., Tau, POK/AKt, APP, PSI, ASF, RCAN-1 , NFAT, p53, ASK1/JNK1, SIRT1 , GluN2A and other NMD A receptors).
  • the invention also relates to the use of compounds for sensitizing cells to additional agent(s), such as agents known to be effective in the treatment of neurodegenerative disorders.
  • the compounds of the invention are useful for the treatment, amelioration, or prevention of disorders associated with DYRKIA activity (e.g., AD, DS, Parkinson's disease, Huntington's disease, glioblastoma), such as those responsive to DYRKIA activity inhibition.
  • the compounds can be used to treat, ameliorate, or prevent cancer that is associated with DYRKIA activity (e.g., glioblastoma).
  • the compounds can be used to treat, ameliorate, or prevent autoimmune diseases.
  • the compounds can be used to treat, ameliorate, or prevent inflammatory disorders (e.g., airway inflammation).
  • the invention also provides pharmaceutical compositions comprising the compounds of the invention in a pharmaceutically acceptable carrier.
  • kits comprising a compound of the invention and instructions for administering the compound to an animal.
  • the kits may optionally contain other therapeutic agents, e.g., agents useful in treating neurodegenerative disorders and/or anticancer agents.
  • each of the "*" substituents is independently Carbon or Nitrogen, and X, Y, Z, Rl, R2, R3, and R4 are as described above.
  • FIG. 1 presents a flow chart describing a screening paradigm for DYRKIA inhibitor progression.
  • DYRKIA is a member of the DYRK family containing 5 kinases (DYRKIA, DYRK1B, DYRK2, DYRK3 and DYRK4).
  • DYRKs belong to the CMGC group of proline-directed kinases, which also includes cyclin-dependent kinases (CDKs), mitogen-activated protein kinases (MAPKs), glycogen synthase kinases (GSKs) and CDC2-like kinases (CLKs). While the signaling pathways of CDK and MAPK families have been extensively studied, much less is known on how DYRKs and CLKs are linked to other proteins and various physiological or pathological processes.
  • CDKs cyclin-dependent kinases
  • MAPKs mitogen-activated protein kinases
  • GSKs glycogen synthase kinases
  • CLKs CDC2-like kinases
  • the DYRKIA gene is located on chromosome 21 (21q22.2), a region known as the Down-Syndrome Critical Region (DSCR) (see, e.g., Hammerle et al., 2011 Development 138, 2543-2554).
  • DSCR Down-Syndrome Critical Region
  • the under- or over-expression of the Dyrkla gene in mammals or of its orthologous gene minibrain (mnb) in Drosophila causes severe retardation of central nervous system development and maturation.
  • DYRKIA phosphorylates the nuclear factor of activated T cells (NFAT), counteracting the effect of calcium signaling and maintaining inactive NFAT (see, e.g., Arron et al, 2006 Nature 411, 595-600).
  • DYRKIA has been identified as a negative regulator of the cell cycle that promotes the switch to a quiescent state or differentiation (see, e.g., Chen et al., 2013 Mol. Cell 52, 87-100). In malignant cells, DYRKIA promotes survival via inhibition of pro-apoptotic proteins (see, e.g., Guo et al., 2010 J. Bio. Chem. 285, 13223-13232; Seifert et al, 2008 FEBS J. 275, 6268-6280).
  • the DYRKIA inhibitors of the present invention provide a new avenue for pharmaceutical intervention of mental impairment associated with AD and other neurodegenerative diseases, and address a critical unmet medical need and significantly changing treatment paradigm for AD.
  • Benzimidazole and imidazopyridine compounds as inhibitors of the dual specificity tyrosine phosphorylation regulated kinase- 1 A (DYRKIA) and their potential for use as therapeutics against AD and other disorders related to DYRK-1A activity (e.g., DS, other neuropathology, cancer (e.g., glioblastoma)).
  • DYRKIA dual specificity tyrosine phosphorylation regulated kinase- 1 A
  • Many benzimidazole and imidazopyridine compounds were also shown to exhibit activity against DYRK1B and exhibit some degree of activity against other kinases implicated in a variety of disease states.
  • the DYRKIA inhibitors of the present invention can be used for treating other cellular pathways involved in mental impairment and neurodegenerative dementia.
  • the DYRKIA inhibitors of the present invention can be used for inhibiting DYRKIA activated PI3K/Akt signaling, a pathway largely involved in neuronal development, growth, and survival.
  • the DYRKIA inhibitors of the present invention DYRKIA can be used for inhibiting DYRKIA stimulated ASK1/JNK1 activity, thereby inducing neuronal death and apoptosis.
  • the DYRKIA inhibitors of the present invention DYRKIA can be used to inhibit DYRKIA phosphorylation of p53 during embryonic brain development, thereby preventing neuronal proliferation alteration.
  • the DYRKIA inhibitors of the present invention can be used to inhibit DYRKIA phosphorylation of synaptic proteins Amph 1, Dynamin 1, and Synaptojanin, involved in the regulation of endocytosis, thereby retaining synaptic plasticity through preventing alteration of the number, size, and morphology of dendritic spines.
  • the DYRKIA inhibitors of the present invention can be used to inhibit presenilin 1 (the catalytic sub-unit of ⁇ -secretase).
  • the present invention addresses the need for effective therapies for AD and DS by providing potent and selective DYRKIA inhibitors able to permeate the blood-brain barrier (BBB) and elicit on-mechanism therapeutic responses in AD animal models.
  • BBB blood-brain barrier
  • this invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRKIA protein, and their use as therapeutics for the treatment of Alzheimer's disease, Down syndrome, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), and other diseases.
  • Formula I is not limited to a particular chemical moiety for Rl , R2, R3, R4, X, Y and Z.
  • the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit DYRKIA activity.
  • the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit one or more of: DYRKIA related PI3K/Akt signaling; DYRKIA related tau
  • X-Y-Z is , thereby rendering the resulting
  • X-Y-Z is , hereby rendering the compound an
  • imidazopyridine compound having the following formula:
  • Rl is selected from hydrogen, aryl and substituted aryl
  • R5 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
  • R6 is selected from hydrogen, C1-C4 alkyl, heterocyclyl alkyl, heteroaryl alkyl, aryl alkyl, aryl, heterocyclyl, and heteroaryl.
  • R7 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
  • each of R2 and R3 is independentl from hydrogen, aryl,
  • R4 is selected from hydrogen, NH 2 ,
  • the compound is one or more of the compounds shown in
  • the invention further provides processes for preparing any of the compounds of the present invention.
  • compositions and methods of the present invention are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in an animal (e.g. , a mammalian patient including, but not limited to, humans and veterinary animals).
  • an animal e.g. , a mammalian patient including, but not limited to, humans and veterinary animals.
  • various diseases and pathologies are amenable to treatment or prophylaxis using the present methods and compositions.
  • a non-limiting exemplary list of these diseases and conditions includes, but is not limited to, Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), any neurodegenerative disorder related to DYRK1 A activity, and any type of cancer related to DYRK1A activity.
  • Some embodiments of the present invention provide methods for administering an effective amount of a compound of the invention and at least one additional therapeutic agent (including, but not limited to, any agent useful in treating Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), any neurodegenerative disorder related to DYRKl A activity, and any type of cancer characterized related to DYRKl A activity).
  • additional therapeutic agent including, but not limited to, any agent useful in treating Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), any neurodegenerative disorder related to DYRKl A activity, and any type of cancer characterized related to DYRKl A activity).
  • compositions within the scope of this invention include all compositions wherein the compounds of the present invention are contained in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art.
  • the compounds may be administered to mammals, e.g. humans, orally at a dose of 0.0025 to 50 mg/kg, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated for disorders responsive to induction of apoptosis. In one embodiment, about 0.01 to about 25 mg/kg is orally administered to treat, ameliorate, or prevent such disorders.
  • the dose is generally about one-half of the oral dose.
  • a suitable intramuscular dose would be about 0.0025 to about 25 mg/kg, or from about
  • the unit oral dose may comprise from about 0.01 to about 1000 mg, for example, about 0.1 to about 100 mg of the compound.
  • the unit dose may be administered one or more times daily as one or more tablets or capsules each containing from about 0.1 to about 10 mg, conveniently about 0.25 to 50 mg of the compound or its solvates.
  • the compound may be present at a concentration of about 0.01 to
  • the compound is present at a concentration of about 0.07-1.0 mg/ml, for example, about 0.1-0.5 mg/ml, and in one embodiment, about 0.4 mg/ml.
  • the compounds of the invention may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compounds into preparations which can be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compounds into preparations which can be used pharmaceutically.
  • the preparations particularly those preparations which can be administered orally or topically and which can be used for one type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by intravenous infusion, injection, topically or orally, contain from about 0.01 to 99 percent, in one embodiment from about 0.25 to 75 percent of active compound(s), together with the excipient.
  • compositions of the invention may be administered to any patient which may experience the beneficial effects of the compounds of the invention.
  • mammals e.g., humans, although the invention is not intended to be so limited.
  • Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).
  • the compounds and pharmaceutical compositions thereof may be administered by any means that achieve their intended purpose.
  • administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes.
  • administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • compositions of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee- making, dissolving, or lyophilizing processes.
  • pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone.
  • fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose,
  • disintegrating agents may be added such as the above- mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate.
  • Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate, are used.
  • Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
  • Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are in one embodiment dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin.
  • stabilizers may be added.
  • Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts and alkaline solutions.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxy methyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers.
  • the topical compositions of this invention are formulated in one embodiment as oils, creams, lotions, ointments and the like by choice of appropriate carriers.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ).
  • the carriers may be those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired.
  • transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762; each herein
  • Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool.
  • a vegetable oil such as almond oil
  • a typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight.
  • Lotions may be conveniently prepared by dissolving the active ingredient, in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.
  • compositions, and methods of the present invention are compositions, and methods of the present invention.
  • Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.
  • HSA21 Human chromosome 21 trisomy results in Down syndrome (DS) (OMIM 190685) and is one of the most common human chromosomal disorders, occurring in ⁇ 1 in 700 live births. While the trisomy usually affects every tissue, reduced cognitive ability is among the most limiting features (see, Lott IT, et al., Lancet neurology 2010;9:623-633; Megarbane A, et al., Genet Med 2009; 1 1 :61 1 -616). Therapies that address cognitive restrictions of DS could have a significant impact for individuals living with this disorder. To this end, normalizing expression levels or the function of critical genes on chromosome 21 could prevent or reverse the deleterious effects of gene overdose.
  • Ts65Dn Three main murine models for DS have been developed (Ts65Dn (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184), TslCje (see, Sago H, et al, PNAS 1998;95:6256-6261), and TslRhr (see, Belichenko NP, et al, J Neurosci 2009;29:5938-5948)) that exhibit partial trisomy of chromosome 16, the murine ortholog to human chromosome 21.
  • the trisomic region of MMU16 contains the gene for dual-specificity tyrosine-(Y)- phosphorylation regulated kinase 1A (DYRK1A), a member of the CMGC family of kinases.
  • DRRK1A dual-specificity tyrosine-(Y)- phosphorylation regulated kinase 1A
  • mice show characteristic symptoms of DS including learning and behavioral deficits (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184; Sago H, et al, PNAS 1998;95:6256-6261 ; Belichenko NP, et al, J Neurosci 2009;29:5938-5948) and alterations in their dendritic spines within the hippocampus and cortical regions of the brain (see, Belichenko NP, et al, J Neurosci 2009;29:5938-5948; Belichenko PV, et al, J Comp Neurol 2004;480:281-298;
  • mice have also been prepared using a yeast artificial chromosome YAC 152F7 bearing extra copies of five different genes found in the DS critical region (DSCR) of human chromosome 21 including DYRKIA, PIGP, TTC3, DSCR9, and DSCR3. These mice demonstrate that
  • mice transgenic for the yeast artificial chromosome YAC 141G6 which contains all genes encompassed in YAC 152F7 except DYRKIA do not exhibit noticeable cognitive impairment.
  • mouse models that are transgenic for the human B AC gene (DYRKIA BAC Tg) or a murine BAC clone (TgDYRKlA) have been generated that specifically overexpress either human or murine DYRKIA, respectively.
  • mice similarly exhibit DS phenotypes including hippocampal-dependent spatial learning and motor deficits and developmental delays, which is highly suggestive of a central function for DYRKIA in mental deficits associated with DS (see, Smith DJ, et al., Nat Genet 1997;16:28-36; Ahn KJ, et al.,Neurobiol Dis 2006;22:463-472; Altafaj X, et al, Hum Mol Genet 2001 ;10: 1915-1923).
  • Harmine 3 is a ⁇ -carboline alkaloid with a reported IC50 of 80 nM against DYRKIA (see, Bain J, et al, Biochem J 2003;371 : 199-204; Bain J, et al, Biochem J 2007;408:297-315).
  • DYRKIA ⁇ -carboline analogues, including harmine, have significant drawbacks to consider when exploring their potential therapeutic applications.
  • MPTP is converted to MPP+ in the brain, affecting the extrapyramidal dopaminergic system leading to permanent Parkinson' s-like symptoms (see, Albores R, et al, PNAS 1990;87:9368-9372). Due to their convulsive properties observed in vivo, it has been suggested that harmine and its analogues are likely susceptible to similar metabolic pathways. Harmine 3 also exhibits potent inhibition of monoamine oxidase-A (MAO-A) reuptake, leading to behavioral side effects (Ki 5 nM, IC50 2 nM) (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184).
  • MAO-A monoamine oxidase-A
  • H-bond donor count (CSF2, Box 2, Figure 1) (see, Hitchcock SA, J. Med. Chem, 2006, 49(26), 7559- 83).
  • DYRK1B is over-expressed in certain cancer cells including colon, ovarian and pancreatic and could be an attractive target for cancer therapy (see, Deng X, et al., Genes & Cancer (2014), 5(9-10), 337-347; Berger F, et al, Ger. Offen. (2014), DE 102014009011 Al 20141218; Anderson K, et al, Bioorg. Med. Chem. Lett., 2013, 23(24), 6610-6615).
  • DYRKIA kinase assay utilizes an EZ Reader Electrophoresis Mobility Chip Instrument (Caliper Life Science).
  • 8.7 nM recombinant DYRKIA enzyme is pre-incubated with the small molecule inhibitor, or control buffer, for 5-10 min. To date, these classes of inhibitors are directly competitive with ATP.
  • a substrate mix containing 100 ⁇ ATP, 20 mM MgCl2, and 3 ⁇ fluorescently labeled substrate peptide (Caliper LS, FL-peptide 24, KKISGRLSPIM) is added to give a final concentration of 0.8 nM enzyme, 45 ⁇ ATP, 9.1 mM MgCl2, and 1.4 ⁇ substrate peptide within each reaction well.
  • the phosphorylation level of the substrate peptide may then be determined.
  • the assay is performed in a 384-well-plate format in which 112 compounds can be evaluated at a single concentration in triplicate, or 14 compounds can be evaluated at 12 concentrations in duplicate for IC50 determination.
  • Biol, 2005, 12(7), 811-823) are kinases that also catalyze tau phosphorylation in the brain,
  • CK15 mediates various processes in the brain including possible involvement in the glutamate deficiency associated with several neurodegenerative diseases (see, Perez DI, et al, Med. Res. Rev., 2011, 31(6), 924-954).
  • Clk-1 although not part of the existing panel, will be added as in recent years many DYRK1A inhibitors have been shown to exhibit equipotent affinity for this kinase (see, Tahtouh, T., et al, J Med Chem 2012, 55, 9312-9330; Coombs, T.
  • KinomeScanTM may be found at http://www.discoverx.com/services/drug-discovery- development-services/kinase-profiling/kinomescan).
  • the 110 kinases comprised, the 96 diversity kinase set from DiscoveRx ⁇ , with an additional 14 kinases hand-picked for their involvement in CNS diseases. Note that members of the internal selectivity panel are included in the larger KinomeScanTM set.
  • Standard MTT assays are used to measure cytotoxicity (loss of viable cells) of potential DYRKIA inhibitors.
  • the MTT assay is a rapid, high-throughput assay for assessing cell metabolic activity.
  • NAD(P)H- dependent cellular oxidoreductase enzymes reflect the number of viable cells present. These enzymes are capable of reducing the tetrazolium dye MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to its insoluble formazan, which has a purple color. Cell viability is, therefore, directly proportional to the production of the purple formazan reaction product (CSF8a, Figure 1).
  • the assay involves the use of recombinant DYRKIA protein and recombinant tau protein in a standard in vitro phosphorylation assay to assess the direct phosphorylation of tau by DYRKIA (see, Frost D, et al, PLoS One 2011;6:el9264).
  • Cellular Activity Assay (CSF7, Box 10): Methodological details of the assay is previously described (see, Frost D, et al, PLoS One 2011 ;6:el9264).
  • Harmine and EGCg improve cognitive performance in wild-type mice and Ts65Dn mice and their mode of action is postulated to be through DYRKIA inhibition (see, De la Torre R, et al, Mol Nutr Food Res 2014;58:278-288). Hence, it was proposed that inhibition of DYRKIA will improve cognitive performance in the Ts65Dn mice which experiments will directly evaluate using a battery of behavioral tests.
  • DYRKIA kinase phosphorylates several cellular substrates in vivo (see, Smith B, et al, ACS Chem Neurosci 2012;3:857-872), including the microtubule associated protein tau (see, Ryoo SR, et al, J Biol Chem 2007;282:34850-34857) and the amyloid precursor protein (see, Ryoo SR, et al, J Neurochem 2008;104: 1333-1344).
  • the combined effect of these phosphorylation events are to increase forms of hyperphosphorylated tau protein that are associated with neurodegeneration and to increase the production of neurotoxic amyloid beta peptides.
  • This example provides information for specific benzimidazole compounds of the present invention: l-(4-methoxyphenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazole, 1.
  • reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 10 mL), washed with NaHC0 3 (1 x 30 mL), H 2 0 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC ⁇ ) and concentrated under reduced pressure.
  • the product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%).
  • reaction was filtered through a celite pad, diluted with DCMTPA (3: 1 , 10 mL), washed with NaHC0 3 (1 x 30 mL), H 2 0 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC ⁇ ) and concentrated under reduced pressure.
  • the product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%).
  • reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 10 mL), washed with NaHC0 3 (1 x 30 mL), H 2 0 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC ⁇ ) and concentrated under reduced pressure.
  • the product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%).
  • Methyl 4-(6-bromo-lH-benzo[d]imidazol-l-yl) benzoate and reagents specified above were placed in a microwave vial, followed by addition of the designated solvent.
  • the capped vial was purged with argon gas for 5 minutes.
  • the reaction was then heated via microwave irradiation for 30 minutes at 130°C.
  • the crude material was filtered through celite, and the celite flushed with methanol. The remaining solvent was evaporated in vacuo via azeotropic removal with toluene.
  • 6-bromo-l-(3,4-dimethoxyphenyl)-lH-benzo[d]imidazole (0.300 mmol, 100 mg)
  • pyridine-4- boronic acid 1.0 eq., 0.300 mmol, 38.8 mg
  • Na2C03 4.0 eq., 1.20 mmol, 127 mg
  • a mixture of 4: 1 DMF/water (3 mL) were added to a microwave vial.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.0 ⁇ , 2.9 mg) and tricyclohexylphosphine (0.03 eq., 9.0 ⁇ , 2.6 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the crude material was purified via flash chromatography eluting with a
  • 6-bromo-l-(3,4-ethylenedioxyphenyl)-lH-benzo[d]imidazole (0.151 mmol, 50.0 mg)
  • pyridine- 4-boronic acid 1.0 eq., 0.151 mmol, 19.6 mg
  • Na2C03 4.0 eq., 0.604 mmol, 64.0 mg
  • a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.6 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • 6-bromo-l-(3,4-methylenedioxyphenyl)-lH-benzo[d] imidazole 0.315 mmol, 100 mg
  • 2-fluro- pyridine-4-boronic acid 1.0 eq., 0.315 mmol, 45.8 mg
  • Na2C03 4.0 eq., 1.26 mmol, 134 mg
  • a mixture of 4: 1 DMF/water 3.5 mL
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 ⁇ , 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 ⁇ , 2.8 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • 6-bromo-l-(3,4-methylenedioxyphenyl)-lH-benzo[d] imidazole 0.315 mmol, 100 mg
  • 2-fluro- pyridine-4-boronic acid 1.0 eq., 0.315 mmol, 43.2 mg
  • Na2C03 4.0 eq., 1.26 mmol, 134 mg
  • a mixture of 4: 1 DMF/water 3.5 mL
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 ⁇ , 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 ⁇ , 2.8 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 ⁇ , 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 ⁇ , 2.8 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • 6-bromoimidazo[l,2-a]pyridine (7.61 mmol, 1.5 g), pyridine-4-boronic acid (1.0 eq., 7.61 mmol, 985 mg), Na 2 C0 3 (4.0 eq., 30.5mmol, 3.23 g), and a mixture of 4: 1 DMF/water (80 mL) were added to a microwave vial.
  • the vial was purged with Ar(g) then
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd 2 (dba)3, 0.01 eq., 2.4 ⁇ , 2.3 mg) and tricyclohexylphosphine (0.03 eq., 7.1 ⁇ , 2.1 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with
  • 6-bromoimidazo[l,2-a]pyridine (4.06 mmol, 800 mg)
  • lH-pyrazol-4-yl boronic acid pinacol ester (1.0 eq., 4.06 mmol, 829mg)
  • Na 2 C0 3 (4.0 eq., 16.2 mmol, 1.72 g)
  • a mixture of 4: 1 DMF/water (40 mL) were added to a microwave vial.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 0.041 mmol, 38.3 mg) and tricyclohexylphosphine (0.03 eq., 0.122 mmol, 35.2 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130°C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd 2 (dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ 1, 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130°C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 ° C for 30 min. then filtered through celite washing down with CH 2 CI 2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.
  • the vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 ⁇ , 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 ⁇ , 1.4 mg) was added.
  • the reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene.

Abstract

This invention is in the field of medicinal chemistry. In particular, the invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRK1A protein, and their use as therapeutics for the treatment of Alzheimer's disease, Down syndrome, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), and other diseases.

Description

SMALL MOLECULE INHIBITORS OF DYRK1A AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to and the benefit of U. S. Provisional Application No. 62/213,904, filed September 3, 2015, which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
This invention is in the field of medicinal chemistry. In particular, the invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRK1A protein, and their use as therapeutics for the treatment of Alzheimer's disease, Down syndrome, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), and other diseases.
INTRODUCTION
With 24.3 million people affected in 2005 and an estimated rise to 42.3 million in 2020, dementia is currently a leading unmet medical need and costly burden on public health. Seventy percent of these cases have been attributed to Alzheimer's disease (AD), a neurodegenerative pathology whose most evident symptom is a progressive decline in cognitive functions.
The underlying treatment of learning and/or memory disorders is a huge and significantly unmet medical need and also included learning and memory repair after, for example, incidents of stroke or significant brain damage. As such, an improved understanding of the dementia (and other neuropathology) and related improved treatment methods are needed.
SUMMARY OF THE INVENTION
In addition to the overwhelmingly prominent /^-amyloid hypothesis being evaluated in a multitude of clinical trials through small molecule modulation of y- and ?-secretases and numerous immune-based approaches, aberrant phosphorylation of the tau protein is believed to significantly contribute to the development of AD and thus affords an alternate approach for therapeutic development. Tau is a cytoplasmic protein involved in the stabilization of microtubules under normal conditions. In AD, neuronal tau has been found to be excessively phosphorylated, with subsequent generation of aggregates of phosphorylated tau protein, known as "neurofibrillary tangles" (NFTs). NFTs and amyloid plaques are considered the most common hallmarks of AD and are correlated with neurofibrillary degeneration, neuronal death, and dementia.
Interestingly, several protein kinases have been implicated in neuronal development and, in particular, their overexpression and aberrant activation have been shown to play a significant role in the development of AD via tau phosphorylation. Dual specificity tyrosine
phosphorylation regulated kinase-1 A (DYRKIA) is important in neuronal development and plays a variety of functional roles within the adult central nervous system. The DYRKIA gene is located within the Down syndrome critical region (DSCR) on human chromosome 21 and current research suggests that overexpression of DYRKIA may be a significant factor leading to cognitive deficits in people with Alzheimer's disease (AD) and Down syndrome (DS).
Currently, treatment options for cognitive deficiencies associated with Down syndrome, as well as Alzheimer's disease, are extremely limited and represent a major unmet therapeutic need. Small molecule inhibition of DYRKIA activity in the brain may provide an avenue for pharmaceutical intervention of mental impairment associated with AD and other
neurodegenerative diseases.
Increased expression of the DYRKIA gene has been implicated in both the cognitive deficits of Down syndrome (DS) and the early onset of tau and amyloid neuropathologies that are associated with this genetic disorder. DYRKIA levels are increased in transgenic mouse models of DS and develop DS-like phenotypes including hippocampal-dependent spatial learning and memory deficits and developmental delays. Together these data strongly support a central function for DYRKIA in cognitive deficits associated with DS. Moreover, inhibition of excess DYRKIA activity has been shown to improve these DYRKlA-mediated cognitive deficits after administration of the natural products epigallocatechin-3-gallate (EGCg) and harmine, the standards for DYRKIA inhibition at the on-set of this translational campaign. However, these probes are not significantly selective and have numerous off-target effects that reduce their practical long-term use. To circumvent many of the detrimental issues observed, in particular with harmine, knowledge-based design efforts herein have unearthed novel small molecule series of structurally unique benzimidazole and imidazopyridine DYRKIA inhibitors, amenable for use as probes to test the benefits of selective DYRKIA inhibition in mouse models of DS/AD and a variety of other disease states including Parkinson's disease, Pick's disease, Huntington's and additional tauopathies.
Experiments conducted during the course of developing embodiments for the present invention designed, synthesized and biologically evaluated benzimidazole and imidazopyridine compounds as inhibitors of the dual specificity tyrosine phosphorylation regulated kinase- 1 A (DYRKIA) and their potential for use as therapeutics against AD and other disorders related to DYRK-IA activity (e.g., DS, other neuropathology, cancer (e.g., glioblastoma), cognitive enhancement). Many benzimidazole and imidazopyridine
compounds were also shown to exhibit activity against DYRKIB and exhibit some degree of activity against other kinases implicated in a variety of disease states.
The DYRKIA inhibitors described herein can also be considered as potential
therapeutics for the treatment of developmental diseases such as Down syndrome, and neurodegenerative diseases such as Parkinson's disease, and Huntington's disease. Moreover, the DYRKIA inhibitors of the present invention have been also implicated as potential therapeutics for the treatment of glioblastomas and further potential utility is highlighted in the oncology arena (see, e.g., Ionescu et al, Mini-reviews in Medicinal Chemistry, 2012, 12, 1315-1329).
These novel DYRKIA inhibitors may also have utility as general cognitive enhancers, given the published findings that DYRKIA can phosphorylate sirtuin 1 , a key regulator of learning and memory (see, e.g., Michan et al, J. Neurosci. 2010, 30(29), 9695-9707; Guo et al, J Biol. Chem. 2010, 285 (17), 13223-13232). The potential utility of these DYRKIA compound series is further reinforced by findings that harmine, a potent, but relatively less selective DYRKIA inhibitor, enhances memory performance in wild-type rodents (Mennenga et al, Physiol. Behav. 2015, 138, 260-265).
These novel DYRKIA inhibitors may also have further utility as results identify
DYRKIA as a physiologically relevant regulator of Treg cell differentiation and suggest a broader role for other DYRK family members in immune homeostasis. As such, new roles may be found in autoimmune diseases such as inflammatory bowel disease and type 1 diabetes (see, e.g., Khor B, et al, eLife 2015;4: e05920).
Accordingly, this invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRKIA protein, and their use as therapeutics for the treatment of disorders related to DYRKIA activity (e.g., AD, DS, neuropathology, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation)). a particular embodiment, benzimidazole and imidazo ridine compounds
encompassed within Formula I are provided:
Figure imgf000005_0001
, including
pharmaceutically acceptable salts, solvates, and/or prodrugs thereof.
Formula I is not limited to a particular chemical moiety for Rl, R2, R3, R4, X, Y and Z. In some embodiments, the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit DYRKIA activity. In some embodiments, the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit one or more of: DYRKIA related PI3K/Akt signaling; DYRKIA related tau
phosphorylation; DYRKIA related NFAT phosphorylation; DYRKIA related ASK1/JNK1 pathway activation; DYRKIA related p53 phosphorylation; DYRKIA related Amph 1 phosphorylation; DYRKIA related Dynamin 1 phosphorylation; DYRKIA related Synaptojanin phosphorylation; DYRKIA related presenilin 1 (the catalytic sub-unit of γ-secretase) activity; DYRKIA related amyloid precursor protein phosphorylation; and DYRKIA related SIRT1 activation.
In some embodiments, X-Y-Z is
Figure imgf000005_0002
, thereby rendering the resulting compound a benzimidazole compound having the following formula:
Figure imgf000005_0003
In some embodiments, X-Y-Z is , thereb rendering the compound
imidazopyridine compound having the following formula:
Figure imgf000005_0004
In some embodiments, Rl is selected from hydrogen, aryl and substituted aryl,
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
In some embodiments, R5 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
Figure imgf000009_0002
Figure imgf000010_0001
In some embodiments, R6 is selected from hydrogen, C1 -C4 alkyl, heterocyclyl alkyl, heteroaryl alkyl, aryl alkyl, aryl, heterocyclyl, and heteroaryl.
In some embodiments, R7 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
Figure imgf000010_0002
In some embodiments, each of R2 and R3 is independently selected from hydrogen, aryl,
substituted aryl, heteroaryl, substituted heteroaryl, ;
Figure imgf000010_0003
Figure imgf000011_0001
Figure imgf000012_0001
In some embodiments, the compound is one or more of the compounds shown in Examples II and/or III.
The invention further provides processes for preparing any of the compounds of the present invention.
The invention also provides the use of compounds to not only inhibit DYRKIA activity but also signaling pathways dependent upon DYRKIA phosphorylation (e.g., Tau, POK/AKt, APP, PSI, ASF, RCAN-1 , NFAT, p53, ASK1/JNK1, SIRT1 , GluN2A and other NMD A receptors). The invention also relates to the use of compounds for sensitizing cells to additional agent(s), such as agents known to be effective in the treatment of neurodegenerative disorders. The compounds of the invention are useful for the treatment, amelioration, or prevention of disorders associated with DYRKIA activity (e.g., AD, DS, Parkinson's disease, Huntington's disease, glioblastoma), such as those responsive to DYRKIA activity inhibition. In certain embodiments, the compounds can be used to treat, ameliorate, or prevent cancer that is associated with DYRKIA activity (e.g., glioblastoma). In certain embodiments, the compounds can be used to treat, ameliorate, or prevent autoimmune diseases. In certain embodiments, the compounds can be used to treat, ameliorate, or prevent inflammatory disorders (e.g., airway inflammation).
The invention also provides pharmaceutical compositions comprising the compounds of the invention in a pharmaceutically acceptable carrier.
The invention also provides kits comprising a compound of the invention and instructions for administering the compound to an animal. The kits may optionally contain other therapeutic agents, e.g., agents useful in treating neurodegenerative disorders and/or anticancer agents.
In a articular embodiment, compounds encompassed within the following formula are
provided:
Figure imgf000013_0001
, including pharmaceutically acceptable salts, solvates, and/or prodrugs thereof. In such embodiments, each of the "*" substituents is independently Carbon or Nitrogen, and X, Y, Z, Rl, R2, R3, and R4 are as described above.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 presents a flow chart describing a screening paradigm for DYRKIA inhibitor progression.
DETAILED DESCRIPTION OF THE INVENTION
DYRKIA is a member of the DYRK family containing 5 kinases (DYRKIA, DYRK1B, DYRK2, DYRK3 and DYRK4). DYRKs belong to the CMGC group of proline-directed kinases, which also includes cyclin-dependent kinases (CDKs), mitogen-activated protein kinases (MAPKs), glycogen synthase kinases (GSKs) and CDC2-like kinases (CLKs). While the signaling pathways of CDK and MAPK families have been extensively studied, much less is known on how DYRKs and CLKs are linked to other proteins and various physiological or pathological processes. The DYRKIA gene is located on chromosome 21 (21q22.2), a region known as the Down-Syndrome Critical Region (DSCR) (see, e.g., Hammerle et al., 2011 Development 138, 2543-2554). The under- or over-expression of the Dyrkla gene in mammals or of its orthologous gene minibrain (mnb) in Drosophila causes severe retardation of central nervous system development and maturation. At the molecular level, DYRKIA phosphorylates the nuclear factor of activated T cells (NFAT), counteracting the effect of calcium signaling and maintaining inactive NFAT (see, e.g., Arron et al, 2006 Nature 411, 595-600). DYRKIA has been identified as a negative regulator of the cell cycle that promotes the switch to a quiescent state or differentiation (see, e.g., Chen et al., 2013 Mol. Cell 52, 87-100). In malignant cells, DYRKIA promotes survival via inhibition of pro-apoptotic proteins (see, e.g., Guo et al., 2010 J. Bio. Chem. 285, 13223-13232; Seifert et al, 2008 FEBS J. 275, 6268-6280).
Currently, treatment options for cognitive deficiencies associated with AD and DS are extremely limited and represent a major, extremely significant unmet therapeutic need. The DYRKIA inhibitors of the present invention provide a new avenue for pharmaceutical intervention of mental impairment associated with AD and other neurodegenerative diseases, and address a critical unmet medical need and significantly changing treatment paradigm for AD.
Experiments conducted during the course of developing embodiments for the present invention designed, synthesized and biologically evaluated benzimidazole and imidazopyridine compounds as inhibitors of the dual specificity tyrosine phosphorylation regulated kinase- 1 A (DYRKIA) and their potential for use as therapeutics against AD and other disorders related to DYRK-1A activity (e.g., DS, other neuropathology, cancer (e.g., glioblastoma)). Many benzimidazole and imidazopyridine compounds were also shown to exhibit activity against DYRK1B and exhibit some degree of activity against other kinases implicated in a variety of disease states.
Moreover, the DYRKIA inhibitors of the present invention can be used for treating other cellular pathways involved in mental impairment and neurodegenerative dementia. Specifically, the DYRKIA inhibitors of the present invention can be used for inhibiting DYRKIA activated PI3K/Akt signaling, a pathway largely involved in neuronal development, growth, and survival. The DYRKIA inhibitors of the present invention DYRKIA can be used for inhibiting DYRKIA stimulated ASK1/JNK1 activity, thereby inducing neuronal death and apoptosis. In addition, the DYRKIA inhibitors of the present invention DYRKIA can be used to inhibit DYRKIA phosphorylation of p53 during embryonic brain development, thereby preventing neuronal proliferation alteration. The DYRKIA inhibitors of the present invention can be used to inhibit DYRKIA phosphorylation of synaptic proteins Amph 1, Dynamin 1, and Synaptojanin, involved in the regulation of endocytosis, thereby retaining synaptic plasticity through preventing alteration of the number, size, and morphology of dendritic spines. The DYRKIA inhibitors of the present invention can be used to inhibit presenilin 1 (the catalytic sub-unit of γ-secretase).
As such, the present invention addresses the need for effective therapies for AD and DS by providing potent and selective DYRKIA inhibitors able to permeate the blood-brain barrier (BBB) and elicit on-mechanism therapeutic responses in AD animal models.
Accordingly, this invention relates to a new class of small-molecules having a benzimidazole or imidazopyridine structure which function as inhibitors of DYRKIA protein, and their use as therapeutics for the treatment of Alzheimer's disease, Down syndrome, glioblastoma, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), and other diseases.
In a particular embodiment, benzimidazole and imidazopyridine compounds
encompassed within Formula I are provided:
Figure imgf000015_0001
, including
pharmaceutically acceptable salts, solvates, and/or prodrugs thereof.
Formula I is not limited to a particular chemical moiety for Rl , R2, R3, R4, X, Y and Z. In some embodiments, the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit DYRKIA activity. In some embodiments, the particular chemical moiety for Rl, R2, R3, R4, X, Y and Z independently include any chemical moiety that permits the resulting compound to inhibit one or more of: DYRKIA related PI3K/Akt signaling; DYRKIA related tau
phosphorylation; DYRKIA related NFAT phosphorylation; DYRKIA related ASK1/JNK1 pathway activation; DYRKIA related p53 phosphorylation; DYRKIA related Amph 1 phosphorylation; DYRKIA related Dynamin 1 phosphorylation; DYRKIA related Synaptojanin phosphorylation; DYRKIA related presenilin 1 (the catalytic sub-unit of γ-secretase) activity; DYRKIA related amyloid precursor protein phosphorylation; and DYRKIA related SIRT1 activation. In some embodiments, X-Y-Z is
Figure imgf000016_0001
, thereby rendering the resulting
compound a benzimidazole compound havin the following formula:
Figure imgf000016_0002
In some embodiments, X-Y-Z is
Figure imgf000016_0003
, hereby rendering the compound an
imidazopyridine compound having the following formula:
Figure imgf000016_0004
In some embodiments, Rl is selected from hydrogen, aryl and substituted aryl,
Figure imgf000016_0005
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001

Figure imgf000020_0001
In some embodiments, R5 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
Figure imgf000020_0002
In some embodiments, R6 is selected from hydrogen, C1-C4 alkyl, heterocyclyl alkyl, heteroaryl alkyl, aryl alkyl, aryl, heterocyclyl, and heteroaryl. In some embodiments, R7 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic moiety comprising
Figure imgf000021_0001
In some embodiments, each of R2 and R3 is independentl from hydrogen, aryl,
Figure imgf000021_0002
Figure imgf000022_0001
21 In some embodiments, R4 is selected from hydrogen, NH2,
Figure imgf000023_0001
Figure imgf000023_0002
In some embodiments, the compound is one or more of the compounds shown in
Examples II and/or III.
The invention further provides processes for preparing any of the compounds of the present invention.
In some embodiments, the compositions and methods of the present invention are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in an animal (e.g. , a mammalian patient including, but not limited to, humans and veterinary animals). In this regard, various diseases and pathologies are amenable to treatment or prophylaxis using the present methods and compositions. A non-limiting exemplary list of these diseases and conditions includes, but is not limited to, Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), any neurodegenerative disorder related to DYRK1 A activity, and any type of cancer related to DYRK1A activity.
Some embodiments of the present invention provide methods for administering an effective amount of a compound of the invention and at least one additional therapeutic agent (including, but not limited to, any agent useful in treating Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune diseases, inflammatory disorders (e.g., airway inflammation), any neurodegenerative disorder related to DYRKl A activity, and any type of cancer characterized related to DYRKl A activity).
Compositions within the scope of this invention include all compositions wherein the compounds of the present invention are contained in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typically, the compounds may be administered to mammals, e.g. humans, orally at a dose of 0.0025 to 50 mg/kg, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated for disorders responsive to induction of apoptosis. In one embodiment, about 0.01 to about 25 mg/kg is orally administered to treat, ameliorate, or prevent such disorders. For intramuscular injection, the dose is generally about one-half of the oral dose. For example, a suitable intramuscular dose would be about 0.0025 to about 25 mg/kg, or from about
0.01 to about 5 mg/kg.
The unit oral dose may comprise from about 0.01 to about 1000 mg, for example, about 0.1 to about 100 mg of the compound. The unit dose may be administered one or more times daily as one or more tablets or capsules each containing from about 0.1 to about 10 mg, conveniently about 0.25 to 50 mg of the compound or its solvates.
In a topical formulation, the compound may be present at a concentration of about 0.01 to
100 mg per gram of carrier. In a one embodiment, the compound is present at a concentration of about 0.07-1.0 mg/ml, for example, about 0.1-0.5 mg/ml, and in one embodiment, about 0.4 mg/ml.
In addition to administering the compound as a raw chemical, the compounds of the invention may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compounds into preparations which can be used pharmaceutically. The preparations, particularly those preparations which can be administered orally or topically and which can be used for one type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by intravenous infusion, injection, topically or orally, contain from about 0.01 to 99 percent, in one embodiment from about 0.25 to 75 percent of active compound(s), together with the excipient. The pharmaceutical compositions of the invention may be administered to any patient which may experience the beneficial effects of the compounds of the invention. Foremost among such patients are mammals, e.g., humans, although the invention is not intended to be so limited. Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).
The compounds and pharmaceutical compositions thereof may be administered by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
The pharmaceutical preparations of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee- making, dissolving, or lyophilizing processes. Thus, pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above- mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate, are used. Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are in one embodiment dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added.
Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxy methyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.
The topical compositions of this invention are formulated in one embodiment as oils, creams, lotions, ointments and the like by choice of appropriate carriers. Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C12). The carriers may be those in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762; each herein
incorporated by reference in its entirety. Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool. A typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight. Lotions may be conveniently prepared by dissolving the active ingredient, in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.
One of ordinary skill in the art will readily recognize that the foregoing represents merely a detailed description of certain preferred embodiments of the present invention. Various modifications and alterations of the compositions and methods described above can readily be achieved using expertise available in the art and are within the scope of the invention.
EXAMPLES
The following examples are illustrative, but not limiting, of the compounds,
compositions, and methods of the present invention. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.
Example I.
Human chromosome 21 trisomy (HSA21) results in Down syndrome (DS) (OMIM 190685) and is one of the most common human chromosomal disorders, occurring in ~1 in 700 live births. While the trisomy usually affects every tissue, reduced cognitive ability is among the most limiting features (see, Lott IT, et al., Lancet neurology 2010;9:623-633; Megarbane A, et al., Genet Med 2009; 1 1 :61 1 -616). Therapies that address cognitive restrictions of DS could have a significant impact for individuals living with this disorder. To this end, normalizing expression levels or the function of critical genes on chromosome 21 could prevent or reverse the deleterious effects of gene overdose.
Three main murine models for DS have been developed (Ts65Dn (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184), TslCje (see, Sago H, et al, PNAS 1998;95:6256-6261), and TslRhr (see, Belichenko NP, et al, J Neurosci 2009;29:5938-5948)) that exhibit partial trisomy of chromosome 16, the murine ortholog to human chromosome 21. In all three models, the trisomic region of MMU16 contains the gene for dual-specificity tyrosine-(Y)- phosphorylation regulated kinase 1A (DYRK1A), a member of the CMGC family of kinases. These mice show characteristic symptoms of DS including learning and behavioral deficits (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184; Sago H, et al, PNAS 1998;95:6256-6261 ; Belichenko NP, et al, J Neurosci 2009;29:5938-5948) and alterations in their dendritic spines within the hippocampus and cortical regions of the brain (see, Belichenko NP, et al, J Neurosci 2009;29:5938-5948; Belichenko PV, et al, J Comp Neurol 2004;480:281-298;
Belichenko PV, et al, J Comp Neurol 2007;504:329-345; Siarey RJ, et al., Neuropharmacology 2005;49: 122-128; Smith DJ, et al, Nat Genet 1997;16:28-36). Transgenic mice have also been prepared using a yeast artificial chromosome YAC 152F7 bearing extra copies of five different genes found in the DS critical region (DSCR) of human chromosome 21 including DYRKIA, PIGP, TTC3, DSCR9, and DSCR3. These mice demonstrate
significantly impaired learning ability and brain abnormalities (see, Smith DJ, et al, Nat
Genet 1997;16:28-36; Branchi I, et al, J Neuropathol Exp Neurol 2004;63:429-440; Chabert C, et al, Behav Genet 2004;34:559-569). In comparison, murine models transgenic for the yeast artificial chromosome YAC 141G6 which contains all genes encompassed in YAC 152F7 except DYRKIA do not exhibit noticeable cognitive impairment. In addition, mouse models that are transgenic for the human B AC gene (DYRKIA BAC Tg) or a murine BAC clone (TgDYRKlA) have been generated that specifically overexpress either human or murine DYRKIA, respectively. These mice similarly exhibit DS phenotypes including hippocampal-dependent spatial learning and motor deficits and developmental delays, which is highly suggestive of a central function for DYRKIA in mental deficits associated with DS (see, Smith DJ, et al., Nat Genet 1997;16:28-36; Ahn KJ, et al.,Neurobiol Dis 2006;22:463-472; Altafaj X, et al, Hum Mol Genet 2001 ;10: 1915-1923). Cognitive impairment and dendritic tree alteration in TgDyrklA recapitulates that of Ts65Dn mice (see, Ahn KJ, et al.,Neurobiol Dis 2006;22:463-472; Altafaj X, et al, Hum Mol Genet 2001;10: 1915- 1923). These combined observations indicate that DYRKIA overexpression is necessary and sufficient for cognitive abnormalities of DS and that normalization of DYRKIA activity is a promising therapeutic approach for this genetic disorder.
Harmine 3 is a β-carboline alkaloid with a reported IC50 of 80 nM against DYRKIA (see, Bain J, et al, Biochem J 2003;371 : 199-204; Bain J, et al, Biochem J 2007;408:297-315). Despite high affinity for DYRKIA, β-carboline analogues, including harmine, have significant drawbacks to consider when exploring their potential therapeutic applications. The hallucinogenic properties of harmine have been exploited historically (see, Callaway JC, et al, J Ethnopharmacol 1999;65:243-256; Gambelunghe C, et al, Biomed Chromatogr 2008;22: 1056- 1059) and more recently shown to be the result of its affinity for the serotonin and tryptamine receptor binding sites (see, Airaksinen MM, et al, Pharmacol Toxicol 1987;60:5-8). Animal studies conducted on the β-carbolines as early as the 1930s revealed a plethora of psychoactive effects including excitation, anxiety, tremors, convulsions, ataxia, pupil dilation, and alterations in the brain's electrical activity (electroencephalographic activity or EEG activity) (see, Fuentes JA, et al, Neuropharmacology 1971;10: 15-23; Kawanishi K, et al, Pharmacol Biochem Behav 1994;47:689-699). In addition, β-carbolines methylated at the pyridine nitrogen can mimic the activity of the powerful neurotoxic metabolite MPTP. MPTP is converted to MPP+ in the brain, affecting the extrapyramidal dopaminergic system leading to permanent Parkinson' s-like symptoms (see, Albores R, et al, PNAS 1990;87:9368-9372). Due to their convulsive properties observed in vivo, it has been suggested that harmine and its analogues are likely susceptible to similar metabolic pathways. Harmine 3 also exhibits potent inhibition of monoamine oxidase-A (MAO-A) reuptake, leading to behavioral side effects (Ki 5 nM, IC50 2 nM) (see, Reeves RH, et al, Nat Genet 1995;11 : 177-184). The deleterious off-target effects of both harmine and EGCg necessitate the development of potent and selective DYRK1 A inhibitors for advancing inhibition of this kinase as a therapeutic target for DS. Experiments conducted during the course of developing embodiments for the present invention generated these much needed DYRK1A inhibitors.
A goal of such experiments was to deliver two advanced leads from innovative, structurally distinct chemical platforms worthy of evaluation in a GLP/GMP modality through critical success factors (CSFs) 15 & 16, Boxes 16-18 (Figure 1). New molecules with the potential to improve the lives of patients with DS and other indications claimed herein have emerged. Figure 1 simply represents one flow chart which can be modified with alternate in vivo studies/models for the other indications claimed herein.
At all times, several guiding principles correlating BBB penetration with
physicochemical properties of CNS-active drugs were closely monitored. Key properties with preferred ranges for small-molecule BBB passive diffusion include molecular weight
(MW≤ 450); polar surface area ^PSA < 7θΑ2); log P or D (2-4), and H-bond donor count (HBD≤1) (CSF2, Box 2, Figure 1) (see, Hitchcock SA, J. Med. Chem, 2006, 49(26), 7559- 83).
At the beginning of this effort, three DYRK1 A inhibitor-bound crystal structures were available: harmine (PDB ID: 3ANR) 3, INDY (PDB ID: 3ANQ) 6, and D15 (PDB ID: 2W06) 7 (www.rcsb.org). Knowledge based approaches utilizing this information ultimately lead to the discovery described herein. Isoform selectivity with DYRK1B is a challenge due to high homology with 1A, although no evidence exists to suggest this will be detrimental to the therapeutic index of a DRYK1A inhibitor and preliminary data suggests selectivity is achievable. Moreover, DYRK1B is over-expressed in certain cancer cells including colon, ovarian and pancreatic and could be an attractive target for cancer therapy (see, Deng X, et al., Genes & Cancer (2014), 5(9-10), 337-347; Berger F, et al, Ger. Offen. (2014), DE 102014009011 Al 20141218; Anderson K, et al, Bioorg. Med. Chem. Lett., 2013, 23(24), 6610-6615). There exists 27 FDA approved small molecule kinase- inhibiting drugs, and encouragingly the 2012 approval of Tofacitinib, a JAK3 inhibitor for rheumatoid arthritis, adds extra impetus to the feasibility of successfully targeting kinases for non-oncology related indications (see, Cohen, P. Nat. Rev. Drug Discov., 2002, 1(4), 309-315; Roskoski, R. USFDA approved protein kinase inhibitors. Blue Ridge Institute for Medical Research. Horse Shoe, NC.
www. brimr . org/PKI/PKIs . htmL) .
To utilize a medium-throughput in vitro phosphorylation assay with recombinant DYRKIA to determine % inhibition and triage actives for IC50 determination. (CSF 3 & 4, Box 7 & 8, Figure 1). The DYRKIA kinase assay utilizes an EZ Reader Electrophoresis Mobility Chip Instrument (Caliper Life Science). In the assay, 8.7 nM recombinant DYRKIA enzyme (Invitrogen) is pre-incubated with the small molecule inhibitor, or control buffer, for 5-10 min. To date, these classes of inhibitors are directly competitive with ATP. After 5-10 minutes of pre-incubation, a substrate mix containing 100 μΜ ATP, 20 mM MgCl2, and 3 μΜ fluorescently labeled substrate peptide (Caliper LS, FL-peptide 24, KKISGRLSPIM) is added to give a final concentration of 0.8 nM enzyme, 45 μΜ ATP, 9.1 mM MgCl2, and 1.4 μΜ substrate peptide within each reaction well. The phosphorylation level of the substrate peptide may then be determined. The assay is performed in a 384-well-plate format in which 112 compounds can be evaluated at a single concentration in triplicate, or 14 compounds can be evaluated at 12 concentrations in duplicate for IC50 determination. To triage and evaluate compounds from Aim 2A in relevant selectivity panels, surrogate BBB penetration and cell viability assays and in silico and in vitro toxicology screens: (a) the established in-house selectivity panel (CSF5, Box 9). (b) PAMPA, Caco-2 studies and MTT assays (CSF8, 9 & 8a, Box 12). (c)
KinomeScan™ (CSF10, Box 13). (d) hERG activity (CSF9a).
(a) All active compounds passing CSF4 are subject to an initial kinase panel selectivity screen against DYRK1B, GSK3 , CDK5/p25 and CK15 respectively using the EZ Reader Electrophoresis Mobility Chip Instrument. Rationale for selection is: (i) DYRK1B shows the highest sequence homology (-95%) with DYRK1A, compared to only 43-45% homology to DYRK2/3/4. (ii) GSK3 (see, Hamann, M,.et al, J. Nat. Prod., 2007, 70(9), 1397-1405) and CDK5 (see, Ahn JS, et al, Chem. Biol, 2005, 12(7), 811-823) are kinases that also catalyze tau phosphorylation in the brain, (iii) CK15 mediates various processes in the brain including possible involvement in the glutamate deficiency associated with several neurodegenerative diseases (see, Perez DI, et al, Med. Res. Rev., 2011, 31(6), 924-954). (iv) Clk-1, although not part of the existing panel, will be added as in recent years many DYRK1A inhibitors have been shown to exhibit equipotent affinity for this kinase (see, Tahtouh, T., et al, J Med Chem 2012, 55, 9312-9330; Coombs, T. C, et al, Bioorg Med Chem Lett 2013, 23, 3654-3661). In addition to the pan-inhibitor staurosporine, known inhibitors SB216763 (GSK3 ) (see, Nemoto T, et al, Brain Research 2006, 1110(1), 1-12), Roscovitine (CDK5/P25) (see, Oumata N, et al, J. Med. Chem., 2008, 51(17), 5229-5242), PF4800567 (CK15) (see, Perez DI, et al, Med. Res. Rev., 2011, 31(6), 924-954), and ML315 (see, Tahtouh, T., et al., J Med Chem 2012, 55, 9312-9330; Coombs, T. C, et al, Bioorg Med Chem Lett 2013, 23, 3654-3661) are/will be employed as standard controls in these assays. Determining activity against these kinases is viewed as key to differentiate functional modes of action of inhibitors in cells.
(b) Compounds progressing to CSF10, Box 13, Figure 1, are subject to evaluation against a panel of 110 kinases via KinomeScan^^ technology (DiscoveRx^^), an active-site directed competition binding assay that measures thermodynamic interaction affinities. A selectivity score is determined as the # hits/# kinases tested (see, Details of
KinomeScan™ may be found at http://www.discoverx.com/services/drug-discovery- development-services/kinase-profiling/kinomescan). The 110 kinases comprised, the 96 diversity kinase set from DiscoveRx^^, with an additional 14 kinases hand-picked for their involvement in CNS diseases. Note that members of the internal selectivity panel are included in the larger KinomeScan™ set.
(c) CACO-2 determinations are performed by Absorption Systems™ using the protocol entitled "P-gp Interaction Assessment in Caco-2 cells" detailed at
http://www.absorption.com/p-gp-interaction-assessment- caco-2-cells/. PAMPA (Parallel Artificial Membrane Permeability Assay) determinations are performed by the CRO
Analiza^^ with assay details described at http://www.analiza.com/adme/pampa.html. An effective permeability above 2 x 10-6 cm/sec correlates with a human fraction absorbed (%FA) above 80% and is a common standard for good membrane permeability (CSF8, Figure 1).
(d) Standard MTT assays are used to measure cytotoxicity (loss of viable cells) of potential DYRKIA inhibitors. The MTT assay is a rapid, high-throughput assay for assessing cell metabolic activity. NAD(P)H- dependent cellular oxidoreductase enzymes reflect the number of viable cells present. These enzymes are capable of reducing the tetrazolium dye MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to its insoluble formazan, which has a purple color. Cell viability is, therefore, directly proportional to the production of the purple formazan reaction product (CSF8a, Figure 1).
(e) Computational assessments of hERG activity (in silico, QikProp-ADME property prediction software Schrodinger Inc.) and patch clamp electrophysiology assays (in vitro) are available on main campus for nominal fees. Note that hERG activity is merely a flag for potential drug induced long QT syndrome and not a "hard" CSF per say (CSF9a, Figure 1).
To triage and evaluate compounds derived from Aim 2B for inhibition of DYRK1A- catalyzed tau phosphorylation (CSF6 & 7, Box 10 & 11, Figure 1).
In Vitro Phosphorylation Assay (CSF6, Box 10): Promising compounds from Aim 2a will be sent for IC50 determination using a physiologically relevant substrate, full-length tau protein. Details of the in vitro phosphorylation assay have been published previously, where experiments have shown the assay to be highly reproducible, able to clearly discriminate affinities down to the low nM range (see, Frost D, et al, PLoS One
2011;6:el9264). The assay involves the use of recombinant DYRKIA protein and recombinant tau protein in a standard in vitro phosphorylation assay to assess the direct phosphorylation of tau by DYRKIA (see, Frost D, et al, PLoS One 2011;6:el9264).
Cellular Activity Assay (CSF7, Box 10): Methodological details of the assay is previously described (see, Frost D, et al, PLoS One 2011 ;6:el9264).
Harmine and EGCg improve cognitive performance in wild-type mice and Ts65Dn mice and their mode of action is postulated to be through DYRKIA inhibition (see, De la Torre R, et al, Mol Nutr Food Res 2014;58:278-288). Hence, it was proposed that inhibition of DYRKIA will improve cognitive performance in the Ts65Dn mice which experiments will directly evaluate using a battery of behavioral tests.
DYRKIA kinase phosphorylates several cellular substrates in vivo (see, Smith B, et al, ACS Chem Neurosci 2012;3:857-872), including the microtubule associated protein tau (see, Ryoo SR, et al, J Biol Chem 2007;282:34850-34857) and the amyloid precursor protein (see, Ryoo SR, et al, J Neurochem 2008;104: 1333-1344). The combined effect of these phosphorylation events are to increase forms of hyperphosphorylated tau protein that are associated with neurodegeneration and to increase the production of neurotoxic amyloid beta peptides. These DYRK1A regulated events are thought to contribute to the early onset Alzheimer's pathology in DS patients and are highly relevant molecular endpoints that will directly assess the extent to which inhibitors ameliorate these deleterious molecular phenotypes. Example II.
This example provides information for specific benzimidazole compounds of the present invention: l-(4-methoxyphenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazole, 1.
Figure imgf000033_0001
Chemical Formula: 019Η 15Ν3Ο
Molecu lar Weight: 301.3419 1
Brown solid. Mp: 135-137°C. 1H NMR (400 MHz, CDC13) δ 8.65 (d, J= 4.7 Hz, 2H), 8.12 (s 1H), 7.97 (d, J= 8.4 Hz, 1H), 7.68 (s, 1H), 7.62 (dd, J= 8.4, 1.4 Hz, 1H), 7.54 (d, J= 4.8 Hz, 2H), 7.46 (dd, J = 6.8, 5.5 Hz, 2H), 7.17 - 7.08 (m, 2H), 3.91 (d, J = 1.3 Hz, 3H). LC/MS [M l]+ = 302 l-(4-methoxyphenyl)-6-(pyrimidin-5-yl)-lH-benzo[d]imidazole, 2.
Figure imgf000033_0002
e 2 XH NMR (400 MHz, CDC13) δ 9.19 (s, IH), 8.97 (s, 2H), 8.14 (s, IH), 8.01 (d, J = 8.4 Hz, IH), 7.59 (d, J= 18.9 Hz, IH), 7.53 (dt, J = 15.9, 7.9 Hz, IH), 7.49 - 7.40 (m, 2H), 7.16 - 7.04 (m, 2H), 3.91 (s, 3H). 1 C NMR (100 MHz, CDCI3) δ 159.75, 157.25, 155.12, 143.95, 134.85, 130.01, 128.57, 125.96, 121.92, 121.55, 115.38, 109.05, 55.71. LC/MS [M + 1]+ = 303
6-(2-chloropyridin-3-yl)- l-(4-methoxyphenyl)- lH-benzo [d] imidazole, 3.
Figure imgf000034_0001
Yellow solid. Mp: 141-143°C. lH NMR (400 MHz, CDCI3) δ 8.46 - 8.35 (m, IH), 8.13 (s, IH), 7.98 - 7.89 (m, IH), 7.77 - 7.68 (m, IH), 7.54 (dd, J = 1.6, 0.6 Hz, IH), 7.47 - 7.41 (m, 2H), 7.41 - 7.38 (m, IH), 7.33 - 7.25 (m, IH), 7.11 - 7.05 (m, 2H), 3.89 (s, 3H). 1 C NMR (100 MHz, CDCI3) δ 159.52, 149.91, 148.27, 143.70, 143.56, 139.97, 137.29, 134.16, 133.00, 128.78, 125.77, 124.18, 122.46, 120.33, 115.26, 111.44, 55.66. LC/MS [M + l]+ = 336
5-(l-(4-methoxyphenyl)-lH-benzo[d]imidazol-6-yl)pyridin-2-amine, 4.
Figure imgf000034_0002
Yellow solid. Mp: 172-174°C. ¾ NMR (400 MHz, CDCI3) δ 8.32 (dd, J= 2.4, 0.7 Hz, IH), 8.06 (s, IH), 7.90 (dd, J= 8.4, 0.6 Hz, IH), 7.69 (dd, J= 8.5, 2.5 Hz, IH), 7.52 (dd, J= 1.7, 0.6 Hz, IH), 7.49 - 7.40 (m, 3H), 7.15 - 7.04 (m, 2H), 6.58 (dd, J = 8.5, 0.7 Hz, IH), 4.54 (s, 2H), 3.90 (s, 3H). 1 C NMR (100 MHz, CDCI3) δ 159.46, 157.37, 146.43, 143.05, 143.01, 137.01, 134.95, 134.35, 129.01, 127.90, 125.86, 121.75, 120.78, 115.22, 108.53, 107.90, 55.68. l-(4-methoxyphenyl)-6-phenyl- lH-benzo [d] imidazole, 5.
Figure imgf000035_0001
Solvent was degassed with N2. The product of the previous reaction was dissolved in DMF:H20 (4: 1,10 mL). To the resulting solution, boronic acid (1 eq.), Na2CC>3 (4 eq.) and Pd(dppf)Cl2 (0.05 eq.) were added and the reaction was heated at 90 °C for 16 h. After cooling down to room temperature, the reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 100 mL), washed with H20 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC^) and concentrated under reduced pressure. The product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%) to afford 5 1-506, 113 mg, 38%; white solid; LC MS [M + 1]+ m/z 302.00; XH NMR (400 MHz, DMSO^) δ: 8.49 (s, 1H, N=CH-N), 7.48 (d, 1Η, J= 8.1, ArH), 7.71-7.76 (m, 4H, ArH), 7.58 (dd, 1H, J= 8.7, 1.5, ArH), 7.48-7.40 (m, 2H, ArH), 7.37-7.30 (m, 1H, ArH), 7.21-7.12 (m, 2H, ArH), 3.85 (s, 3H, OCH3); 1 C NMR (100 MHz, DMSC ) δ:
158.7, 144.1, 143.1, 140.7, 136.0, 134.2, 128.9, 128.7, 127.1, 125.7, 121.7, 120.1, 115.2, 108.5, 55.5.
S nthetic Scheme to 5 & 7. HCOH (neat) reflux, 1 h
Figure imgf000035_0002
1-497, 1.14 gr R = p-H, 1-497 R, = p-H, 1-501
1-498, 1.14 gr R = p-MeO, 1-498 R-, = p-MeO, 1-500
Figure imgf000035_0003
R! = p-H, 1-502 Ri = p-H, X = N, 1-505
R, = p-MeO, 1-503 Ri = p-MeO, x = C, 1-506, Intermediates (5-bromo-2-nitro-iV-phenylaniline) and 5-bromo-N-(4-methoxyphenyl)-2- nitroaniline. A mixture of 4-bromo-2-fluoro-l -nitrobenzene (1.14 g, 1 eq) and aniline (1 eq) in DMF (30 mL) was heated at 80 °C and the reaction progression checked by LC MS. After 18 h the reaction was cooled down to room temperature, diluted with water (50 mL) and extracted with DCM (3 x 50 mL). The organic layers were collected, washed with H20 (2 x 50 mL), brine (1 x 50 mL) and dried (MgSC^). The solvent was removed under reduced pressure and the crude product was used in the next reaction without further purification. 1-497, LC MS [M + 1] + m/z 293.00; 1-498, LC MS [M + 1]+ m/z 323.00. Intermediates 1-500 (5-bromo-Nl-(4-methoxyphenyl)benzene-l,2-diamine) and 501 (5- bromo-Nl-phenylbenzene-l,2-diamine). The crude product from the previous reaction was dissolved in MeOH (10 mL) and the resulting solution was cooled to 0 °C. Zn (7 eq) and formic acid (7 eq) were slowly added keeping the reaction temperature at 0 °C. After stirring for 2 h at room temperature, the solid was filtered off and the crude amine was obtained as a solid after removing the solvent under reduced pressure. The product was used in the following reaction without further purification. LC MS [M + 1]+ m/z 346.00; LC MS [M + 1]+ m/z 293.00.
Intermediates (6-bromo-l-phenyl-lH-benzo[d] imidazole) and (6-bromo-l-(4- methoxyphenyl)-lH-benzo[d] imidazole). The crude product from the previous reaction was dissolved in neat formaldehyde (10 mL) and the resulting solution refluxed at 90 °C for 1 h. After cooling down to room temperature, the solvent was removed under reduced pressure and the product purified by silica gel column chromatography. Final yields: 508 mg, 36%, over 3 steps; 1.2 g, 84%, over 3 steps. LC MS [M + 1]+ m/z 273.00; LC MS [M + 1]+ m/z 303.00. Synthesis of 6
Figure imgf000036_0001
Intermediate 6-bromo-lH-benzo[d]imidazole. A stirring solution of starting material (1 gr, 1 eq) was dissolved in neat HCOH (10 mL) and heated for 2 h at 100 °C. The reaction was cooled down to room temperature, the solvent removed under reduced pressure and the product purified by silica gel column chromatography using an ISCO system (EtOAc-hexane, 0-100%) (831 mg, 79%). LC MS [M + 1]+ m/z 196.00.
6-(pyridin-4-yl)-lH-benzo[d]imidazole, 6. Solvent was degassed with N2. The product of the previous reaction (400 mg, 1 eq) was dissolved in DMF:H20 (4: 1, 5 mL). To the resulting solution, boronic acid (249 mg, 1 eq), Na2C03 (848 mg, 4 eq) and Pd(dppf)Cl2 (81 mg, 0.05 eq) were added and the reaction was heated at 130 °C for 30' under microwave irradiation. After cooling down to room temperature, the reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 10 mL), washed with NaHC03 (1 x 30 mL), H20 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC^) and concentrated under reduced pressure. The product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%). Final yields: 171 mg, 44%; white solid; LC MS [M + 1]+ m/z 198.00; ¾ NMR (400 MHz, CDC13) δ: 10.16 (brs, 1H, NH), 8.27 (s, 1H, N=CH-N), 7.58 (m, 2Η, ArH), 7.34-7.30 (m, 3H, ArH), 7.20- 7.18 (m, 1H, ArH), 7.10-7.04 (m, 1H, ArH); 1 C NMR (100 MHz, CDC13) δ: 163.4, 160.5, 139.2, 139.1, 130.3, 129.8, 124.6, 124.5, 120.0, 118.4. l-phenyl-6-(pyridin-4-yl)- lH-benzo [d] imidazole, 7.
Figure imgf000037_0001
Solvent was degassed with N2. The product of the previous reaction was dissolved in DMF:H20 (4: 1,10 mL). To the resulting solution, boronic acid (1 eq), Na2C03 (4 eq) and Pd(dppf)Cl2 (0.05 eq) were added and the reaction was heated at 90 °C for 16 h. After cooling down to room temperature, the reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 100 mL), washed with H20 (3 x 50 mL) and brine (1 x 100 mL), dried (MgS04) and concentrated under reduced pressure. The product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%) to yield 10 (429 mg, 76% yield); brownish solid; l NMR (400 MHz, CDCI3) δ: 8.70 (brs, 2H, ArH), 8.35 (brs, 1H, ArH), 7.57-7.54 (m, 2H, ArH), 7.37- 7.30 (m, 4H, ArH), 7.20-7.09 (m, 4H, ArH). LC MS [M + 1]+ m/z 272.00 Synthesis route to l-(4-methoxyphenyl)-lH-benzo[d] imidazole, 8.
Figure imgf000038_0001
1 -517 1 -521 1 -522
8 Intermediate N-(4-methoxyphenyl)-2-nitroaniline. Nitrofluorobenzene (1.5 g, 1 eq.) and p- anisidine (1.30 gr, 1 eq.) were mixed in DMF (20 mL) and heated at 80 °C for 2 h. The reaction mixture was diluted with EtOAc (10 mL) and washed with sat. NaHCC (2 x 20 mL), H20 (2 x 20 mL) and brine (20 mL). The organic layers were collected, dried (MgSC^) and concentrated under reduced pressure. The crude product was used in the following reaction without further purification (2 gr); LC MS [M + 1]+ m/z 245.00.
Intermediate Nl-(4-methoxyphenyl)benzene-l,2-diamine. Crude starting material (2 g) was dissolved in MeOH (10 mL). After cooling the resulting solution down to 0 °C, Zn (7 eq.) and HCOOH (7 eq.) were slowly added. The reaction was stirred at room temperature for 2 h. The solid was filtered-off and the resulting solution concentrated under reduced pressure. The product was used in the next reaction without further purification (1 g); LC MS [M + 1]+ m/z 215.00. l-(4-methoxyphenyl)-lH-benzo[d]imidazole. 8
Figure imgf000038_0002
Crude starting material (1 gr) was dissolved in neat HCOH (10 mL) and the resulting solution heated at 100 °C for 15' under microwave irradiation. The reaction was concentrated under reduced pressure and the residue was purified by silica-gel column chromatography using an ISCO system (EtOAc-hexane 0-100%). The product was obtained as a brown solid (500 mg, 47% yield). LC/MS [M + 1]+ m/z 225.00; XH NMR (400 MHz, CDC13) δ: 8.07 (s, 1H, N=CH-N), 7.90-7.84 (m, 1Η, ArH), 7.47-7.42 (m, 1H, ArH), 7.40 (dd, 2H, J= 9.0, AA'BB' system, ArH), 7.34-7.29 (m, 2H, ArH), 7.06 (dd, 2H, J= 9.0, 2.4, ArH), 3.88 (s, 3H, OCH3); 1 C NMR (100 MHz, DMSOdj) <5: 158.7, 143.6, 143.4, 133.6, 128.8, 125.4, 123.2, 122.2, 120.6, 115.1, 113.9, 55.5. l-(4-methoxyphenyl)-6-(pyridin-4-yl-N-oxide)- lH-benzo [d] imidazole, 9.
Figure imgf000039_0001
XH NMR (400 MHz, DMSO-d6) δ 11.27 (s, 1H), 8.17 (dd, J= 26.0, 6.9 Hz, 2H), 7.65 (d, J= 7.1 Hz, 2H), 7.45 (dd, J= 7.3, 4.9 Hz, 3H), 7.12 (dd, J= 19.7, 8.7 Hz, 4H), 3.82 (s, 3H). 1 C NMR (100 MHz, DMSO-d6) 5 158.89, 154.11, 139.13, 137.10, 131.93, 129.54, 129.28, 128.22, 127.20, 123.82, 120.63, 115.13, 110.00, 106.02, 55.85. LC/MS [M + 1]+ = 318. l-(benzo [d] [ 1,3] dioxol-5-yl)-6-(pyridin-4-yl)-lH-benzo [d] imidazole, 10.
Figure imgf000039_0002
¾ NMR (400 MHz, CDC13) δ 8.65 (d, J= 6.0 Hz, 2H), 8.10 (s, 1H), 7.96 (d, J= 8.4 Hz, 1H), 7.70 (dd, J = 8.3, 7.1 Hz, 1H), 7.62 (dd, J= 8.4, 1.7 Hz, 1H), 7.54 (dd, J= 4.5, 1.6 Hz, 2H), 7.04 - 6.95 (m, 3H), 6.12 (s, 2H). 1 C NMR (100 MHz, CDCI3) δ 165.96, 150.56, 147.87, 145.17, 145.07, 140.09, 133.68, 133.67, 131.57, 128.91, 124.03, 122.55, 122.16, 121.15, 109.94, 52.85. l-(2,3-dihydrobenzofuran-5-yl)-6-(pyridin-4-yl)-lH-benzo[d]imidazole, 11.
Figure imgf000040_0001
A suspension of 6-bromo-l-(2,3-dihydrobenzofuran-5-yl)-lH-benzo[d]imidazole (63mg, 0.2 mmol), 4-pyridine boronic acid (37mg, 0.3mmol), Pd(Ph3)4 (lOmg, 0.04 mmol) and Na2CC>3 (42mg, 0.4mmol) in dioxane/H20 (5ml, 4: 1) under argon was irradiated with microwaves at 130°C for 10 minutes. The solvent was evaporated in vacuo and product purified by column chromatography (0 - 100%, hexane/ethyl acetate) to afford 14 as white solid (35mg, 56% yield). 1H NMR (400 MHz, CDC13) δ 8.67-8.63 (m, 2H), 8.12 (s, 1H), 7.99-7.93 (m, 1H), 7.69-7.52 (m, 4H), 7.34-7.23 (m, 2H), 6.98-6.95 (m, 1H), 4.75-4.68 (m, 2H), 3.39-3.31 (m, 2H); 13C NMR (100 MHz, CDC13) δ 160.4, 150.2, 148.8, 144.5, 144.1, 133.9, 129.2, 128.5, 125.0, 121.8, 121.0, 110.3, 109.0, 71.9, 29.7. l-(4-(lH-tetrazol-5-yl)phenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazole, 12.
Figure imgf000040_0002
A suspension of 4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzonitrile (30mg, 0.10 mmol), NaN3 (7mg, 0.13 mmol), NH4C1 (7mg, 0.13 mmol) and LiCl (2mg, 0.01 mmol) in DMF (8ml) was heated to 100°C under argon. The solvent was evaporated in vacuo and the residue was diluted with EtOAc and washed with brine. The organic layer was dried (MgSC^), evaporated in vacuo and product purified by column chromatography (0-40%, hexane/ethylacetate) to afford a white solid 15 (23mg, 67% yield). 1H NMR (400 MHz, DMSO-d6) δ 8.59-8.58 (m, 2H), 8.36- 8.30 (m, 3H), 7.97-7.95 (m, 2H), 7.86-7.85 (m, 1H), 7.69-7.63 (m, 4H), 7.38 (s, 1H); 13C (100 MHz, DMSO-d6) δ 149.6, 144.2, 143.8, 135.7, 134.4, 134.3, 132.2, 132.1, 130.8, 129.0, 128.8, 128.6, 124.7,120.9, 109.7.
6-(pyridin-4-yl)-3'H-l,5'-dibenzo[d]imidazole, 13.
Figure imgf000041_0001
¾ NMR (400 MHz, CDC13) δ 8.56 (d, J = 4.8 Hz, 2H), 8.29 (s, 1H), 8.23 (s, 1H), 7.96 (d, J = 8.5 Hz, 1H), 7.89 (dd, J = 18.4, 6.2 Hz, 2H), 7.79 (d, J = 0.9 Hz, 1H), 7.72 - 7.65 (m, 1H), 7.65 - 7.56 (m, 2H), 7.51 - 7.44 (m, 1H). 1 C NMR (100 MHz, CDC13) δ 149.35, 149.16, 144.10, 143.59, 143.11, 134.86, 133.79, 130.33, 122.25, 122.18, 120.38, 119.61, 109.25.
6-(3-methoxyphenyl)-l-(4-(methylsulfonyl)phenyl)-lH-benzo[d]imidazole, 14.
Figure imgf000041_0002
Brown solid. Mp: 118-120°C. l NMR (400 MHz, CDCI3) δ 8.20 (d, J= 8.3 Hz, 3H), 8.06 - 7.56 (m, 5H), 7.38 (t, J = 7.9 Hz, 1H), 7.24 - 7.11 (m, 2H), 6.92 (ddd, J = 8.3, 2.5, 0.8 Hz, 1H), 3.87 (s, 3H), 3.15 (d, J = 3.5 Hz, 3H). 1 C NMR (100 MHz, CDC13) δ 159.99, 142.71, 139.85, 138.23, 129.91, 129.84, 124.28, 123.48, 121.13, 120.04, 113.68, 112.40, 108.79, 55.40, 44.60. LC/MS [M + 1]+ = 379 6-(3,4-difluorophenyl)-l-(4-(methylsulfonyl)phenyl)-lH-benzo[d]imidazole, 15.
Figure imgf000042_0001
Brown solid. Mp: 211-213°C. XH NMR (400 MHz, CDC13) δ 8.22 (d, J= 8.1 Hz, 3H), 7.95 (d, J = 7.4 Hz, 1H), 7.80 (d, J= 8.3 Hz, 2H), 7.68 (s, 1H), 7.55 (d, J = 6.0 Hz, 1H), 7.40 (ddd, J = 11.2, 7.5, 2.0 Hz, 1H), 7.36 - 7.29 (m, 1H), 7.26 - 7.19 (m, 1H), 3.16 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 151.77, 151.26, 151.13, 149.30, 148.78, 148.65, 140.02, 138.30, 136.20, 129.88, 124.33, 123.45, 123.41, 123.39, 123.35, 123.13, 121.41, 117.72, 117.56, 116.47, 116.30, 108.65, 44.56. LC/MS [M + 1]+ = 385
l-(4-(methylsulfonyl)phenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazole, 16.
Figure imgf000042_0002
White solid. Mp: 209-211OC. XH NMR (400 MHz, CDC13) δ 8.67 (dd, J= 4.6, 1.6 Hz, 2H), 8.34 - 8.17 (m, 3H), 8.01 (d, J= 8.4 Hz, 1H), 7.85 - 7.74 (m, 3H), 7.68 (dd, J= 8.5, 1.6 Hz, 1H), 7.54 (dd, J = 4.5, 1.7 Hz, 2H), 3.17 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 150.31, 148.40, 144.93, 142.89, 140.64, 140.18, 135.06, 133.71, 129.94, 124.40, 123.00, 121.98, 121.72, 108.85, 44.57. LC/MS [M + 1]+ = 350. l-(4-(methylsulfonyl)phenyl)-6-(pyridin-3-yl)-lH-benzo[d]imidazole, 17.
Figure imgf000042_0003
Orange solid. Mp: 203-205°C. l NMR (400 MHz, CDC13) δ 8.88 (d, J = 2.3 Hz, 1H), 8.61 (dd, J = 4.8, 1.5 Hz, 1H), 8.33 - 8.14 (m, 3H), 8.00 (d, J= 8.4 Hz, 1H), 7.95 - 7.88 (m, 1H), 7.84 - 7.78 (m, 2H), 7.77 - 7.73 (m, 1H), 7.61 (dd, J = 8.4, 1.7 Hz, 1H), 7.39 (dd, J = 7.9, 4.8 Hz, 1H), 3.16 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 148.57, 148.55, 144.25, 142.54, 140.71, 140.03, 136.73, 134.75, 134.73, 133.68, 129.89, 124.32, 123.61, 123.26, 121.64, 108.90, 44.58. LC/MS [M + 1]+ = 350 l-(4-(methylsulfonyl)ph )-lH-benzo[d]imidazole, 18.
Figure imgf000043_0001
Brown solid. Mp: 219-221°C. l NMR (400 MHz, CDC13) δ 8.22 (t, J = 7.1 Hz, 3H), 7.96 (d, J = 8.2 Hz, 1H), 7.80 (d, J= 8.5 Hz, 2H), 7.72 (s, 1H), 7.66 - 7.55 (m, 3H), 7.30 (d, J= 8.1 Hz, 2H), 3.16 (s, 3H). 1 C NMR (100 MHz, CDCI3) δ 148.76, 139.99, 136.97, 129.88, 128.87, 124.32, 123.35, 121.38, 108.83, 44.58. LC/MS [M + 1]+ = 433 6-(benzo [d] [ 1,3] dioxol-5-yl)- -(4-(methylsulfonyl)phenyl)- lH-benzo [d] imidazole, 19.
Figure imgf000043_0002
Gray solid. Mp: 182-184°C. XH NMR (400 MHz, CDCI3) δ 8.19 (t, J= 6.7 Hz, 3H), 7.91 (d, J = 8.3 Hz, 1H), 7.78 (t, J= 8.1 Hz, 2H), 7.66 (s, 1H), 7.54 (t, J= 7.5 Hz, 1H), 7.12 - 7.00 (m, 2H), 6.89 (dd, J = 7.6, 0.8 Hz, 1H), 6.01 (d, J = 1.3 Hz, 2H), 3.15 (d, J= 2.7 Hz, 3H). 1 C NMR (100 MHz, CDCI3) δ 148.21, 147.22, 142.02, 140.93, 139.80, 138.15, 135.52, 129.83, 124.24, 123.28, 121.11, 121.03, 108.66, 108.39, 108.01, 101.26, 44.60. LC/MS [M + 1]+ = 393
4-(5-(l-(4-(methylsulfonyl)phenyl)-lH-benzo[d]imidazol-6-yl)pyridin-2-yl)morpholine, 20.
Figure imgf000044_0001
Brown solid. 235-237°C. l NMR (400 MHz, CDC13) δ 8.48 (d, J= 2.2 Hz, 1H), 8.20 (d, J= 8. Hz, 3H), 8.03 - 7.47 (m, 5H), 6.73 (d, J= 8.7 Hz, 2H), 3.98 - 3.78 (m, 4H), 3.55 (dd, J= 19.4, 14.6 Hz, 4H), 3.16 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 139.83, 129.81, 126.75, 124.19, 122.61, 121.39, 109.98, 107.72, 106.77, 66.70, 45.64, 44.59. LC/MS [M + 1]+ = 435.
6-(3-chloro-4-methoxyphenyl)-l-(4-(methylsulfonyl)phenyl)-lH-benzo[d]imidazole, 21.
Figure imgf000044_0002
Gray solid. Mp: 199-201°C. l NMR (400 MHz, CDC13) δ 8.22 (d, J= 7.9 Hz, 3H), 7.93 (s, 1H), 7.80 (d, J= 8.2 Hz, 2H), 7.74 - 7.52 (m, 3H), 7.47 (dd, J = 8.5, 2.2 Hz, 1H), 7.02 (d, J = 8.5 Hz, 1H), 3.95 (s, 3H), 3.16 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 154.54, 139.93, 136.75, 134.62, 129.89, 129.16, 126.71, 124.35, 122.90, 121.28, 112.37, 56.31, 44.61. LC/MS [M + 2]+ = 414
-(furan-3-yl)-l-(4-(methylsulfonyl)phenyl)-lH-benzo[d]imidazole, 22.
Figure imgf000044_0003
White solid. Mp: 194-196°C. XH NMR (400 MHz, CDC13) δ 8.21 (dd, J= 31.2, 22.8 Hz, 3H), 7.89 (d, J= 8.4 Hz, 1H), 7.84 - 7.72 (m, 3H), 7.63 (d, J = 0.6 Hz, 1H), 7.57 - 7.48 (m, 2H), 6.81 - 6.66 (m, 1H), 3.17 (s, 3H). C NMR (100 MHz, CDC13) δ 143.89, 143.47, 141.91, 140.91, 139.87, 138.62, 129.84, 129.42, 126.54, 124.27, 122.30, 121.33, 109.09, 107.21, 44.59. LC/MS [M + 1]+ = 339.
4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzonitrile, 23.
Figure imgf000045_0001
To a suspension of 4-(6-bromo-lH-benzo[d]imidazol-l-yl)benzonitrile (297mg, l .Ommol), 4- pyridine boronic acid (148mg, 1.2mmol) and Na2CC>3 (220mg, 2 mmol) in a 1,4-dioxane and H20 solution (4: 1, 10ml) under argon, was added Pd(PPh3)4 (20mg). The reaction was irradiated with microwave at 130°C for 20 min. The solvent was removed in vacuo and the product was purified by column chromatography to yield 26 as a white solid (169mg, 57%). LC/MS [M + 1]+ m/z 297; XH NMR (400 MHz, DMSC ) δ: 8.64 (s, 1H), 8.23 (s, 1H), 7.97-8.12 (m, 4H), 7.85 (s, 1H), 7.75-7.78 (m, 2H), 7.73 (s, 1H), 7.59-7.61 (m, 1H), 7.04 (s, 1H). 1 C NMR (100 MHz, DMSC g) * 144.63, 144.00, 143.62, 140.18, 139.88, 134.79, 133.45, 128.68, 126.61, 124.62, 121.86, 120.85, 118.79, 110.32, 109.62, 108.02.
4-(6-(furan-3-yl)- lH-benzo [d] imidazol- l-yl)benzonitrile, 24.
Figure imgf000045_0002
To a suspension of 4-(6-bromo-lH-benzo[d]imidazol-l-yl)benzonitrile (297mg, l .Ommol), boronic acid (133 mg, 1.2 mmol), Na2C03 (212mg, 2.0mmol) in l,4-dioxane/H20 (10ml/2ml) under argon was added Pd(PPh3)4 (30mg). The reaction was irradiated with microwaves at 130°C for 30 mins. The reaction mixture was filtered, evaporated in vacuo and the 27 was purified by column chromatography (0-100%, ethyl acetate/hexane) to afford 27 (188mg, 66% yield) as a white solid. LC MS [M + 1]+ m/z 286; XH NMR (400 MHz, DMSC ) δ: 8.44 (s, 1H), 8.20-8.24 (m, 2H), 7.67-7.76 (m, 4H), 7.41 -7.42 (m, 1H), 7.29-7.33 (m, 2H), 6.14 (s, 1H). 1 C NMR (100 MHz, DMSC ) δ: 150.57, 147.86, 145.17, 145.13, 140.01, 134.81, 133.78, 133.54, 124.75, 122.69, 122.19, 121.17, 110.53, 110.05.
4-(6-(lH-pyrazol-4-yl)- lH-benzo [d] imidazol- l- l)benzonitrile, 25.
Figure imgf000046_0001
Synthetic Route to 25 and 37
Figure imgf000046_0002
The bromo-benzimidazole (100 mg, 1 eq) was dissolved in dioxane:H20 (4: 1 , 5 mL). To the resulting solution, boronic acid pinacol ester (97 mg, 1.5 eq), Na2CC>3 (70 mg, 4 eq) and Pd(PPl¾)4 (38 mg, 0.1 eq) were added and the reaction was heated at 130 °C for 30' under microwave irradiation. After cooling down to room temperature, the reaction was filtered through a celite pad, diluted with DCMTPA (3: 1 , 10 mL), washed with NaHC03 (1 x 30 mL), H20 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC^) and concentrated under reduced pressure. The product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%). Yield: 80 mg, 85%; white solid; LC MS [M + 1]+ m/z 286.00; lH NMR (400 MHz, DMSC ) δ: 12.92 (brs, 1H, NH), 8.62 (s, 1H, N=CH-N), 8.39-7.82 (m, 7Η, ArH), 7.76 (d, 1H, J = 8.5, ArH), 7.61 (dd, 1H, J = 8.5, 1.6, ArH); 1 C NMR (100 MHz, OMSOd6) S: 143.1, 142.5, 139.8, 136.5, 134.3, 133.1, 129.3, 125.6, 124.1, 121.5, 121.2, 120.3, 1 18.4, 109.7, 106.8. 4-(6-(lH-pyrazol-4-yl)- lH-benz e, 37.
Figure imgf000047_0001
To a suspension of 25 (41 mg, 1 eq) in MeOH (3 mL), KOH (40 mg, 5 eq) was added and the reaction was heated at 100 °C for 20' under microwave irradiation. The solvent was removed under reduced pressure and the residue was columned on silica gel using an ISCO system
(EtOAc-hexane, 0-100%). Final yields: 14 mg, 33%. white solid; LC MS [M + 1]+ m/z 304.00. lH NMR (400 MHz, DMSC ) δ: 12.90 (br s, 2H, NH2), 8.56 (s, 1H, N=CH-N), 8.30-7.91 (m, 5Η, ArH), 7.87-7.79 (m, 2H, ArH), 7.75 (dd, 1H, J= 8.4, 3.5, ArH), 7.59 (dd, 1H, J = 8.4, 1.4, ArH), 7.50 (br s, 1H, NH); 1 C NMR (100 MHz, DMSO^) δ: 166.9, 143.2, 142.4, 138.3, 133.4, 133.0, 129.4, 129.1, 123.1, 121.6, 121.0, 120.2, 109.5, 109.6, 106.8.
4-(6-(l-methyl-lH-pyrazol-4-yl -lH-benzo[d]imidazol-l-yl)benzonitrile, 26.
Figure imgf000047_0002
Solvent was degassed with N2. The bromo-benzimidazole A (100 mg, 1 eq) was dissolved in dioxane:H20 (4: 1, 3 mL). To the resulting solution, boronic acid (106 mg, 1.5 eq.), Na2C03 (70 mg, 2 eq.) and Pd(PPh3)4 (38 mg, 0.01 eq) were added and the reaction was heated at 130 °C for 30' under microwave irradiation. After cooling down to room temperature, the reaction was filtered through a celite pad, diluted with DCMTPA (3: 1, 10 mL), washed with NaHC03 (1 x 30 mL), H20 (3 x 50 mL) and brine (1 x 100 mL), dried (MgSC^) and concentrated under reduced pressure. The product was purified by silica gel column chromatography using an ISCO system (EtOAc/hexane, 0 to 80%). Yield: 75 mg, 76%; white solid; LC MS [M + 1]+ m/z 300.00; XH NMR [400 MHz, (CD3)2CO] δ: 8.44 (s, 1H, N=CH-N), 8.09-8.00 (m, 4Η, ArH), 7.86 (m, 2H, ArH), 7.75 (d, 2H, J= 8.8, ArH), 7.57 (d, 1H, J= 8.5, ArH), 3.91 (s, 3H, CH3); 1 C NMR [100 MHz, (CD3)2CO] δ: 143.5, 141.3, 137.2, 135.1, 130.6, 128.4, 125.2, 124.0, 122.1, 121.7, 118.9, 111.8, 111.0, 107.8, 38.0.
4-(6-(l-methyl-lH-pyrazol-4- - lH-benzo [d] imidazol- l-yl)benzamide, 38.
Figure imgf000048_0001
To a suspension of 26 (30 mg, 1 eq) in MeOH (3 mL), KOH (28 mg, 5 eq) was added and the reaction was heated at 100 °C for 20' under microwave irradiation. The solvent was removed under reduced pressure and the residue was columned on silica gel using an ISCO system (EtOAc-hexane, 0-100%). Yield: 10 mg, 32%; white solid; LC MS [M + 1]+ m/z 318.00; XH NMR (400 MHz, CDC13) δ: 8.12 (s, 1H, N=CH-N), 8.07 (d, 2Η, J= 8.7, 2.0, A'ABB' system, ArH), 7.86 (d, 1H, J= 8.4, ArH), 7.76 (s, 1H, ArH), 7.64-7.60 (m, 3H, ArH), 7.49 (dd, 1H, J =8.1, 1.4, ArH), 3.96 (s, 3H, CH3).
4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzoic acid, 27.
Figure imgf000048_0002
0.13 g (0.395 mmol) of the methyl ester 32 were mixed with 5% NaOH (1.58 mL) and microwaved for 5 minutes at 90 C with stirring. The solution was titrated to pH 4.0 using IN HC1 until product precipitated. The white solid was filtered using a Buchner funnel and filter paper, then dried under vacuum overnight at 30°C to yield 27 (95% yield). LC/MS [M + 1]+ m/z 316; ¾ NMR [400 MHz, DMSO-d6] δ: 8.68 (s, 1H), 8.60-8.63 (m, 2H), 8.11-8.15 (m, 2H), 7.98 (s, 1H), 7.90-7.92 (m, 1H), 7.77-7.80 (m, 2H), 7.73-7.76 (m, 1H), 7.68-7.72 (m, 2H). 4-(6-(furan-3-yl)-lH-benzo[d]imidazol-l-yl)benzoic acid, 28.
Figure imgf000049_0001
A suspension of 4-(6-(furan-3-yl)-lH-benzo[d]imidazol-l-yl)benzonitrile (57mg, 2.0mmol) and KOH (56mg, lO.OOmmol) in MeOH (6ml) was heated with microwave at 150°C for 20 mins. The reaction mixture was then acidified to ~ pH 4 and solvent was evaporated in vacuo. The product was purified by column chromatography (0 - 80%, hexane/ethylacetate) to afford 28 (52mg, 85% yield) as a white solid. -(6-(lH-pyrazol-4-yl)-lH-benzo[d]imidazol-l-yl)benzoic acid, 29.
Figure imgf000049_0002
To a suspension of 25 (84 mg, 1 eq) in MeOH (1 mL), KOH (79 mg, 5 eq) was added and the resulting mixture was heated at 150 °C for 20' under microwave irradiation. After cooling down to room temperature, MeOH was removed under reduced pressure and the residue suspended in H20 (5 mL) and acidified to pH 2 by slow addition of cone. HC1. The organic layer was extracted with DCM (3 x 10 mL), washed with brine (1 x 10 mL), dried (MgS04) and the solvent removed under reduced pressure. The residue was purified by silica gel column chromatography using an ISCO system (DCM/MeOH, 0-20%) to yield 29: 60 mg, 71%. LC MS [M + 1]+ m/z 305.00; lH NMR (400 MHz, DMSOd6) δ: 9.62 (s, 1H, N=CH-N), 8.32-8.16 (m, 4Η, ArH), 8.06-7.77 (m, 5H, ArH); 1 C NMR (100 MHz, DMSO^) δ: 166.4, 142.1, 137.8, 133.4, 132.2, 131.6, 131.5, 131.4, 131.2 x 2, 127.9, 123.7, 120.7, 117.1, 107.9. -(6-(l-methyl-lH-pyrazol-4-yl)- lH-benzo [d] imidazol- l-yl)benzoic acid, 30.
Figure imgf000050_0001
KOH (75 mg, 5 eq.) was added to a suspension of starting material 26 (81 mg, 1 eq.) in MeOH (1 mL) and the resulting mixture was heated at 150 °C for 20' under microwave irradiation. After cooling down to room temperature, MeOH was removed under reduced pressure, the residue suspended in H20 (5 mL) and acidified to pH 2 by slow addition of cone. HC1. The water layer was extracted with DCM (3 x 10 mL), the organic layers collected and washed with brine (1 x 10 mL), dried (MgSO^ and the solvent removed under reduced pressure. The residue was purified by silica gel column chromatography using an ISCO system (MeOH/DCM, 0- 20%). Yield: 59 mg, 69%; LC/MS [M + 1]+ m/z 319.00; XH NMR (400 MHz, DMSO^) δ: 9.40 (s, 1H, N=CH-N), 8.30-8.21 (m, 3Η, ArH), 8.00-7.85 (m, 5H, ArH), 7.78-7.74 (m, 1H, ArH), 3.86 (s, 3H, CH3); 1 C NMR (100 MHz, DMSO^) δ: 169.9, 143.2, 142.4, 138.3, 133.4, 133.0, 129.4, 129.0, 123.1, 121.6, 121.0, 120.2, 109.6, 106.8, 39.0. Methyl 4-(6-(furan-3-yl)-lH-benzo[d]imidazol-l-yl)benzoate, 31.
Figure imgf000051_0001
To a suspension of 4-(6-(furan-3-yl)-lH-benzo[d]imidazol-l-yl)benzoic acid (30.4mg,
1.Ommol), EDC (38mg, 2 mmol) and DMAP (5mg, 0.2 mmol) in DMF (3ml) was added MeOH dropwise at room temperature. The solvent was removed and product purified by column chromatography (0 - 100%, ethylacetate/hexane) to afford 31 as a white solid (26mg, 83%).1H NMR (400 MHz, CDC13) δ 8.30-8.27 (m, 2H), 8.15 (s, 1H), 7.88-7.85 (m, 1H), 7.75 (s, 1H), 7.65-7.62 (m, 3H), 7.52-7.48 (m, 2H), 6.72 (s, 1H), 3.98 (s, 3H); 13C NMR (100 MHz, CDC13) δ 166.1, 143.9, 142.3, 140.1, 138.6, 133.8, 131.8, 129.7, 129.1, 126.7, 123.5, 122.0, 121.2, 109.2, 107.5, 52.6.
Methyl 4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzoate, 32.
Figure imgf000051_0002
Methyl 4-(6-bromo-lH-benzo[d]imidazol-l-yl) benzoate and reagents specified above were placed in a microwave vial, followed by addition of the designated solvent. The capped vial was purged with argon gas for 5 minutes. The reaction was then heated via microwave irradiation for 30 minutes at 130°C. After reaction completion, the crude material was filtered through celite, and the celite flushed with methanol. The remaining solvent was evaporated in vacuo via azeotropic removal with toluene. The crude material was then reabsorbed onto silica gel and product purified by flash chromatography 0-100% EtO Ac/Hex or 0-100% EtOAC/DCM to afford 32 as an orange-beige solid (39% yield), l NMR (400 MHz, DMSO-d6) δ = 8.77 (s, 1H), 8.66 - 8.59 (m, 2H), 8.24 - 8.16 (m, 2H), 8.05 (dd, J=1.7, 0.7, 1H), 8.03 - 7.88 (m, 3H), 7.83 - 7.72 (m, 3H), 3.92 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ = 165.97, 150.58, 147.89, 145.21, 145.09, 140.11, 133.71, 133.69, 131.58, 128.92, 124.04, 122.56, 122.18, 121.17, 109.96, 52.86; M+l (m/z): 330.
Methyl 4-(6-(l-methyl-lH-pyrazol-4-yl)-lH-benzo[d]imidazol-l-yl)benzoate, 33.
Figure imgf000052_0001
To a suspension of the carboxylic acid 30 (112 mg, 1 eq) in DMF (1 mL), were added DMAP (8 mg, 0.2 eq), EDC (134 mg, 2 eq) and MeOH (2 drops). The reaction was stirred at room temperature for 8 h. The solvent was removed and the final product purified by silica gel column chromatography using an ISCO system (EtOAc-hexane, 0-50%). Final yield: 41 mg, 35%; white solid; LC MS [M + 1]+ m/z 333.00; lH NMR [400 MHz, DMSO^] δ: 8.62 (s, 1H, N=CH-N), 8.21-8.19 (m, 2Η, ArH), 87.93-7.90 (m, 2H, ArH), 7.83-7.75 (m, 2H, ArH), 7.57-7.54 (m, 2H, ArH), 3.96 (s, 3H, OCH3), 3.85 (s, 3H, NCH3); 1 C NMR [400MHz, (CD3)2CO] δ: 165.5, 143.2, 142.4, 140.3, 136.1, 131.2, 129.3, 128.9, 127.2, 123.3, 122.9, 120.9, 120.5, 114.2, 106.7, 51.6, 38.1.
Methyl 4-(6-(lH-pyrazol-4-yl -lH-benzo[d]imidazol-l-yl)benzoate, 34.
Figure imgf000053_0001
To a suspension of the carboxylic acid 29 (35 mg, 1 eq) in DMF (1 mL), DMAP (3 mg, 0.2 eq), EDC (44 mg, 2 eq) and MeOH (2 drops) were added. The reaction was stirred at room temperature for 8 h. The solvent was removed under reduced pressure and the final product 34 purified by silica gel column chromatography using an ISCO system (EtOAc-hexane, 0-50%). Yield: 5 mg, 14%; white solid; LC MS [M + 1]+ m/z 305.00; lH NMR (400 MHz, DMSC ) δ: 12.97 (brs, 1H, NH), 8.61 (s, 1H, N=CH-N), 8.37-8.10 (m, 3Η, ArH), 8.07-7.82 (m, 4H, ArH), 7.66 (d, 1H, J= 8.7, ArH), 7.60 (d, 1H, J= 8.1 Hz, ArH), 3.92 (s, 3H, OCH3); 1 C NMR (100 MHz, DMSCvg) δ: 168.5, 165.6, 143.1, 142.5, 140.0, 133.2, 131.1, 129.2, 128.2, 127.3, 123.4, 121.5, 121.1, 120.3, 106.9, 52.4.
4-(6-(pyridin-4-yl)- lH-benzo [d] imidazol- l-yl)benzamide, 35.
Figure imgf000053_0002
A suspension of 4-(6-(pyridin-4-yl)-lH-benzo[d] imidazol- l-yl)benzonitrile (29.6mg, l.Ommol) and KOH (28mg, 5.0mmol) in MeOH (3ml) was heated in a microwave at 100°C for 20 mins. The solvent was evaporated in vacuo and product purified by column chromatography (0-50%, hexane/ethylacetate) to afford 38 (22mg, 70% yield) as a white solid. 1H NMR (400 MHz, CDCI3) δ 8.67-8.66 (m, 2H), 8.10-7.98 (m, 3H), 7.78 (s, 1H), 7.68-7.65 (m, 3H), 7.54-7.53 (m, 2H), 5.30 (s, 2H).
4-(6-(furan-3-yl)- IH-benzo [d] imidazol- l-yl)benzamide, 36.
Figure imgf000054_0001
'H NMR (400 MHz, MeOD) 8.47 (s, 1H), 8.19-8.21 (m, 2H), 7.91 (s, 1H), 7.20 - 7.27 (4H, 2 x m), 7.5-7.65 (2H, 2 x m), 6.80 (1H, s), 5.5 (2H, s).1 C NMR (100 MHz, MeOD ) δ 169.61, 143.72, 142.77, 142.18, 138.77, 138.74, 133.49, 133.29, 129,39, 129.31, 126.47, 123.52, 123.51, 121.67, 119.56, 108.49, 107.22. [M+H]+ = 304
2-morpholino-N-(4-(6-(pyridin-4-yl)- IH-benzo [d] imidazol- l-yl)phenyl)acetamide, 39.
Figure imgf000054_0002
Yellow solid. Mp: 220-222°C. l NMR (400 MHz, CDC13) δ 9.30 (s, 1H), 8.64 (d, J= 4.5 Hz, 2H), 8.15 (s, 1H), 7.97 (d, J= 8.4 Hz, 1H), 7.84 (d, J = 8.5 Hz, 2H), 7.71 (s, 1H), 7.63 (d, J= 8.4 Hz, 1H), 7.52 (d, J= 8.7 Hz, 3H), 3.98 - 3.69 (m, 4H), 3.17 (d, J = 34.4 Hz, 2H), 2.72 (dd, J = 37.6, 33.6 Hz, 4H). 1 C NMR (100 MHz, CDC13) δ 168.29, 150.10, 148.72, 144.50, 143.52,
137.69, 134.61, 134.17, 131.73, 125.17, 122.17, 121.95, 121.19, 120.93, 108.95, 67.01, 62.41, 53.83. LC/MS [M + 1]+ = 414
2-morpholino-N-(4-(6-(pyridin-3-yl)- IH-benzo [d] imidazol- l-yl)phenyl)acetamide, 40.
Figure imgf000055_0001
Orange solid. Mp: 85-89°C. l NMR (400 MHz, CDC13) δ 9.33 (d, J= 26.4 Hz, 1H), 8.86 (s, 1H), 8.59 (d, J= 4.6 Hz, 1H), 8.14 (s, 1H), 7.97 (d, J = 8.4 Hz, 1H), 7.94 - 7.88 (m, 1H), 7.83 (d, J= 8.4 Hz, 2H), 7.65 (d, J = 0.8 Hz, 1H), 7.55 (ddd, J= 15.5, 8.1, 4.6 Hz, 3H), 7.37 (dd, J = 7.9, 4.8 Hz, 1H), 3.91 - 3.71 (m, 4H), 3.19 (t, J = 15.7 Hz, 2H), 2.76 - 2.59 (m, 4H). 1 C NMR (100 MHz, CDC13) δ 168.16, 148.48, 148.24, 143.86, 143.16, 137.60, 137.03, 134.72, 133.91, 131.86, 125.12, 123.54, 122.46, 121.17, 120.88, 108.95, 66.99, 62.40, 53.82. LC/MS [M + 1]+ =
414
2-Morpholino-N-(4-(6-(pyrimidin-5-yl)-lH-benzo[d]imidazol-l-yl)phenyl)acetamide, 41.
Figure imgf000055_0002
Yellow solid. Mp: 191-193°C. l NMR (400 MHz, CDC13) δ 9.43 - 9.08 (m, 2H), 8.97 (s, 2H) 8.17 (s, 1H), 8.01 (d, J = 8.4 Hz, 1H), 7.83 (t, J= 13.0 Hz, 2H), 7.65 (s, 1H), 7.54 (dd, J= 12.5 8.6 Hz, 2H), 3.91 - 3.73 (m, 4H), 3.22 (s, 2H), 2.85 - 2.60 (m, 4H). 1 C NMR (100 MHz, CDCI3) δ 168.27, 157.28, 155.11, 143.63, 137.83, 134.78, 131.62, 130.18, 125.20, 122.11, 121.68, 120.95, 108.99, 67.02, 62.43, 53.85. LC/MS [M + 1]+ = 415
A/-(4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzyl)acetamide, 42.
Figure imgf000055_0003
Brown solid. Mp: 81-83°C. l NMR (400 MHz, CDC13) δ 8.62 (d, J= 6.0 Hz, 2H), 8.09 (d, J 5.8 Hz, 1H), 7.95 (d, J= 8.5 Hz, 1H), 7.72 (d, J= 1.6 Hz, 1H), 7.62 (dd, J= 8.4, 1.7 Hz, 1H), 7.58 - 7.47 (m, 5H), 6.48 (s, 1H), 4.55 (t, J= 8.1 Hz, 2H), 2.10 (s, 3H). 13C NMR (100 MHz, CDC13) δ 170.26, 150.10, 148.71, 144.57, 143.46, 139.22, 135.03, 134.40, 134.21, 129.53, 124.46, 122.25, 121.98, 121.18, 109.03, 43.06, 23.27. LC/MS [M + 1]+ = 343
N-(4-(6-(pyridin-3-yl)-lH-benzo[d]imidazol-l-yl)benzyl)acetamide, 43.
Figure imgf000056_0001
Brown solid. Mp: 217-219°C. XH NMR (400 MHz, DMSO-d6) δ 7.59 (d, J= 2.0 Hz, 1H), 7.21 (dd, J = 4.7, 1.3 Hz, 1H), 7.12 (t, J = 5.9 Hz, 1H), 6.85 - 6.73 (m, 1H), 6.52 (dd, J= 18.1, 5.0 Hz, 2H), 6.38 (dd, J = 8.7, 2.1 Hz, 2H), 6.30 (dd, J = 8.4, 1.7 Hz, 1H), 6.20 - 6.06 (m, 3H), 3.02 (t, J = 9.0 Hz, 2H), 1.99 (s, 2H), 1.15 (dt, J= 3.5, 1.8 Hz, 1H). 1 C NMR (100 MHz, DMSO-d6) δ 174.45, 153.35, 153.25, 149.50, 148.96, 144.66, 141.37, 139.77, 139.58, 138.92, 138.04, 134.08, 129.00, 128.88, 127.11, 125.70, 114.37, 46.85, 27.79. LC/MS [M + 1]+ = 343
N-(4-(6-(thiophen-3-yl)-lH-benzo[d]imidazol-l-yl)benzyl)acetamide, 44.
Figure imgf000056_0002
Brown solid. Mp: 188-190°C. l NMR (400 MHz, CDCI3) δ 7.98 (s, 1H), 7.86 (d, J = 8.4 Hz, 1H), 7.65 (s, 1H), 7.59 (dd, J = 8.4, 1.2 Hz, 1H), 7.54 - 7.44 (m, 4H), 7.44 - 7.40 (m, 1H), 7.40 - 7.35 (m, 2H), 6.35 (s, 1H), 4.54 (d, J= 5.9 Hz, 2H), 2.10 (s, 3H). 1 C NMR (100 MHz, CDCI3) δ 170.18, 143.10, 142.60, 142.48, 138.84, 135.30, 134.21, 132.27, 129.47, 126.63, 126.32, 124.37, 122.24, 120.65, 120.29, 108.08, 43.08, 23.26. LC/MS [M + 1]+ = 348 N-(4-(6-phenyl- lH-benzo [d] imidazol- l-yl)benzyl)acetamide, 45.
Figure imgf000057_0001
Brown oil. XH NMR (400 MHz, CDC13) δ 8.02 (d, J= 7.0 Hz, 1H), 7.91 (d, J= 8.4 Hz, 1H), 7.68 (dd, J = 16.3, 5.2 Hz, 1H), 7.64 - 7.56 (m, 2H), 7.55 - 7.29 (m, 7H), 6.30 (s, 1H), 4.64 - 4.48 (m, 2H), 2.09 (s, 3H). 1 C NMR (100 MHz, CDC13) δ 170.29, 142.61, 141.42, 138.80, 137.75, 135.33, 134.23, 129.47, 129.42, 129.25, 128.80, 128.53, 127.51, 127.21, 124.39, 124.23, 122.88, 120.56, 120.23, 119.06, 108.93, 43.11, 23.25. LC/MS [M + 1]+ = 342.
N-(4-(6-(pyrimidin-5-yl)- lH-benzo [d] imidazol-l-yl)benzyl)acetamide, 46.
Figure imgf000057_0002
Brown solid. Mp: 205-207°C. XH NMR (400 MHz, CDC13) δ 9.19 (s, 1H), 8.97 (s, 2H), 8.16 (d, J = 14.1 Hz, 1H), 8.01 (d, J= 8.4 Hz, 1H), 7.71 (d, J= 39.9 Hz, 1H), 7.61 - 7.45 (m, 4H), 6.08 (s, 1H), 4.56 (d, J= 6.0 Hz, 2H), 2.10 (s, 3H), 1.87 (s, 1H). 1 C NMR (100 MHz, CDCI3) δ 170.12, 157.31, 155.12, 144.46, 143.52, 139.27, 135.02, 134.76, 134.60, 130.24, 129.60, 124.52, 122.17, 121.69, 109.07, 43.10, 23.30. LC/MS [M + 1]+ = 344 N-(4-(6-(6-methoxypyridin-3-yl)-lH-benzo[d]imidazol-l-yl)benzyl)acetamide, 47.
Figure imgf000057_0003
Brown solid. Mp: 71-73°C. XH NMR (400 MHz, CDC13) δ 8.16 (dt, J= 12.2, 6.1 Hz, 1H), 8.08 (s, 1H), 7.90 (d, J= 8.4 Hz, 1H), 7.67 (d, J = 1.1 Hz, 1H), 7.62 (dd, J= 7.3, 1.9 Hz, 1H), 7.53 (dd, J = 8.4, 1.6 Hz, 1H), 7.50 (s, 4H), 7.02 - 6.94 (m, 1H), 6.02 (s, 1H), 4.54 (d, J= 6.0 Hz, 2H), 3.96 (s, 3H), 2.09 (d, J= 2.8 Hz, 3H). 1 C NMR (100 MHz, CDCI3) δ 170.08, 160.88, 145.72, 142.71, 138.90, 138.65, 135.44, 132.76, 129.42, 124.89, 124.57, 124.30, 120.16, 117.09, 111.15, 53.63, 43.11, 23.31. LC/MS [M + l]+ = 373.
N-(4-(6-(3,4-difluorophenyl)-lH-benzo[d]imidazol-l-yl)benzyl)acetamide, 48.
Figure imgf000058_0001
Brown solid. Mp: 62-64°C. XH NMR (400 MHz, CDCI3) δ 8.09 (s, 1H), 7.91 (t, J = 7.4 Hz, 1H), 7.59 (d, J= 1.4 Hz, 1H), 7.56 - 7.48 (m, 4H), 7.43 - 7.34 (m, 1H), 7.33 - 7.28 (m, 1H), 7.25 - 7.17 (m, 1H), 6.08 (s, 1H), 4.54 (dd, J = 10.6, 6.0 Hz, 2H), 2.10 (s, 3H), 1.86 (brs, 1H). 1 C NMR (100 MHz, CDCI3) δ 170.14, 143.61, 143.02, 138.96, 135.51, 135.23, 134.31, 129.52, 124.48, 123.40, 123.34, 122.49, 120.92, 117.62, 117.45, 116.41, 116.24, 108.82, 43.11, 23.31.
4-(4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)phenyl)morpholine, 49.
Figure imgf000058_0002
Brown solid. Mp: 207-209°C. l NMR (400 MHz, CDCI3) δ 8.65 (d, J= 4.3 Hz, 2H), 8.11 (s, 1H), 7.95 (d, J= 8.4 Hz, 1H), 7.74 - 7.66 (m, 1H), 7.61 (dd, J= 8.4, 1.5 Hz, 1H), 7.53 (d, J = 5.5 Hz, 2H), 7.45 - 7.38 (m, 2H), 7.09 (t, J= 6.0 Hz, 2H), 3.96 - 3.87 (m, 4H), 3.33 - 3.22 (m, 4H). 13C NMR (100 MHz, CDCI3) δ 151.26, 150.14, 148.84, 144.55, 143.93, 135.08, 133.89, 127.56, 125.61, 121.99, 121.90, 121.07, 116.29, 109.06, 66.74, 48.84. LC/MS [M + 1]+ = 357. 4-(4-(6-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzyl)morpholine, 50.
Figure imgf000059_0001
Brown solid. Mp: 166-168°C. XH NMR (400 MHz, DMSO-d6) δ 8.61 (m, 3H), 8.24 (s, 1H), 8.00 - 7.84 (m, 2H), 7.78 - 7.64 (m, 4H), 7.58 (d, J= 8.0 Hz, 2H), 3.62 (m, 4H), 3.33 (s, 2H), 2.59 - 2.39 (m, 4H). 1 C NMR (100 MHz, DMSO-d6) δ 150.39, 148.09, 144.96, 138.23, 134.98, 133.43, 130.80, 124.01, 122.01, 120.95, 109.55, 79.61, 79.28, 78.95, 66.68, 62.34, 53.64.
LC/MS [M + 1]+ = 371
4-(4-(6-phenyl-lH-benzo[d]imidazol-l-yl)benzyl)morpholine, 51.
Figure imgf000059_0002
Yellow solid. Mp: 111-113°C. 'H NMR (400 MHz, CDC13) δ 8.14 (s, 1H), 7.93 (d, J= 8.4 Hz, 1H), 7.70 (t, J= 3.9 Hz, 1H), 7.62 (t, J= 1.7 Hz, 1H), 7.60 (dd, J= 2.2, 1.4 Hz, 1H), 7.59 - 7.56 (m, 1H), 7.55 (d, J= 2.1 Hz, 1H), 7.53-7.40 (m, 5H), 7.39 - 7.31 (m, 1H), 3.74 (dd, J= 15.9, 11.2 Hz, 4H), 3.60 (s, 2H), 2.60 - 2.43 (m, 4H). 1 C NMR (100 MHz, CDC13) δ 142.85, 141.62, 138.32, 137.62, 135.17, 134.30, 130.68, 128.76, 127.56, 127.14, 124.04, 122.77, 120.66, 110.01, 109.00, 67.01, 62.76, 53.67. LC/MS [M + 1]+ = 370
4-(4-(6-(furan-3-yl)- lH-benzo [d] imidazol- l-yl)benzyl)morpholine, 52.
Figure imgf000059_0003
Brown solid. Mp: 119-121°C. XH NMR (400 MHz, CDC13) δ 8.10 (d, J= 10.5 Hz, 1H), 7.86 (dd, J = 8.4, 0.5 Hz, 1H), 7.72 (ddd, J= 9.0, 3.6, 2.1 Hz, 1H), 7.58 (dd, J = 4.8, 4.3 Hz, 3H), 7.52 - 7.45 (m, 4H), 6.74 - 6.70 (m, 1H), 3.75 (dd, J = 13.9, 9.3 Hz, 4H), 3.61 (s, 2H), 2.49 (d, J = 31.1 Hz, 4H). 1 C NMR (100 MHz, CDCI3) δ 143.68, 143.28, 142.64, 138.45, 130.72, 128.53, 126.84, 124.05, 121.54, 120.85, 109.21, 107.50, 106.33, 66.99, 62.76, 53.68. LC/MS [M + 1]+ = 358
4-(4-(6-(l-methyl- lH-pyrazol-4-yl)- IH-benzo [d] imidazol- l-yl)benzyl)morpholine, 53.
Figure imgf000060_0001
Brown solid. Mp: 105-107°C. XH NMR (400 MHz, DMSO-d6) δ 8.35 (d, J= 32.9 Hz, 1H), 8.10 (d, J= 17.5 Hz, 1H), 7.81 (s, 1H), 7.76 - 7.67 (m, 2H), 7.64 (d, J = 8.0 Hz, 2H), 7.57 (d, J= 7.9 Hz, 2H), 7.49 (d, J= 8.4 Hz, 1H), 3.86 (d, J = 13.3 Hz, 3H), 3.61 (m, 4H), 3.33 (s, 2H), 2.49 (m, 4H). 1 C NMR (100 MHz, DMSO-d6) δ 143.40, 142.82, 136.46, 135.24, 134.16, 130.74, 128.84, 128.07, 123.84, 122.88, 120.91, 120.55, 106.88, 66.68, 62.36, 53.64, 39.42. l-(4-methoxyphenyl)-5-(lH-pyrazol-4-yl)-lH-benzo[d]imidazole, 54.
Figure imgf000060_0002
lH NMR (400 MHz, CDC13) δ 8.16 - 7.84 (m, 4H), 7.55 - 7.38 (m, 4H), 7.13 - 7.04 (m, 2H), 3.90 (dd, J = 3.5, 2.3 Hz, 3H). 1 C NMR (100 MHz, CDC13) δ 159.39, 144.43, 143.17, 131.92, 129.05, 128.47, 127.42, 125.63, 122.17, 117.25, 115.17, 110.72, 55.67. LC/MS [M + 1]+ = 291. 4-(5-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzonitrile, 55.
Figure imgf000061_0001
XH NMR (400 MHz, d6-DMSO) δ 8.76 (d, J = 5.7 Hz, 1H), 8.62 (d, J = 5.7 Hz, 2H), 8.25 (s, 1H), 8.12 (d, J= 8.4 Hz, 2H), 7.97 (d, J= 8.4 Hz, 2H), 7.89 - 7.73 (m, 5H). 1 C NMR (100 MHz, d6-DMSO) δ 150.63, 147.60, 145.24, 144.88, 140.01, 134.79, 133.52, 132.82, 124.41, 123.33, 121.84, 118.99, 118.74, 112.16, 110.51. LC/MS [M + 1]+ = 297
4-(5-(pyridin-4-yl)- lH-benzo [d] imidazol- l-yl)benzamide, 56.
Figure imgf000061_0002
XH NMR (400 MHz, d6-DMSO) δ 8.69 (d, J= 5.7 Hz, 1H), 8.57 (d, J= 6.0 Hz, 2H), 8.21 (s, 1H), 8.08 (d, J= 8.6 Hz, 3H), 7.79 (s, 1H), 7.76 (dd, J= 6.1, 2.5 Hz, 5H), 7.47 (s, 1H). 1 C NMR (100 MHz, d6-DMSO) δ 167.28, 150.63, 147.75, 145.13, 144.94, 138.48, 133.90, 133.73, 132.51, 129.83, 123.49, 123.14, 121.84, 118.89, 112.10. LC/MS [M + 1]+ = 315.
4-(5-(pyridin-4-yl)-lH-benzo[d]imidazol-l-yl)benzoic acid, 57.
Figure imgf000061_0003
XH NMR (400 MHz, d6-DMSO) δ 8.75 (s, 1H), 8.62 (d, J= 5.4 Hz, 2H), 8.27 (t, J= 5.9 Hz, 1H), 8.16 (dt, J= 14.6, 4.3 Hz, 2H), 7.84 (ddd, J = 17.0, 9.8, 3.5 Hz, 6H). 1 C NMR (100 MHz, d- DMSO) δ 150.63, 147.72, 145.18, 144.90, 133.81, 132.59, 131.60, 123.58, 123.19, 121.85, 118.94, 112.13. LC/MS [M + l]+ = 316. l-(3,4-dimethoxyphenyl)-6-(4-pyridinyl)-lH-benzo[d]imidazole, 58.
Figure imgf000062_0001
6-bromo-l-(3,4-dimethoxyphenyl)-lH-benzo[d]imidazole (0.300 mmol, 100 mg), pyridine-4- boronic acid (1.0 eq., 0.300 mmol, 38.8 mg), Na2C03 (4.0 eq., 1.20 mmol, 127 mg), and a mixture of 4: 1 DMF/water (3 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.0 μηιοΐ, 2.9 mg) and tricyclohexylphosphine (0.03 eq., 9.0 μηιοΐ, 2.6 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a
CH2Cl2/MeOH gradient to obtain 65 (0.232 mmol, 76.9 mg). Yield: 77%, light red solid. XH NMR (400 MHz, DMSO-d6) δ 8.59 (d, J= 5.4 Hz, 2H), 8.57 (s, 1H), 7.87 (d, J= 8.6 Hz, 2H), 7.73 (d, J= 6.1 Hz, 2H), 7.70 (dd, J= 8.5, 1.6 Hz, 1H), 7.30 (d, J= 2.3 Hz, 1H), 7.25 (dd, J = 8.5, 2.4 Hz, 1H), 7.16 (d, J= 8.6 Hz, 1H), 3.84 (s, 6H). 1 C NMR (100 MHz, DMSO-d6) δ 150.57, 150.03, 148.94, 148.00, 145.50, 144.79, 134.75, 133.08, 129.04, 122.08, 121.90, 120.87, 116.63, 112.76, 109.65, 109.13, 56.30; [M+H]+= 332.0. l-(3,4-ethylenedioxyphenyl)-6-(4-pyridinyl)-lH-benzo[d]imidazole, 59.
Figure imgf000062_0002
6-bromo-l-(3,4-ethylenedioxyphenyl)-lH-benzo[d]imidazole (0.151 mmol, 50.0 mg), pyridine- 4-boronic acid (1.0 eq., 0.151 mmol, 19.6 mg), Na2C03 (4.0 eq., 0.604 mmol, 64.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.6 μιτιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a hexanes/EtOAc gradient to obtain 66 (0.097 mmol, 31.8 mg). Yield: 64%, dark purple solid. XH NMR (400 MHz, DMSO-d6) δ 8.61 (d, J= 4.8 Hz, 2H), 8.55 (d, J= 1.1 Hz, 1H), 7.88 (d, J= 8.4 Hz, 1H), 7.86 (s, 1H), 7.75 (dt, J= 4.5, 1.2 Hz, 2H), 7.71 (dt, J= 8.4, 1.3 Hz, 1H), 7.29 (dd, J = 2.5, 1.1 Hz, 1H), 7.21 (ddd, J= 8.6, 2.5, 1.1 Hz, 1H), 7.10 (dd, J= 8.6, 1.1 Hz, 1H), 4.34 (d, J = 0.9 Hz, 4H). 1 C NMR (100 MHz, DMSO-d6) δ 150.14, 147.61, 144.98, 144.33, 144.09, 143.25, 134.11, 132.75, 128.95, 121.67, 121.54, 120.46, 118.11, 117.17, 113.27, 109.14, 64.23, 64.13; [M+H]+= 330.0. l-(3,4-methylenedioxyphenyl)-6-(2-fluoro-4-pyridinyl)-lH-benzo[d]imidazole, 60.
Figure imgf000063_0001
6-bromo-l-(3,4-methylenedioxyphenyl)-lH-benzo[d] imidazole (0.315 mmol, 100 mg), 2-fluro- pyridine-4-boronic acid (1.0 eq., 0.315 mmol, 45.8 mg), Na2C03 (4.0 eq., 1.26 mmol, 134 mg), and a mixture of 4: 1 DMF/water (3.5 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 μιτιοΐ, 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 μιτιοΐ, 2.8 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a hexanes/EtOAc gradient to obtain 67 (0.270 mmol, 90.5 mg). Yield: 86%, light purple solid. XH NMR (400 MHz, DMSO-d6) δ 8.57 (s, 1H), 8.27 (d, J= 5.3 Hz, 1H), 7.94 (d, J= 1.7 Hz, 1H), 7.88 (d, J= 8.5 Hz, 1H), 7.79 - 7.72 (m, 2H), 7.60 (s, 1H), 7.41 (d, J= 2.2 Hz, 1H), 7.20 (dd, J= 8.2, 2.2 Hz, 1H), 7.14 (d, J= 8.2 Hz, 1H), 6.17 (s, 2H). 1 C NMR (100 MHz, DMSO- d6) δ 165.25, 162.93, 153.81 (d, J= 8.6 Hz), 148.30, 147.91 (d, J= 15.9 Hz), 147.04, 145.36, 144.71, 134.12, 131.50 (d, J= 3.5 Hz), 129.50, 121.74, 120.42, 120.09 (d, J= 3.7 Hz), 117.89, 109.76, 108.89, 107.10, 106.72, 105.92, 102.03; [M+H]+ = 334.0. l-(3,4-methylenedioxyphenyl)-6-(2- enzo[d]imidazole, 61.
Figure imgf000064_0001
6-bromo-l-(3,4-methylenedioxyphenyl)-lH-benzo[d] imidazole (0.315 mmol, 100 mg), 2-fluro- pyridine-4-boronic acid (1.0 eq., 0.315 mmol, 43.2 mg), Na2C03 (4.0 eq., 1.26 mmol, 134 mg), and a mixture of 4: 1 DMF/water (3.5 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 μηιοΐ, 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 μηιοΐ, 2.8 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a hexanes/EtOAc gradient to obtain 68 (0.180 mmol, 58.9 mg). Yield: 57%, off-white oil. A portion of the product was further purified through preparative reverse phase LCMS eluting with a water/acetonitrile gradient to obtain an analytically pure sample. XH NMR (400 MHz, DMSO- d6) δ 8.73 (br. s, 2H), 7.93 (br. s, 2H), 7.70 (br. s, 2H), 7.61 (br. s, 1H), 7.40 (s, 1H), 7.17 (q, J = 8.2 Hz, 2H), 6.17 (s, 2H), 2.54 (s, 3H). [M+H]+ = 330.0. l-(3,4-methylenedioxyphenyl)-6-(lH-pyrazol-4-yl)-lH-benzo[d]imidazole, 62.
Figure imgf000064_0002
6-bromo-l-(3,4-methylenedioxyphenyl)-lH-benzo[d] imidazole (0.315 mmol, 100 mg), (1H- pyrazol-4-yl) boronic acid pinacol ester (1.0 eq., 0.315 mmol, 64.4 mg), Na2CC>3 (4.0 eq., 1.26 mmol, 134 mg), and a mixture of 4: 1 DMF/water (3.5 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 3.2 μιτιοΐ, 3.0 mg) and tricyclohexylphosphine (0.03 eq., 9.5 μιτιοΐ, 2.8 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a CmCh/MeOH gradient to obtain 69 (0.270 mmol, 82.0 mg). Yield: 85%, pink solid. 'H NMR (400 MHz, DMSO-d6) δ 12.91 (br. s, 1H), 8.38 (s, 1H), 8.08 (br. s, 2H), 7.72 (d, J = 8.4 Hz, 1H), 7.68 (d, J= 1.3 Hz, 1H), 7.55 (dd, J= 8.4, 1.6 Hz, 1H), 7.34 (t, J= 1.3 Hz, 1H), 7.14 (d, J= 1.4 Hz, 2H), 6.17 (s, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 148.27, 146.85, 143.55, 142.08, 134.23, 129.89, 128.70, 121.68, 120.66, 120.03, 117.75, 108.89, 106.55, 105.87, 101.98; [M+H]+= 305.0. l-(4-methoxyphenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazol-2-amine, 63.
Figure imgf000065_0001
¾ NMR (400 MHz, CDC13) δ 8.56 (d, J = 5.9 Hz, 2H), 7.51 (d, J = 8.2 Hz, 1H), 7.49 - 7.44 (m 3H), 7.44 - 7.36 (m, 2H), 7.18 (d, J= 1.6 Hz, 1H), 7.16 - 7.10 (m, 2H), 5.13 (d, J= 10.8 Hz, 2H), 3.91 (d, J= 0.5 Hz, 3H). 1 C NMR (100 MHz, CDC13) δ 160.10, 150.01, 149.02, 143.20, 130.19, 128.34, 126.67, 121.42, 121.25, 116.60, 115.71, 106.72, 55.66.
N-(l-(4-methoxyphenyl)-6-(pyri azol-2-yl)acetamide, 64.
Figure imgf000065_0002
XH NMR (400 MHz, CDC13) δ 8.55-8.60 (m, 2H), 7.5-7.6 (2xm, 4H), 7.25-7.39 (m, 3H), 7.05- 7.25 (m, 2H), 3.91 (s, 3H), 2.33 (s, 3H). 1 C NMR (100 MHz, CDC13) 5160.17, 150.21, 150.15, 150.03, 148.31, 128.43, 128.37, 122.28, 121.67, 121.44, 121.35, 116.41, 115.76, 115.32, 108.35, 106.81, 55.69; [M+H]+ = 359.0. l-acetyl-N-(l-(4-methoxyphenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazol-2-yl)piperidine-4- carboxamide, 65.
Figure imgf000065_0003
65 'H NMR (400 MHz, CDCI3) δ 8.61-8.64 (m, 2H), 7.50-7.60 (m, 1H), 7.4-7.5 (m, 4H), 7.3 (s, 1H), 7.25 (s, 1H), 7.09-7.12 (m, 2H), 4.47-4.50 (m, 1H), 3.95 (s, 3H), 3.48-3.52 (m, 1H), 3.1-3.2 (m, 1H), 2.72-2.78 (m, 1H), 2.6 (br s, 1H), 2.09 (s, 3H), 1.8-2.0 (m, 2H), 1.55-1.75 (m, 2H). 1 C NMR (100 MHz, CDCI3) 5168.84, 159.74, 150.20, 148.03, 133.79, 128.25, 126.20, 122.63, 121.66, 114.92, 108.55, 55.61, 46.09, 41.26, 29.16, 28.71, 21.54; [M+H]+ = 470.0.
2-(3,5-difluorophenyl)-N-(l-(4-methoxyphenyl)-6-(pyridin-4-yl)-lH-benzo[d]imidazol-2- yl)acetamide, 66.
Figure imgf000066_0001
66
lH NMR (400 MHz, CDC13) δ 12.1-12.5 (br s, 1H), 7.25-7.55 (3xm, 7H), 7.41-7.19 (m, 2H), 6.83-6.91 (m, 2H), 6.60-6.75 (m, 1H), 3.93 (s, 3H), 3.70-3.74 (m, 2H). 1 C NMR (100 MHz, CDCI3) 5164.03, 163.90, 161.57, 161.44, 159.86, 150.27, 147.93, 133.95, 128.31, 125.96, 122.73, 121.65, 114.94, 112.64, 112.39, 108.61, 101.79, 55.62.
2-amino-N-(l-(4-methoxyph benzo[d]imidazol-2-yl)acetamide, 67.
Figure imgf000066_0002
'H NMR (400 MHz, CDCI3) δ 8.55-8.61 (br s, 2H), 7.30-7.55 (3xm, 7H), 7.02-7.21 (2xm, 4H), 5.25 (br s, 2H), 3.89 (s, 3H). 1 C NMR (100 MHz, CDCI3) 5171.42, 160.22, 154.15, 150.05, 148.87, 135.89, 130.58, 128.44, 128.35, 126.28, 121.50, 121.46, 121.43, 121.16, 116.72, 116.37, 115.84, 106.89, 55.68, 46.04, 29.71. [M+H]+ = 374.0.
6-(4-pyridinyl)imidazo[l,2-a]
Figure imgf000066_0003
6-bromoimidazo[l,2-a]pyridine (7.61 mmol, 1.5 g), pyridine-4-boronic acid (1.0 eq., 7.61 mmol, 985 mg), Na2C03 (4.0 eq., 30.5mmol, 3.23 g), and a mixture of 4: 1 DMF/water (80 mL) were added to a microwave vial. The vial was purged with Ar(g) then
tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 0.076 mmol, 72.0 mg) and tricyclohexylphosphine (0.03 eq., 0.228 mmol, 66.0 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a CH2Cl2/MeOH gradient to obtain 68 (6.8 mmol, 1.326 g). Yield: 89%, light brown solid. XH NMR (400 MHz, DMSO-d6) δ 9.15 (s, 1H), 8.66 (dd, J = 4.5, 1.6 Hz, 2H), 7.99 (s, 1H), 7.74 (dd, J = 4.5, 1.6 Hz, 2H), 7.73 - 7.68 (m, 3H). 1 C NMR (100 MHz, DMSO-d6) δ 150.31, 144.11, 143.92, 134.14, 125.60, 123.38, 122.01, 120.81, 117.19, 113.98; [M+H]+ = 196.0.
Intermediate: 3-iodo-6-(4-pyridinyl)imidazo[l,2-a]pyridine
A solution of 6-(4-pyridinyl)imidazo[l,2-a]pyridine (68, 5.63 mmol, 1.10 g) in DMF (13 mL) was prepared and N-iodosuccinamide (1.05 eq., 5.92 mmol, 1.372 g) was added. The reaction was allowed to stir at rt overnight then was evaporated in vacuo and azeotroped with toluene to remove the DMF. The crude residue was purified by flash chromatography eluting with a CH2Cl2/MeOH gradient followed by recrystallization from EtOH to obtain 3-iodo-6-(4- pyridinyl)imidazo[l,2-a] pyridine (5.40 mmol, 1.729 g). Yield: 96%, pale yellow solid. ¾ NMR (400 MHz, DMSO-d6) δ 8.69 (s, 1H), 8.59 (s, 1H), 7.83 (d, J= 5.8 Hz, 1H), 7.79 (d, J = 11.9 Hz, 1H), 7.76 - 7.70 (m, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 150.32, 143.63, 140.90, 124.51, 124.32, 123.79, 121.41, 117.59; [M+H]+ = 322.0.
Intermediate: 3-iodo-6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine
Figure imgf000067_0002
A solution of 6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine (2.71 mmol, 500 mg) in DMF (11 mL) was prepared and N-iodosuccinamide (1.05 eq., 2.85 mmol, 661 mg) was added. The reaction was allowed to stir at rt overnight then was evaporated in vacuo and azeotroped with toluene to remove the DMF. The crude residue was purified by flash chromatography eluting with a CH2Cl2/MeOH gradient to obtain 3-iodo-6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine (1.56 mmol, 484.2 mg). Yield: 58%, yellow solid. ¾ NMR (400 MHz, DMSO-d6) δ 13.08 (br. s, 1H), 8.39 (t, J= 1.3 Hz, 1H), 8.20 (br. s, 2H), 7.71 - 7.65 (m, 1H), 7.65 - 7.58 (m, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 145.92, 140.02, 124.87, 120.65, 119.72, 117.31, 117.19, 64.66; [M+H]+ = 311.0.
3-(3,4-methylenedioxyphenyl)- -(4-pyridinyl)imidazo[l,2-a]pyridine, 69.
Figure imgf000068_0001
3-iodo-6-(4-pyridinyl)imidazo[l,2-a]pyridine (0.235 mmol, 75mg), 3,4- methylenedioxyphenyl boronic acid (1.0 eq., 0.235 mmol, 39 mg), Na2C03 (4.0 eq., 0.940 mmol, 100 mg), and a mixture of 4: 1 DMF/water (2.5 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 2.4 μιτιοΐ, 2.3 mg) and tricyclohexylphosphine (0.03 eq., 7.1 μιτιοΐ, 2.1 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with
CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a
CH2Cl2/MeOH gradient to obtain 69 (0.121 mmol, 38mg). Yield: 51%, yellow oil, Ti NMR (400 MHz, DMSO-d6) δ 8.74 (s, 1H), 8.65 (d, J = 4.7 Hz, 2H), 7.81 - 7.73 (m, 4H), 7.70 (dd, J = 9.4, 1.7 Hz, 1H), 7.36 (d, J = 1.7 Hz, 1H), 7.22 (dd, J = 8.0, 1.6 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H),
6.12 (s, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 150.23, 148.06, 147.35, 144.74, 144.15, 133.15, 125.97, 123.62, 122.98, 122.34, 122.14, 122.01, 121.35, 117.80, 109.15, 108.51, 101.39;
[M+H]+ = 316.0. 6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine, 70.
Figure imgf000069_0001
6-bromoimidazo[l,2-a]pyridine (4.06 mmol, 800 mg), lH-pyrazol-4-yl boronic acid pinacol ester (1.0 eq., 4.06 mmol, 829mg), Na2C03 (4.0 eq., 16.2 mmol, 1.72 g), and a mixture of 4: 1 DMF/water (40 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 0.041 mmol, 38.3 mg) and tricyclohexylphosphine (0.03 eq., 0.122 mmol, 35.2 mg) was added. The reaction was microwave irradiated under Ar (g) at 130°C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via flash chromatography eluting with a CH2Cl2/MeOH gradient to obtain 70 (3.61 mmol, 665 mg). Yield: 89%, light brown solid. XH NMR (400 MHz, DMSO-d6) δ 8.91 (t, J = 1.3 Hz, 1H), 8.10 (br. s, 2H), 7.91 (s, 1H), 7.64 -7.54 (m, 3H). 13C NMR (100 MHz, DMSO-d6) δ 143.14, 132.56, 124.77, 121.82, 118.42, 117.45, 116.59, 113.27; [M+H]+ = 185.0. -(4-methoxyphenyl)-6-(4-pyridinyl)imidazo[ 1,2- a] pyridine, 71.
Figure imgf000069_0002
3 -iodo-6-(4-pyridinyl)imidazo[l,2-a] pyridine (0.156 mmol, 50.0 mg), 4-methoxyphenyl boronic acid (1.0 eq., 0.156 mmol, 23.7 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μιτιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with water/acetonitrile gradient to obtain 71 (0.042 mmol, 12.6 mg). Yield: 27%, yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 8.85 - 8.56 (m, 3H), 7.90 - 7.70 (m, 4H), 7.69 (d, J = 8.8 Hz, 2H), 7.14 (d, J = 8.8 Hz, 2H), 3.84 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 159.26, 150.22, 144.11, 132.76, 129.47, 123.60, 123.06, 122.18, 121.41, 120.72, 117.84, 114.84, 55.29; [M+H] = 302.0.
3-(4-methylsulfonylphenyl)-6-(4- ,2-a] pyridine, 72.
Figure imgf000070_0001
72
3 -iodo-6-(4-pyridinyl)imidazo[ 1,2-a] pyridine (0.156 mmol, 50.0 mg), 4-(methylsufonyl)phenyl boronic acid (1.0 eq., 0.156 mmol, 32.1 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μηιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μηιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 72 (0.0.54 mmol, 19.0 mg). Yield: 35%, yellow oil. XH
NMR (400 MHz, DMSO-d6) δ 9.13 (s, 1H), 8.81 (d, J = 4.2 Hz, 2H), 8.26 (s, 1H), 8.14 - 8.06 (m, 7H), 8.02 (d, J = 9.5 Hz, 1H), 3.31 (s, 3H). 1 C NMR (100 MHz, DMSO-d6) δ 147.45, 146.56, 143.88, 140.42, 132.38, 131.30, 128.71, 128.02, 127.41, 125.42, 124.71, 124.11, 122.64, 116.48, 43.48; [M+H]+ = 350.0.
3-(4-cyanophenyl)-6-(4-pyridinyl)imidazo [1,2-a] pyridine, 73.
Figure imgf000070_0002
3-iodo-6-(4-pyridinyl)imidazo[l,2-a]pyridine (0.156 mmol, 50.0 mg), 4-cyanophenyl boronic acid (1.0 eq., 0.156 mmol, 23.4 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7μηιο1, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 73 (0.045 mmol, 13.4 mg). Yield: 29%, yellow solid. XH NMR (400 MHz, DMSO-d6) δ 8.94 (dd, J = 1.7, 0.8 Hz, 1H), 8.67 (dd, J = 4.6, 1.6 Hz, 2H), 8.05 - 7.98 (m, 5H), 7.86 (dd, J = 9.4, 0.8 Hz, 1H), 7.82 (dd, J = 4.6, 1.6 Hz, 2H), 7.79 (dd, J = 9.4, 1.7 Hz, 1H). 1 C NMR (100 MHz, DMSO-d6) δ 150.24, 146.01, 143.93, 135.37, 133.38, 133.21, 127.86, 124.81, 124.70, 123.62, 122.92, 121.49, 118.83, 117.99, 109.91; [M+H]+ = 297.0.
4-(6-(4-pyridinyl)imidazo[l,2- d, 74.
Figure imgf000071_0001
3-iodo-6-(4-pyridinyl)imidazo[l,2-a]pyridine (0.156 mmol, 50.0 mg), 4-boronobenzoic acid (1.0 eq., 0.156 mmol, 25.8 mg), Na2C03 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μιτιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130°C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 74 (0.031 mmol, 9.9 mg). Yield: 20%, yellow solid. XH NMR (400 MHz, DMSO-d6) δ 13.19 (br. s, 1H), 9.13 (dd, J = 1.7, 0.7 Hz, 1H), 8.84 (d, J = 5.9 Hz, 2H), 8.33 (s, 1H), 8.19 (dd, J = 9.4, 1.7 Hz, 1H), 8.17 - 8.11 (m, 4H), 8.08 (dd, J = 9.4, 0.7 Hz, 1H), 7.97 (d, J = 8.6 Hz, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 166.84, 147.08, 146.73, 142.66, 130.99, 130.89, 130.29, 128.67, 128.44, 126.31, 125.06, 124.67, 122.88, 115.66;
[M+H]+ = 316.0.
4-(6-(pyridin-4-yl)imidazo [ 1,2-a] pyridin-3-yl)benzamide, 75.
Figure imgf000072_0001
3 -iodo-6-(4-pyridinyl)imidazo[l,2-a] pyridine (0.156 mmol, 50.0 mg), 4-carbamoylphenyl boronic acid (1.0 eq., 0.156 mmol, 26.5 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιηοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μηιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 75 (0.016 mmol, 5.1 mg). Yield: 10%, light orange solid. XH NMR (400 MHz, DMSO-d6) δ 8.99 (br. s, 1H), 8.68 (br. s, 1H), 8.09 (s, 1H), 8.06 (d, J = 8.1 Hz, 2H), 7.98 - 7.72 (m, 8H), 7.45 (s, 1H). 1 C NMR (100 MHz, DMSO-d6) δ 168.05, 167.29, 150.17, 135.42, 133.81, 133.39, 128.52, 127.18, 126.29, 124.14, 123.46, 109.55; [M+H]+ = 315.0.
3-(4-methylbenzoate)-6-(4-py ridine, 76.
Figure imgf000072_0002
76
3 -iodo-6-(4-pyridinyl)imidazo[l,2-a] pyridine (0.156 mmol, 50.0 mg), (4 - methoxycarbonyl)phenyl boronic acid (1.0 eq., 0.156 mmol, 28.0 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μιτιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 76 (0.024 mmol, 7.8 mg). Yield: 15%, pale yellow solid. XH NMR (400 MHz, DMSO-d6) δ 8.94 (s, 1H), 8.66 (br. s, 2H), 8.12 (d, J = 8.6 Hz, 2H), 8.00 (s, 1H), 7.96 (d, J = 8.6 Hz, 2H), 7.85 (d, J = 9.4 Hz, 1H), 7.82 (d, J = 5.9 Hz, 2H), 7.78 (dd, J = 9.4, 1.7 Hz, 1H), 3.90 (s, 3H). 1 C NMR (100 MHz, DMSO-d6) 5165.89, 150.25, 143.94, 134.87, 133.32, 130.17, 128.49, 127.38, 125.17, 124.54, 123.54, 122.71, 121.45, 118.03, 52.28; [M+H]+ = 330.0.
3-(2,3-dihydrobenzofuran-5- -6-(pyridin-4-yl)imidazo [ 1,2- a] pyridine, 77.
Figure imgf000073_0001
3-Iodo-6-(4-pyridinyl)imidazo[l,2-a]pyridine (0.156 mmol, 50.0 mg), (2,3-dihydrobenzofuran- 5-yl) boronic acid (1.0 eq., 0.156 mmol, 25.5 mg), Na2C03 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μηιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μηιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 77 (0.050 mmol, 15.8 mg). Yield: 32%, pale yellow solid. lH NMR (400 MHz, DMSO-d6) δ 8.72 (s, 1H), 8.64 (s, 2H), 7.81 - 7.74 (m, 3H), 7.71 (s, 1H), 7.69 (dd, J = 9.4, 1.7 Hz, 1H), 7.59 (s, 1H), 7.46 (dd, J = 8.2, 1.9 Hz, 1H), 6.95 (d, J = 8.2 Hz, 1H), 4.61 (t, J = 8.7 Hz, 2H), 3.28 (t, J= 8.7 Hz, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 159.91, 150.24, 144.17, 132.75, 128.71, 128.16, 125.18, 123.37, 122.94, 122.08, 121.35, 120.47, 117.87, 109.68, 71.33, 29.09; [M+H]+ = 314.0.
3-(3,4-methylenedioxyphenyl)-6-(l o[l,2-a]pyridine, 78.
Figure imgf000073_0002
78 3- iodo-6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine (0.156 mmol, 50.0 mg), 3,4- methylenedioxyphenyl boronic acid (1.0 eq., 0.156 mmol, 26.8 mg), Na2CC>3 (4.0 eq., 0.623 mmol, 66.0 mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μηιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μηιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 78 (0.048 mmol, 14.5 mg). Yield: 30%, white solid. lH NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 8.37 (br. s, 1H), 8.12 (br. s, 2H), 7.69 - 7.62 (m, 2H), 7.56 (dd, J = 9.3, 1.6 Hz, 1H), 7.27 (d, J = 1.6 Hz, 1H), 7.17 (dd, J = 8.0, 1.7 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 6.12 (s, 2H). 1 C NMR (100 MHz, DMSO-d6) δ 148.02, 147.15, 144.19, 132.49, 125.17, 124.26, 122.54, 121.73, 118.90, 118.37, 117.58, 109.12, 108.44, 101.33; [M+H]+ = 305.0.
4- (6-(lH-pyrazol-4-yl)imid -a] pyridin-3-yl)benzoic acid, 79.
Figure imgf000074_0001
3-iodo-6-(lH-pyrazol-4-yl)imidazo[l,2-a]pyridine (0.156 mmol, 50.0 mg), 4-boronobenzoic acid (1.0 eq., 0.156 mmol, 26.8 mg), Na2C03 (4.0 eq., 0.623 mmol, 66.0mg), and a mixture of 4: 1 DMF/water (2 mL) were added to a microwave vial. The vial was purged with Ar(g) then tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3, 0.01 eq., 1.6 μιτιοΐ, 1.5 mg) and tricyclohexylphosphine (0.03 eq., 4.7 μιτιοΐ, 1.4 mg) was added. The reaction was microwave irradiated under Ar (g) at 130 °C for 30 min. then filtered through celite washing down with CH2CI2 and MeOH. The filtrate was evaporated in vacuo and DMF was removed by azeotroping with toluene. The crude material was purified via reverse phase preparative LCMS eluting with a water/acetonitrile gradient to obtain 79 (0.001 mmol, 1.7 mg). Yield: 4%, pale yellow solid. XH NMR (400 MHz, DMSO-d6) δ 8.74 (s, 1H), 8.17 (br. s, 1H), 8.10 (d, J = 8.4 Hz, 2H), 7.91 - 7.82 (m, 4H), 7.72 (d, J = 9.3 Hz, 1H), 7.64 (dd, J = 9.3, 1.6 Hz, 1H). 1 C NMR (100 MHz, DMSO-d6) δ 145.55, 134.45, 134.34, 130.71, 128.49, 127.34, 125.38, 125.12, 119.90, 119.04, 118.21, 117.88; [M+H]+ =305.0. Example III.
This example shows shows DYRKIA inhibitory activity of the compounds of the present invention. [Key (+ = 1 - 40% inhibition @10uM, ++ = 40 - 60% inhibition @10uM, +++ = 60% - 100% inhibition @10uM)]a.
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000079_0001
Figure imgf000080_0001
Example IV.
This example provides general procedures (Schemes 1, 2, and 3 (note the numbering scheme for Schemes 1, 2, and 3 is applicable only within Example IV)) for the synthesis of compounds of the present invention.
Scheme 1
Figure imgf000080_0002
Figure imgf000080_0003
Scheme 2
Figure imgf000080_0004
2a Ar= 4-Pyridyl, R = 4-OMe
ArCHO KOH, DMF
Figure imgf000080_0005
Figure imgf000080_0006
2b Scheme 3 (General Synthesis of imidazopyridine series)
2CI2 ight
Figure imgf000081_0001
INCORPORATION BY REFERENCE
The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. The following references are herein incorporated by reference in their entireties:
EQUIVALENTS
The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

What Is Claimed Is:
1. A com ound having Formula I:
Figure imgf000082_0001
, including pharmaceutically acceptable salts, solvates, and/or prodrugs thereof;
wherein each of Rl . R2. R3, R4, X, Y and Z. independently include any chemical moi that permits the resulting compound to inhibit DYRK1 A activity.
2. The compound of Claim 1, wherein each Rl, R2, R3, R4, X, Y and Z, independently include any chemical moiety that permits the resulting compound to inhibit one or more of:
DYR 1A related PI3K/Akt signaling;
DYRK1A related tau phosphorylation;
DYRK1 A related NFAT phosphorylation;
DYRK1A related ASK1/JNK1 pathway activation;
DYRK1A related p53 phosphorylation;
DYR 1A related Amph 1 phosphorylation;
DYRK1A related Dynamin 1 phosphorylation;
DYRK1A related Synaptojanin phosphorylation;
DYRK1A related presenillin 1 (the catalytic sub-unit of γ-secretase) activity;
DYRK1A related Amyloid precursor protein phosphorylation; and
DYR 1A related SIRT1 activation.
The compound of Claim 1, wherein X-Y-Z is either:
Figure imgf000082_0002
Figure imgf000082_0003
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
5. The compound of Claim 4, wherein R5 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic
moiety comprising ether functionality, methyl, ethyl, (0¾)3,
Figure imgf000085_0002
Figure imgf000085_0003
X, Y,
Figure imgf000086_0001
6. The compound of Claim 4, wherein R6 is selected from hydrogen, C1 -C4 alkyl, heterocyclyl alkyl, heteroaryl alkyl, aryl alkyl, aryl, heterocyclyl, and heteroaryl.
7. The compound of Claim 4, wherein R7 is selected from hydrogen, alkoxy, alkylsulfonyl, cyano, carboxy, ester, amido, substituted amido, sulfonamide, substituted sulfonamide, methylenedioxy, heterocyclyl alkyl, heterocyclyl, and heterocyclyl alkyl amido, a lipophilic
moiety comprising ether functionality, methyl, ethyl, (CH2)3,
Figure imgf000086_0002
Figure imgf000086_0003
8. The compound of Claim 1, wherein each of R2 and R3 is independently selected from hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl,
Figure imgf000087_0001
Figure imgf000087_0002
Figure imgf000088_0001
11. The compound of Claim 1 having Formula III:
Figure imgf000089_0001
, including pharmaceutically acceptable salts, solvates, and/or prodrugs thereof. 12. The compound of Claim 1, wherein said compound is selected any of the compounds recited in Examples II and III.
13. A pharmaceutical composition comprising a compound of Claim 1. 14. A method of treating, ameliorating, or preventing a disorder related to DYRK1A activity in a patient comprising administering to said patient a therapeutically effective amount of the pharmaceutical composition of Claim 13.
15. The method of Claim 14, wherein said disorder related to DYRK1A activity is
Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, an autoimmune disease, an inflammatory disorder (e.g., airway inflammation), or cancer (e.g., glioblastoma).
16. The method of Claim 15, wherein said patient is a human patient.
17. The method of Claim 15, further comprising administering to said patient one or more agents for treating Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune disease, an inflammatory disorder (e.g., airway inflammation), or cancer (e.g., glioblastoma).
18. A kit comprising a compound of Claim 1 and instructions for administering said compound to a patient having a disorder related to DYRK1 activity.
19. The kit of Claim 18, wherein the disorder related to DYRK1 activity is Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune disease, inflammatory disorder (e.g., airway inflammation), or cancer (e.g., glioblastoma).
20. The kit of Claim 18, further comprising one or more agents for treating Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune disease, an inflammatory disorder (e.g., airway inflammation), or cancer (e.g., glioblastoma).
21. A method for inhibiting DYRK1A related activity in a subject, comprising administering to the subject a compound of Claim 1.
22. The method of Claim 21, wherein administration of the compound results in inhibition of one or more DYRK1A related activities in the subject:
DYRK1A related PBK/Akt signaling;
DYRK1A related tau phosphorylation;
DYRK1 A related NFAT phosphorylation;
DYRK1A related ASK1/JNK1 pathway activation;
DYRK1A related p53 phosphorylation;
DYRK1A related Amph 1 phosphorylation;
DYRK1A related Dynamin 1 phosphorylation;
DYRK1A related Synaptojanin phosphorylation;
DYRK1A related presenilin 1 (the catalytic sub-unit of γ-secretase) activity;
DYRK1A related Amyloid precursor protein phosphorylation; and
DYRK1A related SIRT1 activation.
23. The method of Claim 21, wherein the subject is human subject suffering from or at risk for developing a disorder related to DYRK1A activity.
24. The method of Claim 23, wherein the disorder related to DYRK1 A activity is
Alzheimer's disease, Down syndrome, Huntington's disease, Parkinson's disease, autoimmune disease, an inflammatory disorder (e.g., airway inflammation), or cancer (e.g., glioblastoma).
PCT/US2016/050198 2015-09-03 2016-09-02 Small molecule inhibitors of dyrk1a and uses thereof WO2017040993A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP16843107.0A EP3344039A4 (en) 2015-09-03 2016-09-02 Small molecule inhibitors of dyrk1a and uses thereof
US15/756,917 US10730842B2 (en) 2015-09-03 2016-09-02 Small molecule inhibitors of DYRK1A and uses thereof
JP2018531299A JP2018526452A (en) 2015-09-03 2016-09-02 DYRK1A small molecule inhibitor and use thereof
MX2018002631A MX2018002631A (en) 2015-09-03 2016-09-02 Small molecule inhibitors of dyrk1a and uses thereof.
AU2016315881A AU2016315881B2 (en) 2015-09-03 2016-09-02 Small molecule inhibitors of DYRK1A and uses thereof
CA2997556A CA2997556C (en) 2015-09-03 2016-09-02 Small molecule inhibitors of dyrk1a and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562213904P 2015-09-03 2015-09-03
US62/213,904 2015-09-03

Publications (2)

Publication Number Publication Date
WO2017040993A1 true WO2017040993A1 (en) 2017-03-09
WO2017040993A4 WO2017040993A4 (en) 2017-05-26

Family

ID=58188489

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/050198 WO2017040993A1 (en) 2015-09-03 2016-09-02 Small molecule inhibitors of dyrk1a and uses thereof

Country Status (7)

Country Link
US (1) US10730842B2 (en)
EP (1) EP3344039A4 (en)
JP (1) JP2018526452A (en)
AU (1) AU2016315881B2 (en)
CA (1) CA2997556C (en)
MX (1) MX2018002631A (en)
WO (1) WO2017040993A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019031990A1 (en) * 2017-08-07 2019-02-14 Закрытое Акционерное Общество "Биокад" Novel heterocyclic compounds as cdk8/19 inhibitors
WO2019105886A1 (en) * 2017-12-02 2019-06-06 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
WO2020069418A1 (en) * 2018-09-28 2020-04-02 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of dyrk1/clk and uses thereof
WO2020106685A1 (en) 2018-11-19 2020-05-28 Dana-Farber Cancer Institute, Inc. Macrocyclic inhibitors of dyrk1a
WO2020142486A1 (en) * 2018-12-31 2020-07-09 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
RU2739489C2 (en) * 2018-03-06 2020-12-24 Закрытое Акционерное Общество "Биокад" Novel heterocyclic compounds as cdk8/19 inhibitors
JP2021503486A (en) * 2017-11-20 2021-02-12 アイカーン スクール オブ メディシン アット マウント サイナイ Kinase Inhibitor Compounds and Compositions and Usage
WO2021096314A1 (en) * 2019-11-15 2021-05-20 가천대학교 산학협력단 Novel benzimidazole derivative and use thereof
WO2021126923A1 (en) * 2019-12-19 2021-06-24 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Cd206 modulators their use and methods for preparation
US11352328B2 (en) 2016-07-12 2022-06-07 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus
RU2782375C2 (en) * 2017-12-02 2022-10-26 Галапагос Нв New compounds and their pharmaceutical compositions for treatment of diseases
WO2023064366A1 (en) * 2021-10-12 2023-04-20 Biosplice Therapeutics, Inc. Pyrrolo[2,1-f][1,2,4]triazines derivatives as inhibitors of dyrk1a
WO2023107722A1 (en) * 2021-12-10 2023-06-15 Prothena Biosciences Limited Compounds for use in treating neurological disorders
WO2023107723A3 (en) * 2021-12-10 2023-07-20 Prothena Biosciences Limited Cyclic compounds and their use for the treatment of neurological disorders
US11788064B2 (en) 2018-01-05 2023-10-17 Icahn School Of Medicine At Mount Sinai Method of increasing proliferation of pancreatic beta cells, treatment method, and composition
US11866427B2 (en) 2018-03-20 2024-01-09 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114466840A (en) * 2019-06-12 2022-05-10 威斯达研究所 acetyl-CoA synthetase 2(ACSS2) inhibitors and methods of use thereof
WO2022051291A2 (en) * 2020-09-02 2022-03-10 Ankh Life Sciences Limited Inhibition of dyrk1a kinase
CA3229864A1 (en) * 2021-09-13 2023-03-16 Provention Bio, Inc. Methods and compositions comprising anti-cd3 antibodies and dyrk1a inhibitors for treating diabetes

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6218388B1 (en) * 1997-08-20 2001-04-17 Warner-Lambert Company Benzimidazoles for inhibiting protein tyrosine kinase mediated cellular proliferation
US20050026960A1 (en) * 2003-07-30 2005-02-03 Agouron Pharmaceuticals, Inc. 3, 5 disubstituted indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation
US7666879B2 (en) * 2005-02-24 2010-02-23 Eli Lilly And Company Imidazo[1,2-A]pyridine compounds as VEGF-R2 inhibitors
WO2013147711A1 (en) * 2012-03-30 2013-10-03 Agency For Science, Technology And Research Bicyclic heterocyclic derivatives as mnk1 and mnk2 modulators and uses thereof
WO2014096388A2 (en) * 2012-12-21 2014-06-26 Selvita S.A. Novel benzimidazole derivatives as kinase inhibitors

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4331671A (en) 1981-01-09 1982-05-25 Sterling Drug Inc. 5-(Pyridinyl)-1H-benzimidazoles and 1-hydroxy-6-(pyridinyl)-1H-benzimidazoles and their cardiotonic use
EP1017682A4 (en) * 1997-09-26 2000-11-08 Merck & Co Inc Novel angiogenesis inhibitors
DE60109148T2 (en) * 2000-04-28 2006-01-05 Acadia Pharmaceuticals Inc., San Diego muscarinic receptors
PL366690A1 (en) * 2004-03-29 2005-10-03 Fundacja Rozwoju Diagnostyki I Terapii New derivatives of 4,5,6,7-tetrabromobenzimidazole and method for their obtaining
PE20081152A1 (en) 2006-10-06 2008-08-10 Wyeth Corp N-SUBSTITUTED AZACYCLYLAMINES AS HISTAMINE-3 ANTAGONISTS
JP2011528338A (en) * 2008-07-18 2011-11-17 サノフイ−アベンテイス Novel imidazo [1,2-a] pyridine derivatives, process for their preparation, their use as pharmaceuticals, pharmaceutical compositions, and new uses, especially as MET inhibitors
KR101685718B1 (en) 2008-12-19 2016-12-12 제넨테크, 인크. Heterocyclic compounds and methods of use
JP5819831B2 (en) * 2009-08-17 2015-11-24 インテリカイン, エルエルシー Heterocyclic compounds and their use
US20110230481A2 (en) * 2009-11-12 2011-09-22 Selvita S.A. Derivatives of tetrabromobenzimidazole, a process for the preparation thereof, a pharmaceutical composition comprising the same, a method of using the same, a method for modulating or regulating serine/threonine kinases, and serine/threonine kinases modulating agent
WO2012174312A2 (en) * 2011-06-15 2012-12-20 Glaxosmithkline Llc Benzimidazole derivatives as antiviral agents
AU2012298510B2 (en) * 2011-08-19 2016-10-27 Diaxonhit DYRK1 inhibitors and uses thereof
JP5947465B2 (en) * 2012-10-01 2016-07-06 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Benzimidazoles as CNS activators
CA2906542A1 (en) 2013-03-15 2014-09-25 Intellikine, Llc Combination of kinase inhibitors and uses thereof
GB2519623A (en) 2013-06-18 2015-04-29 4Sc Discovery Gmbh Method of inhibiting DYRK1B
CN104447567B (en) 2014-11-02 2016-06-29 湖南华腾制药有限公司 A kind of preparation method of 1 substituted benzimidazole derivant

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6218388B1 (en) * 1997-08-20 2001-04-17 Warner-Lambert Company Benzimidazoles for inhibiting protein tyrosine kinase mediated cellular proliferation
US20050026960A1 (en) * 2003-07-30 2005-02-03 Agouron Pharmaceuticals, Inc. 3, 5 disubstituted indazole compounds, pharmaceutical compositions, and methods for mediating or inhibiting cell proliferation
US7666879B2 (en) * 2005-02-24 2010-02-23 Eli Lilly And Company Imidazo[1,2-A]pyridine compounds as VEGF-R2 inhibitors
WO2013147711A1 (en) * 2012-03-30 2013-10-03 Agency For Science, Technology And Research Bicyclic heterocyclic derivatives as mnk1 and mnk2 modulators and uses thereof
WO2014096388A2 (en) * 2012-12-21 2014-06-26 Selvita S.A. Novel benzimidazole derivatives as kinase inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BECKER ET AL.: "Activation, regulation, and inhibition of DYRK1A", FEBS JOURNAL., vol. 278, 3 December 2011 (2011-12-03), pages 246 - 256, XP055053004 *
LONESCU ET AL.: "DYRK1A Kinase Inhibitors with Emphasis on Cancer", MINI REVIEWS IN MEDICINE CHEMISTRY., vol. 12, September 2012 (2012-09-01), pages 1 - 14, XP009180029 *
See also references of EP3344039A4 *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11352328B2 (en) 2016-07-12 2022-06-07 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus
CN111670183A (en) * 2017-08-07 2020-09-15 Biocad股份公司 Novel heterocyclic compounds as CDK8/19 inhibitors
AU2018312836B2 (en) * 2017-08-07 2022-12-08 Joint Stock Company "Biocad" Novel heterocyclic compounds as CDK8/19 inhibitors
JP7365332B2 (en) 2017-08-07 2023-10-19 ジョイント・ストック・カンパニー “バイオキャド” Novel heterocyclic compounds as CDK8/19 inhibitors
WO2019031990A1 (en) * 2017-08-07 2019-02-14 Закрытое Акционерное Общество "Биокад" Novel heterocyclic compounds as cdk8/19 inhibitors
JP2020530020A (en) * 2017-08-07 2020-10-15 ジョイント・ストック・カンパニー “バイオキャド” A novel heterocyclic compound as a CDK8 / 19 inhibitor
US11547712B2 (en) 2017-11-20 2023-01-10 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
JP2021503486A (en) * 2017-11-20 2021-02-12 アイカーン スクール オブ メディシン アット マウント サイナイ Kinase Inhibitor Compounds and Compositions and Usage
CN111433200A (en) * 2017-12-02 2020-07-17 加拉帕戈斯股份有限公司 Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
JP2021505553A (en) * 2017-12-02 2021-02-18 ガラパゴス・ナムローゼ・フェンノートシャップGalapagos N.V. New compounds and their pharmaceutical compositions for the treatment of diseases
JP7326276B2 (en) 2017-12-02 2023-08-15 ガラパゴス・ナムローゼ・フェンノートシャップ Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
CN111433200B (en) * 2017-12-02 2024-03-22 加拉帕戈斯股份有限公司 Compounds and pharmaceutical compositions for the treatment of diseases
WO2019105886A1 (en) * 2017-12-02 2019-06-06 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
RU2782375C2 (en) * 2017-12-02 2022-10-26 Галапагос Нв New compounds and their pharmaceutical compositions for treatment of diseases
AU2018374456B2 (en) * 2017-12-02 2022-07-07 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of diseases
US11339166B2 (en) 2017-12-02 2022-05-24 Galapagos Nv Compounds and pharmaceutical compositions thereof for the treatment of diseases
US11788064B2 (en) 2018-01-05 2023-10-17 Icahn School Of Medicine At Mount Sinai Method of increasing proliferation of pancreatic beta cells, treatment method, and composition
RU2739489C2 (en) * 2018-03-06 2020-12-24 Закрытое Акционерное Общество "Биокад" Novel heterocyclic compounds as cdk8/19 inhibitors
US11866427B2 (en) 2018-03-20 2024-01-09 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
US20220041590A1 (en) * 2018-09-28 2022-02-10 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of dyrk/clk and uses thereof
CN113164476A (en) * 2018-09-28 2021-07-23 代表亚利桑那大学的亚利桑那校董会 Small molecule inhibitor of DYRK1/CLK and application thereof
WO2020069418A1 (en) * 2018-09-28 2020-04-02 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of dyrk1/clk and uses thereof
EP3856186A4 (en) * 2018-09-28 2022-07-06 Arizona Board of Regents on behalf of the University of Arizona Small molecule inhibitors of dyrk1/clk and uses thereof
WO2020106685A1 (en) 2018-11-19 2020-05-28 Dana-Farber Cancer Institute, Inc. Macrocyclic inhibitors of dyrk1a
EP3906028A4 (en) * 2018-12-31 2022-10-12 Icahn School of Medicine at Mount Sinai Kinase inhibitor compounds and compositions and methods of use
CN113543783A (en) * 2018-12-31 2021-10-22 西奈山伊坎医学院 Kinase inhibitor compounds and compositions and methods of use
WO2020142486A1 (en) * 2018-12-31 2020-07-09 Icahn School Of Medicine At Mount Sinai Kinase inhibitor compounds and compositions and methods of use
WO2021096314A1 (en) * 2019-11-15 2021-05-20 가천대학교 산학협력단 Novel benzimidazole derivative and use thereof
CN115768757A (en) * 2019-12-19 2023-03-07 ***合众国,由健康及人类服务部部长代表 CD206 modulators and uses and methods of making thereof
WO2021126923A1 (en) * 2019-12-19 2021-06-24 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Cd206 modulators their use and methods for preparation
WO2023064366A1 (en) * 2021-10-12 2023-04-20 Biosplice Therapeutics, Inc. Pyrrolo[2,1-f][1,2,4]triazines derivatives as inhibitors of dyrk1a
WO2023107723A3 (en) * 2021-12-10 2023-07-20 Prothena Biosciences Limited Cyclic compounds and their use for the treatment of neurological disorders
WO2023107722A1 (en) * 2021-12-10 2023-06-15 Prothena Biosciences Limited Compounds for use in treating neurological disorders

Also Published As

Publication number Publication date
CA2997556C (en) 2020-12-22
AU2016315881A1 (en) 2018-03-29
MX2018002631A (en) 2019-02-07
JP2018526452A (en) 2018-09-13
EP3344039A4 (en) 2019-08-28
CA2997556A1 (en) 2017-03-09
US10730842B2 (en) 2020-08-04
US20190062284A1 (en) 2019-02-28
EP3344039A1 (en) 2018-07-11
AU2016315881B2 (en) 2019-09-19
WO2017040993A4 (en) 2017-05-26

Similar Documents

Publication Publication Date Title
AU2016315881B2 (en) Small molecule inhibitors of DYRK1A and uses thereof
JP6982343B2 (en) Therapeutic inhibitor compound
JP6768857B2 (en) Lysine-specific inhibitor of demethylase-1
KR102400920B1 (en) Inhibitors of lysine specific demethylase-1
RU2637936C2 (en) Inhibitors of lrrk2 kinases activity
KR102534962B1 (en) 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6H)-ketone compound
EP3318561B1 (en) Heteroaryl compounds and methods of use thereof
TWI527800B (en) 1-(arylmethyl)quinazoline-2,4(1h,3h)-diones as parp inhibitors and the use thereof
EP2909193B1 (en) Phenyl linked quinolinyl modulators of ror-gamma-t
CA2982588A1 (en) Bromodomain inhibitors
EP3394052B1 (en) Bicyclic hydroxamic acids useful as inhibitors of mammalian histone deacetylase activity
US10287293B2 (en) Bicyclic heterocyclic compounds as PDE2 inhibitors
CN103570625A (en) N-(3-aryl-heteroaryl)-4-aryl-aryl carboxamide and analog as hedgehog pathway inhibitors and application thereof
Garofalo et al. Selective inhibitors of G2019S-LRRK2 kinase activity
DK2817302T3 (en) 1H-indazole-3-carboxamide compounds as glycogen synthase kinase-3 beta inhibitors
JP2018516278A (en) Use of pteridinone derivatives as EGFR inhibitors
AU2015276699A1 (en) Pyridino[1,2-a]pyrimidone analogue used as PI3K inhibitor
EP3476846A1 (en) Novel heterocyclic derivative compound and use thereof
Gao et al. Discovery and anti-inflammatory evaluation of benzothiazepinones (BTZs) as novel non-ATP competitive inhibitors of glycogen synthase kinase-3β (GSK-3β)
Liu et al. Discovery of novel β-carboline-1, 2, 3-triazole hybrids as AChE/GSK-3β dual inhibitors for Alzheimer's disease treatment
KR20150021120A (en) Heteroaryl compounds and methods of use thereof
CN114133394B (en) Compound selectively aiming at activity of cell cycle dependent kinase 12, preparation method and medical application
US20170305913A1 (en) Substituted pyrazoloquinazolinones and pyrroloquinazolinones as allosteric modulators of group ii metabotropic glutamate receptors
WO2008022747A1 (en) Tricyclic lactam derivatives, their manufacture and use as pharmaceutical agents
EP2825166A1 (en) Method of treating ophthalmic conditions with kinase inhibitors

Legal Events

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

Ref document number: 16843107

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: MX/A/2018/002631

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2997556

Country of ref document: CA

Ref document number: 2018531299

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016315881

Country of ref document: AU

Date of ref document: 20160902

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2016843107

Country of ref document: EP