US20090018112A1 - Compounds and Uses Thereof - Google Patents

Compounds and Uses Thereof Download PDF

Info

Publication number
US20090018112A1
US20090018112A1 US12/199,034 US19903408A US2009018112A1 US 20090018112 A1 US20090018112 A1 US 20090018112A1 US 19903408 A US19903408 A US 19903408A US 2009018112 A1 US2009018112 A1 US 2009018112A1
Authority
US
United States
Prior art keywords
alkyl
arylalkyl
amino
carboxamide
disorder
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/199,034
Inventor
Marc J. Chapdelaine
Cyrus J. Ohnmacht
Christopher Becker
Hui-Fang Chang
Bruce T. Dembofsky
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AstraZeneca AB
Original Assignee
AstraZeneca AB
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 AstraZeneca AB filed Critical AstraZeneca AB
Priority to US12/199,034 priority Critical patent/US20090018112A1/en
Publication of US20090018112A1 publication Critical patent/US20090018112A1/en
Priority to US13/031,763 priority patent/US20110144331A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/28Cinnolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • 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
    • C07D403/10Heterocyclic 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 linked by a carbon chain containing aromatic rings
    • 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
    • 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

Definitions

  • the present invention relates to novel cinnoline compounds, their pharmaceutical compositions, methods of use and processes to make such compounds.
  • the present invention relates to therapeutic methods for the treatment and/or prevention of anxiety disorders, cognitive disorders, and/or mood disorders.
  • the present invention comprises, inter alia, cinnoline compounds, their use as central nervous system (CNS) depressants (especially anxiolytics), and pharmacological tools, methods for their preparation, pharmaceutical compositions containing the same, and intermediates used in their preparation.
  • CNS central nervous system
  • Some cinnoline compounds including selected 4-amino- and 4-oxo-cinnoline-3-carboxamides are disclosed in East German Patent 123525 (Verfahren zur compassion von substitu believing 4-Aminocinnolinen); U.S. Pat. No. 4,379,929 to Conrad et al; U.S. Pat. Nos. 4,886,800 and 4,925,844 to Resch; Daunis et al., “Preparation et proprietes de cinnolones-3 et cinnolones-4,” Bull. de la Societe Chimique de France, 8:3198-3202 (1972); Lunt et al. “A New Cinnoline Synthesis,” J. Chem.
  • GABA gamma-Aminobutyric acid
  • GABAA GABA type A receptors
  • GABAB GABA type B receptors
  • GABAC GABA type C receptors
  • GABAA receptors function as ligand-gated ion channels to mediate fast inhibitory synaptic transmissions that regulate neuronal excitability involved in such responses as seizure threshold, skeletal muscle tone, and emotional status.
  • GABAA receptors are targets of many sedating drugs, such as benzodiazepines, barbiturates and neurosteroids.
  • GABAA receptors are pentameric, ligand-gated chloride ion (Cl ⁇ ) channels belonging to a superfamily of ligand-gated ionotropic receptors that includes the nicotinic acetylcholine receptor. GABAA receptors are very heterogeneous, with at least 16 different subunits producing potentially thousands of different receptor types.
  • GABAA receptor subunits aggregate into complexes that form chloride ion selective channels and contain sites that bind GABA along with a variety of pharmacologically active substances.
  • the anion channel is activated, causing it to open and allowing chloride ions (Cl ⁇ ) to enter the neuron.
  • This influx of Cl ⁇ ions hyperpolarizes the neuron, making it less excitable.
  • the resultant decrease in neuronal activity following activation of the GABAA receptor complex can rapidly alter brain function, to such an extent that consciousness and motor control may be impaired.
  • GABAA receptor subunits and the widespread distribution of these receptors in the nervous system likely contributes to the diverse and variable physiological functions of GABAA receptors, which have been implicated in many neurological and psychiatric disorders, and related conditions, including: stroke, head trauma, epilepsy, pain, migraine, mood disorders, anxiety, post traumatic stress disorder, obsessive compulsive disorders, schizophrenia, seizures, convulsions, tinnitus, neurodegenerative disorders including Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's Chorea, Parkinson's disease, depression, bipolar disorders, mania, trigeminal and other neuralgia, neuropathic pain, hypertension, cerebral ischemia, cardiac arrhythmia, myotonia, substance abuse, myoclonus, essential tremor, dyskinesia and other movement disorders, neonatal cerebral hemorrhage, and spasticity. GABAA receptors are also believed to play a role in cognition, consciousness, and sleep.
  • Benzodiazepines such as diazepam, chlordiazepoxide and midazolam.
  • Barbiturates can directly activate GABAA receptors, significantly increasing Cl ⁇ currents in the absence of further intervention by GABA itself and can also indirectly augment GABAergic neural transmission.
  • benzodiazepines act as indirect allosteric modulators, and are largely incapable of increasing Cl ⁇ Currents in the absence of GABA, but enhance GABA-activated increases in Cl ⁇ conductance.
  • This latter property is thought to be responsible for the usefulness of benzodiazepines for treating a number of disorders, including generalized anxiety disorder, panic disorder, seizures, movement disorders, epilepsy, psychosis, mood disorders, and muscle spasms, as well as the relative safety of benzodiazepines compared to barbiturates.
  • barbiturates and benzodiazepines are addictive and can cause drowsiness, poor concentration, ataxia, dysarthria, motor incoordination, diplopia, muscle weakness, vertigo and mental confusion. These side effects can interfere with an individual's ability to perform daily routines such as driving, operating heavy machinery or performing other complex motor tasks while under therapy, making barbiturates and benzodiazepines less than optimal for treating chronic disorders involving GABA and GABAA receptors.
  • GABAA receptors and GABAergic neural transmissions are implicated as targets for therapeutic intervention in a myriad of neurological and psychiatric disorders.
  • the present invention is also, inter alia, directed toward this end.
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R 7 ;
  • R 2 is H, C( ⁇ O)R b , C( ⁇ O)NR c R d , C( ⁇ O)OR a , S( ⁇ O) 2 R b , C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R 8 ;
  • R 3 , R 4 and R 5 are each, independently, H, halo, Si(C 1-10 alkyl) 3 , CN, NO 2 , OR a , SR a , OC( ⁇ O)R a , OC( ⁇ O)OR b , OC( ⁇ O)NR c R d , C( ⁇ O)R a , C( ⁇ O)OR b , C( ⁇ O)NR c R d , NR c NR d , NR c C( ⁇ O)R a , NR c C( ⁇ O)OR b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R a , S( ⁇ O)NR c R d , S( ⁇ O 2 R a , S( ⁇ O) 2 NR c R d , C( ⁇ O 2 R a , S( ⁇ O) 2 NR c R d , C 1-6 alkyl, C
  • R 6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 ;
  • R 7 , R 8 and R 9 are each, independently, halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OR a′ , SR a′ , C( ⁇ O)R b′ , C( ⁇ O)NR c′ R d′ , C( ⁇ O)OR a′ , OC( ⁇ O)R b′ , OC( ⁇ O)NR c′ R d′ , NR c′ R d′ , NR c′C( ⁇ O)R b′ , NR c′ C( ⁇ O)OR a′ , NR c′ S( ⁇ O) 2 R b′ , S( ⁇ O)R b′ , S( ⁇ O)NR c′ R d′ , S( ⁇ O) 2 R b′ , or S( ⁇ O) 2 NR c′ R
  • a 1 is halo, CN, NO 2 , OR a , SR a , C( ⁇ O)R b , C( ⁇ O)NR c R d C( ⁇ O)OR a , OC( ⁇ O) b , OC( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R d , NR c C( ⁇ O)OR a , NR c S( ⁇ O)R b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R b , S( ⁇ O)NR c R d , S( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialky
  • R a and R a′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cyclo
  • R b and R b′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl,
  • R c and R d are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl, hetero
  • R c′ and R d′ are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl
  • R c′ and R d′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R 7 .
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SR a′ , C( ⁇ O)R b′ , C( ⁇ O)NR c′ R d′ , C( ⁇ O)OR a′ , OC( ⁇ O)R b′ , OC( ⁇ O)NR c′ R d′ , NR c′ C( ⁇ O)R b′ ,
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 ,
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C(C(C
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl.
  • R 1 is C 1-6 alkyl.
  • R 1 is n-propyl
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substitu
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , or C 1-6 alkyl.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , or C 1-3 alkyl.
  • R 2 is H.
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OR a , SR a , OC( ⁇ O)R a , OC( ⁇ O)OR b , OC( ⁇ O)NR c R d , C( ⁇ O)R a , C( ⁇ O)OR b , C( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R a , NR c C( ⁇ O)O, NR c S( ⁇ O) 2 R b , S( ⁇ O)NR c R d , S( ⁇ O) 2 NR c R d , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroaryl
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O)—(C 1-4 alkyl), OC( ⁇ O)-(arylalkyl), OC( ⁇ O)NH 2 , OC( ⁇ O)NH(C 1-4 alkyl),
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O)—(C 1-4 alkyl), NHC( ⁇ O)—(C 1-4 alkyl), NHC( ⁇ O)O-(arylalkyl), NHC( ⁇ O)O—(C 1-4 alkyl), NHC( ⁇ O)O—
  • R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, halo, C 1-6 alkyl or C 1-6 haloalkyl.
  • R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, halo or C 1-3 haloalkyl.
  • R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, or halo.
  • R 6 is aryl or heteroaryl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is aryl optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is aryl substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is heteroaryl optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, 2-naphthyl, 3-pyridyl, 4-pyridyl, pyrimidin-5-yl, pyrazin-2-yl, pyrazol-3-yl, pyrazol-4-yl, 3-quinolyl, 6-quinolyl, or indol-5-yl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)( ⁇
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group
  • R c′ and R d′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H,
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alky
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 or C 1-6 alkyl;
  • R 5 is H, C 1-4 alkoxy, halo, C 1-6 alkyl or C 1-6 haloalkyl;
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is other than unsubstituted phenyl.
  • R 1 is C 1-6 alkyl.
  • R 1 is n-propyl
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl) or C 1-6 alkyl.
  • R 2 is H.
  • R 5 is H, C 1-4 alkoxy or halo.
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 1, 2 or 3 C 1-4 alkoxy or C 1-4 alkyl.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 2 C 1-4 alkoxy or C 1-4 alkyl.
  • R 1 is n-propyl and R 2 is H.
  • compositions comprising a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • the present invention further provides methods of treating or preventing an anxiety disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • the present invention further provides methods of treating or preventing a cognitive disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • the present invention further provides methods of treating or preventing a mood disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • the present invention further provides a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, described herein for use as a medicament.
  • the present invention further provides a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, described herein for the manufacture of a medicament.
  • the present invention further provides methods of modulating activity of GABAA receptor comprising contacting the GABAA receptor with a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • the present invention further provides synthetic methods of making a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R 7 ;
  • R 2 is H, C( ⁇ O)R b , C( ⁇ O)NR c R d , C( ⁇ O)OR a , S( ⁇ O) 2 R b , C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R 8 ;
  • R 3 , R 4 and R 5 are each, independently, H, halo, Si(C 1-10 alkyl) 3 , CN, NO 2 , OR a , SR a , OC( ⁇ O)R a , OC( ⁇ O)OR b , OC( ⁇ O)NR c R d , C( ⁇ O)R a , C( ⁇ O)OR b , C( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R a , NR c C( ⁇ O)OR b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R a , S( ⁇ O)NR c R d , S( ⁇ O) 2 NR c R d , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl,
  • R 6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 ;
  • R 7 , R 5 and R 9 are each, independently, halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OR a , SR a′ , C( ⁇ O)R b′ , C( ⁇ O)NR c′ R d′ , C( ⁇ O)OR a′ , OC( ⁇ O)R b′ , OC( ⁇ O)NR c′ R d′ , NR c′ R d′ , NR c′ C( ⁇ O)R b′ , NR c′ C( ⁇ O)OR a′ , NR c′ S( ⁇ O) 2 R b′ , S( ⁇ O)R b′ , S( ⁇ O)NR c′ R d′ , S( ⁇ O) 2 R b′ , or S( ⁇ O) 2 NR c′ R d
  • a 1 is halo, CN, NO 2 , OR a , SR a , C( ⁇ O)R b , C( ⁇ O)NR c R d , C( ⁇ O)OR a , OC( ⁇ O)R b , OC( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R d , NR c C( ⁇ O)OR a , NR c S( ⁇ O)R b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R b , S( ⁇ O)NR c R d , S( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2
  • R a and R a′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cyclo
  • R b and R b′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl,
  • R c and R d are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl, hetero
  • R c′ and R d′ are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl
  • R c′ and R d′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R 7 , or any subgroup thereof.
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R 7 .
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SR a′ , C( ⁇ O)R b′ , C( ⁇ O)NR c′ R d′ , C( ⁇ O)OR a′ , OC( ⁇ O)R b′ , OC( ⁇ O)NR c′ R d′ , NR c′ C( ⁇ O)R b′ ,
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 ,
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C(C(C
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C
  • R 1 is C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R 1 is C 1-6 alkyl or C 1-6 haloalkyl. In some embodiments, R 1 is C 1-6 alkyl. In some embodiments, R 1 is n-propyl.
  • R 2 is H, C( ⁇ O)R b , C( ⁇ O)NR c R d , C( ⁇ O)OR a , S( ⁇ O) 2 R b , C 1-6 alkyl, C 1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein each of the C 1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R 8 , or any subgroup thereof.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C 1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substitu
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C 1-6 alkyl, C 1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 or C 1-6 alkyl.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , or C 1-3 alkyl. In some embodiments, R 2 is H.
  • R 3 , R 4 and R 5 are each, independently, H, halo, Si(C 1-10 alkyl) 3 , CN, NO 2 , OR a , SR a , OC( ⁇ O)R a , OC( ⁇ O)OR b , OC( ⁇ O)NR c R d , C( ⁇ O)R 3 , C( ⁇ O)OR b , C( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R a , NR c C( ⁇ O)OR b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R a , S( ⁇ O)NR c R d , S( ⁇ O) 2 R a , S( ⁇ O) 2 NR c R d , C( ⁇ O) 2 R a , S( ⁇ O) 2 NR c R d , C 1-6 alkyl, C
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OR 3 , SR 3 , OC( ⁇ O)R a , OC( ⁇ O)OR b , OC( ⁇ O)NR c R d , C( ⁇ O)R a , C( ⁇ O)OR b , C( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R a , NR c C( ⁇ O)OR b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R a , S( ⁇ O)NR c R d , S( ⁇ O) 2 R a , S( ⁇ O) 2 NR c R d , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O)—(C 1-4 alkyl), OC( ⁇ O)-(arylalkyl), OC( ⁇ O)NH 2 , OC( ⁇ O)NH(C 1-4 alkyl),
  • R 3 , R 4 and R 5 are each, independently, H, halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O)—(C 1-4 alkyl), NHC( ⁇ O)—(C 1-4 alkyl), NHC( ⁇ O)O-(arylalkyl), NHC( ⁇ O)O—(C 1-4 alkyl), NHC( ⁇ O)O—
  • R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, halo, C 1-6 alkyl or C 1-6 haloalkyl. In some embodiments, R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, halo or C 1-3 haloalkyl. In some embodiments, R 3 , R 4 and R 5 are each, independently, H, C 1-4 alkoxy, or halo.
  • R 6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, or any subgroup thereof, each optionally substituted by 1, 2, 3, 4 or 5 A 1 , or any subgroup thereof.
  • R 6 is aryl or heteroaryl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is aryl optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is aryl substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is heteroaryl optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, 2-naphthyl, 3-pyridyl, 4-pyridyl, pyrimidin-5-yl, pyrazin-2-yl, pyrazol-3-yl, pyrazol-4-yl, 3-quinolyl, 6-quinolyl, or indol-5-yl, each optionally substituted by 1, 2, 3, 4 or 5 A 1 .
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)( ⁇
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R 6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alky
  • R 7 , R 8 and R 9 are each, independently, halo, C 1-4 alkyl, C 1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO 2 , OR a′ , SR a′ , C( ⁇ O)R b′ , C( ⁇ O)NR c′ R d′ , C( ⁇ O)OR a′ , OC( ⁇ O)R b′ , OC( ⁇ O)NR c′ R d′ , NR c′ R d′ , NR c′ C( ⁇ O)R b′ , NR c′ C( ⁇ O)OR a′ , NR c′ S( ⁇ O) 2 R b′ , S( ⁇ O)R b′ , S( ⁇ O)NR c′ R d′ , S( ⁇ O) 2 R b′ , or S( ⁇ O) 2 NR
  • a 1 is halo, CN, NO 2 , OR a , SR a , C( ⁇ O)R b , C( ⁇ O)NR c R d , C( ⁇ O)OR a , OC( ⁇ O)R b , OC( ⁇ O)NR c R d , NR c R d , NR c C( ⁇ O)R d , NR c C( ⁇ O)OR a , NR c S( ⁇ O)R b , NR c S( ⁇ O) 2 R b , S( ⁇ O)R b , S( ⁇ O)NR c R d , S( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C( ⁇ O) 2 R b , S( ⁇ O) 2 NR c R d , C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2
  • R a and R a′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, aryl, arylalkyl, heteroaryl,
  • R b and R b′ are each, independently, H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 haloalkyl, aryl,
  • R c and R d are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 halo alkyl, C 1-6 halo alkyl, aryl, ary
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • R c′ and R d′ are each, independently, H, C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C 1-10 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C 1-6 alkyl, C 1-6 halo alkyl, C 1-6 halo alkyl, aryl,
  • R c′ and R d′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • R 6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO 2 , OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , SH, —S—(C 1-4 alkyl), C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)( ⁇
  • R c′ and R d′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H,
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R 6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alky
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 or C 1-6 alkyl;
  • R 5 is H, C 1-4 alkoxy, halo, C 1-6 alkyl or C 1-6 haloalkyl;
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC( ⁇ O)H, OC( ⁇ O
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • R 6 is other than unsubstituted phenyl.
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl, or any subgroup thereof.
  • R 1 is C 1-6 alkyl. In some embodiments, R 1 is n-propyl.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , or C 1-6 alkyl, or any subgroup thereof.
  • R 2 is H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl) or C 1-6 alkyl.
  • R 2 is H.
  • R 5 is H, C 1-4 alkoxy, halo, C 1-6 alkyl or C 1-6 haloalkyl, or any subgroup thereof. In some embodiments, R 5 is H, C 1-4 alkoxy or halo.
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, or any subgroup thereof, each optionally substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), OC(
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 1, 2 or 3 C 1-4 alkoxy or C 1-4 alkyl. In some embodiments, R 6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 2 C 1-4 alkoxy or C 1-4 alkyl.
  • R c and R d together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • R 6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, C 2-8 dialkylamino, NR c R d , C( ⁇ O)H, C( ⁇ O)—(C 1-4 alkyl), C( ⁇ O)-(arylalkyl), C( ⁇ O)NH 2 , C( ⁇ O)NH(C 1-4 alkyl), C( ⁇ O)N(C 1-4 alkyl) 2 , C( ⁇ O)NR c R d , C( ⁇ O)OH, C( ⁇ O)O—(C 1-4 alkyl), C( ⁇ O)O-(arylalkyl), S( ⁇ O) 2 —(C 1-4 alkyl), S( ⁇ O) 2 -(arylalkyl), S( ⁇ O) 2 NH(C 1-4 alkyl), S( ⁇ O) 2 NH(C 1-4 alky
  • R 1 is n-propyl and R 2 is H.
  • the present invention provides the following compounds:
  • the present invention provides the following compounds: 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide; 4-amino-8-(2,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide; 4-amino-8-(4-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide; and 4-amino-8-(2-methoxy-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide; or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • the present invention provides 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide, or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • the present invention provides the following compounds:
  • the present invention provides the following compounds:
  • Compounds of the present invention also include pharmaceutically acceptable salts, tautomers and in vivo-hydrolysable precursors of the compounds of any of the formulas described herein.
  • Compounds of the invention further include hydrates and solvates.
  • the present invention provides compounds of any of the formulas described herein, or pharmaceutically acceptable salts, tautomers or in vivo-hydrolysable precursors thereof, for use as medicaments.
  • the present invention provides compounds described herein for use as medicaments for treating or preventing an anxiety disorder, cognitive disorder, or mood disorder.
  • the present invention provides compounds of any of the formulas described herein, or pharmaceutically acceptable salts, tautomers or in vivo-hydrolysable precursors thereof, in the manufacture of a medicament for the treatment or prophylaxis of an anxiety disorder, cognitive disorder, or mood disorder.
  • the present invention provides a method for the treatment or prophylaxis of an anxiety disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof.
  • anxiety disorder includes, but is not limited to, one or more of the following: panic disorder, panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, social anxiety disorder, obsessive-compulsive disorder, posttraumatic stress disorder, acute stress disorder, generalized anxiety disorder, generalized anxiety disorder due to a general medical condition, and the like.
  • the present invention provides a method for the treatment or prophylaxis of a cognitive disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof.
  • a mammal including a human
  • a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof includes, but is not limited to, one or more of the following: Alzheimer's disease, dementia, dementia due to Alzheimer's disease, dementia due to Parkinson's disease, and the like.
  • the present invention provides a method for the treatment or prophylaxis of a mood disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof.
  • the phrase “mood disorder” is a depressive disorder including, but is not limited to, one or more of the following: major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, mixed episodes associated with bipolar disorder, and the like.
  • Anxiety disorders, cognitive disorders, and mood disorders are defined, for example, in the American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision, Washington, D.C., American Psychiatric Association, 2000.
  • the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including a human) a compound of any of the formulas described herein or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursors and a cognitive and/or memory enhancing agent.
  • the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including a human) a compound of any of the formulas described herein or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursors thereof wherein constituent members are provided herein, and a choline esterase inhibitor or anti-inflammatory agent.
  • the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including human) a compound of the present invention, and an atypical antipsychotic agent.
  • Atypical antipsychotic agents include, but not limited to, Olanzapine (marketed as Zyprexa), Aripiprazole (marketed as Abilify), Risperidone (marketed as Risperdal), Quetiapine (marketed as Seroquel), Clozapine (marketed as Clozaril), Ziprasidone (marketed as Geodon) and Olanzapine/Fluoxetine (marketed as Symbyax).
  • the mammal or human being treated with a compound of the present invention has been diagnosed with a particular disease or disorder, such as those described herein. In these cases, the mammal or human being treated is in need of such treatment. Diagnosis, however, need not be previously performed.
  • the present invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of the invention herein together with at least one pharmaceutically acceptable carrier, diluent or excipient.
  • compounds of the present invention When used for pharmaceutical compositions, medicaments, manufacture of a medicament, or treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), compounds of the present invention include the compounds of any of the formulas described herein, and pharmaceutically acceptable salts, tautomers and in vivo-hydrolysable precursors thereof. Compounds of the present invention further include hydrates and solvates.
  • substitution means that substitution is optional and therefore it is possible for the designated atom or moiety to be unsubstituted. In the event a substitution is desired then such substitution means that any number of hydrogens on the designated atom or moiety is replaced with a selection from the indicated group, provided that the normal valency of the designated atom or moiety is not exceeded, and that the substitution results in a stable compound. For example, if a methyl group (i.e., CH 3 ) is optionally substituted, then 3 hydrogens on the carbon atom can be replaced.
  • a methyl group i.e., CH 3
  • substituents include, but are not limited to: halogen, CN, NH 2 , OH, SO, SO 2 , COOH, OC 1-6 alkyl, CH 2 OH, SO 2 H, C 1-6 alkyl, OC 1-6 alkyl, C( ⁇ O)C 1-6 alkyl, C( ⁇ O)OC 1-6 alkyl, C( ⁇ O)NH 2 , C( ⁇ O)NHC 1-6 alkyl, C( ⁇ O)N(C 1-6 alkyl) 2 , SO 2 C 1-6 alkyl, SO 2 NHC 1-6 alkyl, SO 2 N(C 1-6 alkyl) 2 , NH(C 1-6 alkyl), N(C 1-6 alkyl) 2 , NHC( ⁇ O)C 1-6 alkyl, NC( ⁇ O)(C 1-6 alkyl) 2 , C 5-6 aryl, OC 5-6 aryl, C( ⁇ O)C 5-6 aryl, C( ⁇ O)OC 5-6 aryl
  • a variety of compounds in the present invention may exist in particular stereoisomeric forms.
  • the present invention takes into account all such compounds, including cis- and trans isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as being covered within the scope of this invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • the compounds herein described may have asymmetric centers. Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms.
  • optically active forms such as by resolution of racemic forms or by synthesis from optically active starting materials.
  • separation of the racemic material can be achieved by methods known in the art.
  • Many stereoisomers of olefins, C ⁇ N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention.
  • Cis and trans isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. All chiral, diastereomeric, racemic forms and all stereoisomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated.
  • the compounds of the invention may form isolable atropisomers in certain solvents (e.g. supercritical CO 2 containing 25-35% methanol) at room temperature.
  • the atropisomers of the compounds may be isolated using chiral LC. All atropisomers of a structure are intended, unless the specific atropisomer is specifically indicated.
  • C m-n or “C m-n group” used alone or as a prefix, refers to any group having m to n carbon atoms.
  • alkyl used alone or as a suffix or prefix, refers to a saturated monovalent straight or branched chain hydrocarbon radical comprising 1 to about 12 carbon atoms.
  • alkyls include, but are not limited to, C 1-6 alkyl groups, such as methyl, ethyl, propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, pentyl,
  • alkylene used alone or as suffix or prefix, refers to divalent straight or branched chain hydrocarbon radicals comprising 1 to about 12 carbon atoms, which serves to links two structures together.
  • alkenyl refers to an alkyl group having one or more double carbon-carbon bonds.
  • Example alkenyl groups include ethenyl, propenyl, cyclohexenyl, and the like.
  • alkenylenyl refers to a divalent linking alkenyl group.
  • alkynyl refers to an alkyl group having one or more triple carbon-carbon bonds.
  • Example alkynyl groups include ethynyl, propynyl, and the like.
  • alkynylenyl refers to a divalent linking alkynyl group.
  • aromatic refers to hydrocarbyl groups having one or more polyunsaturated carbon rings having aromatic characters, (e.g., 4n+2 delocalized electrons) and comprising up to about 14 carbon atoms.
  • aryl refers to an aromatic ring structure made up of from 5 to 14 carbon atoms. Ring structures containing 5, 6, 7 and 8 carbon atoms would be single-ring aromatic groups, for example, phenyl. Ring structures containing 8, 9, 10, 11, 12, 13, or 14 would be a polycyclic moiety in which at least one carbon is common to any two adjoining rings therein (for example, the rings are “fused rings”), for example naphthyl.
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, for example, the other cyclic rings can be cycloalkyls, cycloalkenyls or cycloalkynyls.
  • ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively.
  • the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
  • cycloalkyl refers to a saturated monovalent ring-containing hydrocarbon radical comprising at least 3 up to about 12 carbon atoms.
  • cycloalkyls include, but are not limited to, C 3-7 cycloalkyl groups, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl, and saturated cyclic and bicyclic terpenes.
  • a cycloalkyl can be unsubstituted or substituted by one or two suitable substituents.
  • the cycloalkyl is a monocyclic ring or bicyclic ring.
  • cycloalkenyl refers to ring-containing hydrocarbyl groups having at least one carbon-carbon double bond in the ring, and having from 3 to 12 carbons atoms.
  • halo or “halogen” refers to fluoro, chloro, bromo, and iodo.
  • Counterion is used to represent a small, negatively or positively charged species such as chloride (Cl ⁇ ), bromide (Br ⁇ ), hydroxide (OH ⁇ ), acetate (CH 3 COO ⁇ ), sulfate (SO 4 2 ⁇ ), tosylate (CH 3 -phenyl-SO 3 ⁇ ), benezensulfonate (phenyl-SO 3 —), sodium ion (Na + ), potassium (R + ), ammonium (NH 4 + ), and the like.
  • heterocycle used alone or as a suffix or prefix, refers to a ring-containing structure or molecule having one or more multivalent heteroatoms, independently selected from N, O, P and S, as a part of the ring structure and including at least 3 and up to about 20 atoms in the ring(s).
  • Heterocycle may be saturated or unsaturated, containing one or more double bonds, and heterocycle may contain more than one ring.
  • the rings may be fused or unfused.
  • Fused rings generally refer to at least two rings share two atoms therebetween.
  • Heterocycle may have aromatic character or may not have aromatic character.
  • heteromatic used alone or as a suffix or prefix, refers to a ring-containing structure or molecule having one or more multivalent heteroatoms, independently selected from N, O, P and S, as a part of the ring structure and including at least 3 and up to about 20 atoms in the ring(s), wherein the ring-containing structure or molecule has an aromatic character (e.g., 4n+2 delocalized electrons).
  • heterocyclic group refers to a radical derived from a heterocycle by removing one or more hydrogens therefrom.
  • heterocyclyl used alone or as a suffix or prefix, refers a monovalent radical derived from a heterocycle by removing one hydrogen therefrom.
  • heterocyclylene used alone or as a suffix or prefix, refers to a divalent radical derived from a heterocycle by removing two hydrogens therefrom, which serves to links two structures together.
  • heteroaryl used alone or as a suffix or prefix, refers to a heterocyclyl having aromatic character.
  • heterocycloalkyl used alone or as a suffix or prefix, refers to a monocyclic or polycyclic ring comprising carbon and hydrogen atoms and at least one heteroatom, preferably, 1 to 3 heteroatoms selected from nitrogen, oxygen, and sulfur, and having no unsaturation.
  • heterocycloalkyl groups include pyrrolidinyl, pyrrolidino, piperidinyl, piperidino, piperazinyl, piperazino, morpholinyl, morpholino, thiomorpholinyl, thiomorpholino, and pyranyl.
  • a heterocycloalkyl group can be unsubstituted or substituted with one or two suitable substituents.
  • the heterocycloalkyl group is a monocyclic or bicyclic ring, more preferably, a monocyclic ring, wherein the ring comprises from 3 to 6 carbon atoms and form 1 to 3 heteroatoms, referred to herein as C 3-6 heterocycloalkyl.
  • heteroarylene used alone or as a suffix or prefix, refers to a heterocyclylene having aromatic character.
  • heterocycloalkylene used alone or as a suffix or prefix, refers to a heterocyclylene that does not have aromatic character.
  • five-membered used as prefix refers to a group having a ring that contains five ring atoms.
  • a five-membered ring heteroaryl is a heteroaryl with a ring having five ring atoms wherein 1, 2 or 3 ring atoms are independently selected from N, O and S.
  • Exemplary five-membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4-thiadiazolyl, and 1,3,4-oxadiazolyl.
  • a six-membered ring heteroaryl is a heteroaryl with a ring having six ring atoms wherein 1, 2 or 3 ring atoms are independently selected from N, O and S.
  • Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl and pyridazinyl.
  • heterocyclyls include, but are not limited to, 1H-indazole, 2-pyrrolidonyl, 2H, 6H-1,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azabicyclo, azetidine, azepane, aziridine, azocinyl, benzimidazolyl, benzodioxol, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carcino
  • alkoxy or “alkyloxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, n-pentoxy, isopentoxy, cyclopropylmethoxy, allyloxy and propargyloxy.
  • alkylthio or “thioalkoxy” represent an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge.
  • Halogenated used as a prefix of a group, means one or more hydrogens on the group is replaced with one or more halogens.
  • carbonyl is art recognized and includes the —C( ⁇ O) groups of such moieties as can be represented by the general formula:
  • X is a bond or represents an oxygen or sulfur
  • R represents a hydrogen, an alkyl, an alkenyl, —(CH 2 ) m —R′′ or a pharmaceutically acceptable salt
  • R′ represents a hydrogen, an alkyl, an alkenyl or —(CH 2 ) m —R′′, where m is an integer less than or equal to ten
  • R′′ is alkyl, cycloalkyl, alkenyl, aryl, or heteroaryl.
  • X is an oxygen, and R is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R′ is a hydrogen, the formula represents a “carboxylic acid.” Where X is oxygen, and R′ is a hydrogen, the formula represents a “formate.” In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a “thiolcarbonyl” group.
  • sulfonyl refers to the —S( ⁇ O) 2 — of a moiety that can be represented by the general formula:
  • R is represented by but not limited to hydrogen, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, aralkyl, or heteroaralkyl.
  • p is 1, 2, 3, 4, 5, 6 or 7 (i.e., C 3-9 cycloalkyl); the C 3-9 cycloalkyl is substituted by R d ; and the point of attachment of the “C( ⁇ O)C 3-9 cycloalkylR d ” is through the carbon atom of the carbonyl group, which is on the left of the expression.
  • protecting group means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations.
  • protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones respectively.
  • the field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 3 rd ed.; Wiley: New York, 1999).
  • “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof (i.e., also include counterions).
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, phosphoric, and the like; and the salts prepared from organic acids such as lactic, maleic, citric, benzoic, methanesulfonic, and the like.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile can be used.
  • in vivo hydrolysable precursors means an in vivo hydrolysable (or cleavable) ester of a compound of any of the formulas described herein that contains a carboxy or a hydroxy group.
  • amino acid esters C 1-6 alkoxymethyl esters like methoxymethyl; C 1-6 alkanoyloxymethyl esters like pivaloyloxymethyl; C 3-8 cycloalkoxycarbonyloxy C 1-6 alkyl esters like 1-cyclohexylcarbonyloxyethyl, acetoxymethoxy, or phosphoramidic cyclic esters.
  • tautomer means other structural isomers that exist in equilibrium resulting from the migration of a hydrogen atom. For example, keto-enol tautomerism where the resulting compound has the properties of both a ketone and an unsaturated alcohol.
  • stable compound and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • the present invention further includes isotopically-labeled compounds of the invention.
  • An “isotopically” or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2H (also written as D for deuterium), 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 18 F, 35 S, 36 Cl, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 , 125 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro receptor labeling and competition assays, compounds that incorporate 3 H, 14 C, 82 Br, 125 I, 131 I, 35 S or will generally be most useful. For radio-imaging applications 11 C, 18 F, 125 I, 123 I, 124 I, 131 I, 75 Br, 76 Br or 77 Br will generally be most useful.
  • a “radio-labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 I, 35 S and 82 Br.
  • the antidementia treatment defined herein may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional chemotherapy.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention.
  • Compounds of the present invention may be administered orally, parenteral, buccal, vaginal, rectal, inhalation, insufflation, sublingually, intramuscularly, subcutaneously, topically, intranasally, intraperitoneally, intrathoracially, intravenously, epidurally, intrathecally, intracerebroventricularly and by injection into the joints.
  • the dosage will depend on the route of administration, the severity of the disease, age and weight of the patient and other factors normally considered by the attending physician, when determining the individual regimen and dosage level as the most appropriate for a particular patient.
  • An effective amount of a compound of the present invention for use in therapy of dementia is an amount sufficient to symptomatically relieve in a warm-blooded animal, particularly a human the symptoms of dementia, to slow the progression of dementia, or to reduce in patients with symptoms of dementia the risk of getting worse.
  • inert, pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • a solid carrier can be one or more substances, which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material.
  • the carrier is a finely divided solid, which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • a low-melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient sized molds and allowed to cool and solidify.
  • Suitable carriers include magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low-melting wax, cocoa butter, and the like.
  • Some of the compounds of the present invention are capable of forming salts with various inorganic and organic acids and bases and such salts are also within the scope of this invention.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, phosphoric, and the like; and the salts prepared from organic acids such as lactic, maleic, citric, benzoic, methanesulfonic, trifluoroacetate and the like.
  • the present invention provides a compound of any of the formulas described herein or a pharmaceutically acceptable salt thereof for the therapeutic treatment (including prophylactic treatment) of mammals including humans, it is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
  • the pharmaceutical composition of this invention may also contain, or be co-administered (simultaneously or sequentially) with, one or more pharmacological agents of value in treating one or more disease conditions referred to herein.
  • composition is intended to include the formulation of the active component or a pharmaceutically acceptable salt with a pharmaceutically acceptable carrier.
  • this invention may be formulated by means known in the art into the form of, for example, tablets, capsules, aqueous or oily solutions, suspensions, emulsions, creams, ointments, gels, nasal sprays, suppositories, finely divided powders or aerosols or nebulisers for inhalation, and for parenteral use (including intravenous, intramuscular or infusion) sterile aqueous or oily solutions or suspensions or sterile emulsions.
  • Liquid form compositions include solutions, suspensions, and emulsions.
  • Sterile water or water-propylene glycol solutions of the active compounds may be mentioned as an example of liquid preparations suitable for parenteral administration.
  • Liquid compositions can also be formulated in solution in aqueous polyethylene glycol solution.
  • Aqueous solutions for oral administration can be prepared by dissolving the active component in water and adding suitable colorants, flavoring agents, stabilizers, and thickening agents as desired.
  • Aqueous suspensions for oral use can be made by dispersing the finely divided active component in water together with a viscous material such as natural synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other suspending agents known to the pharmaceutical formulation art.
  • the pharmaceutical compositions can be in unit dosage form.
  • the composition is divided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparations, for example, packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can also be a capsule, cachet, or tablet itself, or it can be the appropriate number of any of these packaged forms.
  • compositions may be formulated for any suitable route and means of administration.
  • Pharmaceutically acceptable carriers or diluents include those used in formulations suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • conventional non-toxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, cellulose, cellulose derivatives, starch, magnesium stearate, sodium saccharin, talcum, glucose, sucrose, magnesium carbonate, and the like may be used.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc, an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension.
  • the pharmaceutical composition to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, sorbitan monolaurate, triethanolamine oleate, etc.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, sorbitan monolaurate, triethanolamine oleate, etc.
  • the compounds of the invention may be derivatised in various ways.
  • “derivatives” of the compounds includes salts (e.g. pharmaceutically acceptable salts), any complexes (e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or coordination complexes with metal ions such as Mn 2+ and Zn 2+ ), esters such as in vivo hydrolysable esters, free acids or bases, polymorphic forms of the compounds, solvates (e.g. hydrates), prodrugs or lipids, coupling partners and protecting groups.
  • prodrugs is meant for example any compound that is converted in vivo into a biologically active compound.
  • Salts of the compounds of the invention are preferably physiologically well tolerated and non toxic. Many examples of salts are known to those skilled in the art. All such salts are within the scope of this invention, and references to compounds include the salt forms of the compounds.
  • Compounds having acidic groups can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl)amine. Salts can be formed between compounds with basic groups, e.g. amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid. Compounds having both acidic and basic groups can form internal salts.
  • Acid addition salts may be formed with a wide variety of acids, both inorganic and organic.
  • acid addition salts include salts formed with hydrochloric, hydriodic, phosphoric, nitric, sulphuric, citric, lactic, succinic, maleic, malic, isethionic, fumaric, benzenesulphonic, toluenesulphonic, methanesulphonic, ethanesulphonic, naphthalenesulphonic, valeric, acetic, propanoic, butanoic, malonic, glucuronic and lactobionic acids.
  • a salt may be formed with a suitable cation.
  • suitable inorganic cations include, but are not limited to, alkali metal ions such as Na + and K + , alkaline earth cations such as Ca 2+ and Mg 2+ , and other cations such as Al 3+ .
  • suitable organic cations include, but are not limited to, ammonium ion (i.e., NH 4 + ) and substituted ammonium ions (e.g., NH 3 R + , NH 2 R 2 + , NHR 3 + , NR 4 + ).
  • Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine.
  • An example of a common quaternary ammonium ion is N(CH 3 ) 4 + .
  • the compounds may contain an amine function, these may form quaternary ammonium salts, for example by reaction with an alkylating agent according to methods well known to the skilled person. Such quaternary ammonium compounds are within the scope of the invention.
  • Compounds containing an amine function may also form N-oxides.
  • a reference herein to a compound that contains an amine function also includes the N-oxide.
  • N-oxides are the N-oxides of a tertiary amine or a nitrogen atom of a nitrogen-containing heterocycle.
  • N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a per-acid (e.g. a peroxycarboxylic acid), see for example Advanced Organic Chemistry , by Jerry March, 4 th Edition, Wiley Interscience, pages. More particularly, N-oxides can be made by the procedure of L. W. Deady ( Syn. Comm. 1977, 7, 509-514) in which the amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.
  • MCPBA m-chloroperoxybenzoic acid
  • Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques well known in the art.
  • esters are compounds containing the group C( ⁇ O)OR, wherein R is an ester substituent, for example, a C 1-7 alkyl group, a C 3-20 heterocyclyl group, or a C 5-20 aryl group, preferably a C 1-7 alkyl group.
  • Particular examples of ester groups include, but are not limited to, C( ⁇ O)OCH 3 , C( ⁇ O)OCH 2 CH 3 , C( ⁇ O)OC(CH 3 ) 3 , and —C( ⁇ O)OPh.
  • acyloxy (reverse ester) groups are represented by OC( ⁇ O)R, wherein R is an acyloxy substituent, for example, a C 17 alkyl group, a C 320 heterocyclyl group, or a C 520 aryl group, preferably a C 17 alkyl group.
  • R is an acyloxy substituent, for example, a C 17 alkyl group, a C 320 heterocyclyl group, or a C 520 aryl group, preferably a C 17 alkyl group.
  • Particular examples of acyloxy groups include, but are not limited to, OC( ⁇ O)CH 3 (acetoxy), OC( ⁇ O)CH 2 CH 3 , OC( ⁇ O)C(CH 3 ) 3 , OC( ⁇ O)Ph, and OC( ⁇ O)CH 2 Ph.
  • prodrugs which are prodrugs of the compounds are convertible in vivo or in vitro into one of the parent compounds. Typically, at least one of the biological activities of compound will be reduced in the prodrug form of the compound, and can be activated by conversion of the prodrug to release the compound or a metabolite of it.
  • Some prodrugs are esters of the active compound (e.g., a physiologically acceptable metabolically labile ester). During metabolism, the ester group (—C( ⁇ O)OR) is cleaved to yield the active drug.
  • esters may be formed by esterification, for example, of any of the carboxylic acid groups (—C( ⁇ O)OH) in the parent compound, with, where appropriate, prior protection of any other reactive groups present in the parent compound, followed by deprotection if required.
  • Examples of such metabolically labile esters include those of the formula —C( ⁇ O)OR wherein R is: C 17 alkyl (e.g., Me, Et, -nPr, -iPr, -nBu, -sBu, -iBu, tBu); C 17 aminoalkyl (e.g., aminoethyl; 2-(N,N-diethylamino)ethyl; 2(4morpholino)ethyl); and acyloxy-C 17 alkyl (e.g., acyloxymethyl; acyloxyethyl; pivaloyloxymethyl; acetoxymethyl; 1acetoxyethyl; 1-(1-methoxy-1-methyl)ethyl-carbonyloxyethyl; 1-(benzoyloxy)ethyl; isopropoxy-carbonyloxymethyl; 1isopropoxy-carbonyloxyethyl; cyclohexyl-carbonyloxymethyl
  • prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound (for example, as in ADEPT, GDEPT, LIDEPT, etc.).
  • the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid ester derivative.
  • Coupled derivatives include coupling partners of the compounds in which the compounds is linked to a coupling partner, e.g. by being chemically coupled to the compound or physically associated with it.
  • Examples of coupling partners include a label or reporter molecule, a supporting substrate, a carrier or transport molecule, an effector, a drug, an antibody or an inhibitor.
  • Coupling partners can be covalently linked to compounds of the invention via an appropriate functional group on the compound such as a hydroxyl group, a carboxyl group or an amino group.
  • Other derivatives include formulating the compounds with liposomes.
  • the quantity of the compound to be administered will vary for the patient being treated and will vary from about 100 ng/kg of body weight to 100 mg/kg of body weight per day and preferably will be from 10 pg/kg to 10 mg/kg per day.
  • dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art.
  • the skilled artisan can readily determine the amount of compound and optional additives, vehicles, and/or carrier in compositions and to be administered in methods of the invention.
  • the compounds described herein are central nervous system depressants and may be used as tranquilizers or ataractic agents for the relief of anxiety and tension states, for example, in mice, cats, rats, dogs and other mammalian species such as humans, in the same manner as chlordiazepoxide.
  • a compound or mixture of compounds of any of the formulas described herein, or non-toxic physiologically acceptable salts, such as acid addition salts thereof may be administered orally or parenterally in a conventional dosage form such as tablet, pill, capsule, injectable or the like.
  • the dosage in mg/kg of body weight of compounds of the present invention in mammals will vary according to the size of the animal and particularly with respect to the brain/body weight ratio.
  • a minimum effective dosage for a compound of formula (I) will be at least about 0.1 mg/kg of body weight per day for mammals with a maximum dosage for a small mammal such as a dog, of about 100 mg/kg per day.
  • a dosage of about 0.1 to 12 mg/kg per day will be effective, for example, about 5 to 600 mg/day for an average man.
  • the dosage can be given once daily or in divided doses, for example, 2 to 4 doses daily, and such dosage will depend on the duration and maximum level of activity of a particular compound.
  • the dose may be conventionally formulated in an oral or parenteral dosage form by compounding about 5 to 250 mg per unit of dosage of conventional vehicle, excipient, binder, preservative, stabilizer, flavor or the like as called for by accepted pharmaceutical practice, for example, as described in U.S. Pat. No. 3,755,340.
  • the compounds of this invention may be used in pharmaceutical compositions comprising a compound of any of the formulas described herein or can be contained in the same formulation with or co-administered with one or more known drugs.
  • Some example tests that can be conducted to demonstrate the anxiolytic activity of the present compounds include binding tests of GABAA receptors.
  • the binding test was directed to a subtype of GABAA receptors, such as GABAA1 receptors (i.e., those containing the ⁇ 1 subunit), GABAA2 receptors (i.e., those containing the ⁇ 2 subunit), GABAA3 receptors (i.e., those containing the ⁇ 3 subunit) and GABAA5 receptors (i.e., those containing the ⁇ 5 subunit).
  • GABAA modulator anxiolytics work via interactions at the classical benzodiazepine binding site. To a large degree these anxiolytics lack GABAA receptor subtype-selectivity.
  • the subtype-selective GABAA receptor modulators may offer more advantages. For example, a growing body of work suggests that desirable anxiolytic activity is driven primarily by interactions with GABAA receptors containing the ⁇ 2 subunit. Sedation, a side-effect common to all marketed benzodiazepines, is believed to be mediated by interactions at GABAARs containing the ⁇ 1 subunit. To develop anxiolytics with minimal liabilities due to interactions with other subunits, an electrophysiological assay was developed to screen modulatory effects of various compounds on different GABA subunit combinations heterologously expressed in Xenopus oocytes.
  • GABAA receptors were heterologously expressed in Xenopus oocytes by injecting cRNA corresponding to human ⁇ 1 , ⁇ 2 , ⁇ 3 , ⁇ 5 , ⁇ 2 , ⁇ 3 and ⁇ 2 subunits of the GABAA receptor genes.
  • the specific subunit combinations (subtypes) were as follows: ⁇ 1 ⁇ 2 ⁇ 2 , ⁇ 2 ⁇ 3 ⁇ 2 , ⁇ 3 ⁇ 3 ⁇ 2 , and ⁇ 5 ⁇ 3 ⁇ 2 .
  • the EC10 of GABA was approximated for each cell. Stability of GABA-mediated (EC10) current was established. Modulatory effect of test compound was determined and compared across subtypes.
  • the assay developed has reproducibility which allows discrimination of modulatory activity down to minimal effect of about 25% potentiation (prior to normalization to standard) for all four subtypes.
  • the assay can characterize modulatory effects and determine subtype selectivity of test compounds on major subtypes of GABAA receptors.
  • a compound can selectively bind to one subtype of GABAA receptor (by showing about 25% or more of binding comparing to another subtype of GABAA receptor).
  • Anxiolytic activity is indicated in the GABAA binding test by a displacement of the flunitrazepam such as is exhibited by benzodiazepines or by enhancement of the binding such as is shown by cartazolate and tracazolate.
  • the compounds of the invention can bind to GABAA receptors. In some embodiments, the compounds of the invention can bind to GABAA receptors by displacement of benzodiazepines. Accordingly, the compounds of the invention can be used to modulate activities of GABAA receptors. In some embodiments, the compounds of the invention can selectively bind to a subtype of GABAA receptors, such as such as GABAA1 receptors (i.e., those containing the ⁇ 1 subunit), GABAA2 receptors (i.e., those containing the ⁇ 2 subunit), GABAA3 receptors (i.e., those containing the ⁇ 3 subunit) or GABAA5 receptors (i.e., those containing the ⁇ 5 subunit).
  • GABAA1 receptors i.e., those containing the ⁇ 1 subunit
  • GABAA2 receptors i.e., those containing the ⁇ 2 subunit
  • GABAA3 receptors i.e., those containing the ⁇ 3 sub
  • the compounds of the invention can selectively bind to a subtype of GABAA receptors by displacement of benzodiazepines. Accordingly, the compounds of the invention can be used to selectively modulate activities of a subtype of GABAA receptors, such as GABAA1 receptors, GABAA2 receptors, GABAA3 receptors or GABAA5 receptors.
  • a subtype of GABAA receptors such as GABAA1 receptors, GABAA2 receptors, GABAA3 receptors or GABAA5 receptors.
  • certain compounds of the invention are GABAA1 receptor antagonists and GABAA2 receptor agonists.
  • the compounds of the invention can be used to modulate activities of GABAA receptors, or to selectively modulate activities of a subtype of GABAA receptors, the compounds of the invention are envisioned to be useful for treating or preventing diseases mediated by GABAA receptors or a subtype of GABAA receptors.
  • Such disease include, but is not limited to, stroke, head trauma, epilepsy, pain, migraine, mood disorders, anxiety, post traumatic stress disorder, obsessive compulsive disorders, schizophrenia, seizures, convulsions, tinnitus, neurodegenerative disorders including Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's Chorea, Parkinson's disease, depression, bipolar disorders, mania, trigeminal and other neuralgia, neuropathic pain, hypertension, cerebral ischemia, cardiac arrhythmia, myotonia, substance abuse, myoclonus, essential tremor, dyskinesia and other movement disorders, neonatal cerebral hemorrhage, spasticity, cognitive disorder, and sleeping disorder.
  • melatonin receptor agonists are effective in treating depression.
  • the compounds of the invention can selectively modulate activities of a subtype of melatonin receptors, melatonin receptor 1 (MT-1).
  • certain compounds of the invention are MT1 agonists.
  • the compounds of the invention may be effective in treating depression disorders such as major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, or mixed episodes associated with bipolar disorder.
  • an effective amount of one or more compounds of the invention is administered to a patient with such a need.
  • certain compounds of the present invention may be effective in treating insomnia.
  • a compound of formula I or a pharmaceutically acceptable salt, solvate or in vivo hydrolysable ester thereof, or a pharmaceutical composition or formulation comprising a compound of formula I may be administered concurrently, simultaneously, sequentially or separately with one or more pharmaceutically active compound(s) selected from the following:
  • antidepressants such as amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, protriptyline, reboxetine, robalzotan, sertraline, sibutramine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • atypical antipsychotics including for example quetiapine and pharmaceutically active isomer(s) and metabolite(s) thereof, amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, lithium, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, risperidone, quetiapine, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine,
  • antipsychotics including for example amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone and
  • anxiolytics including for example alnespirone, azapirones, benzodiazepines, barbiturates such as adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, zolazepam and equivalents and pharmaceutically
  • anticonvulsants including, for example, carbamazepine, valproate, lamotrogine, gabapentin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • Alzheimer's therapies including, for example, donepezil, memantine, tacrine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • Parkinson's therapies including, for example, deprenyl, L-dopa, Requip, Mirapex, MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • migraine therapies including, for example, almotriptan, amantadine, bromocriptine, butalbital, cabergoline, dichloralphenazone, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, zolmitriptan, zomitriptan, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (ix) stroke therapies including, for example, abciximab, activase, NXY-059, citicoline, crobenetine, desmoteplase, repinotan, traxoprodil and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • neuropathic pain therapies including, for example, gabapentin, lidoderm, pregablin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, paracetamol and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • insomnia therapies including, for example, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, etomidate, glutethimide, halazepam, hydroxyzine, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, roletamide, triclofos, secobarbital, zaleplon, zolpidem and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof, and
  • mood stabilizers including, for example, carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, verapamil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • Such combinations employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within approved dosage ranges and/or the dosage described in the publication reference.
  • the compounds of the present invention can be prepared in a number of ways well known to one skilled in the art of organic synthesis.
  • the compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art.
  • the starting materials and precursors used in the processes described herein were either commercially available or readily prepared by established organic synthesis methods. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule must be compatible with the reagents and reactions proposed. Such restrictions to the substituents which are compatible with the reaction conditions will be readily apparent to one skilled in the art and alternate methods should then be used.
  • DMSO dimethylsulfoxide
  • THF tetrahydrofuran
  • DMF denotes N,N-dimethylformamide.
  • reaction progress was monitored by HPLC, LC-MS or TLC. Oven-dried standard laboratory glassware was used and routine manipulations were done at ambient temperature under a blanket of nitrogen unless otherwise indicated.
  • Commercially available reagents and anhydrous solvents were typically used as received. Evaporations were typically performed under reduced pressure using a rotary evaporator. Preparative chromatography was performed using ICN silica gel 60, 32-63 ⁇ or a suitable equivalent. Products were dried under reduced pressure at 40° C. or a suitable temperature.
  • a compound 1-3 can be made by coupling of a halogenated cinnoline derivative 1-1 (wherein X 1 is halo such as bromo or iodo) to a boron compound 1-2 wherein R 6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.), R 101 and R 102 are each, independently, hydrogen or C 1-6 alkyl; or R 101 and R 102 , together with the two oxygen atoms to which they are attached and the boron atom to which the two oxygen atoms are attached, form a 4-7 membered heterocyclic ring whose ring-forming atoms comprises B, O and C atoms and which is optionally substituted by 1, 2, 3, or 4 C 1-6 alkyl (i.e., a moiety shown as 1-2B-R wherein t1 is 0, 1, 2 or 3; t2 is 0, 1, 2, 3 or 4; and R 400 is each, independently
  • the coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst.
  • a suitable catalyst such as a metal catalyst.
  • metal catalysts include palladium catalyst, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0).
  • the coupling reaction can be carried out in the presence of a suitable base such as an inorganic base.
  • a suitable inorganic base include cesium carbonate, sodium carbonate, and potassium phosphate.
  • the coupling reaction can be carried out in a suitable solvent such as an organic solvent.
  • suitable organic solvent include polar organic solvents, such as an ether and an alcohol.
  • Suitable ethers include 1,2-dimethoxyethane and tetrahydrofuran.
  • Suitable alcohols include ethanol, propanol and isopropanol.
  • a suitable solvent also includes a mixture of two or more individual solvents. Suitable solvents can further contain water.
  • the coupling reaction can be carried out at a suitable temperature to afford the compound 1-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature).
  • the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C.
  • the reaction progress can be monitored by conventional methods such as TLC or NMR.
  • a compound 2-3 can be made by coupling of a halogenated cinnoline derivative 2-1 (wherein X 2 is halo such as bromo or iodo) to a tin compound 2-2 wherein R 6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.), R 201 , R 202 and R 203 are each, independently, C 1-6 alkyl.
  • the coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst.
  • Some exemplary metal catalysts include palladium catalysts, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0).
  • the coupling reaction can be carried out in a suitable organic solvent.
  • Some suitable organic solvent include polar organic solvent.
  • Some suitable organic solvent include aprotic solvent.
  • Some suitable organic solvent include polar aprotic organic solvent such as N,N-dimethylformamide.
  • the coupling reaction can be carried out at a suitable temperature for a time sufficient to afford the compound 2-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature).
  • the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C.
  • the reaction progress can be monitored by conventional methods such as TLC or NMR.
  • a compound 3-3 can be made by coupling of a trialkylstannyl-cinnoline derivative 3-1 (wherein R 301 , R 302 and R 303 are each, independently, C 1-6 alkyl) to a halogenated compound R 6 X 3 3-2 wherein X 3 is halo such as bromo or iodo, and wherein R 6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.).
  • the coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst.
  • Some exemplary metal catalysts include palladium catalysts, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0).
  • the coupling reaction can be carried out in a suitable organic solvent.
  • Some suitable organic solvent include polar organic solvent.
  • Some suitable organic solvents include aprotic organic solvent.
  • Some suitable organic solvents include polar aprotic organic solvents such as N,N-dimethylformamide.
  • the coupling reaction can be carried out at a suitable temperature for a time sufficient to afford the compound 2-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature).
  • the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C.
  • the reaction progress can be monitored by conventional methods such as TLC or NMR.
  • the trialkylstannyl-cinnoline derivative 3-1 can be made by coupling of a halogenated cinnoline derivative 3-0-1 (wherein X 4 is halo such as bromo or iodo) to a di-tin compound 3-0-2 (wherein R 301 , R 302 and R 303 are each, independently, C 1-6 alkyl) in the presence of a suitable catalyst, such as a palladium catalyst.
  • a suitable catalyst such as a palladium catalyst.
  • palladium catalysts include bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0).
  • a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to an amide; a carboxylic acid can be converted to a ester, which in turn can be reduced to an alcohol, which in turn can be further modified.
  • an OH group can be converted into a better leaving group such as mesylate, which in turn is suitable for nucleophilic substitution, such as by CN.
  • reacting refers to the bringing together of designated chemical reactants such that a chemical transformation takes place generating a compound different from any initially introduced into the system. Reacting can take place in the presence or absence of solvent.
  • the mixture was stirred for 30 minutes and then filtered through a pad of celite.
  • the celite was washed with ethyl acetate (3 ⁇ 150 mL).
  • the filtrates were placed in a separatory funnel and the water layer was removed.
  • the organic layer was concentrated under reduced pressure to a volume of 200 mL, placed in a separatory funnel, diluted with ethyl acetate (400 mL), washed with brine, dried over sodium sulfate, filtered and concentrated to dryness.
  • the crude product was taken up in ether (300 mL) and made acidic to pH 1 with 2M hydrochloric acid/ether (Aldrich). After 1 hour, the tan solid was isolated by filtration (39.2 g, 80%).
  • the crude solid (188 g) was then dissolved in hot methanol (6 L), treated with activated charcoal (19 g), stirred 15 minutes at reflux, filtered through celite while hot, concentrated to approximately 3 L, and allowed to crystallize overnight.
  • the solids were collected, washed with diethyl ether (400 mL) and dried in a vacuum oven at 50° C. to give a white crystalline solid.
  • the filtrate was concentrated to approximately 1 L and a second crop obtained.
  • the mother liquors were stripped and a third and fourth crop were obtained from additional recrystallizations to afford a total of 164.6 g of the desired compound as a white crystalline solid (84%).
  • the mixture was diluted with chloroform (2 L) until all of the precipitate was dissolved, washed twice with water, dried through magnesium sulfate, and concentrated to a volume of approximately 200 mL to leave a suspension of the product.
  • the title compound as a light beige solid (11.06 g) was collected by filtration and washed with methylene chloride (50 mL ⁇ 2), methanol (50 mL) and hexane (100 mL ⁇ 2).
  • the mother liquor was concentrated, and purified by flash chromatography using a gradient of ethyl acetate in hexane to give an additional 400 mg of the title compound as a beige solid.
  • Solution A To a mechanically stirred solution of 2-fluoroaniline (11.51 g, 100.34 mmol) in acetic acid (50 mL) was added water (30 mL) at ambient temperature. The mixture was cooled to 0° C., and then concentrated aqueous HCl (25 mL) added. A precipitate was formed as soon as the concentrated HCl was added, and the suspension was stirred at 0° C. for 20 minutes. To this suspension was added dropwise a solution of sodium nitrite (7.72 g, 111.88 mmol) in water (30 mL), maintaining the internal temperature below 5° C. The resulting clear orange solution was stirred at 0° C. for another 30 minutes.
  • Solution B To a mechanically stirred solution of N-propyl-2-cyanoacetamide (15.69 g, 124.37 mmol) in ethanol (220 mL) was added a solution of sodium acetate (136.00 g, 1.66 moles) in water (600 mL), and chilled to between 0° C. and ⁇ 5° C.
  • Solution A was poured into solution B, maintaining the internal temperature below 0° C. An orange precipitate was formed gradually after 10 minutes. The mixture was stirred below 0° ° C. for another hour, and was then diluted with water (500 mL). After 30 minutes, the orange precipitate was collected by filtration, washed with water (100 mL ⁇ 3), and dried at 50° C. under high vacuum to remove water. An orange solid (9.50 g) was obtained, which was the “E” isomer, and used for the next step without further purification.
  • the aqueous layer contained a thick white precipitate and was quickly removed.
  • the organic layer was washed with Rochelle's salt and brine, dried over magnesium sulfate, filtered and concentrated to give 2.6 g slightly crude product which was purified on silica gel using a gradient of 20 to 60% ethyl acetate in hexane. Recrystallization from ethyl acetate/hexanes (10 mL each, 0° C. overnight) afforded the title compound as a white solid (650 mg, 26%).
  • Method A The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (typically 2-3 molar equivalents), cesium carbonate (2 molar equivalents) and bis(triphenylphosphine)palladium(II) dichloride (0.025 molar equivalents) were placed in a microwave reaction vessel and dissolved in 7:3:2 (v/v/v) 1,2-dimethoxyethane:water:ethanol (5 mL/mmol cinnoline-halide) at ambient temperature.
  • reaction vessel was capped, the head-space purged with dry nitrogen and the stirred mixture was heated on a Biotage Optimizer (300 W) microwave system maintaining a reaction temperature of 150° C. for 30-90 minutes, reaction pressures of 7 bar were typically observed.
  • the reaction was then cooled to ambient temperature and extracted with ethyl acetate.
  • the residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method B To a solution of the cinnoline-halide in 1,2-dimethoxyethane (10 mL/mmol cinnoline-halide) under nitrogen at ambient temperature was added tetrakis(triphenylphosphine)palladium (0) (0.05-0.15 molar equivalents). After stirring 10-20 min an arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (1-4 molar equivalents) was added followed by a solution of sodium carbonate (2.5 molar equivalents) in water (3 mL/mmol halide). The resulting mixture was heated at reflux for 2-24 h.
  • reaction was then cooled to ambient temperature and extracted with ethyl acetate.
  • residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method C To a stirred solution of the cinnoline-halide in anhydrous N,N-dimethylformamide (2 mL/mmol cinnoline-halide) at ambient temperature was added an optionally substituted aryl- or heteroaryl-tin reagent (1.2 molar equivalents) and tetrakis(triphenylphosphine)palladium(0) (0.05 molar equivalents). The mixture was heated at 100° C. for 8-48 h. The reaction was then cooled to ambient temperature and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with an increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method D The cinnoline-halide, an optionally substituted aryl- or heteroaryl-tin reagent (1.2-3 molar equivalents) and tetrakis(triphenylphosphine) palladium(0) (0.05-0.10 molar equivalents) were placed in a microwave reaction vessel and dissolved in 2-4 mL of anhydrous N,N-dimethylformamide at ambient temperature. The reaction vessel was purged with nitrogen, capped, and the stirred mixture was heated on a Biotage Optimizer (300 W) microwave system maintaining a reaction temperature of 150° C. for 30 minutes.
  • a Biotage Optimizer 300 W
  • the reaction was cooled to ambient temperature, diluted with methylene chloride, washed with water, dried over magnesium sulfate and the solvent was evaporated. The residue was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method E To a stirred solution of 8-trimethylstannyl-cinnoline derivative and tetrakis(triphenylphosphine) palladium(0) (0.05-0.10 molar equivalents) in anhydrous N,N-dimethylformamide at ambient temperature under nitrogen was added an optionally substituted aryl- or heteroaryl bromide (1.2-3 molar equivalents). The reaction was heated to 150° C. for 4-16 hours. The reaction mixture was evaporated under reduced pressure. The residue was dissolved in methylene chloride, washed with water twice, dried through MgSO 4 , and then the solvent was evaporated. The residue was purified by flash chromatography on silica gel eluting with an increasingly polar gradient of ethyl acetate in hexane to afford the desired compound.
  • Method F To a solution of the cinnoline-halide in anhydrous tetrahydrofuran (10 mL/mmol cinnoline-halide) under nitrogen at ambient temperature was added (triphenylphosphine)palladium(II) dichloride (0.10 molar equivalents) followed by an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (2-4 molar equivalents) followed by freshly ground potassium phosphate (2.0 molar equivalents). The resulting mixture was heated at reflux for 2-40 h. The reaction was then cooled to ambient temperature and diluted with saturated sodium bicarbonate and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with 5% ether in chloroform to afford the desired compound.
  • Method G The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (4-5 molar equivalents), cesium carbonate (4-5 molar equivalents), 2-dicyclohexylphosphino-2′,4′,6′-trisopropylbiphenyl (0.24 molar equivalents) and tris(dibenzylidene-acetone)dipalladium(0) (0.06 molar equivalents) were placed in a 3-neck flask under N 2 and dissolved in 7:3:2 (v/v/v) THF:water:2-propanol (5 mL/mmol cinnoline-halide) at ambient temperature.
  • reaction vessel was fitted with a reflux condenser, capped, vacuum degassed (3 ⁇ ) backfilling with N 2 and placed in a preheated oil bath (70° C.) and heated for 20 hours. (* if reaction not complete more boronic acid and cesium carbonate in equal proportions were added with additional heating time). The reaction was then cooled to ambient temperature, decanted organic layer and concentrated under reduced pressure. Residue partitioned between ethyl acetate and 5% sodium bicarbonate (aq).
  • Method H The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (3-5 molar equivalents), sodium carbonate (4-5 molar equivalents), [1,1′-bis(diphenylphospino)-ferrocene] dichloropalladium(II) complex with dichloromethane (1:1) (0.075 molar equivalents) were placed in a 3-neck flask under N 2 and dissolved in 7:3:2 (v/v/v) THF:water:2-propanol (5 mL/mmol cinnoline-halide) at ambient temperature.
  • reaction vessel was fitted with a reflux condenser, under N 2 and placed in a preheated oil bath (85° C.) and refluxed 2-20 hours (* if reaction not complete added more boronic acid with additional heating time).
  • the reaction was then cooled to ambient temperature, reduced volume under reduced pressure, partitioned between ethyl acetate and water.
  • the residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • the reaction mixture was diluted with methylene chloride, quenched with water, washed with 10% potassium carbonate aqueous solution twice, dried through magnesium sulfate, and the solvent was evaporated to dry.
  • the residue was purified by flash chromatography using a gradient of methanol in methylene chloride to give a yellow liquid as 3-bromo-5-(azetidin-1-ylcarbonyl)-pyridine (846 mg, 70.9% yield).
  • This precursor was prepared according to the method of A. V. Ivanchatchenko as described in the Journal of Heterocyclic Chemistry (2004) vol. 41 p. 931
  • Tetrahydrofuran (515 mL, anhydrous) and isopropanol (147 mL, anhydrous) were added and the resulting red suspension was stirred at room temperature for 15 minutes.
  • a solution of sodium carbonate (57.0 g, 537.7 mmol) in water (220 mL) was added rapidly through the addition funnel and the resulting mixture immediately placed into a pre-heated 80° C. oil bath. After 90 minutes at reflux (observed internal temperature 65° C.), the reaction mixture was cooled to room temperature and filtered though a bed of Celite supported on a sintered glass funnel topped with Norite decolorizing carbon (30 g).
  • the titled compound can be separated into two atropisomers using Supercritical Fluid Chromatography.
  • supercritical CO 2 modified with methanol these atropisomers are stable and hence are separable on a chiral support.
  • the atropisomers inter-conversion is greatly facilitated.
  • the mixture was stirred for 30 minutes and then filtered through a pad of Celite.
  • the Celite was washed with EtOAc (3 ⁇ 150 mL).
  • the filtrates were placed in a separatory funnel and the water layer was removed.
  • the organic layer was concentrated under reduced pressure to reduce volume to ⁇ 200 mL, placed in a separatory funnel, diluted with EtOAc (400 mL), washed organic with brine, dried over sodium sulfate, filtered and concentrated to dryness.
  • the crude product was taken up in ether (300 mL) and made acidic to pH 1 with 2M HCl/ether (Aldrich). After 1 hour, the tan solid was isolated by filtration (39.2 g, 80%).
  • the title compound may form isolable atropisomers in certain organic solvents (e.g. 25-35% methanol) at room temperature.
  • the two atropisomers of the title compound may be isolated using chiral LC. However, these isomers will racimize rapidly under neutral or acidic aqueous solutions.
  • the title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (200 mg, 0.535 mmol) and 2,5-dimethoxyphenyl boronic acid (194 mg, 1.07 mmol) according to Method A.
  • the off-white solid from chromatography was slurried in ether, filtered and dried under vacuum at room temperature to afford the desired product (147 mg, 71%).
  • the title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (220 mg, 58.8 mmol) and 2,4-dimethoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine (312 mg, 1.62 mmol) according to Method B to afford a white solid (123 mg, 54%).
  • the title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (70.0 mg, 0.237 mmol) and 4-methoxy-3-pyridine boronic acid (153.0 mg, 0.3439 mmol) according to Method A except that the extraction was carried out with methylene chloride rather than ethyl acetate and the flash column was eluted with a gradient of 10 to 60% methanol in dichloromethane. The concentrated product was then recrystallized from chloroform (with a few drops of methanol) and hexanes to afford the title compound as a yellow solid (19.6 mg, 26% yield).
  • the title compound was prepared from 4-amino-8-bromo-N-butyl-cinnoline-3-carboxamide (100 mg, 0.31 mmol) and 2,5-dimethoxyphenyl boronic acid (112.6 mg, 0.62 mmol) according to Method A to afford a white solid (96.3 mg, 82%).
  • the title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (100.0 mg, 0.339 mmol) and 2,5-dimethoxyphenyl boronic acid (123.3 mg, 0.678 mmol) according to Method A.
  • the solid obtained after chromatography was washed with diethyl ether and dried overnight at 40° C. to afford the title compound as a fluffy white solid (64.4 mg, 54% yield).
  • the title compound was prepared from 4-amino-8-bromo-N-methyl-cinnoline-3-carboxamide (20.0 g, 63.1 mmol) and 2,5-dimethoxyphenyl boronic acid (22.3 g, 122.4 mmol) according to Method B except that potassium carbonate was used as the base and tetrahydrofuran:ethanol:water (1:1:1) was used as the solvent system.
  • the reaction mixture was filtered and the yellow solids were slurried in 10% methanol in chloroform and filtered. The combined filtrates were concentrated to a solid, slurried in hot ethyl acetate, and filtered.
  • the title compound was prepared from 4-amino-8-bromo-N-butyl-cinnoline-3-carboxamide (200.0 mg, 0.62 mmol) and 2,4-dimethoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine (987.9 mg, 3.72 mmol) according to Method B to afford a white solid (162.1 mg, 69%).
  • the title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (200.0 mg, 0.678 mmol) and 2,4-dimethoxyprimidine-5-boronic acid pinacol ester (363.1 mg, 1.362 mmol) according to Method B except that the reaction was heated at 90° C. to fully dissolve the starting materials. After 4 hours, additional 2,4-dimethoxyprimidine-5-boronic acid pinacol ester (363.1 mg, 1.362 mmol) was added and the reaction was refluxed overnight.
  • the title compound was prepared from 4-amino-8-bromo-cinnoline-3-carboxylic acid allylamide (273 mg, 0.89 mmol) and 2,5-dimethoxyphenyl boronic acid (201.1 mg, 1.11 mmol) according to Method A to afford an off-white solid (105 mg, 32%).
  • the title compound was prepared from 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (450 mg, 1.46 mmol) and 3-methylphenyl boronic acid (408 mg, 3.00 mmol) according to Method A to afford an off-white solid (321 mg, 69%).
  • the title compound was prepared from 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (300.0 mg, 0.97 mmol) and 2-fluoro-6-methylpyridine-3-boronic acid (426.7 mg, 2.75 mmol) according to Method A to afford a white solid (124.2 mg, 38%).
  • the title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (200.0 mg, 0.53 mmol) and 2-fluoro-5-methoxyphenyl boronic acid (181.7 mg, 1.07 mmol) according to Method A to afford a yellow crystalline solid (111.0 mg, 56%).
  • the title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (250 mg, 0.67 mmol) and 2-chloro-5-methoxyphenyl boronic acid (279 mg, 1.50 mmol) according to Method A to afford a solid (181 mg, 72%).

Abstract

This invention relates to novel compounds having the structural formula I below:
Figure US20090018112A1-20090115-C00001
and their pharmaceutically acceptable salts, tautomers or in vivo-hydrolysable precursors, compositions and methods of use thereof. These novel compounds provide a treatment or prophylaxis of anxiety disorders, cognitive disorders, and/or mood disorders.

Description

  • This application is a divisional of U.S. application Ser. No. 11/611,943, filed on Dec. 18, 2006, which claims the benefit of United State Provisional Applications 60/752,137, filed Dec. 20, 2005 and 60/823,693, filed Aug. 28, 2006 under 35 U.S.C. § 119(e), the entireties of which are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to novel cinnoline compounds, their pharmaceutical compositions, methods of use and processes to make such compounds. In addition, the present invention relates to therapeutic methods for the treatment and/or prevention of anxiety disorders, cognitive disorders, and/or mood disorders.
  • BACKGROUND OF THE INVENTION
  • The present invention comprises, inter alia, cinnoline compounds, their use as central nervous system (CNS) depressants (especially anxiolytics), and pharmacological tools, methods for their preparation, pharmaceutical compositions containing the same, and intermediates used in their preparation.
  • Some cinnoline compounds including selected 4-amino- and 4-oxo-cinnoline-3-carboxamides are disclosed in East German Patent 123525 (Verfahren zur Herstellung von substituierten 4-Aminocinnolinen); U.S. Pat. No. 4,379,929 to Conrad et al; U.S. Pat. Nos. 4,886,800 and 4,925,844 to Resch; Daunis et al., “Preparation et proprietes de cinnolones-3 et cinnolones-4,” Bull. de la Societe Chimique de France, 8:3198-3202 (1972); Lunt et al. “A New Cinnoline Synthesis,” J. Chem. Soc. (C), 687-695 (1968); Gewald, et al., “Synthese von 4-Aminocinnolinen aus (Arylhydrazono) (cyan)-essigsaurederivaten,” Liebigs Ann. Chem., 1390-1394 (1984); and U.S. Pat. No. 3,657,241 to Kurihara. Additionally, selected cinnoline compounds, including 3-acyl-4-substituted cinnoline derivatives are disclosed in Liebigs Ann. Chem. 1390-1394 (1984) supra and Sandison, et al., “A New Heterocyclisation Reaction Leading to Cinnolin-4(1H)-one Derivatives,” J. Chem. Soc. Chem. Comm., 752-753 (1974). Additionally, cinnoline compounds are also disclosed in EP205272 and EP 328282. However, none of the foregoing discloses or suggests the novel compounds of the present invention or suggests their use as CNS depressants.
  • gamma-Aminobutyric acid (GABA) is a common inhibitory neurotransmitter in the mammalian brain and is estimated to be present at about one third of all synapses. When GABA binds to a GABA receptor, it affects the ability of neurons expressing the receptors to conduct neural impulses. In the adult mammalian nervous system, GABA typically inhibits neuron firing (depolarization). Neurons in the brain express three main types of GABA receptors: GABA type A receptors (GABAA), GABA type B receptors (GABAB), and GABA type C receptors (GABAC). GABAA receptors function as ligand-gated ion channels to mediate fast inhibitory synaptic transmissions that regulate neuronal excitability involved in such responses as seizure threshold, skeletal muscle tone, and emotional status. GABAA receptors are targets of many sedating drugs, such as benzodiazepines, barbiturates and neurosteroids.
  • The intrinsic inhibitory signal of GABA is transduced principally by GABAA receptors. GABAA receptors are pentameric, ligand-gated chloride ion (Cl) channels belonging to a superfamily of ligand-gated ionotropic receptors that includes the nicotinic acetylcholine receptor. GABAA receptors are very heterogeneous, with at least 16 different subunits producing potentially thousands of different receptor types.
  • GABAA receptor subunits aggregate into complexes that form chloride ion selective channels and contain sites that bind GABA along with a variety of pharmacologically active substances. When GABA binds to this receptor, the anion channel is activated, causing it to open and allowing chloride ions (Cl) to enter the neuron. This influx of Cl ions hyperpolarizes the neuron, making it less excitable. The resultant decrease in neuronal activity following activation of the GABAA receptor complex can rapidly alter brain function, to such an extent that consciousness and motor control may be impaired.
  • The numerous possible combinations of GABAA receptor subunits and the widespread distribution of these receptors in the nervous system likely contributes to the diverse and variable physiological functions of GABAA receptors, which have been implicated in many neurological and psychiatric disorders, and related conditions, including: stroke, head trauma, epilepsy, pain, migraine, mood disorders, anxiety, post traumatic stress disorder, obsessive compulsive disorders, schizophrenia, seizures, convulsions, tinnitus, neurodegenerative disorders including Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's Chorea, Parkinson's disease, depression, bipolar disorders, mania, trigeminal and other neuralgia, neuropathic pain, hypertension, cerebral ischemia, cardiac arrhythmia, myotonia, substance abuse, myoclonus, essential tremor, dyskinesia and other movement disorders, neonatal cerebral hemorrhage, and spasticity. GABAA receptors are also believed to play a role in cognition, consciousness, and sleep.
  • Currently available drugs for modulating GABAA receptor activity include barbiturates, such as pentobarbital and secobarbital, and benzodiazepines such as diazepam, chlordiazepoxide and midazolam. Barbiturates can directly activate GABAA receptors, significantly increasing Cl currents in the absence of further intervention by GABA itself and can also indirectly augment GABAergic neural transmission. In contrast, benzodiazepines act as indirect allosteric modulators, and are largely incapable of increasing Cl Currents in the absence of GABA, but enhance GABA-activated increases in Cl conductance. This latter property is thought to be responsible for the usefulness of benzodiazepines for treating a number of disorders, including generalized anxiety disorder, panic disorder, seizures, movement disorders, epilepsy, psychosis, mood disorders, and muscle spasms, as well as the relative safety of benzodiazepines compared to barbiturates.
  • Both barbiturates and benzodiazepines are addictive and can cause drowsiness, poor concentration, ataxia, dysarthria, motor incoordination, diplopia, muscle weakness, vertigo and mental confusion. These side effects can interfere with an individual's ability to perform daily routines such as driving, operating heavy machinery or performing other complex motor tasks while under therapy, making barbiturates and benzodiazepines less than optimal for treating chronic disorders involving GABA and GABAA receptors.
  • GABAA receptors and GABAergic neural transmissions are implicated as targets for therapeutic intervention in a myriad of neurological and psychiatric disorders. Adverse side effects, including addictive properties exhibited by currently available GABA and GABAA receptor modulating drugs, make these drugs unsuitable in many therapeutic contexts. Accordingly, there remains an important, unmet need in the art for alternative compositions, methods and tools that will be useful in broad clinical applications to modulate the function and activity of GABA and GABA receptors in mammalian subjects, including humans, and/or to target GABAergic neural transmission. The present invention is also, inter alia, directed toward this end.
  • DESCRIPTION OF EMBODIMENTS
  • Provided herein are novel compounds of structural formula J:
  • Figure US20090018112A1-20090115-C00002
  • or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, wherein:
  • R1 is C1-6 alkyl, C1-6haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R7;
  • R2 is H, C(═O)Rb, C(═O)NRcRd, C(═O)ORa, S(═O)2Rb, C1-6 alkyl, C1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R8;
  • R3, R4 and R5 are each, independently, H, halo, Si(C1-10 alkyl)3, CN, NO2, ORa, SRa, OC(═O)Ra, OC(═O)ORb, OC(═O)NRcRd, C(═O)Ra, C(═O)ORb, C(═O)NRcRd, NRcNRd, NRcC(═O)Ra, NRcC(═O)ORb, NRcS(═O)2Rb, S(═O)Ra, S(═O)NRcRd, S(═O2Ra, S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 R9;
  • R6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, each optionally substituted by 1, 2, 3, 4 or 5 A1;
  • R7, R8 and R9 are each, independently, halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa′, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′ Rd′;
  • A1 is halo, CN, NO2, ORa, SRa, C(═O)Rb, C(═O)NRcRdC(═O)ORa, OC(═O)b, OC(═O)NRcRd, NRcRd, NRcC(═O)Rd, NRcC(═O)ORa, NRcS(═O)Rb, NRcS(═O)2Rb, S(═O)Rb, S(═O)NRcRd, S(═O)2Rb, S(═O)2NRcRd, C1-4 alkoxy, C1-4haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl, wherein each of the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl is optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa′, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)Rb′, NRc′S(═O)2Rb′, S(═O)Rc′Rd′, S(═O)NRc′R d′, S(═O)2Rb′, or S(═O)2NRc′Rd′;
  • Ra and Ra′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • Rb and Rb′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • Rc and Rd are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • or Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; and
  • Rc′ and Rd′ are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • or Rc′ and Rd′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, when R2, R3, R4 and R5 are each H, then R6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R7.
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′.
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)—(C1-4 alkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • In some embodiments, R1 is C1-6 alkyl or C1-6 haloalkyl.
  • In some embodiments, R1 is C1-6 alkyl.
  • In some embodiments, R1 is n-propyl.
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, or C1-6 alkyl.
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, or C1-3 alkyl.
  • In some embodiments, R2 is H.
  • In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, ORa, SRa, OC(═O)Ra, OC(═O)ORb, OC(═O)NRcRd, C(═O)Ra, C(═O)ORb, C(═O)NRcRd, NRcRd, NRcC(═O)Ra, NRcC(═O)O, NRcS(═O)2Rb, S(═O)NRcRd, S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, OH, C1-4 alkoxy, C1-4haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, OH, C1-4 alkoxy, C1-4haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl).
  • In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, halo, C1-6 alkyl or C1-6 haloalkyl.
  • In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, halo or C1-3 haloalkyl.
  • In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, or halo.
  • In some embodiments, R6 is aryl or heteroaryl, each optionally substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is aryl optionally substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is aryl substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is heteroaryl optionally substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is phenyl, 2-naphthyl, 3-pyridyl, 4-pyridyl, pyrimidin-5-yl, pyrazin-2-yl, pyrazol-3-yl, pyrazol-4-yl, 3-quinolyl, 6-quinolyl, or indol-5-yl, each optionally substituted by 1, 2, 3, 4 or 5 A1.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, Rc′Rd′, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)Rc′Rd′, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl) and S(═O)2NRc′Rd′;
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; and
  • Rc′ and Rd′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • Also provided herein are novel compounds of structural formula II:
  • Figure US20090018112A1-20090115-C00003
  • or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, wherein:
  • R1 is C1-6 alkyl or C1-6haloalkyl;
  • R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2 or C1-6 alkyl;
  • R5 is H, C1-4alkoxy, halo, C1-6alkyl or C1-6haloalkyl;
  • R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, when R2 and R5 are each H, then R6 is other than unsubstituted phenyl.
  • In some embodiments, R1 is C1-6 alkyl.
  • In some embodiments, R1 is n-propyl.
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl) or C1-6 alkyl.
  • In some embodiments, R2 is H.
  • In some embodiments, R5 is H, C1-4 alkoxy or halo.
  • In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 1, 2 or 3 C1-4 alkoxy or C1-4 alkyl.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 2 C1-4 alkoxy or C1-4 alkyl.
  • In some embodiments, R1 is n-propyl and R2 is H.
  • The present invention further provides compositions comprising a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • The present invention further provides methods of treating or preventing an anxiety disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • The present invention further provides methods of treating or preventing a cognitive disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • The present invention further provides methods of treating or preventing a mood disorder in a patient, comprising administering to the patient a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • The present invention further provides a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, described herein for use as a medicament.
  • The present invention further provides a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, described herein for the manufacture of a medicament.
  • The present invention further provides methods of modulating activity of GABAA receptor comprising contacting the GABAA receptor with a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • The present invention further provides synthetic methods of making a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof.
  • Provided herein are novel compounds of structural formula J:
  • Figure US20090018112A1-20090115-C00004
  • or a pharmaceutically acceptable salt, tautomer, atropisomer, or in vivo-hydrolysable precursor thereof, wherein:
  • R1 is C1-6 alkyl, C1-6haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R7;
  • R2 is H, C(═O)Rb, C(═O)NRcRd, C(═O)ORa, S(═O)2Rb, C1-6 alkyl, C1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R8;
  • R3, R4 and R5 are each, independently, H, halo, Si(C1-10 alkyl)3, CN, NO2, ORa, SRa, OC(═O)Ra, OC(═O)ORb, OC(═O)NRcRd, C(═O)Ra, C(═O)ORb, C(═O)NRcRd, NRcRd, NRcC(═O)Ra, NRcC(═O)ORb, NRcS(═O)2Rb, S(═O)Ra, S(═O)NRcRd, S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 R9;
  • R6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, each optionally substituted by 1, 2, 3, 4 or 5 A1;
  • R7, R5 and R9 are each, independently, halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′;
  • A1 is halo, CN, NO2, ORa, SRa, C(═O)Rb, C(═O)NRcRd, C(═O)ORa, OC(═O)Rb, OC(═O)NRcRd, NRcRd, NRcC(═O)Rd, NRcC(═O)ORa, NRcS(═O)Rb, NRcS(═O)2Rb, S(═O)Rb, S(═O)NRcRd, S(═O)2Rb, S(═O)2NRcRd, C1-4 alkoxy, C1-4haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl, wherein each of the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl is optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa′, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)Rb′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′;
  • Ra and Ra′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • Rb and Rb′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • Rc and Rd are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • or Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; and
  • Rc′ and Rd′ are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6alkyl, C1-6haloalkyl, C1-6haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl;
  • or Rc′ and Rd′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, when R2, R3, R4 and R5 are each H, then R6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R7, or any subgroup thereof. In some embodiments, R1 is C1-6 alkyl, C1-6haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 R7. In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′. In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R1 is C1-6 alkyl, C1-6haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R1 is C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, each optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)—(C1-4 alkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R1 is C1-6 alkyl, C1-6haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R1 is C1-6 alkyl or C1-6 haloalkyl. In some embodiments, R1 is C1-6 alkyl. In some embodiments, R1 is n-propyl.
  • In some embodiments, R2 is H, C(═O)Rb, C(═O)NRcRd, C(═O)ORa, S(═O)2Rb, C1-6 alkyl, C1-6 haloalkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein each of the C1-6 alkyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 R8, or any subgroup thereof. In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C1-6 alkyl, C1-6 haloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2 or C1-6 alkyl. In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, or C1-3 alkyl. In some embodiments, R2 is H.
  • In some embodiments, R3, R4 and R5 are each, independently, H, halo, Si(C1-10 alkyl)3, CN, NO2, ORa, SRa, OC(═O)Ra, OC(═O)ORb, OC(═O)NRcRd, C(═O)R3, C(═O)ORb, C(═O)NRcRd, NRcRd, NRcC(═O)Ra, NRcC(═O)ORb, NRcS(═O)2Rb, S(═O)Ra, S(═O)NRcRd, S(═O)2Ra, S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 R9, or any subgroup thereof. In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, OR3, SR3, OC(═O)Ra, OC(═O)ORb, OC(═O)NRcRd, C(═O)Ra, C(═O)ORb, C(═O)NRcRd, NRcRd, NRcC(═O)Ra, NRcC(═O)ORb, NRcS(═O)2Rb, S(═O)Ra, S(═O)NRcRd, S(═O)2Ra, S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-14 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R3, R4 and R5 are each, independently, H, halo, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl) and S(═O)2NH(arylalkyl). In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, halo, C1-6 alkyl or C1-6 haloalkyl. In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, halo or C1-3 haloalkyl. In some embodiments, R3, R4 and R5 are each, independently, H, C1-4 alkoxy, or halo.
  • In some embodiments, R6 is aryl, cycloalkyl, heteroaryl or heterocycloalkyl, or any subgroup thereof, each optionally substituted by 1, 2, 3, 4 or 5 A1, or any subgroup thereof. In some embodiments, R6 is aryl or heteroaryl, each optionally substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is aryl optionally substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is aryl substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is heteroaryl optionally substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is phenyl, 2-naphthyl, 3-pyridyl, 4-pyridyl, pyrimidin-5-yl, pyrazin-2-yl, pyrazol-3-yl, pyrazol-4-yl, 3-quinolyl, 6-quinolyl, or indol-5-yl, each optionally substituted by 1, 2, 3, 4 or 5 A1. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, Rc′Rd′, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)Rc′Rd′, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl) and S(═O)2NRc′Rd′. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl.
  • In some embodiments, R7, R8 and R9 are each, independently, halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa′, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′, or any subgroup thereof.
  • In some embodiments, A1 is halo, CN, NO2, ORa, SRa, C(═O)Rb, C(═O)NRcRd, C(═O)ORa, OC(═O)Rb, OC(═O)NRcRd, NRcRd, NRcC(═O)Rd, NRcC(═O)ORa, NRcS(═O)Rb, NRcS(═O)2Rb, S(═O)Rb, S(═O)NRcRd, S(═O)2Rb, S(═O)2NRcRd, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl, or any subgroup thereof, wherein each of the C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocycloalkylalkyl, aryl, cycloalkyl, heteroaryl or heterocycloalkyl is optionally substituted by 1, 2, 3, 4 or 5 substituents independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, ORa′, SRa′, C(═O)Rb′, C(═O)NRc′Rd′, C(═O)ORa′, OC(═O)Rb′, OC(═O)NRc′Rd′, NRc′Rd′, NRc′C(═O)Rb′, NRc′C(═O)ORa′, NRc′S(═O)Rb′, NRc′S(═O)2Rb′, S(═O)Rb′, S(═O)NRc′Rd′, S(═O)2Rb′, or S(═O)2NRc′Rd′, or any subgroup thereof.
  • In some embodiments, Ra and Ra′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6 haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl, or any subgroup thereof.
  • In some embodiments, Rb and Rb′ are each, independently, H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6 haloalkyl, C1-6 haloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl, or any subgroup thereof.
  • In some embodiments, Rc and Rd are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6 halo alkyl, C1-6 halo alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl, or any subgroup thereof.
  • In some embodiments, Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • In some embodiments, Rc′ and Rd′ are each, independently, H, C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof, wherein the C1-10 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted with OH, amino, halo, C1-6 alkyl, C1-6 halo alkyl, C1-6 halo alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl or heterocycloalkyl, or any subgroup thereof.
  • In some embodiments, Rc′ and Rd′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • In some embodiments, when R2, R3, R4 and R5 are each H, then R6 is other than unsubstituted phenyl or unsubstituted cycloalkyl.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, wherein each of the C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl is optionally substituted by 1, 2 or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, CN, NO2, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, Rc′Rd′, SH, —S—(C1-4 alkyl), C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)Rc′Rd′, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl) and S(═O)2NRc′Rd′; Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group; and
  • Rc′ and Rd′ together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2, 3, 4 or 5 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R6 is naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • Also provided herein are novel compounds of structural formula II:
  • Figure US20090018112A1-20090115-C00005
  • or a pharmaceutically acceptable salt, tautomer, or in vivo-hydrolysable precursor thereof, wherein:
  • R1 is C1-6 alkyl or C1-6haloalkyl;
  • R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2 or C1-6 alkyl;
  • R5 is H, C1-4 alkoxy, halo, C1-6 alkyl or C1-6 haloalkyl;
  • R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and
  • Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, when R2 and R5 are each H, then R6 is other than unsubstituted phenyl.
  • In some embodiments, R1 is C1-6 alkyl or C1-6haloalkyl, or any subgroup thereof.
  • In some embodiments, R1 is C1-6 alkyl. In some embodiments, R1 is n-propyl.
  • In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, or C1-6 alkyl, or any subgroup thereof. In some embodiments, R2 is H, C(═O)—(C1-4 alkyl), C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl) or C1-6 alkyl. In some embodiments, R2 is H.
  • In some embodiments, R5 is H, C1-4 alkoxy, halo, C1-6 alkyl or C1-6 haloalkyl, or any subgroup thereof. In some embodiments, R5 is H, C1-4 alkoxy or halo.
  • In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, or any subgroup thereof, each optionally substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), OC(═O)H, OC(═O)—(C1-4 alkyl), OC(═O)-(arylalkyl), OC(═O)NH2, OC(═O)NH(C1-4 alkyl), OC(═O)NH-(arylalkyl), OC(═O)N(C1-4 alkyl)2, NHC(═O)—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHC(═O)O—(C1-4 alkyl), NHC(═O)O-(arylalkyl), NHS(═O)2—(C1-4 alkyl), NHS(═O)2-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl, or any subgroup thereof. In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 1, 2 or 3 C1-4 alkoxy or C1-4 alkyl. In some embodiments, R6 is phenyl, naphthyl, pyridyl, pyrimidinyl, pyrazinyl, pyrazolyl, quinolyl or indolyl, each substituted by 2 C1-4 alkoxy or C1-4 alkyl.
  • In some embodiments, Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group, or any subgroup thereof.
  • In some embodiments, R6 is phenyl substituted by 1, 2 or 3 halo, CN, OH, C1-4 alkoxy, C1-4 haloalkoxy, amino, C1-4 alkylamino, C2-8 dialkylamino, NRcRd, C(═O)H, C(═O)—(C1-4 alkyl), C(═O)-(arylalkyl), C(═O)NH2, C(═O)NH(C1-4 alkyl), C(═O)N(C1-4 alkyl)2, C(═O)NRcRd, C(═O)OH, C(═O)O—(C1-4 alkyl), C(═O)O-(arylalkyl), S(═O)2—(C1-4 alkyl), S(═O)2-(arylalkyl), S(═O)2NH(C1-4 alkyl), S(═O)2NH(arylalkyl), S(═O)2NRcRd, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, cycloalkylalkyl or heterocycloalkylalkyl; and RcC and Rd together with the N atom to which they are attached form a 4-, 5-, 6- or 7-membered heterocycloalkyl group.
  • In some embodiments, R1 is n-propyl and R2 is H.
  • In some embodiments, the present invention provides the following compounds:
    • 4-amino-7-fluoro-8-phenyl-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-7-chloro-8-phenyl-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-7-methoxy-8-phenyl-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-7-chloro-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-methoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-[4-methoxy-2-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-difluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-fluoro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-chloro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-azetidin-1-ylcarbonyl-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,3-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,3-difluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,3-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(6-quinolyl)cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(3-quinolyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2-naphthyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(1H-indol-5-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(3,4,5-trimethoxyphenyl)-cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(2,3,4-trimethoxyphenyl)-cinnoline-3-carboxamide;
    • 4-amino-8-(2-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,6-dimethoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 3-[4-amino-3-(propylcarbamoyl)cinnolin-8-yl]benzoic acid;
    • 4-amino-8-(3-azetidin-1-ylcarbonylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-pyrazin-2-yl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(3-pyridyl)cinnoline-3-carboxamide;
    • 4-amino-8-(3-methylsulfonylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3-cyanophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(2-pyridyl)cinnoline-3-carboxamide;
    • 4-amino-8-[3,5-bis(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(1H-pyrazol-4-yl)cinnoline-3-carboxamide;
    • 4-amino-8-[2-chloro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-methoxy-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-[2-(trifluoromethyl)phenyl]-cinnoline-3-carboxamide;
    • 4-amino-8-(5-chloro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(4-pyridyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(1-methyl-1H-pyrazol-4-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-3-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethyl-2H-pyrazol-3-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-[2-fluoro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-4-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-fluoro-2-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-5-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(5-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-fluorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-[4-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-[3-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide;
    • 4-amino-8-(6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxy-3,5-dimethyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxy-3-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(6-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(4-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(5-methoxy-2-methylphenyl)-N-propylcinnoline-3-carboxamide; and
    • 4-Amino-8-(2,4-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide;
      or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • In some embodiments, the present invention provides the following compounds: 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide; 4-amino-8-(2,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide; 4-amino-8-(4-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide; and 4-amino-8-(2-methoxy-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide; or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • In some embodiments, the present invention provides 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide, or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • In some embodiments, the present invention provides the following compounds:
    • 4-amino-8-(3,5-dimethyl-1H-pyrazol-4-yl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-[5-(azetidin-1-ylcarbonyl)-2-methoxyphenyl]-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(6-methoxy-2-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(1,3,5-trimethyl-1H-pyrazol-4-yl)cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(2,4,6-trifluoro-3-methoxyphenyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-5-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(1,3-dimethyl-1H-pyrazol-5-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2-fluoro-4,6-dimethoxyphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(3,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2,3-dihydro-1,4-benzodioxin-6-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(4,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(1,3-benzodioxol-4-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-[5-(azetidin-1-ylcarbonyl)-2-methylphenyl]-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(6-methoxy-4-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-7-chloro-8-(4-methoxypyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-7-fluoro-8-(4-methoxypyridin-3-yl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-7-chloro-8-(2-methoxy-5-methylphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-7-fluoro-8-(2-methoxy-5-methylphenyl)-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-7-chloro-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-7-chloro-N-propylcinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-N-propylcinnoline-3-carboxamide;
    • 4-amino-N-butyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide;
    • 4-amino-N-ethyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxypyridin-3-yl)-N-methylcinnoline-3-carboxamide;
    • 4-amino-N-butyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide;
    • 4-amino-N-ethyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2-methoxy-5-methylphenyl)-N-methylcinnoline-3-carboxamide;
    • 4-amino-N-butyl-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-ethylcinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-methylcinnoline-3-carboxamide;
    • 4-amino-N-butyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-ethylcinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-methylcinnoline-3-carboxamide;
    • 4-amino-8-(4-methoxypyridin-3-yl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide;
    • 4-amino-N-isobutyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide;
    • 4-amino-N-(2-hydroxypropyl)-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide;
    • 4-amino-8-(2-methoxy-5-methylphenyl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide;
    • 4-amino-N-isobutyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide;
    • 4-amino-N-(2-hydroxypropyl)-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-isobutylcinnoline-3-carboxamide;
    • 4-amino-8-(2,5-dimethoxyphenyl)-N-(2-hydroxypropyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-isobutylcinnoline-3-carboxamide; and
    • 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-(2-hydroxypropyl)cinnoline-3-carboxamide;
      or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • In some embodiments, the present invention provides the following compounds:
    • 4-amino-8-(2,3-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(2,4-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(3,4-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-(cyclopropylmethyl)-8-phenyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(p-tolyl)cinnoline-3-carboxamide;
    • 4-amino-8-(3-chlorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(4-chlorophenyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(o-tolyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-8-(m-tolyl)-N-propyl-cinnoline-3-carboxamide;
    • 4-amino-N-propyl-8-(3-thienyl)cinnoline-3-carboxamide; and
    • 4-amino-8-(2,6-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide;
      or a pharmaceutically acceptable salt thereof, or any subgroup thereof.
  • Compounds of the present invention also include pharmaceutically acceptable salts, tautomers and in vivo-hydrolysable precursors of the compounds of any of the formulas described herein. Compounds of the invention further include hydrates and solvates.
  • Compounds of the invention can be used as medicaments. In some embodiments, the present invention provides compounds of any of the formulas described herein, or pharmaceutically acceptable salts, tautomers or in vivo-hydrolysable precursors thereof, for use as medicaments. In some embodiments, the present invention provides compounds described herein for use as medicaments for treating or preventing an anxiety disorder, cognitive disorder, or mood disorder.
  • In some embodiments, the present invention provides compounds of any of the formulas described herein, or pharmaceutically acceptable salts, tautomers or in vivo-hydrolysable precursors thereof, in the manufacture of a medicament for the treatment or prophylaxis of an anxiety disorder, cognitive disorder, or mood disorder.
  • In some embodiments, the present invention provides a method for the treatment or prophylaxis of an anxiety disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof. As used herein, the phrase “anxiety disorder” includes, but is not limited to, one or more of the following: panic disorder, panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, social anxiety disorder, obsessive-compulsive disorder, posttraumatic stress disorder, acute stress disorder, generalized anxiety disorder, generalized anxiety disorder due to a general medical condition, and the like.
  • In some embodiments, the present invention provides a method for the treatment or prophylaxis of a cognitive disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof. As used herein, the phrase “cognitive disorder” includes, but is not limited to, one or more of the following: Alzheimer's disease, dementia, dementia due to Alzheimer's disease, dementia due to Parkinson's disease, and the like.
  • In some embodiments, the present invention provides a method for the treatment or prophylaxis of a mood disorder comprising administering to a mammal (including a human) a therapeutically effective amount of a compound of any of the formulas described herein, or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursor thereof. As used herein, the phrase “mood disorder” is a depressive disorder including, but is not limited to, one or more of the following: major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, mixed episodes associated with bipolar disorder, and the like.
  • Anxiety disorders, cognitive disorders, and mood disorders are defined, for example, in the American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision, Washington, D.C., American Psychiatric Association, 2000.
  • In some embodiments, the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including a human) a compound of any of the formulas described herein or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursors and a cognitive and/or memory enhancing agent.
  • In some embodiments, the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including a human) a compound of any of the formulas described herein or a pharmaceutically acceptable salt, tautomer or in vivo-hydrolysable precursors thereof wherein constituent members are provided herein, and a choline esterase inhibitor or anti-inflammatory agent.
  • In some embodiments, the present invention provides a method of treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), by administering to a mammal (including human) a compound of the present invention, and an atypical antipsychotic agent. Atypical antipsychotic agents include, but not limited to, Olanzapine (marketed as Zyprexa), Aripiprazole (marketed as Abilify), Risperidone (marketed as Risperdal), Quetiapine (marketed as Seroquel), Clozapine (marketed as Clozaril), Ziprasidone (marketed as Geodon) and Olanzapine/Fluoxetine (marketed as Symbyax).
  • In some embodiments, the mammal or human being treated with a compound of the present invention, has been diagnosed with a particular disease or disorder, such as those described herein. In these cases, the mammal or human being treated is in need of such treatment. Diagnosis, however, need not be previously performed.
  • The present invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of the invention herein together with at least one pharmaceutically acceptable carrier, diluent or excipient.
  • When used for pharmaceutical compositions, medicaments, manufacture of a medicament, or treating or preventing an anxiety disorder, cognitive disorder, or mood disorder (such as any of those described herein), compounds of the present invention include the compounds of any of the formulas described herein, and pharmaceutically acceptable salts, tautomers and in vivo-hydrolysable precursors thereof. Compounds of the present invention further include hydrates and solvates.
  • The definitions set forth in this application are intended to clarify terms used throughout this application. The term “herein” means the entire application.
  • As used in this application, the term “optionally substituted,” as used herein, means that substitution is optional and therefore it is possible for the designated atom or moiety to be unsubstituted. In the event a substitution is desired then such substitution means that any number of hydrogens on the designated atom or moiety is replaced with a selection from the indicated group, provided that the normal valency of the designated atom or moiety is not exceeded, and that the substitution results in a stable compound. For example, if a methyl group (i.e., CH3) is optionally substituted, then 3 hydrogens on the carbon atom can be replaced. Examples of suitable substituents include, but are not limited to: halogen, CN, NH2, OH, SO, SO2, COOH, OC1-6alkyl, CH2OH, SO2H, C1-6alkyl, OC1-6alkyl, C(═O)C1-6alkyl, C(═O)OC1-6alkyl, C(═O)NH2, C(═O)NHC1-6alkyl, C(═O)N(C1-6alkyl)2, SO2C1-6alkyl, SO2NHC1-6alkyl, SO2N(C1-6alkyl)2, NH(C1-6alkyl), N(C1-6alkyl)2, NHC(═O)C1-6alkyl, NC(═O)(C1-6alkyl)2, C5-6aryl, OC5-6aryl, C(═O)C5-6aryl, C(═O)OC5-6aryl, C(═O)NHC5-6aryl, C(═O)N(C5-6aryl)2, SO2C5-6aryl, SO2NHC5-6aryl, SO2N(C5-6aryl)2, NH(C5-6aryl), N(C5-6aryl)2, NC(═O)C5-6aryl, NC(═O)(C5-6aryl)2, C5-6heterocyclyl, OC5-6heterocyclyl, C(═O)C5-6heterocyclyl, C(═O)OC5-6heterocyclyl, C(═O)NHC5-6heterocyclyl, C(═O)N(C5-6heterocyclyl)2, SO2C5-6heterocyclyl, SO2NHC5-6heterocyclyl, SO2N(C5-6heterocyclyl)2, NH(C5-6heterocyclyl), N(C5-6heterocyclyl)2, NC(═O)C5-6heterocyclyl, NC(═O)(C5-6heterocyclyl)2.
  • A variety of compounds in the present invention may exist in particular stereoisomeric forms. The present invention takes into account all such compounds, including cis- and trans isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as being covered within the scope of this invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention. The compounds herein described may have asymmetric centers. Compounds of the present invention containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis from optically active starting materials. When required, separation of the racemic material can be achieved by methods known in the art. Many stereoisomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. All chiral, diastereomeric, racemic forms and all stereoisomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated.
  • The compounds of the invention may form isolable atropisomers in certain solvents (e.g. supercritical CO2 containing 25-35% methanol) at room temperature. The atropisomers of the compounds may be isolated using chiral LC. All atropisomers of a structure are intended, unless the specific atropisomer is specifically indicated.
  • When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • The term “Cm-n” or “Cm-n group” used alone or as a prefix, refers to any group having m to n carbon atoms.
  • The term “alkyl” used alone or as a suffix or prefix, refers to a saturated monovalent straight or branched chain hydrocarbon radical comprising 1 to about 12 carbon atoms. Illustrative examples of alkyls include, but are not limited to, C1-6alkyl groups, such as methyl, ethyl, propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, and hexyl, and longer alkyl groups, such as heptyl, and octyl.
  • The term “alkylene” used alone or as suffix or prefix, refers to divalent straight or branched chain hydrocarbon radicals comprising 1 to about 12 carbon atoms, which serves to links two structures together.
  • As used herein, “alkenyl” refers to an alkyl group having one or more double carbon-carbon bonds. Example alkenyl groups include ethenyl, propenyl, cyclohexenyl, and the like. The term “alkenylenyl” refers to a divalent linking alkenyl group.
  • As used herein, “alkynyl” refers to an alkyl group having one or more triple carbon-carbon bonds. Example alkynyl groups include ethynyl, propynyl, and the like. The term “alkynylenyl” refers to a divalent linking alkynyl group.
  • As used herein, “aromatic” refers to hydrocarbyl groups having one or more polyunsaturated carbon rings having aromatic characters, (e.g., 4n+2 delocalized electrons) and comprising up to about 14 carbon atoms.
  • As used herein, the term “aryl” refers to an aromatic ring structure made up of from 5 to 14 carbon atoms. Ring structures containing 5, 6, 7 and 8 carbon atoms would be single-ring aromatic groups, for example, phenyl. Ring structures containing 8, 9, 10, 11, 12, 13, or 14 would be a polycyclic moiety in which at least one carbon is common to any two adjoining rings therein (for example, the rings are “fused rings”), for example naphthyl. The aromatic ring can be substituted at one or more ring positions with such substituents as described above. The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are “fused rings”) wherein at least one of the rings is aromatic, for example, the other cyclic rings can be cycloalkyls, cycloalkenyls or cycloalkynyls. The terms ortho, meta and para apply to 1,2-, 1,3- and 1,4-disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and ortho-dimethylbenzene are synonymous.
  • The term “cycloalkyl,” used alone or as suffix or prefix, refers to a saturated monovalent ring-containing hydrocarbon radical comprising at least 3 up to about 12 carbon atoms. Examples of cycloalkyls include, but are not limited to, C3-7cycloalkyl groups, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl, and saturated cyclic and bicyclic terpenes. A cycloalkyl can be unsubstituted or substituted by one or two suitable substituents. Preferably, the cycloalkyl is a monocyclic ring or bicyclic ring.
  • As used herein, “cycloalkenyl” refers to ring-containing hydrocarbyl groups having at least one carbon-carbon double bond in the ring, and having from 3 to 12 carbons atoms.
  • As used herein, “halo” or “halogen” refers to fluoro, chloro, bromo, and iodo.
  • “Counterion” is used to represent a small, negatively or positively charged species such as chloride (Cl), bromide (Br), hydroxide (OH), acetate (CH3COO), sulfate (SO4 2−), tosylate (CH3-phenyl-SO3 ), benezensulfonate (phenyl-SO3—), sodium ion (Na+), potassium (R+), ammonium (NH4 +), and the like.
  • The term “heterocycle” used alone or as a suffix or prefix, refers to a ring-containing structure or molecule having one or more multivalent heteroatoms, independently selected from N, O, P and S, as a part of the ring structure and including at least 3 and up to about 20 atoms in the ring(s). Heterocycle may be saturated or unsaturated, containing one or more double bonds, and heterocycle may contain more than one ring. When a heterocycle contains more than one ring, the rings may be fused or unfused. Fused rings generally refer to at least two rings share two atoms therebetween. Heterocycle may have aromatic character or may not have aromatic character.
  • The term “heteroaromatic” used alone or as a suffix or prefix, refers to a ring-containing structure or molecule having one or more multivalent heteroatoms, independently selected from N, O, P and S, as a part of the ring structure and including at least 3 and up to about 20 atoms in the ring(s), wherein the ring-containing structure or molecule has an aromatic character (e.g., 4n+2 delocalized electrons).
  • The term “heterocyclic group,” “heterocyclic moiety,” “heterocyclic,” or “heterocyclo” used alone or as a suffix or prefix, refers to a radical derived from a heterocycle by removing one or more hydrogens therefrom.
  • The term “heterocyclyl” used alone or as a suffix or prefix, refers a monovalent radical derived from a heterocycle by removing one hydrogen therefrom.
  • The term “heterocyclylene” used alone or as a suffix or prefix, refers to a divalent radical derived from a heterocycle by removing two hydrogens therefrom, which serves to links two structures together.
  • The term “heteroaryl” used alone or as a suffix or prefix, refers to a heterocyclyl having aromatic character.
  • The term “heterocylcoalkyl” used alone or as a suffix or prefix, refers to a monocyclic or polycyclic ring comprising carbon and hydrogen atoms and at least one heteroatom, preferably, 1 to 3 heteroatoms selected from nitrogen, oxygen, and sulfur, and having no unsaturation. Examples of heterocycloalkyl groups include pyrrolidinyl, pyrrolidino, piperidinyl, piperidino, piperazinyl, piperazino, morpholinyl, morpholino, thiomorpholinyl, thiomorpholino, and pyranyl. A heterocycloalkyl group can be unsubstituted or substituted with one or two suitable substituents. Preferably, the heterocycloalkyl group is a monocyclic or bicyclic ring, more preferably, a monocyclic ring, wherein the ring comprises from 3 to 6 carbon atoms and form 1 to 3 heteroatoms, referred to herein as C3-6heterocycloalkyl.
  • The term “heteroarylene” used alone or as a suffix or prefix, refers to a heterocyclylene having aromatic character.
  • The term “heterocycloalkylene” used alone or as a suffix or prefix, refers to a heterocyclylene that does not have aromatic character.
  • The term “six-membered” used as prefix refers to a group having a ring that contains six ring atoms.
  • The term “five-membered” used as prefix refers to a group having a ring that contains five ring atoms.
  • A five-membered ring heteroaryl is a heteroaryl with a ring having five ring atoms wherein 1, 2 or 3 ring atoms are independently selected from N, O and S.
  • Exemplary five-membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4-thiadiazolyl, and 1,3,4-oxadiazolyl.
  • A six-membered ring heteroaryl is a heteroaryl with a ring having six ring atoms wherein 1, 2 or 3 ring atoms are independently selected from N, O and S.
  • Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl and pyridazinyl.
  • Examples of heterocyclyls include, but are not limited to, 1H-indazole, 2-pyrrolidonyl, 2H, 6H-1,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azabicyclo, azetidine, azepane, aziridine, azocinyl, benzimidazolyl, benzodioxol, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, b-carbolinyl, chromanyl, chromenyl, cinnolinyl, diazepane, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dioxolane, furyl, 2,3-dihydrofuran, 2,5-dihydrofuran, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, homopiperidinyl, imidazolidine, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxirane, oxazolidinylperimidinyl, phenanthridinyl, phenanthrolinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, piperidonyl, 4-piperidonyl, purinyl, pyranyl, pyrrolidinyl, pyrroline, pyrrolidine, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, N-oxide-pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolidinyl dione, pyrrolinyl, pyrrolyl, pyridine, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, carbolinyl, tetrahydrofuranyl, tetramethylpiperidinyl, tetrahydroquinoline, tetrahydroisoquinolinyl, thiophane, thiotetrahydroquinolinyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, thiirane, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, xanthenyl.
  • As used herein, “alkoxy” or “alkyloxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, n-pentoxy, isopentoxy, cyclopropylmethoxy, allyloxy and propargyloxy. Similarly, “alkylthio” or “thioalkoxy” represent an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge.
  • “Halogenated,” used as a prefix of a group, means one or more hydrogens on the group is replaced with one or more halogens.
  • As used herein, the term “carbonyl” is art recognized and includes the —C(═O) groups of such moieties as can be represented by the general formula:
  • Figure US20090018112A1-20090115-C00006
  • wherein X is a bond or represents an oxygen or sulfur, and R represents a hydrogen, an alkyl, an alkenyl, —(CH2)m—R″ or a pharmaceutically acceptable salt, R′ represents a hydrogen, an alkyl, an alkenyl or —(CH2)m—R″, where m is an integer less than or equal to ten, and R″ is alkyl, cycloalkyl, alkenyl, aryl, or heteroaryl. Where X is an oxygen and R and R′ is not hydrogen, the formula represents an “ester”. Where X is an oxygen, and R is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R′ is a hydrogen, the formula represents a “carboxylic acid.” Where X is oxygen, and R′ is a hydrogen, the formula represents a “formate.” In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a “thiolcarbonyl” group. Where X is a sulfur and R and R′ is not hydrogen, the formula represents a “thiolester.” Where X is sulfur and R is hydrogen, the formula represents a “thiolcarboxylic acid.” Where X is sulfur and R′ is hydrogen, the formula represents a “thiolformate.” On the other hand, where X is a bond, and R is not a hydrogen, the above formula represents a “ketone” group. Where X is a bond, and R is hydrogen, the above formula is represents an “aldehyde” group.
  • As used herein, the term “sulfonyl” refers to the —S(═O)2— of a moiety that can be represented by the general formula:
  • Figure US20090018112A1-20090115-C00007
  • wherein R is represented by but not limited to hydrogen, alkyl, cycloalkyl, alkenyl, aryl, heteroaryl, aralkyl, or heteroaralkyl.
  • As used herein, some substituents are described in a combination of two or more groups. For example, the expression of “C(═O)C3-9cycloalkylRd” is meant to refer to a structure:
  • Figure US20090018112A1-20090115-C00008
  • wherein p is 1, 2, 3, 4, 5, 6 or 7 (i.e., C3-9cycloalkyl); the C3-9cycloalkyl is substituted by Rd; and the point of attachment of the “C(═O)C3-9cycloalkylRd” is through the carbon atom of the carbonyl group, which is on the left of the expression.
  • As used herein, the phrase “protecting group” means temporary substituents which protect a potentially reactive functional group from undesired chemical transformations. Examples of such protecting groups include esters of carboxylic acids, silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones respectively. The field of protecting group chemistry has been reviewed (Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 3rd ed.; Wiley: New York, 1999).
  • As used herein, “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • As used herein, “pharmaceutically acceptable salts” refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof (i.e., also include counterions). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, phosphoric, and the like; and the salts prepared from organic acids such as lactic, maleic, citric, benzoic, methanesulfonic, and the like.
  • The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile can be used.
  • As used herein, “in vivo hydrolysable precursors” means an in vivo hydrolysable (or cleavable) ester of a compound of any of the formulas described herein that contains a carboxy or a hydroxy group. For example amino acid esters, C1-6 alkoxymethyl esters like methoxymethyl; C1-6alkanoyloxymethyl esters like pivaloyloxymethyl; C3-8cycloalkoxycarbonyloxy C1-6alkyl esters like 1-cyclohexylcarbonyloxyethyl, acetoxymethoxy, or phosphoramidic cyclic esters.
  • As used herein, “tautomer” means other structural isomers that exist in equilibrium resulting from the migration of a hydrogen atom. For example, keto-enol tautomerism where the resulting compound has the properties of both a ketone and an unsaturated alcohol.
  • As used herein “stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • The present invention further includes isotopically-labeled compounds of the invention. An “isotopically” or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 18F, 35S, 36Cl, 82Br, 75Br, 76Br, 77Br, 123I, 124, 125I and 131I. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro receptor labeling and competition assays, compounds that incorporate 3H, 14C, 82Br, 125I, 131I, 35S or will generally be most useful. For radio-imaging applications 11C, 18F, 125I, 123I, 124I, 131I, 75Br, 76Br or 77Br will generally be most useful.
  • It is understood that a “radio-labeled compound” is a compound that has incorporated at least one radionuclide. In some embodiments the radionuclide is selected from the group consisting of 3H, 14C, 125I, 35S and 82Br.
  • The antidementia treatment defined herein may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional chemotherapy.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Such combination products employ the compounds of this invention.
  • Compounds of the present invention may be administered orally, parenteral, buccal, vaginal, rectal, inhalation, insufflation, sublingually, intramuscularly, subcutaneously, topically, intranasally, intraperitoneally, intrathoracially, intravenously, epidurally, intrathecally, intracerebroventricularly and by injection into the joints.
  • The dosage will depend on the route of administration, the severity of the disease, age and weight of the patient and other factors normally considered by the attending physician, when determining the individual regimen and dosage level as the most appropriate for a particular patient.
  • An effective amount of a compound of the present invention for use in therapy of dementia is an amount sufficient to symptomatically relieve in a warm-blooded animal, particularly a human the symptoms of dementia, to slow the progression of dementia, or to reduce in patients with symptoms of dementia the risk of getting worse.
  • For preparing pharmaceutical compositions from the compounds of this invention, inert, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • A solid carrier can be one or more substances, which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material.
  • In powders, the carrier is a finely divided solid, which is in a mixture with the finely divided active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • For preparing suppository compositions, a low-melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient sized molds and allowed to cool and solidify.
  • Suitable carriers include magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low-melting wax, cocoa butter, and the like.
  • Some of the compounds of the present invention are capable of forming salts with various inorganic and organic acids and bases and such salts are also within the scope of this invention. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, phosphoric, and the like; and the salts prepared from organic acids such as lactic, maleic, citric, benzoic, methanesulfonic, trifluoroacetate and the like.
  • In some embodiments, the present invention provides a compound of any of the formulas described herein or a pharmaceutically acceptable salt thereof for the therapeutic treatment (including prophylactic treatment) of mammals including humans, it is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
  • In addition to the compounds of the present invention, the pharmaceutical composition of this invention may also contain, or be co-administered (simultaneously or sequentially) with, one or more pharmacological agents of value in treating one or more disease conditions referred to herein.
  • The term composition is intended to include the formulation of the active component or a pharmaceutically acceptable salt with a pharmaceutically acceptable carrier. For example this invention may be formulated by means known in the art into the form of, for example, tablets, capsules, aqueous or oily solutions, suspensions, emulsions, creams, ointments, gels, nasal sprays, suppositories, finely divided powders or aerosols or nebulisers for inhalation, and for parenteral use (including intravenous, intramuscular or infusion) sterile aqueous or oily solutions or suspensions or sterile emulsions.
  • Liquid form compositions include solutions, suspensions, and emulsions. Sterile water or water-propylene glycol solutions of the active compounds may be mentioned as an example of liquid preparations suitable for parenteral administration. Liquid compositions can also be formulated in solution in aqueous polyethylene glycol solution. Aqueous solutions for oral administration can be prepared by dissolving the active component in water and adding suitable colorants, flavoring agents, stabilizers, and thickening agents as desired. Aqueous suspensions for oral use can be made by dispersing the finely divided active component in water together with a viscous material such as natural synthetic gums, resins, methyl cellulose, sodium carboxymethyl cellulose, and other suspending agents known to the pharmaceutical formulation art.
  • The pharmaceutical compositions can be in unit dosage form. In such form, the composition is divided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparations, for example, packeted tablets, capsules, and powders in vials or ampoules. The unit dosage form can also be a capsule, cachet, or tablet itself, or it can be the appropriate number of any of these packaged forms.
  • Compositions may be formulated for any suitable route and means of administration. Pharmaceutically acceptable carriers or diluents include those used in formulations suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural) administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • For solid compositions, conventional non-toxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, cellulose, cellulose derivatives, starch, magnesium stearate, sodium saccharin, talcum, glucose, sucrose, magnesium carbonate, and the like may be used. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc, an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, sorbitan monolaurate, triethanolamine oleate, etc. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975.
  • The compounds of the invention may be derivatised in various ways. As used herein “derivatives” of the compounds includes salts (e.g. pharmaceutically acceptable salts), any complexes (e.g. inclusion complexes or clathrates with compounds such as cyclodextrins, or coordination complexes with metal ions such as Mn2+ and Zn2+), esters such as in vivo hydrolysable esters, free acids or bases, polymorphic forms of the compounds, solvates (e.g. hydrates), prodrugs or lipids, coupling partners and protecting groups. By “prodrugs” is meant for example any compound that is converted in vivo into a biologically active compound.
  • Salts of the compounds of the invention are preferably physiologically well tolerated and non toxic. Many examples of salts are known to those skilled in the art. All such salts are within the scope of this invention, and references to compounds include the salt forms of the compounds.
  • Compounds having acidic groups, such as carboxylate, phosphates or sulfates, can form salts with alkaline or alkaline earth metals such as Na, K, Mg and Ca, and with organic amines such as triethylamine and Tris (2-hydroxyethyl)amine. Salts can be formed between compounds with basic groups, e.g. amines, with inorganic acids such as hydrochloric acid, phosphoric acid or sulfuric acid, or organic acids such as acetic acid, citric acid, benzoic acid, fumaric acid, or tartaric acid. Compounds having both acidic and basic groups can form internal salts.
  • Acid addition salts may be formed with a wide variety of acids, both inorganic and organic. Examples of acid addition salts include salts formed with hydrochloric, hydriodic, phosphoric, nitric, sulphuric, citric, lactic, succinic, maleic, malic, isethionic, fumaric, benzenesulphonic, toluenesulphonic, methanesulphonic, ethanesulphonic, naphthalenesulphonic, valeric, acetic, propanoic, butanoic, malonic, glucuronic and lactobionic acids.
  • If the compound is anionic, or has a functional group which may be anionic (e.g., COOH may be COO), then a salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Na+ and K+, alkaline earth cations such as Ca2+ and Mg2+, and other cations such as Al3+. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e., NH4 +) and substituted ammonium ions (e.g., NH3R+, NH2R2 +, NHR3 +, NR4 +). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4 +.
  • Where the compounds contain an amine function, these may form quaternary ammonium salts, for example by reaction with an alkylating agent according to methods well known to the skilled person. Such quaternary ammonium compounds are within the scope of the invention.
  • Compounds containing an amine function may also form N-oxides. A reference herein to a compound that contains an amine function also includes the N-oxide.
  • Where a compound contains several amine functions, one or more than one nitrogen atom may be oxidised to form an N-oxide. Particular examples of N-oxides are the N-oxides of a tertiary amine or a nitrogen atom of a nitrogen-containing heterocycle.
  • N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a per-acid (e.g. a peroxycarboxylic acid), see for example Advanced Organic Chemistry, by Jerry March, 4th Edition, Wiley Interscience, pages. More particularly, N-oxides can be made by the procedure of L. W. Deady (Syn. Comm. 1977, 7, 509-514) in which the amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.
  • Esters can be formed between hydroxyl or carboxylic acid groups present in the compound and an appropriate carboxylic acid or alcohol reaction partner, using techniques well known in the art. Examples of esters are compounds containing the group C(═O)OR, wherein R is an ester substituent, for example, a C1-7 alkyl group, a C3-20 heterocyclyl group, or a C5-20 aryl group, preferably a C1-7 alkyl group. Particular examples of ester groups include, but are not limited to, C(═O)OCH3, C(═O)OCH2CH3, C(═O)OC(CH3)3, and —C(═O)OPh. Examples of acyloxy (reverse ester) groups are represented by OC(═O)R, wherein R is an acyloxy substituent, for example, a C17 alkyl group, a C320 heterocyclyl group, or a C520 aryl group, preferably a C17 alkyl group. Particular examples of acyloxy groups include, but are not limited to, OC(═O)CH3 (acetoxy), OC(═O)CH2CH3, OC(═O)C(CH3)3, OC(═O)Ph, and OC(═O)CH2Ph.
  • Derivatives which are prodrugs of the compounds are convertible in vivo or in vitro into one of the parent compounds. Typically, at least one of the biological activities of compound will be reduced in the prodrug form of the compound, and can be activated by conversion of the prodrug to release the compound or a metabolite of it. Some prodrugs are esters of the active compound (e.g., a physiologically acceptable metabolically labile ester). During metabolism, the ester group (—C(═O)OR) is cleaved to yield the active drug. Such esters may be formed by esterification, for example, of any of the carboxylic acid groups (—C(═O)OH) in the parent compound, with, where appropriate, prior protection of any other reactive groups present in the parent compound, followed by deprotection if required.
  • Examples of such metabolically labile esters include those of the formula —C(═O)OR wherein R is: C17alkyl (e.g., Me, Et, -nPr, -iPr, -nBu, -sBu, -iBu, tBu); C17aminoalkyl (e.g., aminoethyl; 2-(N,N-diethylamino)ethyl; 2(4morpholino)ethyl); and acyloxy-C17alkyl (e.g., acyloxymethyl; acyloxyethyl; pivaloyloxymethyl; acetoxymethyl; 1acetoxyethyl; 1-(1-methoxy-1-methyl)ethyl-carbonyloxyethyl; 1-(benzoyloxy)ethyl; isopropoxy-carbonyloxymethyl; 1isopropoxy-carbonyloxyethyl; cyclohexyl-carbonyloxymethyl; 1cyclohexyl-carbonyloxyethyl; cyclohexyloxy-carbonyloxymethyl; 1-cyclohexyloxy-carbonyloxyethyl; (4-tetrahydropyranyloxy) carbonyloxymethyl; 1-(4-tetrahydropyranyloxy)carbonyloxyethyl; (4-tetrahydropyranyl)carbonyloxymethyl; and 1(4-tetrahydropyranyl)carbonyloxyethyl).
  • Also, some prodrugs are activated enzymatically to yield the active compound, or a compound which, upon further chemical reaction, yields the active compound (for example, as in ADEPT, GDEPT, LIDEPT, etc.). For example, the prodrug may be a sugar derivative or other glycoside conjugate, or may be an amino acid ester derivative.
  • Other derivatives include coupling partners of the compounds in which the compounds is linked to a coupling partner, e.g. by being chemically coupled to the compound or physically associated with it. Examples of coupling partners include a label or reporter molecule, a supporting substrate, a carrier or transport molecule, an effector, a drug, an antibody or an inhibitor. Coupling partners can be covalently linked to compounds of the invention via an appropriate functional group on the compound such as a hydroxyl group, a carboxyl group or an amino group. Other derivatives include formulating the compounds with liposomes.
  • Where the compounds contain chiral centres, all individual optical forms such as enantiomers, epimers, atropisomers and diastereoisomers, as well as racemic mixtures of the compounds are within the scope of the invention.
  • Compounds may exist in a number of tautomeric forms and references to compounds include all such forms. For the avoidance of doubt, where a compound can exist in one of several tautomeric forms and only one is specifically described or shown, all others are nevertheless embraced by the scope of this invention.
  • The quantity of the compound to be administered will vary for the patient being treated and will vary from about 100 ng/kg of body weight to 100 mg/kg of body weight per day and preferably will be from 10 pg/kg to 10 mg/kg per day. For instance, dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art. Thus, the skilled artisan can readily determine the amount of compound and optional additives, vehicles, and/or carrier in compositions and to be administered in methods of the invention.
  • In some embodiments, the compounds described herein are central nervous system depressants and may be used as tranquilizers or ataractic agents for the relief of anxiety and tension states, for example, in mice, cats, rats, dogs and other mammalian species such as humans, in the same manner as chlordiazepoxide. For this purpose a compound or mixture of compounds of any of the formulas described herein, or non-toxic physiologically acceptable salts, such as acid addition salts thereof, may be administered orally or parenterally in a conventional dosage form such as tablet, pill, capsule, injectable or the like. The dosage in mg/kg of body weight of compounds of the present invention in mammals will vary according to the size of the animal and particularly with respect to the brain/body weight ratio. In general, a higher mg/kg dosage for a small animal such as a dog will have the same effect as a lower mg/kg dosage in an adult human. A minimum effective dosage for a compound of formula (I) will be at least about 0.1 mg/kg of body weight per day for mammals with a maximum dosage for a small mammal such as a dog, of about 100 mg/kg per day. For humans, a dosage of about 0.1 to 12 mg/kg per day will be effective, for example, about 5 to 600 mg/day for an average man. The dosage can be given once daily or in divided doses, for example, 2 to 4 doses daily, and such dosage will depend on the duration and maximum level of activity of a particular compound. The dose may be conventionally formulated in an oral or parenteral dosage form by compounding about 5 to 250 mg per unit of dosage of conventional vehicle, excipient, binder, preservative, stabilizer, flavor or the like as called for by accepted pharmaceutical practice, for example, as described in U.S. Pat. No. 3,755,340. The compounds of this invention may be used in pharmaceutical compositions comprising a compound of any of the formulas described herein or can be contained in the same formulation with or co-administered with one or more known drugs.
  • Some example tests that can be conducted to demonstrate the anxiolytic activity of the present compounds include binding tests of GABAA receptors. In some embodiments, the binding test was directed to a subtype of GABAA receptors, such as GABAA1 receptors (i.e., those containing the α1 subunit), GABAA2 receptors (i.e., those containing the α2 subunit), GABAA3 receptors (i.e., those containing the α3 subunit) and GABAA5 receptors (i.e., those containing the α5 subunit).
  • Presently available GABAA modulator anxiolytics work via interactions at the classical benzodiazepine binding site. To a large degree these anxiolytics lack GABAA receptor subtype-selectivity. The subtype-selective GABAA receptor modulators may offer more advantages. For example, a growing body of work suggests that desirable anxiolytic activity is driven primarily by interactions with GABAA receptors containing the α2 subunit. Sedation, a side-effect common to all marketed benzodiazepines, is believed to be mediated by interactions at GABAARs containing the α1 subunit. To develop anxiolytics with minimal liabilities due to interactions with other subunits, an electrophysiological assay was developed to screen modulatory effects of various compounds on different GABA subunit combinations heterologously expressed in Xenopus oocytes.
  • GABAA receptors were heterologously expressed in Xenopus oocytes by injecting cRNA corresponding to human α1, α2, α3, α5, β2, β3 and γ2 subunits of the GABAA receptor genes. The specific subunit combinations (subtypes) were as follows: α1β2γ2, α2β3γ2, α3β3γ2, and α5β3γ2. The EC10 of GABA was approximated for each cell. Stability of GABA-mediated (EC10) current was established. Modulatory effect of test compound was determined and compared across subtypes. The assay developed has reproducibility which allows discrimination of modulatory activity down to minimal effect of about 25% potentiation (prior to normalization to standard) for all four subtypes. Thus, the assay can characterize modulatory effects and determine subtype selectivity of test compounds on major subtypes of GABAA receptors. In some embodiments, a compound can selectively bind to one subtype of GABAA receptor (by showing about 25% or more of binding comparing to another subtype of GABAA receptor).
  • Anxiolytic activity is indicated in the GABAA binding test by a displacement of the flunitrazepam such as is exhibited by benzodiazepines or by enhancement of the binding such as is shown by cartazolate and tracazolate.
  • In some embodiments, the compounds of the invention can bind to GABAA receptors. In some embodiments, the compounds of the invention can bind to GABAA receptors by displacement of benzodiazepines. Accordingly, the compounds of the invention can be used to modulate activities of GABAA receptors. In some embodiments, the compounds of the invention can selectively bind to a subtype of GABAA receptors, such as such as GABAA1 receptors (i.e., those containing the α1 subunit), GABAA2 receptors (i.e., those containing the γ2 subunit), GABAA3 receptors (i.e., those containing the γ3 subunit) or GABAA5 receptors (i.e., those containing the γ5 subunit). In some embodiments, the compounds of the invention can selectively bind to a subtype of GABAA receptors by displacement of benzodiazepines. Accordingly, the compounds of the invention can be used to selectively modulate activities of a subtype of GABAA receptors, such as GABAA1 receptors, GABAA2 receptors, GABAA3 receptors or GABAA5 receptors.
  • In some embodiments, certain compounds of the invention are GABAA1 receptor antagonists and GABAA2 receptor agonists.
  • Because the compounds of the invention can be used to modulate activities of GABAA receptors, or to selectively modulate activities of a subtype of GABAA receptors, the compounds of the invention are envisioned to be useful for treating or preventing diseases mediated by GABAA receptors or a subtype of GABAA receptors. Such disease, include, but is not limited to, stroke, head trauma, epilepsy, pain, migraine, mood disorders, anxiety, post traumatic stress disorder, obsessive compulsive disorders, schizophrenia, seizures, convulsions, tinnitus, neurodegenerative disorders including Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's Chorea, Parkinson's disease, depression, bipolar disorders, mania, trigeminal and other neuralgia, neuropathic pain, hypertension, cerebral ischemia, cardiac arrhythmia, myotonia, substance abuse, myoclonus, essential tremor, dyskinesia and other movement disorders, neonatal cerebral hemorrhage, spasticity, cognitive disorder, and sleeping disorder.
  • It is known that melatonin receptor agonists are effective in treating depression. We find that the compounds of the invention can selectively modulate activities of a subtype of melatonin receptors, melatonin receptor 1 (MT-1). In certain embodiments, certain compounds of the invention are MT1 agonists. As a results, the compounds of the invention may be effective in treating depression disorders such as major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, or mixed episodes associated with bipolar disorder. To treat depression disorders, an effective amount of one or more compounds of the invention is administered to a patient with such a need.
  • In another embodiment, certain compounds of the present invention may be effective in treating insomnia.
  • In a further embodiment, a compound of formula I or a pharmaceutically acceptable salt, solvate or in vivo hydrolysable ester thereof, or a pharmaceutical composition or formulation comprising a compound of formula I may be administered concurrently, simultaneously, sequentially or separately with one or more pharmaceutically active compound(s) selected from the following:
  • (i) antidepressants such as amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, protriptyline, reboxetine, robalzotan, sertraline, sibutramine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (ii) atypical antipsychotics including for example quetiapine and pharmaceutically active isomer(s) and metabolite(s) thereof, amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, lithium, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, risperidone, quetiapine, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone and equivalents thereof;
  • (iii) antipsychotics including for example amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (iv) anxiolytics including for example alnespirone, azapirones, benzodiazepines, barbiturates such as adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, zolazepam and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (v) anticonvulsants including, for example, carbamazepine, valproate, lamotrogine, gabapentin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (vi) Alzheimer's therapies including, for example, donepezil, memantine, tacrine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (vii) Parkinson's therapies including, for example, deprenyl, L-dopa, Requip, Mirapex, MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (viii) migraine therapies including, for example, almotriptan, amantadine, bromocriptine, butalbital, cabergoline, dichloralphenazone, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, zolmitriptan, zomitriptan, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (ix) stroke therapies including, for example, abciximab, activase, NXY-059, citicoline, crobenetine, desmoteplase, repinotan, traxoprodil and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (x) over active bladder urinary incontinence therapies including, for example, darafenacin, falvoxate, oxybutynin, propiverine, robalzotan, solifenacin, tolterodine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (xi) neuropathic pain therapies including, for example, gabapentin, lidoderm, pregablin and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (xii) nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, paracetamol and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
  • (xiii) insomnia therapies including, for example, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, etomidate, glutethimide, halazepam, hydroxyzine, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, roletamide, triclofos, secobarbital, zaleplon, zolpidem and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof, and
  • (xiv) mood stabilizers including, for example, carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, verapamil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • Such combinations employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within approved dosage ranges and/or the dosage described in the publication reference.
  • General Procedures for Making the Compounds of the Invention is as Follows:
  • The invention will now be illustrated by the following non-limiting examples, in which, unless stated otherwise:
  • In order that the invention disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the invention in any manner.
  • Some example compounds of the invention in table 1 were made according to the methods described herein below.
  • TABLE 1
    Example Synthesis
    Number Method Compound Name Structure
    1 F 4-amino-7-fluoro-8-phenyl-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00009
    2 F 4-amino-7-chloro-8-phenyl-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00010
    3 F 4-amino-7-methoxy-8-phenyl-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00011
    4 A 4-amino-7-chloro-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00012
    5 A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00013
    6 A 4-amino-8-(5-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00014
    7 A 4-amino-8-(2-methoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00015
    8 A 4-amino-8-(3-fluoro-2-methoxy-phenyl)-N-propyl-N-cinnoline-3 -carboxamide
    Figure US20090018112A1-20090115-C00016
    9 A 4-amino-8-[4-methoxy-2-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00017
    10 A 4-amino-8-(2,5-difluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00018
    11 A 4-amino-8-(5-fluoro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00019
    12 A 4-amino-8-(5-chloro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00020
    13 B 4-amino-8-(3,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00021
    14 B 4-amino-8-(3,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00022
    15 D 4-amino-8-(5-azetidin-1-ylcarbonyl-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00023
    16 A 4-amino-8-(2,3-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00024
    17 A 4-amino-8-(4-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00025
    18 A 4-amino-8-(3-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00026
    19 A 4-amino-8-(3,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00027
    20 B 4-amino-8-(2,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00028
    21 A 4-amino-8-(3,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00029
    22 A 4-amino-8-(2,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00030
    23 E 4-amino-8-(2-fluoro-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00031
    24 A 4-amino-8-(2,3-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00032
    25 A 4-amino-8-(2,3-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00033
    26 A 4-amino-N-propyl-8-(6-quinolyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00034
    27 A 4-amino-N-propyl-8-(3-quinolyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00035
    28 A 4-amino-8-(2-naphthyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00036
    29 A 4-amino-8-(1H-indol-5-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00037
    30 B 4-amino-8-(4-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00038
    31 A 4-amino-8-(3-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00039
    32 A 4-amino-N-propyl-8-(3,4,5-trimethoxyphenyl)-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00040
    33 A 4-amino-8-(2,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00041
    34 A 4-amino-8-(3,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00042
    35 A 4-amino-N-propyl-8-(2,3,4-trimethoxyphenyl)-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00043
    36 A 4-amino-8-(2-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00044
    37 A 4-amino-8-(2,6-dimethoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00045
    38 B 4-amino-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00046
    39 B 3-[4-amino-3-(propylcarbamoyl)cinnolin-8-yl]benzoic acid
    Figure US20090018112A1-20090115-C00047
    40 4-amino-8-(3-azetidin-1-ylcarbonylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00048
    41 C 4-amino-N-propyl-8-pyrazin-2-yl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00049
    42 A 4-amino-N-propyl-8-(3-pyridyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00050
    43 A 4-amino-8-(3-methylsulfonylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00051
    44 A 4-amino-8-(3-cyanophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00052
    45 C 4-amino-N-propyl-8-(2-pyridyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00053
    46 A 4-amino-8-[3,5-bis(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00054
    47 A 4-amino-N-propyl-8-(1H-pyrazol-4-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00055
    48 A 4-amino-8-[2-chloro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00056
    49 A 4-amino-8-(2-methoxy-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00057
    50 A 4-amino-N-propyl-8-[2-(trifluoromethyl)phenyl]-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00058
    51 A 4-amino-8-(5-chloro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00059
    52 C 4-amino-N-propyl-8-(4-pyridyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00060
    53 A 4-amino-8-(2,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00061
    54 A 4-amino-8-(2,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00062
    55 A 4-amino-8-(1-methyl-1H-pyrazol-4-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00063
    56 A 4-amino-8-(2-fluoro-3-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00064
    57 A 4-amino-8-(2,5-dimethyl-2H-pyrazol-3-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00065
    58 A 4-amino-8-[2-fluoro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00066
    59 A 4-amino-8-(2-fluoro-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00067
    60 A 4-amino-8-(2-fluoro-4-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00068
    61 A 4-amino-8-(5-fluoro-2-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00069
    62 A 4-amino-8-(4-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00070
    63 A 4-amino-8-(3-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00071
    64 A 4-amino-8-(2-fluoro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00072
    65 A 4-amino-8-(2-fluoro-5-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00073
    66 A 4-amino-8-(5-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00074
    67 F 4-amino-8-(4-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00075
    68 F 4-amino-8-(4-fluorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00076
    69 F 4-amino-N-propyl-8-[4-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00077
    70 F 4-amino-N-propyl-8-[3-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00078
    71 A 4-amino-8-(6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00079
    72 A 4-amino-8-(4-methoxy-3,5-dimethyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00080
    73 A 4-amino-8-(4-methoxy-3-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00081
    74 A 4-amino-8-(2-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00082
    75 C 4-amino-8-(6-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00083
    76 A 4-amino-8-(4-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00084
    77 A 4-amino-8-(5-methoxy-2-methylphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00085
    78 A 4-Amino-8-(2,4-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00086
    79 A 4-amino-8-(2,5-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00087
    80 A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00088
    81 A 4-amino-N-ethyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00089
    82 A 4-amino-N-butyl-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00090
    83 A 4-amino-8-(2,5-dimethoxyphenyl)-N-ethylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00091
    84 B 4-amino-8-(2,5-dimethoxyphenyl)-N-methylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00092
    85 B 4-amino-N-butyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00093
    86 B 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-ethylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00094
    87 A 4-Amino-8-(2,5-dimethoxy-phenyl)-cinnoline-3-carboxylic acid allylamide
    Figure US20090018112A1-20090115-C00095
    88 4-amino-N-(cyclopropylmethyl)-8-phenyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00096
    89 A 4-amino-8-(m-tolyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00097
    90 A 4-Amino-8-(2-fluoro-6-methylpyridin-3-yl)-cinnoline-3-carboxylic acidpropylamide
    Figure US20090018112A1-20090115-C00098
    91 A 4-Amino-7-fluoro-8-(5-fluoro-2-methoxyphenyl)-cinnoline-3-carboxylic acidpropylamide
    Figure US20090018112A1-20090115-C00099
    92 A 4-Amino-8-(2-chloro-5-methoxyphenyl)-7-fluoro-cinnoline-3-carboxylic acidpropylamide
    Figure US20090018112A1-20090115-C00100
    93 A 4-amino-N-cyclopropyl-8-(2,6-dimethoxypyridin-3-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00101
    94 A 4-amino-N-cyclopropyl-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00102
    95 A 4-amino-N-cyclopropyl-8-(2,4-dimethoxyphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00103
    96 A 4-amino-N-cyclopropyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00104
    97 A 4-amino-N-cyclopropyl-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00105
    98 A 4-amino-N-ethyl-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00106
    99 G 4-amino-7-fluoro-8-(2-fluoro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00107
    100 G 4-amino-7-cyano-8-(2,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00108
    101 H 4-amino-N-cyclobutyl-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00109
    102 H 4-amino-N-cyclopropyl-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00110
    103 H 4-amino-8-(2-chloro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00111
    104 A 4-amino-7-fluoro-8-(2-fluoro-3-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00112
    105 A 4-amino-7-fluoro-8-(3-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00113
    106 A 4-amino-8-(3,5-difluoro-2-methoxy-phenyl)-7-fluoro-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00114
    107 A 4-amino-7-fluoro-8-(4-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00115
    108 A 4-amino-7-fluoro-8-(2-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00116
    109 A 4-amino-8-(4-chlorophenyl)-7-fluoro-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00117
    110 A 4-amino-7-fluoro-8-(5-fluoro-2-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00118
    111 A 4-amino-8-(2,3-dimethylphenyl)-7-fluoro-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00119
    112 A 4-amino-8-(2,5-dimethoxyphenyl)-N-(3,3,3-trifluoropropyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00120
    113 A 4-amino-8-(2,5-difluorophenyl)-7-fluoro-N--propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00121
  • The compounds in Table 2 can also be made according to the methods described herein below.
  • TABLE 2
    Synthesis
    Method Compound Name Structure
    E 4-amino-8-(3,5-dimethyl-1H-pyrazol-4-yl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00122
    E 4-amino-8-(3,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00123
    E 4-amino-8-[5-(azetidin-1-ylcarbonyl)-2-methoxyphenyl]-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00124
    E 4-amino-8-(6-methoxy-2-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00125
    E 4-amino-N-propyl-8-(1,3,5-trimethyl-1H-pyrazol-4-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00126
    A 4-amino-N-propyl-8-(2,4,6-trifluoro-3-methoxyphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00127
    E 4-amino-8-(2-fluoro-5-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00128
    E 4-amino-8-(1,3-dimethyl-1H-pyrazol-5-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00129
    E 4-amino-8-(2-fluoro-4,6-dimethoxyphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00130
    A 4-amino-8-(3,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00131
    A 4-amino-8-(2,3-dihydro-1,4-benzodioxin-6-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00132
    E 4-amino-8-(4,5-difluoro-2-methoxyphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00133
    E 4-amino-8-(1,3-benzodioxol-4-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00134
    E 4-amino-8-[5-(azetidin-1-ylcarbonyl)-2-methylphenyl]-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00135
    E 4-amino-8-(6-methoxy-4-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00136
    A 4-amino-7-chloro-8-(4-methoxypyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00137
    A 4-amino-7-fluoro-8-(4-methoxypyridin-3-yl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00138
    A 4-amino-7-chloro-8-(2-methoxy-5-methylphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00139
    A 4-amino-7-fluoro-8-(2-methoxy-5-methylphenyl)-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00140
    A 4-amino-8-(2,5-dimethoxyphenyl)-7-chloro-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00141
    A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-7-chloro-N-propylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00142
    A 4-amino-N-butyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00143
    A 4-amino-8-(4-methoxypyridin-3-yl)-N-methylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00144
    A 4-amino-N-butyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00145
    A 4-amino-N-ethyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00146
    A 4-amino-8-(2-methoxy-5-methylphenyl)-N-methylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00147
    A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-methylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00148
    A 4-amino-8-(4-methoxypyridin-3-yl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00149
    A 4-amino-N-isobutyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00150
    A 4-amino-N-(2-hydroxypropyl)-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00151
    A 4-amino-8-(2-methoxy-5-methylphenyl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00152
    A 4-amino-N-isobutyl-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00153
    A 4-amino-N-(2-hydroxypropyl)-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00154
    A 4-amino-8-(2,5-dimethoxyphenyl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00155
    A 4-amino-8-(2,5-dimethoxyphenyl)-N-isobutylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00156
    A 4-amino-8-(2,5-dimethoxyphenyl)-N-(2-hydroxypropyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00157
    A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-(tetrahydrofuran-2-ylmethyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00158
    A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-isobutylcinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00159
    A 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-(2-hydroxypropyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00160
  • The compounds in Table 3 were also made according to the methods described herein below.
  • TABLE 3
    Example
    Number Compound Name Structure
    114 4-amino-8-(2,3-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00161
    115 4-amino-8-(3,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00162
    116 4-amino-8-(2,4-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00163
    117 4-amino-8-(3,4-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00164
    118 4-amino-N-propyl-8-(p-tolyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00165
    119 4-amino-8-(3-chlorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00166
    120 4-amino-8-(4-chlorophenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00167
    121 4-amino-8-(o-tolyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00168
    122 4-amino-N-propyl-8-(3-thienyl)cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00169
    123 4-amino-8-(2,6-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
    Figure US20090018112A1-20090115-C00170
  • Additional example compounds of the invention in table 4 were made according to the methods described herein below.
  • TABLE 4
    Mass
    Synthesis Spectrum
    Example No. Structure Compound Name Method m/z
    124
    Figure US20090018112A1-20090115-C00171
    4-amino-N-cyclobutyl-7-fluoro-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide A 381
    125
    Figure US20090018112A1-20090115-C00172
    4-amino-N-cyclobutyl-7-fluoro-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 385
    126
    Figure US20090018112A1-20090115-C00173
    4-amino-N-cyclobutyl-7-fluoro-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide G 385
    127
    Figure US20090018112A1-20090115-C00174
    4-amino-N-cyclobutyl-7-fluoro-8-(2-fluoro-3-methoxy-phenyl)cinnoline-3-carboxamide A 385
    128
    Figure US20090018112A1-20090115-C00175
    4-amino-N-cyclobutyl-8-(2,4-dimethoxyphenyl)-7-fluoro-cinnoline-3-carboxamide A 397
    129
    Figure US20090018112A1-20090115-C00176
    4-amino-N-cyclobutyl-7-fluoro-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide A 368
    130
    Figure US20090018112A1-20090115-C00177
    4-amino-N-cyclobutyl-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-cinnoline-3-carboxamide A 399
    131
    Figure US20090018112A1-20090115-C00178
    4-amino-N-cyclobutyl-8-(2,6-dimethoxypyridin-3-yl)-7-fluoro-cinnoline-3-carboxamide A 398
    132
    Figure US20090018112A1-20090115-C00179
    4-amino-N-cyclobutyl-7-fluoro-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 352
    133
    Figure US20090018112A1-20090115-C00180
    4-amino-N-cyclobutyl-7-fluoro-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 368
    134
    Figure US20090018112A1-20090115-C00181
    4-amino-N-cyclobutyl-8-(3,5-dimethylphenyl)-7-fluoro-cinnoline-3-carboxamide A 365
    135
    Figure US20090018112A1-20090115-C00182
    4-amino-N-cyclobutyl-8-(2,5-difluorophenyl)-7-fluoro-cinnoline-3-carboxamide A 373
    136
    Figure US20090018112A1-20090115-C00183
    4-amino-N-cyclobutyl-7-fluoro-8-(3-methylphenyl)cinnoline-3-carboxamide A 351
    137
    Figure US20090018112A1-20090115-C00184
    4-amino-N-cyclobutyl-8-(2,3-dimethoxyphenyl)-7-fluoro-cinnoline-3-carboxamide A 397
    138
    Figure US20090018112A1-20090115-C00185
    4-amino-N-cyclobutyl-7-fluoro-8-(2-methoxyphenyl)cinnoline-3-carboxamide A 367
    139
    Figure US20090018112A1-20090115-C00186
    4-amino-N-cyclobutyl-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide A 363
    140
    Figure US20090018112A1-20090115-C00187
    4-amino-N-cyclobutyl-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 367
    141
    Figure US20090018112A1-20090115-C00188
    4-amino-N-cyclobutyl-8-(2-fluoro-3-methoxy-phenyl)cinnoline-3-carboxamide A 367
    142
    Figure US20090018112A1-20090115-C00189
    4-amino-N-cyclobutyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide A 350
    143
    Figure US20090018112A1-20090115-C00190
    4-amino-N-cyclobutyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide A 381
    144
    Figure US20090018112A1-20090115-C00191
    4-amino-N-cyclobutyl-8-(2,6-dimethoxypyridin-3-yl)cinnoline-3-carboxamide A 380
    145
    Figure US20090018112A1-20090115-C00192
    4-amino-N-cyclobutyl-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 334
    145
    Figure US20090018112A1-20090115-C00193
    4-amino-N-cyclobutyl-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 350
    147
    Figure US20090018112A1-20090115-C00194
    4-amino-N-cyclobutyl-8-(3,5-dimethylphenyl)cinnoline-3-carboxamide A 347
    148
    Figure US20090018112A1-20090115-C00195
    4-amino-N-cyclobutyl-8-(2,5-difluorophenyl)cinnoline-3-carboxamide A 355
    149
    Figure US20090018112A1-20090115-C00196
    4-amino-N-cyclobutyl-8-(3-methylphenyl)cinnoline-3-carboxamide A 333
    150
    Figure US20090018112A1-20090115-C00197
    4-amino-N-cyclobutyl-8-(2,3-dimethoxyphenyl)cinnoline-3-carboxamide A 379
    151
    Figure US20090018112A1-20090115-C00198
    4-amino-N-cyclobutyl-8-(2-methoxyphenyl)cinnoline-3-carboxamide A 349
    152
    Figure US20090018112A1-20090115-C00199
    4-amino-N-cyclobutyl-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide A 334
    153
    Figure US20090018112A1-20090115-C00200
    4-amino-N-cyclobutyl-8-(2,3,4-trimethoxyphenyl)cinnoline-3-carboxamide A 409
    154
    Figure US20090018112A1-20090115-C00201
    4-amino-8-(4-chlorophenyl)-N-cyclobutyl-cinnoline-3-carboxamide A 353
    155
    Figure US20090018112A1-20090115-C00202
    4-amino-N-cyclobutyl-8-(3,4-dimethoxyphenyl)cinnoline-3-carboxamide A 379
    156
    Figure US20090018112A1-20090115-C00203
    4-amino-N-cyclopropyl-8-(2-fluoro-6-methyl-pyridin-3-yl)cinnoline-3-carboxamide A 338
    157
    Figure US20090018112A1-20090115-C00204
    4-amino-N-cyclopropyl-7-fluoro-8-(5-fluoro-6-methoxy-pyridin-3-yl)cinnoline-3-carboxamide A 372
    158
    Figure US20090018112A1-20090115-C00205
    4-amino-N-cyclopropyl-7-fluoro-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 354
    159
    Figure US20090018112A1-20090115-C00206
    4-amino-N-cyclopropyl-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide A 320
    160
    Figure US20090018112A1-20090115-C00207
    4-amino-N-cyclopropyl-7-fluoro-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide A 338
    161
    Figure US20090018112A1-20090115-C00208
    4-amino-N-cyclopropyl-8-(2,6-dimethoxypyridin-3-yl)-7-fluoro-cinnoline-3-carboxamide A 384
    162
    Figure US20090018112A1-20090115-C00209
    4-amino-N-cyclopropyl-7-fluoro-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 338
    163
    Figure US20090018112A1-20090115-C00210
    4-amino-N-cyclopropyl-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-cinnoline-3-carboxamide A 385
    164
    Figure US20090018112A1-20090115-C00211
    4-amino-N-cyclopropyl-8-(2,5-dimethoxyphenyl)-7-fluoro-cinnoline-3-carboxamide A 383
    165
    Figure US20090018112A1-20090115-C00212
    4-amino-N-cyclopropyl-7-fluoro-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 371
    166
    Figure US20090018112A1-20090115-C00213
    4-amino-N-cyclopropyl-7-fluoro-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide G 371
    167
    Figure US20090018112A1-20090115-C00214
    4-amino-N-cyclopropyl-7-fluoro-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide A 367
    168
    Figure US20090018112A1-20090115-C00215
    4-amino-N-cyclopropyl-8-(2,4-dimethoxyphenyl)-7-fluoro-cinnoline-3-carboxamide A 383
    169
    Figure US20090018112A1-20090115-C00216
    4-amino-8-(2,4-dimethoxyphenyl)-N-ethyl-7-fluoro-cinnoline-3-carboxamide A 371
    170
    Figure US20090018112A1-20090115-C00217
    4-amino-N-ethyl-8-(2-fluoro-3-methoxy-phenyl)cinnoline-3-carboxamide A 341
    171
    Figure US20090018112A1-20090115-C00218
    4-amino-N-ethyl-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 324
    172
    Figure US20090018112A1-20090115-C00219
    4-amino-N-ethyl-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 308
    173
    Figure US20090018112A1-20090115-C00220
    4-amino-N-ethyl-8-(5-fluoro-6-methoxy-pyridin-3-yl)cinnoline-3-carboxamide A 342
    174
    Figure US20090018112A1-20090115-C00221
    4-amino-N-cyclopropyl-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 353
    175
    Figure US20090018112A1-20090115-C00222
    4-amino-N-cyclopropyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide A 336
    176
    Figure US20090018112A1-20090115-C00223
    4-amino-N-cyclopropyl-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 336
    177
    Figure US20090018112A1-20090115-C00224
    4-amino-N-cyclobutyl-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide A 363
    178
    Figure US20090018112A1-20090115-C00225
    4-amino-N-cyclobutyl-8-(2,4-dimethoxyphenyl)cinnoline-3-carboxamide A 379
    179
    Figure US20090018112A1-20090115-C00226
    4-amino-8-(2,6-dimethoxypyridin-3-yl)-N-ethyl-cinnoline-3-carboxamide A 354
    180
    Figure US20090018112A1-20090115-C00227
    4-amino-N-cyclopropyl-8-(5-fluoro-6-methoxy-pyridin-3-yl)cinnoline-3-carboxamide A 354
    181
    Figure US20090018112A1-20090115-C00228
    4-amino-N-cyclopropyl-8-(2-fluoro-3-methoxy-phenyl)cinnoline-3-carboxamide A 353
    182
    Figure US20090018112A1-20090115-C00229
    4-amino-N-cyclopropyl-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 320
    183
    Figure US20090018112A1-20090115-C00230
    4-amino-N-ethyl-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 341
    184
    Figure US20090018112A1-20090115-C00231
    4-amino-8-(2,4-dimethoxyphenyl)-N-ethyl-cinnoline-3-carboxamide A 353
    185
    Figure US20090018112A1-20090115-C00232
    4-amino-N-cyclopropyl-7-fluoro-8-(2-fluoro-3-methoxyphenyl)cinnoline-3-carboxamide A 371
    186
    Figure US20090018112A1-20090115-C00233
    4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-ethyl-7-fluoro-cinnoline-3-carboxamide A 373
    187
    Figure US20090018112A1-20090115-C00234
    4-amino-N-ethyl-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide A 308
    188
    Figure US20090018112A1-20090115-C00235
    4-amino-N-ethyl-7-fluoro-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide A 359
    189
    Figure US20090018112A1-20090115-C00236
    4-amino-8-(2,6-dimethoxypyridin-3-yl)-N-ethyl-7-fluoro-cinnoline-3-carboxamide A 372
    190
    Figure US20090018112A1-20090115-C00237
    4-amino-N-ethyl-7-fluoro-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide A 359
    191
    Figure US20090018112A1-20090115-C00238
    4-amino-N-ethyl-7-fluoro-8-(5-fluoro-6-methoxy-pyridin-3-yl)cinnoline-3-carboxamide A 360
    192
    Figure US20090018112A1-20090115-C00239
    4-amino-N-ethyl-7-fluoro-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide A 326
    193
    Figure US20090018112A1-20090115-C00240
    4-amino-N-ethyl-7-fluoro-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide A 342
    194
    Figure US20090018112A1-20090115-C00241
    4-amino-N-ethyl-7-fluoro-8-(2-fluoro-3-methoxy-phenyl)cinnoline-3-carboxamide A 359
    195
    Figure US20090018112A1-20090115-C00242
    4-amino-8-(2,5-dimethoxyphenyl)-N-ethyl-7-fluoro-cinnoline-3-carboxamide A 371
  • Synthesis
  • The compounds of the present invention can be prepared in a number of ways well known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. The starting materials and precursors used in the processes described herein were either commercially available or readily prepared by established organic synthesis methods. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule must be compatible with the reagents and reactions proposed. Such restrictions to the substituents which are compatible with the reaction conditions will be readily apparent to one skilled in the art and alternate methods should then be used.
  • Chemical abbreviations used in the Examples are defined as follows: “DMSO” denotes dimethylsulfoxide, “THF” denotes tetrahydrofuran, “DMF” denotes N,N-dimethylformamide. Unless otherwise stated reaction progress was monitored by HPLC, LC-MS or TLC. Oven-dried standard laboratory glassware was used and routine manipulations were done at ambient temperature under a blanket of nitrogen unless otherwise indicated. Commercially available reagents and anhydrous solvents were typically used as received. Evaporations were typically performed under reduced pressure using a rotary evaporator. Preparative chromatography was performed using ICN silica gel 60, 32-63μ or a suitable equivalent. Products were dried under reduced pressure at 40° C. or a suitable temperature.
  • HPLC-Mass Spectroscopy data were collected utilizing an Agilent Zorbax 5μ SB-C8 column 2.1 mm×5 cm. with a column temperature of 30° C. Solvents: A=98:2 Water:Acetonitrile with 0.1% formic acid added, B=98:2 Acetonitrile:Water with 0.05% formic acid added. Flow rate 1.4 mL/min, injection volume 2.0 μL, initial conditions 5% B, eluting with a linear gradient from 5 to 90% B from time zero to 3 minutes holding at 90% B until 4 minutes. Photodiode array UV detection was used averaging signal from 210 through 400 nm. Mass Spectral data were collected using Full Scan APCI (+), base peak index, 150.0 to 900.0 amu., 30 cone volts with a probe temperature of 450° C.
  • 1H NMR data (δ, ppm) were obtained on a Bruker 300 MHz instrument at 30° C. with tetramethylsilane as an internal standard set at 0.00 ppm. The multiplicities of the NMR spectra absorptions may be abbreviated by: s, singlet; br, broad peak; bs, broad singlet; d, doublet; t, triplet; q, quartet; dd, doublet of doublets; dt, doublet of triplets; m, multiplet. In many cases proton resonances associated with the cinnoline 4-amino group protons were not readily observable in the proton NMR spectra recorded at 30° C. in chloroform-d due to severe broadening into the baseline. These protons can be clearly observed by recording the spectrum at −20° C.
  • As shown in Scheme 1, a compound 1-3 can be made by coupling of a halogenated cinnoline derivative 1-1 (wherein X1 is halo such as bromo or iodo) to a boron compound 1-2 wherein R6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.), R101 and R102 are each, independently, hydrogen or C1-6 alkyl; or R101 and R102, together with the two oxygen atoms to which they are attached and the boron atom to which the two oxygen atoms are attached, form a 4-7 membered heterocyclic ring whose ring-forming atoms comprises B, O and C atoms and which is optionally substituted by 1, 2, 3, or 4 C1-6 alkyl (i.e., a moiety shown as 1-2B-R wherein t1 is 0, 1, 2 or 3; t2 is 0, 1, 2, 3 or 4; and R400 is each, independently, C1-6 alkyl). Two examples of the boron compound 1-2 are 1-2A (a boronic acid derivative) and 1-2B (a 4,4,5,5-tetramethyl-1,3,2-dioxoborolane derivative). The coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst. Some exemplary metal catalysts include palladium catalyst, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0). The coupling reaction can be carried out in the presence of a suitable base such as an inorganic base. Some exemplar suitable inorganic base include cesium carbonate, sodium carbonate, and potassium phosphate. The coupling reaction can be carried out in a suitable solvent such as an organic solvent. Some suitable organic solvent include polar organic solvents, such as an ether and an alcohol. Suitable ethers include 1,2-dimethoxyethane and tetrahydrofuran. Suitable alcohols include ethanol, propanol and isopropanol. A suitable solvent also includes a mixture of two or more individual solvents. Suitable solvents can further contain water. The coupling reaction can be carried out at a suitable temperature to afford the compound 1-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature). In some embodiments, the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C. The reaction progress can be monitored by conventional methods such as TLC or NMR.
  • Figure US20090018112A1-20090115-C00243
  • As shown in Scheme 2, a compound 2-3 can be made by coupling of a halogenated cinnoline derivative 2-1 (wherein X2 is halo such as bromo or iodo) to a tin compound 2-2 wherein R6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.), R201, R202 and R203 are each, independently, C1-6 alkyl. The coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst. Some exemplary metal catalysts include palladium catalysts, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0). The coupling reaction can be carried out in a suitable organic solvent. Some suitable organic solvent include polar organic solvent. Some suitable organic solvent include aprotic solvent. Some suitable organic solvent include polar aprotic organic solvent such as N,N-dimethylformamide. The coupling reaction can be carried out at a suitable temperature for a time sufficient to afford the compound 2-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature). In some embodiments, the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C. The reaction progress can be monitored by conventional methods such as TLC or NMR.
  • Figure US20090018112A1-20090115-C00244
  • As shown in Scheme 3, a compound 3-3 can be made by coupling of a trialkylstannyl-cinnoline derivative 3-1 (wherein R301, R302 and R303 are each, independently, C1-6 alkyl) to a halogenated compound R6X3 3-2 wherein X3 is halo such as bromo or iodo, and wherein R6 can be an optionally substituted aryl or heteroaryl (suitable substituents can be alkyl, CN etc.). The coupling reaction can be carried out in the presence of a suitable catalyst, such as a metal catalyst. Some exemplary metal catalysts include palladium catalysts, such as bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0). The coupling reaction can be carried out in a suitable organic solvent. Some suitable organic solvent include polar organic solvent. Some suitable organic solvents include aprotic organic solvent. Some suitable organic solvents include polar aprotic organic solvents such as N,N-dimethylformamide. The coupling reaction can be carried out at a suitable temperature for a time sufficient to afford the compound 2-3. In some embodiments, the reaction mixture is heated to an elevated temperature (i.e., above the room temperature). In some embodiments, the reaction mixture is heated to a temperature of about 40° C., about 50° C., about 60° C., about 70° C., about 80° C., about 90° C., about 100° C., about 110° C., about 120° C., about 130° C., about 140° C., about 150° C., about 160° C. The reaction progress can be monitored by conventional methods such as TLC or NMR.
  • Also as shown in Scheme 3, the trialkylstannyl-cinnoline derivative 3-1 can be made by coupling of a halogenated cinnoline derivative 3-0-1 (wherein X4 is halo such as bromo or iodo) to a di-tin compound 3-0-2 (wherein R301, R302 and R303 are each, independently, C1-6 alkyl) in the presence of a suitable catalyst, such as a palladium catalyst. Some exemplar palladium catalysts include bis(triphenylphosphine)palladium(II) dichloride and tetrakis(triphenylphosphine)palladium(0).
  • Figure US20090018112A1-20090115-C00245
  • It should noted that in all of the schemes described herein, if there are functional (reactive) groups present on a substituent group such as R1, R2, R3, R4, R5, R6, etc., further modification can be made if appropriate and/or desired. For example, a CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to an amide; a carboxylic acid can be converted to a ester, which in turn can be reduced to an alcohol, which in turn can be further modified. In another example, an OH group can be converted into a better leaving group such as mesylate, which in turn is suitable for nucleophilic substitution, such as by CN. One skilled in the art will recognize further such modifications. Thus, a compound of formula I (such as compound 1-3 of Scheme 1, compound 2-3 in Scheme 2 and compound 3-3 of Scheme 3) having a substituent which contains a function group can be converted to another compound of formula I having a different substituent group.
  • As used herein, the term “reacting” refers to the bringing together of designated chemical reactants such that a chemical transformation takes place generating a compound different from any initially introduced into the system. Reacting can take place in the presence or absence of solvent.
  • Some more detailed methods, procedures and precursors as outlined in Schemes 1-3 and additional detailed procedures and characterization data for certain above exemplified compounds are further described herein below.
  • Precursor 1 4-Amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide
  • To a 1 L, 3-necked flask equipped with a mechanical stirrer charged with (2E)-2-cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-propylacetamide (43.9 g, 117 mmol) in anhydrous toluene (Aldrich, 600 mL) under N2 was added portion-wise aluminum chloride (Aldrich, 46.8 g, 352 mmol) over 20 minutes. The mixture was heated to 60° C. with vigorous stirring for 2 hours and then cooled to −15° C. Ethyl acetate (30 mL) was carefully added while maintaining the internal temperature between 20-25° C. Additional ethyl acetate (900 mL) was then added, followed by careful addition of Rochelle's salt (saturated aqueous potassium sodium tartrate, 500 mL). Upon addition of the first 50 mL, the temperature rose from 20 to 36° C. The reaction was heated with stirring at 60° C. for 30 minutes. The aqueous layer contained a thick white precipitate and the organic layer slowly solubilized the brownish yellow solid. Note: If a non-white (brown/yellow) solid still existed at the aqueous/organic interface, the hot extraction was repeated. The mixture was placed in a separatory funnel and the aqueous layer was removed. The organic layer was washed with Rochelle's salt (500 mL) and brine, dried over magnesium sulfate, filtered and concentrated to give 38 g of product (86.5%). Further purification by trituration with ethyl acetate/hexanes was carried out when appropriate. An analytically pure sample was obtained by recrystallization from ethyl acetate. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.84 (dd, J=5.3, 9.2 Hz, 1H), 7.39 (dd, J=7.0, 9.2 Hz, 1H), 3.47 (apparent q, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.03 (t, J=7.4 Hz, 3H). MS APCI, m/z=375 (M+H). HPLC 2.13 min.
  • The intermediate compounds were prepared as follows:
  • 3-Fluoro-2-iodoaniline hydrochloride
  • To a 1 L, 3 necked round bottom flask fitted with a mechanical stirrer was added 3-fluoro-2-iodonitrobenzene (3B Medical, 47.7 g, 179 mmol) and 500 mL absolute ethanol. To this stirred solution was added iron powder (325 mesh, Aldrich, 30 g, 537 mmol) followed by dropwise addition of concentrated HCl (30 mL, 360 mmol). The internal temperature rose from 23 to −60° C. over the addition. The flask was fitted with a heating mantle and heated with vigorous stirring for 90 minutes. After cooling to room temperature, 1 N sodium carbonate (300 mL) was added followed by ethyl acetate (200 mL). The mixture was stirred for 30 minutes and then filtered through a pad of celite. The celite was washed with ethyl acetate (3×150 mL). The filtrates were placed in a separatory funnel and the water layer was removed. The organic layer was concentrated under reduced pressure to a volume of 200 mL, placed in a separatory funnel, diluted with ethyl acetate (400 mL), washed with brine, dried over sodium sulfate, filtered and concentrated to dryness. The crude product was taken up in ether (300 mL) and made acidic to pH 1 with 2M hydrochloric acid/ether (Aldrich). After 1 hour, the tan solid was isolated by filtration (39.2 g, 80%). The above aqueous layers were extracted with diethyl ether (300 mL), dried over sodium sulfate, combined with the filtrate of the 1st crop, made acidic to pH 1, and isolated as above to give additional tan solid (9.0 g, 18%) for an overall yield of 98%. 1H NMR (300 MHz, CDCl3) δ 7.06 (m, 1H), 6.58 (m, 1H), 6.39 (m, 1H), 5.73 (bm, 1H). MS APCI, m/z=238 (M+H). HPLC 2.19 min.
  • (2E)-2-Cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-propylacetamide
  • Using the procedure outlined in the U.S. Pat. No. 4,886,800 example 89b substituting 3-fluoro-2-iodoaniline hydrochloride (8.8 g, 32.5 mmol) for 2-iodoaniline, the title compound (2E)-2-cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-propylacetamide (8.5 g, 70% yield) was obtained as a light brown solid. An analytically pure sample was obtained by recrystallization from ethyl acetate as a yellow crystalline solid.
  • 1H NMR (300 MHz, CDCl3) δ 14.39, (s, 1H), 8.67 (bm, 1H), 7.45 (m, 1H), 7.32 (m, 1H), 7.03 (m, 1H), 3.1 (apparent q, J=6.6 Hz, 2H), 1.53 (apparent sextet, J=7.4 Hz, 2H), 0.88 (t, J=7.4 Hz, 3H).
  • Precursor 2 4-Amino-7-chloro-8-iodo-N-propyl-cinnoline-3-carboxamide
  • Using a procedure similar to that used in the synthesis of 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide, the title compound 4-amino-7-chloro-8-iodo-N-propyl-cinnoline-3-carboxamide (2.75 g, 67% yield) was obtained from (2E)-2-cyano-2-[(3-chloro-2-iodophenyl)hydrazono]-N-propylacetamide (4.1 g, 10.5 mmol) as a white solid.
  • 1H NMR (300 MHz, CDCl3) δ 8.54 (bs, 1H), 7.76 (d, J=9.0 Hz, 1H), 7.66 (d, J=9.0 Hz, 1H), 3.47 (apparent q, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.03 (t, 7.4 Hz, 3H). MS APCI, m/z=391 (M+H) HPLC 2.38 min. The intermediate compounds were prepared as follows:
  • (2E)-2-Cyano-2-[(3-chloro-2-iodophenyl)hydrazono]-N-propylacetamide
  • Prepared according to the method described in U.S. Pat. No. 4,886,800 example 89b substituting 3-chloro-2-iodoaniline (7.1 g, 28.1 mmol) for 2-iodoaniline, the title compound (2E)-2-cyano-2-[(3-chloro-2-iodophenyl)hydrazono]-N-propylacetamide (4.2 g, 38% yield) was obtained as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 14.30, (s, 1H), 7.48 (m, 1H), 7.24-7.33 (m, 3H), 6.28 (bm, 1H), 3.37 (apparent q, J=7.0 Hz, 2H), 1.64 (apparent sextet, J=7.4 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=391 (M+H) HPLC 3.00 min.
  • 3-Chloro-2-iodoaniline
  • Prepared according to the method described in U.S. Pat. No. 4,822,781, process 1, substituting 2-chloro-6-nitrophenol for 2-fluoro-6-nitrophenol, the title compound, 3-chloro-2-iodoaniline (7.1 g, 3 step overall yield 55% yield) was obtained from 2-chloro-6-nitrophenol as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 7.04 (t, J=8.0 Hz, 1H), 6.84 (dd, J=8.0, 1.3 Hz, 1H), 6.60 (dd, J=8.0, 1.3 Hz, 1H), 4.31 (bs, 2H). MS APCI, m/z=254 (M+H) HPLC 2.38 min
  • Precursor 3 4-Amino-8-bromo-N-propyl-cinnoline-3-carboxamide
  • A 22 L, 3-necked flask equipped with a mechanical stirrer, thermometer, nitrogen inlet, reflux condenser, and addition funnel was charged with N-propyl-2-cyano-2-[(2-bromophenyl)hydrazono]acetamide (195.4 g, 0.632 mol) in toluene (4 L). Aluminum chloride (295 g, 2.21 mol) was added in three portions. The mixture was heated with a mantle to 90° C. in approximately 30 minutes. After 2.5 hours, the heat was removed and the reaction mixture was allowed to cool to room temperature overnight. The reaction mixture was cooled in an ice bath to ≦10° C. and celite was added. Water (680 mL) was added dropwise over 1 hr at ≦10° C. After stirring for 30 minutes, methylene chloride was added (8 L). The reaction mixture was cooled to ≦10° C. and 10% sodium hydroxide (5.8 L) was added dropwise over 45 minutes at ≦10° C. After stirring for 30 minutes, tetrahydrofuran (2 L) was added and the phases were allowed to separate. The aqueous layer was removed, filtered through celite, and the filter cake washed with 2:1 methylene chloride:tetrahydrofuran (4 L). Note: Addition of fresh portions of methylene chloride helped expediate the rather tedious filtration. The phases of the filtrate were separated and the organic phase was transferred to a separatory funnel. Separation of the organic phase from the aqueous base as quickly as possible helped avoid undue hydrolysis of the propyl amide in the product. The solids remaining in the reaction flask were dissolved with 2:1 tetrahydrofuran:methanol (4 L) and then 10% methanol in chloroform (4 L). The layers were separated and the organic layer was washed with brine (500 mL), dried over magnesium sulfate, filtered, and concentrated under reduced pressure to a dark brown solid. The solid was slurried in diethyl ether, collected by filtration and dried. The crude solid (188 g) was then dissolved in hot methanol (6 L), treated with activated charcoal (19 g), stirred 15 minutes at reflux, filtered through celite while hot, concentrated to approximately 3 L, and allowed to crystallize overnight. The solids were collected, washed with diethyl ether (400 mL) and dried in a vacuum oven at 50° C. to give a white crystalline solid. The filtrate was concentrated to approximately 1 L and a second crop obtained. The mother liquors were stripped and a third and fourth crop were obtained from additional recrystallizations to afford a total of 164.6 g of the desired compound as a white crystalline solid (84%). 1H NMR (300.132 MHz, CDCl3) δ 8.57 (bs, 1H), 8.12 (dd, J=7.6, 1.1 Hz, 1H), 7.83 (dd, J=8.4, 1.0 Hz, 1H), 7.50 (dd, J=8.4, 7.5 Hz, 1H), 3.48 (q, J=6.7 Hz, 2H), 1.69 (sextet, J=7.3 Hz, 2H), 1.03 (t, J=7.4 Hz, 3H). MS APCI, m/z=309/311 (M+H). HPLC 1.66 min.
  • Precursor 4 4-Amino-8-iodo-N-propyl-cinnoline-3-carboxamide
  • Prepared according to the method described in the U.S. Pat. No. 4,886,800 example 36a.
  • Precursor 5 4-Amino-8-fluoro-N-propyl-cinnoline-3-carboxamide
  • To a suspension of N-propyl-2-cyano-2-[(2-fluorophenyl)hydrazono]acetamide (11.1 g, 44.71 mmol) in toluene (275 mL) was added aluminum chloride (20.90 g, 156.74 mmol). The mixture was stirred at 90° C. for 2.5 hours. The reaction mixture was cooled to 0° C., and then diluted with chloroform (1 L). A small amount of water was added to quench the reaction at 0° C. Aqueous sodium hydroxide (750 mL, 20% w/v solution) was poured into the mixture slowly at 0° C., and the mixture stirred at ambient temperature for one hour. A precipitate was formed gradually. The mixture was diluted with chloroform (2 L) until all of the precipitate was dissolved, washed twice with water, dried through magnesium sulfate, and concentrated to a volume of approximately 200 mL to leave a suspension of the product. The title compound as a light beige solid (11.06 g) was collected by filtration and washed with methylene chloride (50 mL×2), methanol (50 mL) and hexane (100 mL×2). The mother liquor was concentrated, and purified by flash chromatography using a gradient of ethyl acetate in hexane to give an additional 400 mg of the title compound as a beige solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.55-7.70 (m, 2H), 3.49 (m, 2H), 1.71 (m, 2H), 1.03 (t, J=7.4 Hz, 3H) MS APCI, m/z=249 (M+H) HPLC 1.30 min.
  • The intermediate compounds were prepared as follows:
  • N-propyl 2-cyano-2-[(2-fluorophenyl)hydrazono] acetamide
  • Solution A: To a mechanically stirred solution of 2-fluoroaniline (11.51 g, 100.34 mmol) in acetic acid (50 mL) was added water (30 mL) at ambient temperature. The mixture was cooled to 0° C., and then concentrated aqueous HCl (25 mL) added. A precipitate was formed as soon as the concentrated HCl was added, and the suspension was stirred at 0° C. for 20 minutes. To this suspension was added dropwise a solution of sodium nitrite (7.72 g, 111.88 mmol) in water (30 mL), maintaining the internal temperature below 5° C. The resulting clear orange solution was stirred at 0° C. for another 30 minutes.
  • Solution B: To a mechanically stirred solution of N-propyl-2-cyanoacetamide (15.69 g, 124.37 mmol) in ethanol (220 mL) was added a solution of sodium acetate (136.00 g, 1.66 moles) in water (600 mL), and chilled to between 0° C. and −5° C.
  • Solution A was poured into solution B, maintaining the internal temperature below 0° C. An orange precipitate was formed gradually after 10 minutes. The mixture was stirred below 0° ° C. for another hour, and was then diluted with water (500 mL). After 30 minutes, the orange precipitate was collected by filtration, washed with water (100 mL×3), and dried at 50° C. under high vacuum to remove water. An orange solid (9.50 g) was obtained, which was the “E” isomer, and used for the next step without further purification. 1H NMR (300 MHz, CDCl3) δ 14.18 (br, 1H), 7.68 (td, 1H, J=7.94 Hz, J′=1.47 Hz), 7.00-7.20 (m, 3H), 6.28 (s, 1H), 3.34 (m, 2H), 1.64 (m, 2H), 0.99 (t, 3H, J=7.40 Hz)
  • Precursor 6 4-amino-8-trimethylstannyl-N-propyl-cinnoline-3-carboxamide
  • To a stirred solution of 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (3.4 g, 9.4 mmol) and tetrakis(triphenylphosphine) palladium(0) (800 mg, 0.69 mmol) in anhydrous N,N-dimethylformamide at ambient temperature under nitrogen was added hexamethylditin (5.0 g, 15.2 mmol). The reaction was heated to 150° C. for 1-1.5 hours. The reaction mixture was filtered through Celite, and the solution evaporated. The residue was dissolved in methylene chloride, washed with water twice, dried through MgSO4, and then the solvent was evaporated. The residue was purified by flash chromatography using an increasingly polar gradient of ethyl acetate in hexane to give a yellow solid as the title compound (2.4 g, 68.4% yield).
  • 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.99 (dd, J=6.6 Hz, J′=1.0 Hz, 1H), 7.83 (dd, J=8.4 Hz, J′=1.1 Hz, 1H), 7.61 (dd, J=8.3 Hz, J′=6.6 Hz, 1H), 3.47 (q, J=6.8 Hz, 2H), 1.70 (m, J=7.3 Hz, 2H), 1.02 (t, 3H, J=7.40 Hz), 0.44 (s, 9H). MS APCI, m/z=391/392/395 (M+H). HPLC 2.75 min.
  • Precursor 7 4-Amino-8-bromo-N-ethyl-cinnoline-3-carboxamide
  • To a stirred solution of 2-[(2-bromophenyl)-hydrazono]-N-ethyl-2-cyanoacetamide (260 mg, 0.88 mmol) in anhydrous toluene (10 mL) was added aluminum chloride (370 mg, 2.78 mmol). The reaction was heated with vigorous stirring at 90° C. for 1.5 hours, cooled, diluted with ethyl acetate (40 mL), and treated with Rochelle's salt (saturated aqueous solution). After stirring for 30 minutes, the organic layer was decanted into a separatory funnel. (The white precipitate was rinsed with ethyl acetate three times.) The organic layer was washed with 1:1 brine:Rochelle's salt solution, dried over sodium sulfate, and concentrated to a light brown solid. The solid was slurried in ether and filtered to afford the title compound as a brown solid (180 mg, 69%). 1H NMR (300.132 MHz, CDCl3) δ 8.52 (s, 1H), 8.13 (dd, J=7.4, 1.1 Hz, 1H), 7.82 (dd, J=8.4, 1.1 Hz, 1H), 7.51 (dd, J=8.4, 7.5 Hz, 1H), 3.56 (dq, J=5.8, 7.3 Hz, 2H), 1.31 (t, J=7.3 Hz, 3H). MS APCI, m/z=395/397 (M+H). HPLC 1.90 min.
  • The intermediate compounds were prepared as follows:
  • 2-[(2-Bromophenyl)-hydrazono]-N-ethyl-2-cyanoacetamide
  • To a solution of [(2-bromophenyl)-hydrazono]-cyanoacetic acid ethyl ester (1.0 g, 3.4 mmol) in methanol (14 mL) was added 70% ethyl amine in water (16 mL, 20.2 mmol) followed by triethyl amine (468 uL, 3.6 mmol). The reaction was stirred at room temperature overnight, concentrated and dried under high vacuum. The material was routinely used crude. Purification on silica gel using a gradient of 10 to 50% ethyl acetate in hexanes afforded the title compound as a yellow solid. 1H NMR (300.132 MHz, CDCl3) δ 14.33 (s, 1H), 7.67 (dd, J=8.3, 1.4 Hz, 1H), 7.53 (dd, J=8.1, 1.3 Hz, 1H), 7.34 (tq, J=7.8, 0.6 Hz, 1H), 7.01 (td, J=7.7, 1.6 Hz, 1H), 3.45 (dq, J=5.9, 7.2 Hz, 2H), 1.26 (t, J=7.3 Hz, 3H). HPLC 4.66 min.
  • Precursor 8 2-(Biphenyl-2-yl-hydrazono)-2-cyano-N-cyclopropylmethyl-acetamide
  • To a stirred solution of 2-aminobiphenyl (2.95 g, 17.4 mmol) in glacial acetic acid (16 mL) and water (14 mL) with cooling was added dropwise concentrated hydrochloric acid (10 mL). Additional water (10 mL) was added to maintain stirring. The mixture was cooled to 0° C. and a solution of sodium nitrite (1.44 g, 20.7 mmol) in water (10 mL) was added dropwise maintaining an internal temperature of ≦5° C. Upon complete addition, the reaction was stirred at 0° C. for 30 minutes, poured portionwise into a mechanically stirred 3-necked round bottomed flask charged with a predissolved solution of 2-cyano-N-cyclopropylmethylacetamide (2.8 g, 20.3 mmol), sodium acetate (12.0 g, 146 mmol), and sodium carbonate (12.8 g, 121 mmol) in 2:1 water:ethanol (180 mL). Vigorous CO2 (g) evolution was observed. After 1 hour at 0° C., the reaction was diluted with water (200 mL) and extracted with ethyl acetate (400 mL). The organic layer was washed with water (200 mL) and brine (200 mL) and dried over sodium sulfate. The mixture was filtered, concentrated, and purified by recrystallization from ethyl acetate/hexanes to afford the title compound as a yellow solid (2.0 g, 36%). 1H NMR (500.133 MHz, CDCl3) δ 9.23 (t, J=5.3 Hz, 1H), 9.13 (bs, 1H), 8.43 (d, J=8.3 Hz, 1H), 8.17 (bs, 1H), 7.86 (d, J=7.2 Hz, 1H), 7.81 (t, J=7.6 Hz, 1H), 7.71 (d, J=7.5 Hz, 2H), 7.49 (t, J=7.2 Hz, 2H), 7.43 (t, J=7.2 Hz, 1H), 3.30 (s, OH), 3.23 (t, J=6.1 Hz, 2H), 1.12 (septet, J=6.4 Hz, 1H), 0.45 (d, J=8.0 Hz, 2H), 0.29 (d, J=4.1 Hz, 2H). MS APCI, m/z=319 (M+H). HPLC 1.84 min.
  • The intermediate compounds were prepared as follows:
  • 2-Cyano-N-cyclopropylmethylacetamide
  • To an ice-cooled flask charged with cyclopropyl methyl amine (4.25 g, 59.8 mmol) was added ethyl cyano acetate (3.17 mL, 29.7 mmol). The reaction was stirred at 0° C. for 1.75 hour at which point a precipitate had formed and 1:1 ether:hexanes (40 mL) was added. The mixture was stirred for 15 minutes, filtered, and the solids washed with hexanes to give the title compound as a white solid (3.44 g, 84%). 1H NMR (300.132 MHz, CDCl3) δ 6.17 (s, 1H), 3.37 (s, 2H), 3.17 (dd, J=7.1, 5.4 Hz, 2H), 1.06-0.92 (m, 1H), 0.62-0.51 (m, 2H), 0.24 (q, J=5.1 Hz, 2H).
  • Precursor 9 4-Amino-8-bromo-N-butyl-cinnoline-3-carboxamide
  • To a solution of 2-[(2-bromophenyl)-hydrazono]-N-butyl-2-cyanoacetamide (2.5 g, 7.7 mmol) in anhydrous toluene (Aldrich, 50 mL) under N2 was added portion-wise aluminum chloride (Aldrich, 3.1 g, 23.2 mmol) over 5 minutes. The mixture was heated to 90° C. with vigorous stirring for 1.5 hours then cooled to −0° C. Water (3 mL) was added dropwise followed by careful addition of Rochelle's salt (saturated aqueous potassium sodium tartrate, 50 mL). The reaction was stirred for 25 minutes and then poured into a separatory funnel. The aqueous layer contained a thick white precipitate and was quickly removed. The organic layer was washed with Rochelle's salt and brine, dried over magnesium sulfate, filtered and concentrated to give 2.6 g slightly crude product which was purified on silica gel using a gradient of 20 to 60% ethyl acetate in hexane. Recrystallization from ethyl acetate/hexanes (10 mL each, 0° C. overnight) afforded the title compound as a white solid (650 mg, 26%). 1H NMR (300.132 MHz, CDCl3) δ 8.55 (bs, 1H), 8.13 (dd, J=7.4, 1.0 Hz, 1H), 7.82 (dd, J=8.5, 1.0 Hz, 1H), 7.50 (dd, J=8.5, 7.6 Hz, 1H), 3.52 (q, J=6.6 Hz, 2H), 1.65 (quintet, J=7.2 Hz, 2H), 1.47 (sextet, J=7.3 Hz, 2H), 0.97 (t, J=7.3 Hz, 3H). MS APCI, m/z=323/325 (M+H). HPLC 1.93 min.
  • The intermediate compounds were prepared as follows:
  • 2-[(2-Bromophenyl)-hydrazono]-N-butyl-2-cyanoacetamide
  • To a microwave vial charged with [(2-bromophenyl)-hydrazono]-cyanoacetic acid ethyl ester (387 mg, 1.31 mmol) was added methanol (3 mL) and n-butylamine (520 uL, 5.24 mmol). The reaction temperature rose approximately 30° C. and everything went into solution. After 25 minutes, additional n-butylamine (260 uL, mg, 2.6 mmol) and triethyl amine (182 uL, 1.3 mmol) were added. The reaction was stirred at room temperature overnight and then concentrated to afford the title compound which was used without further purification (420 mg, 99%). 1H NMR (300.132 MHz, CDCl3) δ 14.33 (s, 1H), 7.67 (dd, J=8.3, 1.5 Hz, 1H), 7.53 (dd, J=8.1, 1.3 Hz, 1H), 7.34 (td, J=7.8, 1.2 Hz, 1H), 7.01 (dt?ddd, J=6.1 Hz, J=8.0 Hz, J=1.5 Hz, 1H), 6.22 (bs, 1H), 3.40 (dt, J=5.9, 7.2 Hz, 2H), 1.65-1.35 (m, 4H), 0.96 (td, J=7.3, 1.9 Hz, 3H). MS APCI, m/z=323/325 (M+H). HPLC 2.94 min
  • Precursor 10 4-Amino-8-bromo-N-methyl-cinnoline-3-carboxamide hydrochloric acid salt
  • A 250 mL round-bottomed flask was charged with 2-[(2-bromophenyl)-hydrazono]-N-methyl-2-cyanoacetamide (2.00 g, 7.12 mmol), aluminum chloride (3.46 g, 25.97 mmol), and anhydrous toluene (68 mL). The reaction was gently refluxed for 45 minutes, cooled to room temperature, and slowly treated with 2N HCl (68 mL). A precipitate formed. The mixture was heated to 90° C. for 10 minutes, cooled to room temperature, and filtered. The solids were dried under high vacuum at 50° C. to afford the title compound (2.02 g, 90%). 1H NMR (300.132 MHz, DMSO) δ 9.08 (d, J=4.7 Hz, 1H), 8.56 (dd, J=8.4, 0.8 Hz, 1H), 8.28 (dd, J=7.6, 0.7 Hz, 1H), 7.65 (t, J=8.0 Hz, 1H), 2.89 (d, J=4.7 Hz, 3H). MS APCI, m/z=281/283 (M+H). HPLC 1.61 min.
  • The intermediate compounds were prepared as follows:
  • 2-[(2-Bromophenyl)-hydrazono]-N-methyl-2-cyanoacetamide
  • [(2-Bromophenyl)-hydrazono]-cyanoacetic acid ethyl ester (15.28 g, 51.60 mmol) was dissolved in 40% methylamine in water (67.5 mL) and stirred at room temperature overnight. The reaction mixture was concentrated to dryness, slurried in diethyl ether, and filtered. After drying under high vacuum at 40° C., the title compound was obtained as a yellow solid (11.16 g, 77%). 1H NMR (300.132 MHz, CDCl3) δ 7.68 (dd, J=8.2, 1.4 Hz, 1H), 7.54 (dd, J=8.1, 1.3 Hz, 1H), 7.35 (t, J=7.9 Hz, 1H), 7.01 (td, J=7.7, 1.5 Hz, 1H), 6.29 (s, 1H), 2.98 (d, J=4.9 Hz, 3H). MS APCI, m/z=281/283 (M+H). HPLC 4.08 min.
  • Precursor 11 4-Amino-8-bromo-cinnoline-3-carboxylic acid allylamide
  • To an ice-cooled suspension of 4-amino-8-bromo-cinnoline-3-carboxylic acid (360 mg, 1.34 mmol) in dimethylformamide (5 mL) was added CDI (370 mg, 2.3 mmol) and the mixture was stirred at room temperature for 1 hour. Additional DMF (14 mL) was added to enable stirring. After an additional 1 hour at room temperature, the mixture was treated with allyl amine (120 uL, 91 mg, 1.60 mmol) in one portion. The reaction was stirred at room temperature for 1 hour and then concentrated. Purification on silica gel using a gradient of 20 to 80% ethyl acetate in hexanes afforded the title compound (300 mg, 73%). 1H NMR (300.132 MHz, CDCl3) δ 8.14 (dd, J=7.4, 1.0 Hz, 1H), 7.83 (dd, J=8.4, 1.0 Hz, 1H), 7.51 (dd, J=8.4, 7.5 Hz, 1H), 6.04-5.91 (m, 1H), 5.33 (dq, J=17.1, 1.6 Hz, 1H), 5.21 (dq, J=10.4, 1.4 Hz, 1H), 4.15 (ddt, J=6.2, 5.8, 1.6 Hz, 2H). MS APCI, m/z=307/309 (M+H). HPLC 1.59 min.
  • Precursor 12 2,4-Dimethoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine
  • To a 100 mL round-bottomed flask charged with 4 Å molecular sieves (approximately 1 g) was added the 2,4-dimethoxypyrimidine-5-boronic acid (5.34 g, 29.0 mmol) and anhydrous tetrahydrofuran (25 mL). The pinocol (2.98 g, 25.3 mmol) was added and the reaction stirred at room temperature for 1.5 hours. Additional 2,4-dimethoxypyrimidine-5-boronic acid (662.2 mg, 3.6 mmol) was added and the reaction stirred overnight. Molecular sieves and 2,4-dimethoxypyrimidine-5-boronic acid (1.53 g, 8.3 mmol) were added and the reaction stirred for 0.5 hours. Pinacol (0.613 g, 5.2 mmol) was then added. After 2 hours, the molecular sieves were removed by filtration and the filtrate was concentrated. After drying under high vacuum, the title compound was obtained as a fine yellow solid (8.61 g, 79%). 1H NMR (300.132 MHz, CDCl3) δ 8.56 (s, 1H), 4.01 (d, J=1.5 Hz, 6H), 1.34 (s, 12H).
  • Precursor 13 4-Amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide
  • A 500 mL, 3-necked flask equipped with a mechanical stirrer, thermometer, nitrogen inlet, reflux condenser, and addition funnel was charged with N-cyclopropyl-2-cyano-2-[(2-bromophenyl)hydrazono]acetamide (1.7 g, 5.6 mmol) in anhydrous toluene (0.2 L). The reaction mixture was cooled with stirring in an ice bath. Aluminum chloride (1.6 g, 12.0 mmol) was added in three portions. Removed ice bath and heated at 70-75° C. for 60 hours. The reaction mixture was allowed to cool to room temperature, diluted with ethyl acetate (200 mL), added saturated Rochelle's salt (100 mL), stirred vigorously for 1 hour (until purple color dissipated to orange/yellow). Decanted organic layer from thick white aqueous layer, washed with additional Rochelle's salt, brine, dried and concentrated to an orange residue. The residue was slurried in ether (20 mL) to give title compound (930 mg, 52% yield). MS APCI, m/z=307/309 (M+H).
  • (2E)-2-Cyano-2-[(2-bromoophenyl)hydrazono]-N-cyclopropylacetamide
  • Using the procedure outlined in the U.S. Pat. No. 4,886,800 example 89b substituting 2-bromoaniline for 2-iodoaniline and 2-cyano-N-cyclopropylacetamide for 2-cyano-N-propylacetamide, to give 11.1 g (85% yield) of the title compound as a yellow solid. MS APCI, m/z=307/309 (M+H). 1H NMR (300 MHz, CDCl3) δ 14.39, (s, 1H), 8.67 (bm, 1H), 7.45 (m, 1H), 7.32 (m, 1H), 7.03 (m, 1H), 3.1 (apparent q, J=6.6 Hz, 2H), 1.53 (apparent sextet, J=7.4 Hz, 2H), 0.88 (t, J=7.4 Hz, 3H). The intermediate compounds were prepared as follows:
  • 2-Cyano-N-cyclopropylacetamide
  • To a flask charged with cyclopropylamine (12.3 g, 215.3 mmol) was added ethyl cyanoacetate (9.8 g, 86.1 mmol). The reaction was stirred at 45° C. for 1.5 hour, cooled and concentrated under reduced pressure to give 10.7 g title compound (˜100%) as a light yellow solid. 1H NMR (300.132 MHz, CDCl3) δ 6.20 (bs, 1H), 3.34 (s, 2H), 2.75 (m, 2H), 0.83 (m, 2H), 0.59 (m, 2H).
  • Precursor 14 4-Amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide
  • Using the procedure outline for 4-Amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (precursor 13) substituting N-cyclopropyl-2-cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]acetamide (5.8 g, 15.6 mmol) for N-cyclopropyl-2-cyano-2-[(2-bromophenyl)hydrazono]acetamide to give title compound (3.3 g, 57% yield). MS APCI, m/z=373 (M+H).
  • (2E)-2-Cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-cyclopropylacetamide
  • Using the procedure outlined in the U.S. Pat. No. 4,886,800 example 89b substituting 3-fluoro-2-iodoaniline for 2-iodoaniline and 2-cyano-N-cyclopropylacetamide for 2-cyano-N-propylacetamide, to give 8.9 g (94% yield) of the title compound as a yellow solid. MS APCI, m/z=373 (M+H)
  • Detailed Synthesis Methods/Procedures:
  • Method A: The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (typically 2-3 molar equivalents), cesium carbonate (2 molar equivalents) and bis(triphenylphosphine)palladium(II) dichloride (0.025 molar equivalents) were placed in a microwave reaction vessel and dissolved in 7:3:2 (v/v/v) 1,2-dimethoxyethane:water:ethanol (5 mL/mmol cinnoline-halide) at ambient temperature. The reaction vessel was capped, the head-space purged with dry nitrogen and the stirred mixture was heated on a Biotage Optimizer (300 W) microwave system maintaining a reaction temperature of 150° C. for 30-90 minutes, reaction pressures of 7 bar were typically observed. The reaction was then cooled to ambient temperature and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method B: To a solution of the cinnoline-halide in 1,2-dimethoxyethane (10 mL/mmol cinnoline-halide) under nitrogen at ambient temperature was added tetrakis(triphenylphosphine)palladium (0) (0.05-0.15 molar equivalents). After stirring 10-20 min an arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (1-4 molar equivalents) was added followed by a solution of sodium carbonate (2.5 molar equivalents) in water (3 mL/mmol halide). The resulting mixture was heated at reflux for 2-24 h. The reaction was then cooled to ambient temperature and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method C: To a stirred solution of the cinnoline-halide in anhydrous N,N-dimethylformamide (2 mL/mmol cinnoline-halide) at ambient temperature was added an optionally substituted aryl- or heteroaryl-tin reagent (1.2 molar equivalents) and tetrakis(triphenylphosphine)palladium(0) (0.05 molar equivalents). The mixture was heated at 100° C. for 8-48 h. The reaction was then cooled to ambient temperature and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with an increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method D: The cinnoline-halide, an optionally substituted aryl- or heteroaryl-tin reagent (1.2-3 molar equivalents) and tetrakis(triphenylphosphine) palladium(0) (0.05-0.10 molar equivalents) were placed in a microwave reaction vessel and dissolved in 2-4 mL of anhydrous N,N-dimethylformamide at ambient temperature. The reaction vessel was purged with nitrogen, capped, and the stirred mixture was heated on a Biotage Optimizer (300 W) microwave system maintaining a reaction temperature of 150° C. for 30 minutes. The reaction was cooled to ambient temperature, diluted with methylene chloride, washed with water, dried over magnesium sulfate and the solvent was evaporated. The residue was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Method E: To a stirred solution of 8-trimethylstannyl-cinnoline derivative and tetrakis(triphenylphosphine) palladium(0) (0.05-0.10 molar equivalents) in anhydrous N,N-dimethylformamide at ambient temperature under nitrogen was added an optionally substituted aryl- or heteroaryl bromide (1.2-3 molar equivalents). The reaction was heated to 150° C. for 4-16 hours. The reaction mixture was evaporated under reduced pressure. The residue was dissolved in methylene chloride, washed with water twice, dried through MgSO4, and then the solvent was evaporated. The residue was purified by flash chromatography on silica gel eluting with an increasingly polar gradient of ethyl acetate in hexane to afford the desired compound.
  • Method F: To a solution of the cinnoline-halide in anhydrous tetrahydrofuran (10 mL/mmol cinnoline-halide) under nitrogen at ambient temperature was added (triphenylphosphine)palladium(II) dichloride (0.10 molar equivalents) followed by an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (2-4 molar equivalents) followed by freshly ground potassium phosphate (2.0 molar equivalents). The resulting mixture was heated at reflux for 2-40 h. The reaction was then cooled to ambient temperature and diluted with saturated sodium bicarbonate and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with 5% ether in chloroform to afford the desired compound.
  • Method G: The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (4-5 molar equivalents), cesium carbonate (4-5 molar equivalents), 2-dicyclohexylphosphino-2′,4′,6′-trisopropylbiphenyl (0.24 molar equivalents) and tris(dibenzylidene-acetone)dipalladium(0) (0.06 molar equivalents) were placed in a 3-neck flask under N2 and dissolved in 7:3:2 (v/v/v) THF:water:2-propanol (5 mL/mmol cinnoline-halide) at ambient temperature. The reaction vessel was fitted with a reflux condenser, capped, vacuum degassed (3×) backfilling with N2 and placed in a preheated oil bath (70° C.) and heated for 20 hours. (* if reaction not complete more boronic acid and cesium carbonate in equal proportions were added with additional heating time). The reaction was then cooled to ambient temperature, decanted organic layer and concentrated under reduced pressure. Residue partitioned between ethyl acetate and 5% sodium bicarbonate (aq). The residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes (alternately 1% methanol/dichloromethane) to afford the desired compound.
  • Method H: The cinnoline-halide, an optionally substituted arylboronic acid, heteroaryl boronic acid, or a boron compound 1-2B of Scheme 2 (3-5 molar equivalents), sodium carbonate (4-5 molar equivalents), [1,1′-bis(diphenylphospino)-ferrocene] dichloropalladium(II) complex with dichloromethane (1:1) (0.075 molar equivalents) were placed in a 3-neck flask under N2 and dissolved in 7:3:2 (v/v/v) THF:water:2-propanol (5 mL/mmol cinnoline-halide) at ambient temperature. The reaction vessel was fitted with a reflux condenser, under N2 and placed in a preheated oil bath (85° C.) and refluxed 2-20 hours (* if reaction not complete added more boronic acid with additional heating time). The reaction was then cooled to ambient temperature, reduced volume under reduced pressure, partitioned between ethyl acetate and water. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the desired compound.
  • Example 1 4-amino-7-fluoro-8-phenyl-N-propyl-cinnoline-3-carboxamide
  • Using method F 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (291 mg, 0.78 mmol) and phenylboronic acid (379 mg, 3.11 mmol) were reacted (reflux 4 hours) to afford the title compound (65 mg, 26% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (bs, 1H), 7.89 (dd, J=9.2, 4.6 Hz, 1H), 7.42-7.60 (m, 6H), 3.45 (apparent q, J=6.6 Hz, 2H), 1.65 (apparent sextet, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=325 (M+H) HPLC 1.92 min.
  • Example 2 4-amino-7-chloro-8-phenyl-N-propyl-cinnoline-3-carboxamide
  • Using Method F, 4-amino-7-chloro-8-iodo-N-propyl-cinnoline-3-carboxamide (184 mg, 0.47 mmol) and phenylboronic acid (229 mg, 1.89 mmol) were reacted (refluxed 40 hours) to afford the title compound (90 mg, 56% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.49 (bs, 1H), 7.82 (d, J=9.0 Hz, 1H), 7.75 (d, J=9.0 Hz, 1H), 7.42-7.55 (m, 5H), 3.43 (apparent q, J=6.6 Hz, 2H), 1.63 (apparent sextet, J=7.2 Hz, 2H), 0.98 (t, J=7.4 Hz, 3H). MS APCI, m/z=341 (M+H) HPLC 2.04 min.
  • Example 3 4-amino-7-methoxy-8-phenyl-N-propyl-cinnoline-3-carboxamide
  • Using Method F, 4-amino-7-methoxy-8-iodo-N-propyl-cinnoline-3-carboxamide (311 mg, 0.81 mmol) and phenylboronic acid (394 mg, 3.24 mmol) were reacted (refluxed overnight) to afford the title compound (140 mg, 52% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.51 (bm, 1H), 7.90 (d, J=9.2 Hz, 1H), 7.52 (d, J=9.2 Hz, 1H), 7.36-7.50 (m, 5H), 3.92 (s, 3H), 3.43 (apparent q, J=6.4 Hz, 2H), 1.63 (apparent sextet, J=7.2 Hz, 2H), 0.98 (t, J=7.4 Hz, 3H). MS APCI, m/z=337 (M+H) HPLC 1.76 min.
  • Example 4 4-amino-7-chloro-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-7-chloro-8-iodo-N-propyl-cinnoline-3-carboxamide (78 mg, 0.20 mmol) and (2,5-dimethylphenyl)boronic acid (63 mg, 0.417 mmol) were reacted to afford the title compound (33 mg, 45% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.48 (bm, 1H), 7.85 (bm, 1H), 7.75 (m, 1H), 7.15-7.24 (m, 2H), 6.98 (s, 1H), 3.43 (apparent q, J=6.7 Hz, 2H), 2.36 (s, 3H), 1.95 (s, 3H), 1.63 (apparent sextet, J=7.2 Hz, 2H), 0.98 (t, J=7.4 Hz, 3H). MS APCI, m/z=369 (M+H) HPLC 2.15 min.
  • Example 5 4-amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.324 mmol) and (2,4-dimethoxypyrimidin-5-yl)boronic acid (125 mg, 0.68 mmol) were reacted to afford the title compound (33 mg, 28% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (bm, 1H), 8.33 (s, 1H), 7.91 (dd, J=7.7, 2.0 Hz, 1H), 7.70-7.77 (m, 2H), 4.06 (s, 3H), 3.93 (s, 3H), 3.46 (apparent q, J=6.5 Hz, 2H), 1.67 (apparent sextet, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=369 (M+H) HPLC 1.69 min.
  • Example 6 4-amino-8-(5-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.324 mmol) and 3-methoxy-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (160 mg, 0.68 mmol) were reacted to afford the title compound (84 mg, 77% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.49-8.60 (m, 2H), 8.39 (m, 1H), 7.93 (dd, J=8.2, 1.6 Hz, 1H), 7.73-7.84 (m, 2H), 7.66 (m, 1H), 3.93 (s, 3H), 3.47 (apparent q, J=6.7 Hz, 2H), 1.68 (apparent sextet, J=7.4 Hz, 2H), 0.98 (t, J=7.4 Hz, 3H). MS APCI, m/z=338 (M+H) HPLC 1.52 min.
  • Example 7 4-amino-8-(2-methoxypyrimidin-5-yl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.324 mmol) and (2-methoxypyrimidin-5-yl)boronic acid (104 mg, 0.68 mmol) were reacted to afford the title compound (84 mg, 77% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.1-9.3 (m, 1.5H), 8.96 (s, 2H), 8.47 (dd, J=8.4, 1.0 Hz, 1H), 8.1-8.4 (bm, 0.5H), 7.99 (dd, J=7.2, 1.0 Hz, 1H), 7.83 (dd, J=8.4, 7.2 Hz, 1H), 4.01 (s, 3H), 3.32 (apparent q, J=7.4 Hz, 2H), 1.60 (apparent sextet, J=7.2 Hz, 2H), 0.91 (t, J=7.4 Hz, 3H). MS APCI, m/z=339 (M+H) HPLC 1.75 min.
  • Example 8 4-amino-8-(3-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (116 mg, 0.375 mmol) and (3-fluoro-2-methoxyphenyl)boronic acid (127 mg, 0.75 mmol) were reacted to afford the title compound (117 mg, 88% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.06 (t, J=6.0 Hz, 1H), 8.45 (dd, J=7.5, 2.2 Hz, 1H), 7.74-7.81 (m, 2H), 7.28-7.37 (m, 1H), 7.12-7.21 (m, 2H), 3.54 (s, 3H), 3.31 (apparent q, J=7.0 Hz, 2H), 1.56 (apparent sextet, J=7.0 Hz, 2H), 0.90 (t, J=7.0 Hz, 3H). MS APCI, m/z=355 (M+H) HPLC 1.86 min.
  • Example 9 4-amino-8-[4-methoxy-2-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (116 mg, 0.375 mmol) and [4-methoxy-2-(trifluoromethyl)phenyl]boronic acid (164 mg, 0.75 mmol) were reacted to afford the title compound (124 mg, 82% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.02 (t, J=6.0 Hz, 1H), 8.45 (d, J=8.3 Hz, 1H), 7.67-7.78 (m, 2H), 7.26-7.38 (m, 3H), 3.91 (s, 3H), 3.29 (apparent q, J=7.0 Hz, 2H), 1.57 (apparent sextet, J=7.0 Hz, 2H), 0.90 (t, J=7.0 Hz, 3H). MS APCI, m/z=405 (M+H) HPLC 2.11 min.
  • Example 10 4-amino-8-(2,5-difluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (116 mg, 0.375 mmol) and (2,5-difluoro-4-methoxyphenyl)boronic acid (140 mg, 0.75 mmol) were reacted to afford the title compound (93 mg, 67% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.15 (t, J=6.0 Hz, 1H), 8.46 (dd, J=8.0, 1.7 Hz, 1H), 7.76-7.86 (m, 2H), 7.44 (dd, J=11.8, 6.8 Hz, 1H), 7.22 (dd, J=11.3, 7.4 Hz, 1H), 3.93 (s, 3H), 3.29 (apparent q, J=7.0 Hz, 2H), 1.59 (apparent sextet, J=7.0 Hz, 2H), 0.91 (t, J=7.0 Hz, 3H). MS APCI, m/z=373 (M+H) HPLC 2.03 min.
  • Example 11 4-amino-8-(5-fluoro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (126 mg, 0.408 mmol) and (5-fluoro-6-methoxypyridin-3-yl)boronic acid (138 mg, 0.81 mmol) were reacted to afford the title compound (70 mg, 48% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.22 (t, J=6.0 Hz, 1H), 8.45 (d, J=7.6 Hz, 1H), 8.29 (d, J=1.9 Hz, 1H), 8.13 (dd, J=11.9, 1.9 Hz, 1H), 7.95 (apparent d, J=7.0 Hz, 1H), 7.80 (apparent t, J=8.0 Hz, 1H), 4.03 (s, 3H), 3.31 (apparent q, J=7.0 Hz, 2H), 1.60 (apparent sextet, J=7.0 Hz, 2H), 0.91 (t, J=7.0 Hz, 3H). MS APCI, m/z=356 (M+H) HPLC 1.99 min
  • Example 12 4-amino-8-(5-chloro-6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (119 mg, 0.385 mmol) and (5-chloro-6-methoxypyridin-3-yl)boronic acid (144 mg, 0.77 mmol) were reacted to afford the title compound (59 mg, 42% yield) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.25 (t, J=6.0 Hz, 1H), 8.43-8.46 (m, 2H), 8.34 (d, J=2.0 Hz, 1H), 7.95 (apparent d, J=7.0, Hz, 1H), 7.80 (apparent t, J=7.8 Hz, 1H), 4.03 (s, 3H), 3.31 (apparent q, J=7.0 Hz, 2H), 1.59 (apparent sextet, J=7.0 Hz, 2H), 0.91 (t, J=7.0 Hz, 3H). MS APCI, m/z=372 (M+H) HPLC 2.17 min
  • Example 13 4-amino-8-(3,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method B, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3,5-dichlorophenyl boronic acid (252 mg, 1.32 mmol) were reacted to afford the title compound (65 mg, 52.5% yield) as a pale-yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 9.26 (br, 1H), 8.48 (d, J=8.2 Hz, 1H), 7.95 (d, J=7.2 Hz, 2H), 7.74-7.85 (m, 3H), 7.67 (t, J=2.0 Hz, 1H), 3.31 (m, overlapped with H2O), 1.60 (m, J=7.3 Hz, 2H), 0.91 (t, J=7.4 Hz, 3H). MS APCI, m/z=375/377 (M+H). HPLC 2.52 min.
  • Example 14 4-amino-8-(3,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method B, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (200 mg, 0.65 mmol), 3,5-difluorophenyl boronic acid (300 mg, 1.90 mmol) and bis(triphenylphosphine) palladium(II) dichloride (24 mg, 0.034 mmol) were reacted to afford the title compound (200 mg, 89.7% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.92 (dd, J=8.1, Hz, J′=1.4 Hz, 1H), 7.67-7.82 (m, 2H), 7.22 (m, 2H), 6.88 (m, 1H), 3.47 (q, J=6.7 Hz, 2H), 1.68 (m, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H). MS APCI, m/z=343 (M+H). HPLC 2.14 min.
  • Example 15 4-amino-8-(5-azetidin-1-ylcarbonyl-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method D, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.28 mmol) and 3-trimethylstannyl-5-(azetidin-1-ylcarbonyl)-pyridine (182 mg, of 80%, 0.45 mmol) were reacted to afford the title compound (48 mg, 44.0% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 9.00 (s, 1H), 8.89 (s, 1H), 8.51 (br, 1H), 8.40 (s, 1H), 7.96 (d, J=7.0 Hz, 1H), 7.72-7.88 (m, 2H), 4.46 (br, 2H), 4.28 (br, 2H), 3.48 (q, J=6.7 Hz, 2H), 2.40 (m, J=7.8 Hz, 2H), 1.69 (m, J=7.3 Hz, 2H), 1.02 (t, J=7.4 Hz, 3H). MS APCI, m/z=391 (M+H). HPLC 1.74 min.
  • The reagent, 3-trimethylstannyl-5-(azetidin-1-ylcarbonyl)-pyridine, was synthesized by the following method:
  • To a stirred suspension of 5-bromonicotinic acid (1.0 g, 4.95 mmol) in 15 mL of anhydrous methylene chloride at 0° C. under nitrogen was added oxaylic chloride (817 mg, 6.44 mmol). The reaction mixture was stirred at 0° C. for 30 minutes. Then triethylamine (1.25 g, 12.38 mmol) was added slowly, and followed by the addition of azetidine (565 mg, 9.90 mmol) at 0° C. The reaction was warmed to ambient temperature, and stirred for another hour. The reaction mixture was diluted with methylene chloride, quenched with water, washed with 10% potassium carbonate aqueous solution twice, dried through magnesium sulfate, and the solvent was evaporated to dry. The residue was purified by flash chromatography using a gradient of methanol in methylene chloride to give a yellow liquid as 3-bromo-5-(azetidin-1-ylcarbonyl)-pyridine (846 mg, 70.9% yield). Following, to a stirred solution of 3-bromo-5-(azetidin-1-ylcarbonyl)-pyridine (600 mg, 2.50 mmol) and tetrakis(triphenylphosphine) palladium(0) (240 mg, 0.21 mmol) in 40 mL of xylene at ambient temperature under nitrogen was added hexamethylditin (1.58 g, 4.50 mmol). The reaction was heated to 150° C. overnight. The reaction mixture was filtrated through Celite, and the filtrate was vacuumed to dry. The residual was dissolved in methylene chloride, washed with water twice, dried through MgSO4, and then the solvent was evaporated. The precipitate was purified by flash chromatography using a gradient of ethyl acetate in hexane to give a yellow solid as 3-trimethylstannyl-5-(azetidin-1-ylcarbonyl)-pyridine (846 mg, 83.0% yield). 1H NMR (300 MHz, CDCl3) δ 8.65-8.70 (m, 2H), 8.04 (t, J=1.9 Hz, 1H), 4.31 (t, J=7.63 Hz, 2H), 4.00-4.10 (m, overlapped with H2O), 2.27 (m, J=6.2 Hz, 2H MS APCI, m/z=323/325/327 (M+H) HPLC 1.61 min.
  • Example 16 4-amino-8-(2,3-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,3-dimethoxyphenyl boronic acid (148 mg, 0.97 mmol) were reacted to afford the title compound (106 mg, 89.5% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.89 (d, J=8.1, Hz, 1H), 7.78 (dd, J=7.1 Hz, J′=1.5 Hz, 1H), 7.71 (t, J=7.6 Hz, 1H), 7.15 (t, J=7.9 Hz, 1H), 6.99 (m, 2H), 3.92 (s, 3H), 3.53 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.65 (m, J=7.2 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=367 (M+H) HPLC 1.86 min.
  • Example 17 4-amino-8-(4-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 4-dimethylaminophenyl boronic acid (160 mg, 0.97 mmol) were reacted to afford the title compound (105 mg, 93.0% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.61 (br, 1H), 7.58-7.85 (m, 5H), 6.88 (d, J=8.8 Hz, 2H), 3.47 (q, J=6.7 Hz, 2H), 3.02 (s, 6H), 1.67 (m, J=7.3 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=350 (M+H) HPLC 2.67 min.
  • Example 18 4-amino-8-(3-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3-methoxyphenyl boronic acid (147 mg, 0.97 mmol) were reacted to afford the title compound (69 mg, 64.2% yield) as an off-white crystal. 1H NMR (300 MHz, CDCl3) δ 8.58 (br, 1H), 7.87 (dd, J=8.3 Hz, J′=1.4 Hz, 1H), 7.81 (dd, J=7.1 Hz, J′=1.4 Hz, 1H), 7.72 (t, J=8.1 Hz, 1H), 7.41 (t, J=8.0 Hz, 1H), 7.15-7.35 (m, overlapped with CHCl3), 6.98 (dd, J=8.1 Hz, J′=1.8 Hz, 1H), 3.86 (s, 3H), 3.46 (q, J=6.7 Hz, 2H), 1.67 (m, J=7.3 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=337 (M+H) HPLC 1.89 min.
  • Example 19 4-amino-8-(3,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3,4-dimethoxyphenyl boronic acid (148 mg, 0.97 mmol) were reacted to afford the title compound (91 mg, 77.7% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.58 (br, 1H), 7.76-7.88 (m, 2H), 7.71 (t, J=7.7 Hz, 1H), 7.29 (m, overlapped with CHCl3), 7.23 (d, J=1.9 Hz, 1H), 7.02 (d, J−8.3 Hz, 1H), 3.95 (s, 3H), 3.93 (s, 3H), 3.47 (q, J=6.7 Hz, 2H), 1.68 (m, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=367 (M+H) HPLC 1.78 min.
  • Example 20 4-amino-8-(2,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method B, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (13.0 g, 42.1 mmol), 2,5-dimethoxyphenyl boronic acid (15.4 g, 84.6 mmol) and bis(triphenylphosphine) palladium(II) dichloride (886 mg, 1.3 mmol) were reacted to afford the title compound (13.51 g, 87.7% yield) as an off-white needle. 1H NMR (300 MHz, CDCl3) δ 8.59 (br, 1H), 7.89 (dd, J=7.8 Hz, J′=1.9 Hz, 1H), 7.65-7.77 (m, 2H), 6.85-7.20 (m, 3H), 3.79 (s, 3H), 3.64 (s, 3H), 3.35-3.55 (m, overlapped with H2O), 1.64 (m, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=367 (M+H) HPLC 1.72 min.
  • Example 21 4-amino-8-(3,5-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2-(3,5-dimethoxyphenyl)-4,4,5,5-tetramethyl-(1,3,2)-dioxaborolane (256 mg, 0.97 mmol) were reacted to afford the title compound (110 mg, 93.9% yield) as an off-white solid.
  • 1H NMR (300 MHz, CDCl3) δ 8.57 (br, 1H), 7.87 (d, J=8.2 Hz, 1H), 7.79 (dd, J=7.2 Hz, J′=1.3 Hz, 1H), 7.71 (t, J=7.7 Hz, 1H), 6.79 (d, 3H), 3.83 (s, 6H), 3.47 (q, J=6.7 Hz, 2H), 1.67 (m, J=7.3 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=367 (M+H) HPLC 1.98 min.
  • Example 22 4-amino-8-(2,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,4-dimethoxyphenyl boronic acid (148 mg, 0.97 mmol) were reacted to afford the title compound (88 mg, 75.1% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.57 (br, 1H), 7.84 (dd, J=8.2 Hz, J′=1.4 Hz, 1H), 7.75 (dd, J=7.1 Hz, J′=1.4 Hz, 1H), 7.68 (t, J=7.6 Hz, 1H), 7.29 (m, overlapped with CHCl3), 6.58-6.60 (m, 2H), 3.87 (s, 3H), 3.69 (s, 3H), 3.45 (q, 6.7 Hz, 2H), 1.65 (m, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=367 (M+H) HPLC 1.94 min.
  • Example 23 4-amino-8-(2-fluoro-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method E, 4-amino-8-trimethylstannyl-N-propyl-cinnoline-3-carboxamide (170 mg of 90% purity, 0.37 mmol) and 3-bromo-2-fluoro-3-pyridine (195 mg, 1.11 mmol) were reacted to afford the title compound (45 mg, 37.7% yield) as an off-white solid. 1H NMR (300 MHz, DMSO-d6) δ 9.16 (br, 1H), 8.52 (dd, J=8.4 Hz, J′=1.2 Hz, 1H), 8.33 (m, 1H), 8.11 (m, 1H), 7.92 (d, J=6.1 Hz, 1H), 7.83 (t, J=7.7 Hz, 1H), 7.50 (m, 1H), 3.20-3.35 (m, overlapped with H2O), 1.58 (m, J=7.2 Hz, 2H), 0.90 (t, J=7.4 Hz, 3H) MS APCI, m/z=326 (M+H) HPLC 1.74 min
  • Example 24 4-amino-8-(2,3-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,3-difluorophenyl boronic acid (153 mg, 0.97 mmol) were reacted to afford the title compound (63 mg, 57.6% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.95 (dd, J=8.0 Hz, J′=1.7 Hz, 1H), 7.70-7.85 (m, 2H), 7.15-7.30 (m, overlapped with CHCl3), 3.46 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=343 (M+H) HPLC 2.08 min.
  • Example 25 4-amino-8-(2,3-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,3-dichlorophenyl boronic acid (185 mg, 0.97 mmol) were reacted to afford the title compound (99.8 mg, 83.1% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (br, 1H), 7.96 (dd, J=7.6 Hz, J′=2.1 Hz, 1H), 7.66-7.78 (m, 2H), 7.53 (dd, J=6.3 Hz, J′=3.3 Hz, 1H), 7.28-7.35 (m, 2H), 3.45 (q, J=6.7 Hz, 2H), 1.64 (m, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=375/377 (M+H) HPLC 2.19 min.
  • Example 26 4-amino-N-propyl-8-(6-quinolyl)cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 6-(4,4,5,5-tetramethyl-1,3,2-dioxa-borolane-2-yl)quinoline (247 mg, 0.97 mmol) were reacted to afford the title compound (105 mg, 91.9% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.96 (dd, J=4.2 Hz, J′=1.7 Hz, 1H), 8.56 (br, 1H), 8.23 (d, J=8.4 Hz, 2H), 8.05-8.15 (m, 2H), 7.88-7.96 (m, 2H), 7.78 (t, J=7.7 Hz, 1H), 7.44 (dd, J=8.2 Hz, J′=4.2 Hz, 1H), 3.47 (q, J=6.7 Hz, 2H), 1.67 (m, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H)
  • MS APCI, m/z=358 (M+H) HPLC 1.47 min.
  • Example 27 4-amino-N-propyl-8-(3-quinolyl)cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3-quinoline boronic acid (168 mg, 0.97 mmol) were reacted to afford the title compound (93 mg, 80.5% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 9.24 (d, J=2.2 Hz, 1H), 8.50-8.60 (m, 2H), 8.18 (d, J=8.6 Hz, 1H), 7.88-7.98 (m, 3H), 7.82 (d, J=8.2 Hz, 1H), 7.76 (m, 1H), 7.59 (m, 1H), 3.47 (q, J=6.7 Hz, 2H), 1.68 (m, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=358 (M+H) HPLC 1.88 min.
  • Example 28 4-amino-8-(2-naphthyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2-naphthalene boronic acid (167 mg, 0.97 mmol) were reacted to afford the title compound (99 mg, 86.9% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.58 (br, 1H), 8.11 (s, 1H), 7.83-7.99 (m, 6H), 7.76 (dd, J=8.0 Hz, J′=7.1 Hz, 1H), 7.46-7.55 (m, 2H), 3.46 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=357 (M+H) HPLC 2.11 min.
  • Example 29 4-amino-8-(1H-indol-5-yl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 5-indolyl boronic acid (156 mg, 0.97 mmol) were reacted to afford the title compound (105 mg, 95.1% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.60 (br, 1H), 8.34 (s, 1H), 7.94 (s, 1H), 7.86 (d, J=7.1 Hz, 1H), 7.81 (d, J=8.4 Hz, 1H), 7.69 (t, J=7.7 Hz, 1H), 7.57 (d, J=8.5 Hz, 1H), 7.47 (d, J=8.5 Hz, 1H), 7.22 (s, 1H), 6.61 (s, 1H), 3.46 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.2 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=346 (M+H) HPLC 1.88 min.
  • Example 30 4-amino-8-(4-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • To a stirred solution of 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (1.00 g, 2.81 mmol), sodium bicarbonate (473 mg, 5.62 mmol) and tetrakis(triphenylphosphine) palladium(0) (974 mg, 0.84 mmol) in 1,2-dimethoxyethane (180 mL)/water (30 mL) at 85° C. under nitrogen was added the aqueous solution of 4-methoxy-pyridine-3-boronic acid (25 mg/mL) dropwise, maintaining the internal temperature between 80° C. and 85° C. The reaction was monitored by HPLC until completed. Upon the completion, 2.42 equivalents of 4-methoxy-pyridine-3-boronic acid (1.16 g, 6.80 mmol) were applied. The reaction mixture was diluted with methylene chloride (300 mL), washed with water twice, dried through MgSO4, and then the solvent was evaporated. The residual was purified by flash chromatography using a gradient of methanol in methylene chloride to give a yellow solid. The yellow solid was crystallized from methylene chloride/methanol (2/1) to give an off-white needle crystal as the title compound (570 mg, 60.2% yield). 1H NMR (300 MHz, CDCl3) δ 8.58 (d, J=5.8 Hz, 1H), 8.53 (br, 1H), 5.11 (m, 6H), 8.46 (s, 1H), 7.94 (t, J=4.9 Hz, 1H), 7.74 (d, J=4.8 Hz, 2H), 6.96 (d, J=5.8 Hz, 1H), 3.78 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.66 (m, overlapped with H2O), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=338 (M+H) HPLC 1.28 min.
  • Example 31 4-amino-8-(3-dimethylaminophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3-dimethylaminophenyl boronic acid (160 mg, 0.97 mmol) were reacted to afford the title compound (99 mg, 88.6% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.60 (br, 1H), 7.83 (m, 2H), 7.70 (t, J=7.7 Hz, 1H), 7.35 (t, J=7.9 Hz, 1H), 7.03 (d, J=7.9 Hz, 1H), 6.98 (m, 1H), 6.82 (dd, J=8.3 Hz, J′=2.3 Hz, 1H), 3.46 (q, J=6.7 Hz, 2H), 2.99 (s, 6H), 1.67 (m, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=350 (M+H) HPLC 1.60 min.
  • Example 32 4-amino-N-propyl-8-(3,4,5-trimethoxyphenyl)-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3,4,5-trimethoxyphenyl boronic acid (206 mg, 0.97 mmol) were reacted to afford the title compound (116 mg, 91.5% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.88 (d, J=8.2 Hz, 1H), 7.80 (dd, J=7.2 Hz, J′=1.4 Hz, 1H), 7.72 (t, J=7.7 Hz, 1H), 6.87 (s, 2H), 3.92 (s, 3H), 3.90 (s, 6H), 3.47 (q, J=6.5 Hz, 2H), 1.68 (m, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=397 (M+H) HPLC 1.83 min.
  • Example 33 4-amino-8-(2,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,4-difluorophenyl boronic acid (202 mg, 1.28 mmol) were reacted to afford the title compound (101 mg, 92.3% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.92 (dd, J=8.0 Hz, J′=1.7 Hz, 1H), 7.70-7.80 (m, 2H), 7.49 (m, 1H), 6.90-7.05 (m, 2H), 3.46 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=343 (M+H) HPLC 1.84 min.
  • Example 34 4-amino-8-(3,4-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 3,4-difluorophenyl boronic acid (202 mg, 1.28 mmol) were reacted to afford the title compound (108 mg, 98.7% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.89 (dd, J=7.9 Hz, J′=1.9 Hz, 1H), 7.68-7.80 (m, 2H), 7.48-7.59 (m, 1H), 7.37-7.45 (m, 1H), 7.23-7.33 (m, overlapped with CHCl3), 3.47 (q, J=6.7 Hz, 2H), 1.68 (m, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H) MS APCI, m/z=343 (M+H) HPLC 2.01 min.
  • Example 35 4-amino-N-propyl-8-(2,3,4-trimethoxyphenyl)-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (106 mg, 0.34 mmol) and 2,3,4-trimethoxyphenyl boronic acid (206 mg, 0.97 mmol) were reacted to afford the title compound (124 mg, 92.1% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.88 (dd, J=8.2 Hz, J′=1.5 Hz, 1H), 7.76 d, J=6.3 Hz, 1H), 7.69 (t, J=7.6 Hz, 1H), 7.09 (d, J=8.6 Hz, 1H), 6.80 (d, J=8.6 Hz, 1H), 3.94 (s, 3H), 3.92 (s, 3H), 3.62 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.65 (m, J=7.2 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=397 (M+H) HPLC 1.70 min.
  • Example 36 4-amino-8-(2-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2-methoxy-pyridyl-3-boronic acid (148 mg, 0.97 mmol) were reacted to afford the title compound (92 mg, 85.3% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.53 (br, 1H), 8.26 (dd, J=5.0 Hz, J′=1.9 Hz, 1H), 7.90 (dd, J=8.1 Hz, J′=1.5 Hz, 1H), 7.79 (dd, J=7.1 Hz, J′=1.6 Hz, 1H), 7.74 (d, J=8.0 Hz, 1H), 7.70 (dd, J=7.3 Hz, J′=2.0 Hz, 1H), 7.04 (dd, J=6.8 Hz, J′=5.0 Hz, 1H), 3.88 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=338 (M+H) HPLC 1.49 min.
  • Example 37 4-amino-8-(2,6-dimethoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,6-dimethoxy-pyridyl-3-boronic acid (120 mg, 0.64 mmol) were reacted to afford the title compound (110 mg, 92.6% yield) as a pale-yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.85 (dd, J=8.3 Hz, J′=1.4 Hz, 1H), 7.79 (d, J=7.2 Hz, 1H), 7.60-7.73 (m, 2H), 6.46 (d, J=8.0 Hz, 1H), 3.98 (s, 3H), 3.88 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.66 (m, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H) MS APCI, m/z=368 (M+H) HPLC 1.77 min.
  • Example 38 4-amino-8-(2,5-dimethylphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method B, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (250 mg, 0.702 mmol) and 2,5-dimethylphenylboronic acid (150 mg, 1.00 mmol) were reacted to afford the title compound (195 mg, 83% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.87 (m, 1H), 7.75-7.64 (m, 2H), 7.23-7.07 (m, 3H), 3.44 (apparent quartet, J=7.0 Hz, 2H), 2.35 (s, 3H), 2.01 (s, 3H), 1.64 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H) MS APCI, m/z=335 HPLC 1.98 min.
  • Example 39 3-[4-amino-3-(propylcarbamoyl)cinnolin-8-yl]benzoic acid hydrochloride
  • Using method B, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.421 mmol) and 3-(dihydroxyboryl)benzoic acid (77 mg, 0.464 mmol) were reacted to afford the title compound (117 mg, 79% yield) as a white solid. 1H NMR (300 MHz, Methanol-d4) δ 8.58-8.52 (m, 1H), 8.30-8.22 (m, 2H), 8.07-7.92 (m, 2H), 7.86-7.73 (m, 2H), 3.41 (t, J=7.0 Hz, 2H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=351 (M+H)
  • HPLC 1.65 min.
  • Example 40 4-amino-8-(3-azetidin-1-ylcarbonylphenyl)-N-propyl-cinnoline-3-carboxamide
  • To a stirred solution of 3-[4-amino-3-(propylcarbamoyl)cinnolin-8-yl]benzoic acid (67 mg, 0.191 mmol) dissolved in anhydrous DMF (2 mL) at ambient temperature under argon was added azetidine (16.4 mg, 0.287 mmol), N-methylmorpholine (29 mg, 0.287 mmol), 1-hydroxybenzotriazole (44 mg, 0.287 mmol) and N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (55 mg, 0.287 mmol). The mixture was stirred at ambient temperature for 19 hours then diluted with water and extracted with ethyl acetate. The residue from the organic extracts was purified by flash chromatography on silica gel eluting with increasingly polar gradient of ethyl acetate in hexanes to afford the title compound (61 mg, 82% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 8.03-7.64 (m, 6H), 7.53 (t, J=7.7 Hz, 1H), 4.38 (br, 2H), 4.25 (br, 2H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 2.34 (apparent pentet, J=7.5 Hz, 2H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H)
  • MS APCI, m/z=390 (M+H) HPLC 1.70 min.
  • Example 41 4-amino-N-propyl-8-pyrazin-2-yl-cinnoline-3-carboxamide
  • Using method C, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.500 mmol) and 2-(tributylstannyl)pyrazine (219 mg, 0.600 mmol) were reacted to afford the title compound (70 mg, 45% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 9.41 (m, 1H), 8.74 (m, 1H), 8.61 (d, J=2.5 Hz, 1H), 8.54 (br, 1H), 8.22 (m, 1H), 7.99 (m, 1H), 7.85-7.78 (m, 1H), 3.48 (apparent quartet, J=7.0 Hz, 2H), 1.69 (apparent sextet, J=7.0 Hz, 2H), 1.20 (t, J=7.0 Hz, 3H). MS APCI, m/z=309 (M+H) HPLC 1.49 min.
  • Example 42 4-amino-N-propyl-8-(3-pyridyl)cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (200 mg, 0.647 mmol) and pyridin-3-ylboronic acid (160 mg, 1.301 mmol) were reacted to afford the title compound (153 mg, 74% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.89 (m, 1H), 8.68 (m, 1H), 8.54 (br, 1H), 8.13 (m, 1H), 7.93 (m, 1H), 7.85-7.73 (m, 2H), 7.44 (m, 1H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=308 (M+H) HPLC 1.46 min.
  • Example 43 4-amino-8-(3-methylsulfonylphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 3-(methylsulfonyl)phenylboronic acid (200 mg, 1.000 mmol) were reacted to afford the title compound (155 mg, 83% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.51 (br, 1H), 8.21 (m, 1H), 8.11-7.67 (m, 6H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 3.12 (s, 3H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=385 M+H)
  • HPLC 1.75 min.
  • Example 44 4-amino-8-(3-cyanophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 3-cyanophenylboronic acid (147 mg, 1.000 mmol) were reacted to afford the title compound (138 mg, 86% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 8.06-7.89 (m, 3H), 7.85-7.69 (m, 3H), 7.65-7.57 (m, 1H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=332 M+H)
  • HPLC 1.93 min.
  • Example 45 4-amino-N-propyl-8-(2-pyridyl)cinnoline-3-carboxamide
  • Using method C, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.500 mmol) and 2-(tributylstannyl)pyridine (220 mg, 0.600 mmol) were reacted to afford the title compound (60 mg, 39% yield) as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.79 (m, 1H), 8.52 (br, 1H), 8.26-8.21 (m, 1H), 8.14-8.09 (m, 1H), 8.05-7.98 (m, 1H), 7.88-7.75 (m, 2H), 7.34 (m, 1H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=308 (M+H). HPLC 1.53 min.
  • Example 46 4-amino-8-[3,5-bis(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 3,5-bis(trifluoromethyl)phenylboronic acid (258 mg, 1.000 mmol) were reacted to afford the title compound 200 mg, 94% yield as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.50 (br, 1H), 8.13 (s, 2H), 7.99-7.93 (m, 2H), 7.84-7.73 (m, 2H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=443 (M+H). HPLC 2.85 min.
  • Example 47 4-amino-N-propyl-8-(1H-pyrazol-4-yl)cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (194 mg, 1.000 mmol) were reacted to afford the title compound (35 mg, 25% yield) as an off-white solid. 1H NMR (300 MHz, DMSO-d6) δ 12.97 (br, 1H), 9.20 (broad triplet, 1H), 8.48 (br, 2H), 8.23 (d, J=8.3 Hz, 1H), 8.14 (d, J=6.6 Hz, 1H), 7.70 (m, 1H), 3.32 (m, 2H), 1.62 (apparent sextet, J=7.0 Hz, 2H), 0.93 (t, J=7.0 Hz, 3H). MS APCI, m/z=297 (M+H). HPLC 1.43 min.
  • Example 48 4-amino-8-[2-chloro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-chloro-5-(trifluoromethyl)phenylboronic acid (224 mg, 1.000 mmol) were reacted to afford the title compound (85 mg, 44% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.51 (br, 1H), 7.98 (m, 1H), 7.81-7.63 (m, 5H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=409 (M+H). HPLC 2.46 min.
  • Example 49 4-amino-8-(2-methoxy-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-methoxy-5-methyl-phenylboronic acid (166 mg, 1.000 mmol) were reacted to afford the title compound (141 mg, 83% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.88-7.82 (m, 1H), 7.76-7.65 (m, 2H), 7.22-7.12 (m, 2H), 6.93 (m, 1H), 3.67 (s, 3H), 3.45 (apparent q, J=7.0 Hz, 2H), 2.34 (s, 3H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=351 (M+H). HPLC 1.96 min.
  • Example 50 4-amino-N-propyl-8-[2-(trifluoromethyl)phenyl]-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-trifluororomethyl-phenylboronic acid (190 mg, 1.000 mmol) were reacted to afford the title compound (115 mg, 64% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.50 (br, 1H), 7.93 (m, 1H), 7.82 (m, 1H), 7.71 (m, 1H), 7.66-7.50 (m, 2H), 7.41 (m, 1 h), 3.43 (apparent quartet, J=7.0 Hz, 2H), 1.63 (apparent sextet, J=7.0 Hz, 2H), 0.98 (t, J=7.0 Hz, 3H). MS APCI, m/z=375 (M+H). HPLC 2.05 min.
  • Example 51 4-amino-8-(5-chloro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-methoxy-5-chloro-phenylboronic acid (186 mg, 1.000 mmol) were reacted to afford the title compound (137 mg, 77% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.89 (m, 1H), 7.71 (m, 2H), 7.40-7.29 (m, 2H), 6.95 (m, 1H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 3H). MS APCI, m/z=371 (M+H). HPLC 2.00 min.
  • Example 52 4-amino-N-propyl-8-(4-pyridyl)cinnoline-3-carboxamide
  • Using method C, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.500 mmol) 4-(tributylstannyl)pyridine (220 mg, 0.600 mmol) were reacted to afford the title compound (47 mg, 30% yield) as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 8.75 (m, 2H), 8.54 (br, 1H), 7.95 (m, 1H), 7.86-7.72 (m, 2H), 7.66 (m, 2H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.68 (m, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=308 (M+H) HPLC 1.47 min.
  • Example 53 4-amino-8-(2,5-dichlorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2,5-dichloro-phenylboronic acid (190 mg, 1.00 mmol) were reacted to afford the title compound (85 mg, 47% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (br, 1H), 7.99-7.91 (m, 1H), 7.78-7.67 (m, 2H), 7.48-7.31 (m, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=375 (M+H). HPLC 2.24 min.
  • Example 54 4-amino-8-(2,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2,5-difluoro-phenylboronic acid (160 mg, 1.00 mmol) were reacted to afford the title compound (116 mg, 70% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.97-7.91 (m, 1H), 7.83-7.69 (m, 2H), 7.27-7.05 (m, 3H), 3.46 (apparent quartet, J=7.0 Hz, 2H), 1.74-1.59 (m, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=343 (M+H). HPLC 2.01 min.
  • Example 55 4-amino-8-(1-methyl-1H-pyrazol-4-yl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (212 mg, 1.020 mmol) were reacted to afford the title compound (123 mg, 82% yield) as a white solid.
  • 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 8.50 (s, 1H), 8.05 (s, 1H), 8.02-7.95 (m, 1H), 7.75-0.60 (m, 2H), 4.01 (s, 3H), 3.49 (apparent quartet, J=7.0 Hz, 2H), 1.71 (apparent sextet, J=7.0 Hz, 2H), 1.03 (t, J=7.0 Hz, 3H). MS APCI, m/z=311 (M+H). HPLC 1.56 min.
  • Example 56 4-amino-8-(2-fluoro-3-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-3-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (99 mg, 57% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.93 (m, 1H), 7.83-7.69 (m, 2H), 7.22-7.14 (m, 1H), 7.10-7.00 (m, 2H), 3.93 (s, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=355 (M+H). HPLC 1.74 min.
  • Example 57 4-amino-8-(2,5-dimethyl-2H-pyrazol-3-yl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (200 mg, 0.647 mmol) and 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (288 mg, 1.297 mmol) were reacted to afford the title compound (45 mg, 21% yield) as a white solid.
  • 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.95 (m, 1H), 7.80-7.68 (m, 2H), 6.25 (s, 1H), 3.68 (s, 3H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 2.35 (s, 3H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H). MS APCI, m/z=325 (M+H). HPLC 1.55 min.
  • 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole
  • This precursor was prepared according to the method of A. V. Ivanchatchenko as described in the Journal of Heterocyclic Chemistry (2004) vol. 41 p. 931
  • Example 58 4-amino-8-[2-fluoro-5-(trifluoromethyl)phenyl]-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-5-(trifluoromethyl)phenylboronic acid (208 mg, 1.000 mmol) were reacted to afford the title compound (148 mg, 78% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (br, 1H), 7.97 (m, 1H), 7.83-7.67 (m, 4H), 7.32 (m, 1H), 3.46 (apparent quartet, J=7.0 Hz, 2H), 1.66 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=393 (M+H). HPLC 2.30 min.
  • Example 59 4-amino-8-(2-fluoro-5-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-5-methyl-phenylboronic acid (154 mg, 1.000 mmol) were reacted to afford the title compound (127 mg, 77% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.91 (m, 1H), 7.81-7.68 (m, 2H), 7.28 (m, 1H), 7.24-7.17 (m, 1H), 7.13-7.05 (m, 1H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 2.39 (s, 3H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=339 (M+H). HPLC 1.86 min.
  • Example 60 4-amino-8-(2-fluoro-4-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-4-methyl-phenylboronic acid (154 mg, 1.000 mmol) were reacted to afford the title compound (141 mg, 86% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.91 (m, 1H), 7.81-7.69 (m, 2H), 7.28 (m, 1H), 7.24-7.16 (m, 1H), 7.13-7.04 (m, 1H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 2.39 (s, 3H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=339 (M+H). HPLC 1.86 min.
  • Example 61 4-amino-8-(5-fluoro-2-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 5-fluoro-2-methyl-phenylboronic acid (154 mg, 1.000 mmol) were reacted to afford the title compound (142 mg, 86% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.53 (br, 1H), 7.91 (m, 1H), 7.77-7.63 (m, 2H), 7.26 (m, 1H), 7.07-6.97 (m, 2H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 2.01 (s, 3H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=339 (M+H). HPLC 1.77 min.
  • Example 62 4-amino-8-(4-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 4-fluoro-2-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (143 mg, 83% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.87 (m, 1H), 7.74-7.67 (m, 2H), 7.34-7.27 (m, 1H), 6.83-6.72 (m, 2H), 3.69 (s, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, 3H, J=7.0 Hz). MS APCI, m/z=355 (M+H). HPLC 1.66 min.
  • Example 63 4-amino-8-(3-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 3-fluoro-4-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (135 mg, 78% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.57 (br, 1H), 7.88-7.66 (m, 3H), 7.52-7.41 (m, 2H), 7.10 (m, 1H), 3.96 (s, 3H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H).
  • MS APCI, m/z=355 (M+H). HPLC 1.76 min.
  • Example 64 4-amino-8-(2-fluoro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-6-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (73 mg, 42% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.93 (m, 1H), 7.78-7.69 (m, 2H), 7.42-7.31 (m, 1H), 6.89-6.80 (m, 2H), 3.70 (s, 3H), 3.44 (apparent quartet, J=7.0 Hz, 2H), 1.64 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=355 (M+H). HPLC 1.68 min.
  • Example 64A Large Scale Synthesis of 4-Amino-8-(2-fluoro-6-methoxy-phenyl)-N-propylcinnoline-3-carboxamide
  • Figure US20090018112A1-20090115-C00246
  • A 2 L, 3-necked flask equipped with a reflux condenser, mechanical stirrer, and 250 mL addition funnel was charged with 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (36.80 g, 119.09 mmol), 2-fluoro-6-methoxy-phenylboronic acid (60.70 g, 357.06 mmol), and Pd(dppf)Cl2.CH2Cl2 (7.40 g, 9.06 mmol) under Argon at ambient temperature. A gentle vacuum was applied and the apparatus was back-filled with Argon two times. Tetrahydrofuran (515 mL, anhydrous) and isopropanol (147 mL, anhydrous) were added and the resulting red suspension was stirred at room temperature for 15 minutes. A solution of sodium carbonate (57.0 g, 537.7 mmol) in water (220 mL) was added rapidly through the addition funnel and the resulting mixture immediately placed into a pre-heated 80° C. oil bath. After 90 minutes at reflux (observed internal temperature 65° C.), the reaction mixture was cooled to room temperature and filtered though a bed of Celite supported on a sintered glass funnel topped with Norite decolorizing carbon (30 g). The residual salts and filter-cake were washed with 4:1 (v/v) THF: isopropanol until no additional material could be detected in the eluent by TLC (silica gel, 1:1 (v/v) hexanes:ethyl acetate, UV detection, Rf=0.25). The dark red solution was concentrated to a small volume under reduced pressure and then diluted with ethyl acetate (250 mL). The organic phase was separated and the aqueous phase extracted with ethyl acetate (2×250 mL). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residues were passed through a small pad of silica gel on a sintered glass funnel washing with ethyl acetate until no more material was detected in the eluent. The solution was evaporated to afford the crude product as a foamy red-brown solid. This material was purified by flash chromatography on silica gel using a gradient of 40 to 50% ethyl acetate in hexanes. Product containing fractions were combined and evaporated. The residue was precipitated from dichloromethane by addition of hexanes at room temperature. Recrystallization of this material from hot 1:1 (v/v) ethanol: water afforded the title compound as off-white white crystals (32.78 g, 78% yield). Additional title compound (4.30 g, 10% yield) was isolated by processing the residues form the crystallization liquors through an acid-base extractive workup.
  • 1H NMR (500.3 MHz, CDCl3) δ 8.54 (br, 1H), 7.90 (dd, J=8, 1 Hz, 1H), 7.75-7.67 (m, 2H), 7.37-7.31 (m, 1H), 6.86-6.80 (m, 2H), 3.69 (s, 3H), 3.44 (qd, J=7, 1 Hz, 2H), 1.64 (apparent sextet, J=7 Hz, 2H), 0.99 (t, J=7 Hz, 3H). The 4-Amino protons were not observable in the reported proton NMR spectra recorded at 30° C. due to severe broadening into the baseline. These protons can be clearly observed by recording the spectrum at −20° C. HRMS (C19H19FN4O2) Cal'd=355.1570, Observed=355.1531. HPLC 1.68 min.
  • It is found that the titled compound can be separated into two atropisomers using Supercritical Fluid Chromatography. Generally, in supercritical CO2 modified with methanol, these atropisomers are stable and hence are separable on a chiral support. However, in aqueous media and acidic aqueous media, in particular, the atropisomers inter-conversion is greatly facilitated.
  • 4-Amino-8-bromo-N-propyl-cinnoline-3-carboxamide
  • Prepared according to the method described in the U.S. Pat. No. 4,886,800 example 35a.
  • Example 65 4-amino-8-(2-fluoro-5-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 2-fluoro-5-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (142 mg, 83% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (br, 1H), 7.92 (m, 1H), 7.82-7.69 (m, 2H), 7.12 (m, 1H), 7.02-6.89 (m, 2H), 3.81 (s, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.66 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=355 (M+H). HPLC 1.78 min.
  • Example 66 4-amino-8-(5-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 5-fluoro-2-methoxy-phenylboronic acid (170 mg, 1.000 mmol) were reacted to afford the title compound (140 mg, 81% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.89 (m, 1H), 7.75-7.67 (m, 2H), 7.14-7.04 (m, 2H), 6.99-6.92 (m, 1H), 3.67 (s, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=355 (M+H). HPLC 1.67 min.
  • Example 67 4-amino-8-(4-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method F, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.50 mmol) and 4-methoxyphenylboronic acid (303 mg, 2.00 mmol) were reacted (reflux 14 hours) to afford the title compound (118 mg, 70% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.58 (bm, 1H), 7.69-7.83 (m, 3H), 7.62 (d, J=8.9 Hz, 2H), 7.05 (d, J=8.9 Hz, 2H), 3.87 (s, 3H), 3.45 (apparent q, J=6.6 Hz, 2H), 1.65 (apparent sextet, J=7.4 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H).
  • MS APCI, m/z=337 (M+H) HPLC 1.71 min.
  • Example 68 4-amino-8-(4-fluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method F, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.50 mmol) and 4-fluorophenylboronic acid (280 mg, 2.00 mmol) were reacted (reflux 24 hours) to afford the title compound (76 mg, 47% yield) as a white solid. 1H NMR (300 MHz, CDCl3)) δ8.56 (bm, 1H), 7.86 (dd, J=7.7 Hz, 1H), 7.65-7.80 (m, 4H), 7.19 (t, J=8.6 Hz, 2H), 3.45 (apparent q, J=6.6 Hz, 2H), 1.67 (apparent sextet, J=7.4 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H).
  • MS APCI, m/z=325 (M+H) HPLC 1.74 min.
  • Example 69 4-amino-N-propyl-8-[4-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide
  • Using method F, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.42 mmol) and 4-trifluoromethylphenylboronic acid (347 mg, 1.68 mmol) were reacted (reflux 16 hours) to afford the title compound (119 mg, 73% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.56 (bs, 1H), 7.90 (dd, J=7.9, 1.5 Hz, 1H), 7.71-7.82 (m, 4H), 7.35 (d, J=8.3 Hz, 2H), 3.45 (apparent q, J=6.8 Hz, 2H), 1.67 (apparent sextet, J=7.3 Hz, 2H), 1.01 (t, J=7.3 Hz, 3H).
  • MS APCI, m/z=391 (M+H) HPLC 2.21 min.
  • Example 70 4-amino-N-propyl-8-[3-(trifluoromethoxy)phenyl]-cinnoline-3-carboxamide
  • Using method F, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.42 mmol) and 3-trifluoromethylphenylboronic acid (347 mg, 1.68 mmol) were reacted (reflux 16 hours) to afford the title compound (94 mg, 57% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.56 (bs, 1H), 7.90 (d, J=8.3, 1H), 7.71-7.81 (m, 2H), 7.67 (d, J=7.9, 1H), 7.49-7.57 (m, 2H), 7.28 (m, 1H), 3.47 (apparent q, J=6.7 Hz, 2H), 1.67 (apparent sextet, J=7.3 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H). MS APCI, m/z=391 (M+H) HPLC 2.20 min.
  • Example 71 4-amino-8-(6-methoxy-3-pyridyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.49 mmol) and (6-methoxypyridin-3-yl)boronic acid (153 mg, 1.00 mmol) were reacted to afford the title compound (117 mg, 72% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.55 (bs, 1H), 8.43 (d, J=1.65 Hz, 1H), 8.05 (dd, J=8.5, 1.9 Hz, 1H), 7.87 (d, J=7.9 Hz, 1H), 7.71-7.81 (m, 2H), 6.89 (d, J=8.5 Hz, 1H), 4.01 (s, 3H), 3.46 (apparent q, J=6.6 Hz, 2H), 1.67 (apparent sextet, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H). MS APCI, m/z=338 (M+H) HPLC 1.73 min.
  • Example 72 4-amino-8-(4-methoxy-3,5-dimethyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (125 mg, 0.40 mmol) and (4-methoxy-3,5-dimethylphenyl)boronic acid (144 mg, 0.80 mmol) were reacted to afford the title compound (121 mg, 82% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.57 (bs, 1H), 7.66-7.85 (m, 3H), 7.32 (s, 2H), 3.78 (s, 3H), 3.46 (apparent q, J=6.6 Hz, 2H), 2.36 (s, 6H), 1.67 (apparent sextet, J=7.2 Hz, 2H), 1.01 (t, J=7.4 Hz, 3H). MS APCI, m/z=365 (M+H) HPLC 2.08 min.
  • Example 73 4-amino-8-(4-methoxy-3-methyl-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (125 mg, 0.40 mmol) and (4-methoxy-3-methylphenyl)boronic acid (133 mg, 0.80 mmol) were reacted to afford the title compound (105 mg, 75% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.59 (bs, 1H), 7.76-7.82 (m, 2H), 7.67-7.72 (m, 1H), 7.53 (dd, J=8.2, 2.2 Hz, 1H), 7.46 (m, 1H), 6.96 (d, J=8.2, 1H), 3.89 (s, 3H), 3.46 (apparent q, J=6.7 Hz, 2H), 2.30 (s, 3H), 1.67 (apparent sextet, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=351 (M+H) HPLC 1.88 min.
  • Example 74 4-amino-8-(2-fluoro-4-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (125 mg, 0.40 mmol) and (2-fluoro-4-methoxyphenyl)boronic acid (136 mg, 0.80 mmol) were reacted to afford the title compound (93 mg, 66% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ8.56 (bs, 1H), 7.88 (dd, J=8.2, 1.5 Hz, 1H), 7.69-7.77 (m, 2H), 7.43 (t, J=8.3 Hz, 1H), 6.76-6.86 (m, 2H), 3.86 (s, 3H), 3.45 (apparent q, J=6.6 Hz, 2H), 1.65 (apparent sextet, J=7.2 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=355 (M+H) HPLC 1.79 min.
  • Example 75 4-amino-8-(6-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
  • Using method C, 4-amino-8-iodo-N-propyl-cinnoline-3-carboxamide (178 mg, 0.500 mmol) and 2-methyl-5-(trimethylstannyl)pyridine (270 mg, 1.058 mmol) were reacted to afford the title compound (123 mg, 76% yield). 1H NMR (300 MHz, CDCl3) δ 8.75 (bs, 1H), 8.55 (br, 1H), 8.03 (m, 1H), 7.910 (m, 1H), 7.84-7.70 (m, 2H), 7.30 (m, 1H), 3.47 (apparent quartet, J=7.0 Hz, 2H), 2.64 (s, 3H), 1.67 (apparent sextet, J=7.0 Hz, 2H), 1.01 (t, J=7.0 Hz, 3H).
  • MS APCI, m/z=322 (M+H). HPLC 1.41 min.
  • 2-methyl-5-(trimethylstannyl)pyridine
  • Prepared according to the method described by Li et. al., J. Med. Chem., 1996, 39, 1846-1856.
  • Example 76 4-amino-8-(4-methylpyridin-3-yl)-N-propylcinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and (4-methylpyridin-3-yl)boronic acid (274 mg, 2.000 mmol) were reacted to afford the title compound (134 mg, 86% yield). 1H NMR (300 MHz, CDCl3) δ 8.59-8.45 (m, 3H), 7.98-7.92 (m, 1H), 7.80-7.66 (m, 2H), 7.27 (m, 1H), 3.44 (m, 2H), 2.11 (s, 3H), 1.65 (m, 2H), 1.00 (t, J=7.0 Hz, 3H). MS APCI, m/z=322 (M+H). HPLC 1.27 min.
  • Example 77 4-amino-8-(5-methoxy-2-methylphenyl)-N-propylcinnoline-3-carboxamide
  • Using method A, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.485 mmol) and 5-methoxy-2-methyl-phenylboronic acid (125 mg, 1.000 mmol) were reacted to afford the title compound (125 mg, 73% yield). 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.88 (m, 1H), 7.76-7.64 (m, 2H), 7.21 (m, 1H), 6.92-6.81 (m, 2H), 3.79 (s, 3H), 3.45 (apparent quartet, J=7.0 Hz, 2H), 1.97 (s, 3H), 1.65 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=351 (M+H). HPLC 1.77 min.
  • Example 78 4-Amino-8-(2,4-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide
  • Synthetic Scheme for Making Compound 78:
  • Figure US20090018112A1-20090115-C00247
  • A 2 L, 3-necked flask equipped with a mechanical stirrer was charged with 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (40.5 g, 108.2 mmol), DME (700 mL, anhydrous), and ethanol (200 mL, absolute). A nitrogen dispersion tube was fitted into the suspension and the mixture was stirred until a solution was obtained. Water (300 mL) and PdCl2(PPh3)2 (7.6 g, 10 mol %) were added. After 5 minutes, 2,4-dimethoxyphenyl boronic acid (39.4 g, 216.5 mmol) was added followed by cesium carbonate (70.3 g, 216.5 mmol). Nitrogen was bubbled through the suspension for 5 minutes. The mixture was heated to approximately 80° C. Additional 7:3:2 DME:H2O:EtOH (340 mL) was added as the reflux started. The reaction was refluxed 18 hours and then cooled to room temperature, diluted with ethyl acetate (1.5 L), and washed with water (3×500 mL). The aqueous layers were extracted with ethyl acetate (3×150 mL). The combined organic layers were stirred for 1 hour with 40 g of DARCO, dried over sodium sulfate, and filtered through Celite. The solids were washed with 5% methanol in chloroform (3×200 mL) and the filtrates concentrated to a dark semisolid. This was taken up in 200 mL 1% methanol in chloroform and warmed to solubilize the material. The solution was divided into two portions. Each portion was filtered through Whatman fluted filter paper onto a 330 g silica gel column and eluted with 5% ethyl acetate in dichloromethane. (Note: Some solid catalyst appeared to be removed via the filter paper.) The purest fractions from each column were combined in 5-10% ethyl acetate in dichloromethane. The solution was concentrated to approximately 200 mL, diluted with hexane (200 mL), and let stand at room temperature overnight. The resulting solids were isolated by filtration, washed with ether (3 times), and dried under vacuum at room temperature to afford the desired product (26.4 g, 63%). 1H NMR (500.333 MHz, CDCl3) δ 8.51 (bs, 1H), 7.86 (dd, J=9.4, 5.2 Hz, 1H), 7.50 (t, J=8.8 Hz, 1H), 7.27 (d, J=9.2, 1H), 6.66 (dd, J=8.2, 2.3 Hz, 1H), 6.63 (d, J=2.3 Hz, 1H), 3.87 (s, 3H), 3.71 (s, 3H), 3.44 (q, J=6.7 Hz, 2H), 1.64 (sextet, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H). MS APCI, m/z=385 (M+H). HPLC: 2.61 min.
  • 4-Amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide
  • To a 1 L, 3-necked flask equipped with a mechanical stirrer charged with (2E)-2-cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-propylacetamide (43.9 g, 117 mmol) in anhydrous toluene (Aldrich, 600 mL) under N2 was added portion-wise aluminum chloride (Aldrich, 46.8 g, 352 mmol) over 20 minutes. The mixture was heated to 60° C. with vigorous stirring for 2 hours then cooled to 15° C. Ethyl acetate (30 mL) was carefully added while maintaining the internal temperature between 20-25° C. Additional ethyl acetate (900 mL) was then added, followed by careful addition of Rochelle's salt (saturated aqueous potassium sodium tartrate, 500 mL). Upon addition of the first 50 mL, the temperature rose from 20 to 36° C. The reaction was heated with stirring at 60° C. for 30 minutes. The aqueous layer contained a thick white precipitate and the organic layer slowly solubilized the brownish yellow solid. (Note: If a non-white (brown/yellow) solid still existed at the aqueous/organic interface, the hot extraction was repeated). The mixture was placed in a separatory funnel and the aqueous layer was removed. The organic layer was washed with Rochelle's salt (500 mL), washed with brine, dried over magnesium sulfate, filtered and concentrated to give 38 g slightly crude product (86.5%). Further purification by trituration with ethyl acetate/hexanes was carried out when appropriate. An analytically pure sample was obtained by recrystallization from ethyl acetate. 1H NMR (300 MHz, CDCl3) δ 8.54 (br, 1H), 7.84 (dd, J=5.3, 9.2 Hz, 1H), 7.39 (dd, J=7.0, 9.2 Hz, 1H), 3.47 (apparent q, J=7.0 Hz, 2H), 1.68 (apparent sextet, J=7.0 Hz, 2H), 1.03 (t, J=7.4 Hz, 3H). MS APCI, m/z=375 (M+H). HPLC 2.13 min.
  • (2E)-2-Cyano-2-[(3-fluoro-2-iodophenyl)hydrazonol]-N-propylacetamide
  • Using the procedure outlined in the U.S. Pat. No. 4,886,800 example 89b substituting 3-fluoro-2-iodoaniline hydrochloride (8.8 g, 32.5 mmol) for 2-iodoaniline, the title compound (2E)-2-cyano-2-[(3-fluoro-2-iodophenyl)hydrazono]-N-propylacetamide was obtained as a light brown solid (8.5 g, 70% yield). An analytically pure sample was obtained by recrystallization from ethyl acetate as a yellow crystalline solid.
  • 1H NMR (300 MHz, CDCl3) δ 14.39, (s, 1H), 8.67 (bm, 1H), 7.45 (m, 1H), 7.32 (m, 1H), 7.03 (m, 1H), 3.1 (apparent q, J=6.6 Hz, 2H), 1.53 (apparent sextet, J=7.4 Hz, 2H), 0.88 (t, J=7.4 Hz, 3H).
  • 3-Fluoro-2-iodoaniline hydrochloride
  • To a 1 L, 3 necked round bottom flask fitted with a mechanical stirrer was added 3-fluoro-2-iodonitrobenzene (3B Medical, 47.7 g, 179 mmol) and 500 mL absolute ethanol. To this stirred solution was added iron powder (325 mesh, Aldrich, 30 g, 537 mmol) followed by dropwise addition of concentrated HCl (30 mL, 360 mmol). The internal temperature rose from 23 to −60° C. over the addition. The flask was fitted with a heating mantle and heated with vigorous stirring for 90 minutes. After cooling to room temperature, 1 N Na2CO3 (300 mL) was added followed by EtOAc (200 mL). The mixture was stirred for 30 minutes and then filtered through a pad of Celite. The Celite was washed with EtOAc (3×150 mL). The filtrates were placed in a separatory funnel and the water layer was removed. The organic layer was concentrated under reduced pressure to reduce volume to ˜200 mL, placed in a separatory funnel, diluted with EtOAc (400 mL), washed organic with brine, dried over sodium sulfate, filtered and concentrated to dryness. The crude product was taken up in ether (300 mL) and made acidic to pH 1 with 2M HCl/ether (Aldrich). After 1 hour, the tan solid was isolated by filtration (39.2 g, 80%). The above aqueous layers were extracted with diethyl ether (300 mL), dried over sodium sulfate, combined with the filtrate of the 1st crop, made acidic to pH 1, and isolated as above to give additional tan solid (9.0 g, 18%) for an overall yield of 98%. 1H NMR (300 MHz, CDCl3) δ 7.06 (m, 1H), 6.58 (m, 1H), 6.39 (m, 1H), 5.73 (bm, 1H). MS APCI, m/z=238 (M+H). HPLC 2.19 min.
  • It is found that the title compound may form isolable atropisomers in certain organic solvents (e.g. 25-35% methanol) at room temperature. The two atropisomers of the title compound may be isolated using chiral LC. However, these isomers will racimize rapidly under neutral or acidic aqueous solutions.
  • Example 79 4-amino-8-(2,5-dimethoxyphenyl)-7-fluoro-N-propylcinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (200 mg, 0.535 mmol) and 2,5-dimethoxyphenyl boronic acid (194 mg, 1.07 mmol) according to Method A. The off-white solid from chromatography was slurried in ether, filtered and dried under vacuum at room temperature to afford the desired product (147 mg, 71%). 1H NMR (300.132 MHz, CDCl3) δ 8.50 (t, J=4.7 Hz, 1H), 7.89 (dd, J=9.3, 5.1 Hz, 1H), 7.52 (t, J=8.8 Hz, 1H), 6.98 (m, 2H), 6.91 (dd, J=2.4, 0.9 Hz, 1H), 3.79 (s, 3H), 3.67 (s, 3H), 3.44 (q, J=6.7 Hz, 2H), 1.64 (sextet, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H). MS APCI, m/z=385 (M+H). HPLC 2.62 min.
  • Example 80 4-Amino-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-N-propylcinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (220 mg, 58.8 mmol) and 2,4-dimethoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine (312 mg, 1.62 mmol) according to Method B to afford a white solid (123 mg, 54%). 1H NMR (300.132 MHz, CDCl3) δ 8.47 (t, J=5.4 Hz, 1H), 8.32 (s, 1H), 7.93 (dd, J=9.1, 5.2 Hz, 1H), 7.53 (dd, J=9.2, 8.4 Hz, 1H), 4.07 (s, 3H), 3.94 (s, 3H), 3.45 (q, J=6.7 Hz, 2H), 1.66 (sextet, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=387 (M+H). HPLC 1.87 min.
  • Example 81 4-Amino-N-ethyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (70.0 mg, 0.237 mmol) and 4-methoxy-3-pyridine boronic acid (153.0 mg, 0.3439 mmol) according to Method A except that the extraction was carried out with methylene chloride rather than ethyl acetate and the flash column was eluted with a gradient of 10 to 60% methanol in dichloromethane. The concentrated product was then recrystallized from chloroform (with a few drops of methanol) and hexanes to afford the title compound as a yellow solid (19.6 mg, 26% yield). 1H NMR (300.132 MHz, CDCl3) δ 8.58 (d, J=5.7 Hz, 1H), 8.52-8.43 (bm, 1H), 8.46 (s, 1H), 7.92 (app quintet, J=4.0 Hz, 1H), 7.74 (dd, J=4.9, 0.9 Hz, 2H), 6.96 (d, J=5.8 Hz, 1H), 3.78 (s, 3H), 3.53 (dq, J=5.8, 7.3, Hz, 2H), 1.27 (t, J=7.3 Hz, 3H). MS APCI, m/z=324 (M+H). HPLC 1.42 min.
  • Example 82 4-Amino-N-butyl-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-butyl-cinnoline-3-carboxamide (100 mg, 0.31 mmol) and 2,5-dimethoxyphenyl boronic acid (112.6 mg, 0.62 mmol) according to Method A to afford a white solid (96.3 mg, 82%). 1H NMR 300.132 MHz, CDCl3) δ 8.54 (t, J=4.7 Hz, 1H), 7.86 (dd, J=7.6, 2.2 Hz, 1H), 6.96 (t, J=3.0 Hz, 1H), 6.96 (s, 1H), 6.93 (d, J=1.9 Hz, 1H), 7.76-7.68 (m, 2H), 3.79 (s, 3H), 3.64 (s, 3H), 3.48 (q, J=6.6 Hz, 2H), 1.61 (quintet, J=7.2 Hz, 2H), 1.43 (sextet, J=7.3 Hz, 2H), 0.94 (t, J=7.3 Hz, 3H). MS APCI, m/z=381.2 (M+H). HPLC 2.83 min.
  • Example 83 4-Amino-8-(2,5-dimethoxyphenyl)-N-ethylcinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (100.0 mg, 0.339 mmol) and 2,5-dimethoxyphenyl boronic acid (123.3 mg, 0.678 mmol) according to Method A. The solid obtained after chromatography was washed with diethyl ether and dried overnight at 40° C. to afford the title compound as a fluffy white solid (64.4 mg, 54% yield). 1H NMR (300.132 MHz, CDCl3) δ 8.51 (bt, J=5.2 Hz, 1H), 7.86 (dd, J=7.5, 2.1 Hz, 1H), 7.76-7.68 (m, 2H), 6.97-6.91 (m, 3H), 3.79 (s, 3H), 3.64 (s, 3H), 3.52 (dq, J=5.7, 7.3 Hz, 2H), 1.26 (t, J=7.3 Hz, 3H). MS APCI, m/z=353 (M+H). HPLC 2.41 min.
  • Example 84 4-Amino-8-(2,5-dimethoxyphenyl)-N-methylcinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-methyl-cinnoline-3-carboxamide (20.0 g, 63.1 mmol) and 2,5-dimethoxyphenyl boronic acid (22.3 g, 122.4 mmol) according to Method B except that potassium carbonate was used as the base and tetrahydrofuran:ethanol:water (1:1:1) was used as the solvent system. The reaction mixture was filtered and the yellow solids were slurried in 10% methanol in chloroform and filtered. The combined filtrates were concentrated to a solid, slurried in hot ethyl acetate, and filtered. The combined solids were further purified on silica gel using 5% methanol in chloroform as the eluent. A final crystallization from refluxing ethyl acetate followed by drying under high vacuum at 45° C. afforded the title compound as a light yellow solid (12.65 g, 59%). 1H NMR (500.333 MHz, DMSO) δ 9.07 (d, J=4.6 Hz, 1H), 8.39 (d, J=8.2 Hz, 1H), 7.76-7.70 (m, 2H), 7.03 (d, J=9.1 Hz, 1H), 6.97 (dd, J=8.9, 3.0 Hz, 1H), 6.87 (d, J=3.2 Hz, 1H), 3.73 (s, 3H), 3.55 (s, 3H), 2.86 (d, J=4.8 Hz, 3H). MS APCI, m/z=xx (M+H). HPLC 2.23 min. MP=279.1-279.8.
  • Example 85 4-Amino-N-butyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-butyl-cinnoline-3-carboxamide (200.0 mg, 0.62 mmol) and 2,4-dimethoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine (987.9 mg, 3.72 mmol) according to Method B to afford a white solid (162.1 mg, 69%). 1H NMR (300.132 MHz, CDCl3) δ 8.49 (t, J=5.5 Hz, 1H), 8.33 (s, 1H), 7.90 (dd, J=7.6, 2.1 Hz, 1H), 7.77-7.69 (m, 3H), 4.07 (s, 3H), 3.93 (s, 3H), 3.50 (q, J=6.6 Hz, 2H), 1.63 (quintet, J=7.2 Hz, 2H), 1.44 (sextet, J=7.4 Hz, 2H), 0.95 (t, J=7.3 Hz, 3H). MS APCI, m/z=383.1 (M+H). HPLC 2.52 min.
  • Example 86 4-Amino-8-(2,4-dimethoxypyrimidin-5-yl)-N-ethylcinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide (200.0 mg, 0.678 mmol) and 2,4-dimethoxyprimidine-5-boronic acid pinacol ester (363.1 mg, 1.362 mmol) according to Method B except that the reaction was heated at 90° C. to fully dissolve the starting materials. After 4 hours, additional 2,4-dimethoxyprimidine-5-boronic acid pinacol ester (363.1 mg, 1.362 mmol) was added and the reaction was refluxed overnight. A third addition of 2,4-dimethoxyprimidine-5-boronic acid pinacol ester (363.1 mg, 1.362 mmol) and an additional 5 mol % tetrakis(triphenylphospine) palladium (0) were required to force the reaction to completion. The reaction was then worked up as described in Method B using dichloromethane rather than ethyl acetate for the extraction and crystallizing the material obtained from the flash column from chloroform/hexanes to afford the title compound as a white solid (89.5 mg, 37%). 1H NMR (300.132 MHz, CDCl3) δ 8.47 (s, 1H), 8.32 (s, 1H), 7.90 (dd, J=7.6, 2.1 Hz, 1H), 7.78-7.70 (m, 2H), 4.07 (s, 3H), 3.93 (s, 3H), 3.53 (dq, J=5.7, 7.3 Hz, 2H), 1.28 (t, J=7.3 Hz, 3H). MS APCI, m/z=354 (M+H). HPLC 2.07 min.
  • Example 87 4-Amino-8-(2,5-dimethoxyphenyl)-cinnoline-3-carboxylic acid allylamide
  • The title compound was prepared from 4-amino-8-bromo-cinnoline-3-carboxylic acid allylamide (273 mg, 0.89 mmol) and 2,5-dimethoxyphenyl boronic acid (201.1 mg, 1.11 mmol) according to Method A to afford an off-white solid (105 mg, 32%). 1H NMR (300.132 MHz, CDCl3) δ 8.64 (bs, 1H), 7.87 (d, J=7.3 Hz, 1H), 7.77-7.69 (m, 2H), 6.96 (t, J=2.8 Hz, 1H), 6.96 (s, 1H), 6.93 (d, J=1.8 Hz, 1H), 6.00-5.88 (m, 1H), 5.29 (dq, J=17.2, 1.6 Hz, 1H), 5.17 (dq, J=10.2, 1.4 Hz, 1H), 4.12 (tt, J=5.7, 1.6 Hz, 2H), 3.79 (s, 3H), 3.64 (s, 3H). MS APCI, m/z=365 (M+H). HPLC 1.76 min.
  • Example 88 4-Amino-N-(cyclopropylmethyl)-8-phenyl-cinnoline-3-carboxamide
  • To a suspension of 2-(biphenyl-2-yl-hydrazono)-2-cyano-N-cyclopropylmethylacetamide (1.9 g, 6.0 mmol) in anhydrous toluene (30 mL) was added aluminum chloride (1.72 g, 13.0 mmol). The mixture was heated at 70° C. for 1 hour, cooled to room temperature, and diluted with ethyl acetate (150 mL). Water was added dropwise until no further precipitate formed. Aqueous 10% sodium hydroxide (150 mL) was added and the layers were separated. The organic layer was washed with 10% sodium hydroxide (100 mL), water (100 mL), and brine (100 mL) and dried over sodium sulfate, filtered, and concentrated to a semisolid. The material was dissolved in chloroform and purified on silica gel to give 500 mg of material which was then recrystallized from ethyl acetate/hexanes and then from hot toluene (two times) to afford the pure product as a solid (117 mg, 6.2%). 1H NMR (500.133 MHz, CDCl3) δ 9.23 (t, J=5.3 Hz, 1H), 9.13 (bs, 1H), 8.43 (d, J=8.4 Hz, 1H), 8.17 (bs, 1H), 7.86 (d, J=7.1 Hz, 1H), 7.81 (td, J=7.7, 1.2 Hz, 1H), 7.71 (d, J=7.5 Hz, 2H), 7.49 (t, J=7.3 Hz, 2H), 7.43 (td, J=7.2, 0.9 Hz, 1H), 3.23 (t, J=6.3 Hz, 2H), 1.12 (t, J=5.6 Hz, 1H), 0.45 (dd, J=6.5, 1.5 Hz, 2H), 0.29 (d, J=4.4 Hz, 2H). MS APCI, m/z=319 (M+H). HPLC 1.83 min.
  • Example 89 4-Amino-8-(m-tolyl)-N-propyl-cinnoline-3-carboxamide
  • The title compound was prepared from 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (450 mg, 1.46 mmol) and 3-methylphenyl boronic acid (408 mg, 3.00 mmol) according to Method A to afford an off-white solid (321 mg, 69%). 1H NMR (300.132 MHz, CDCl3) δ 8.58 (t, J=5.0 Hz, 1H), 7.85 (dd, J=8.3, 1.4 Hz, 1H), 7.79 (dd, J=7.3, 1.4 Hz, 1H), 7.71 (dd, J=8.1, 7.3 Hz, 1H), 7.50 (s, 1H), 7.48 (s, 2H), 7.39 (t, J=7.9 Hz, 1H), 7.23 (s, 1H), 3.46 (q, J=6.7 Hz, 2H), 2.44 (s, 3H), 1.67 (sextet, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=321 (M+H). HPLC 1.90 min.
  • Example 90 4-Amino-8-(2-fluoro-6-methylpyridin-3-yl)-cinnoline-3-carboxylic acid propylamide
  • The title compound was prepared from 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (300.0 mg, 0.97 mmol) and 2-fluoro-6-methylpyridine-3-boronic acid (426.7 mg, 2.75 mmol) according to Method A to afford a white solid (124.2 mg, 38%). 1H NMR (300.132 MHz, CDCl3) δ 8.53 (bs, 1H), 7.94 (t, J=1.4 Hz, 1H), 7.92 (dt, J=8.6, 1.4 Hz, 1H), 7.89 (d, J=7.6 Hz, 1H), 7.83 (dt, J=7.1, 1.2 Hz, 1H), 7.74 (dd, J=8.8, 7.3 Hz, 1H), 7.19 (dd, J=7.4, 1.3 Hz, 1H), 3.46 (q, J=6.7 Hz, 2H), 2.59 (s, 3H), 1.66 (sextet, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=340 (M+H). HPLC 2.28 min.
  • Example 91 4-Amino-7-fluoro-8-(5-fluoro-2-methoxyphenyl)-cinnoline-3-carboxylic acid propylamide
  • The title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (200.0 mg, 0.53 mmol) and 2-fluoro-5-methoxyphenyl boronic acid (181.7 mg, 1.07 mmol) according to Method A to afford a yellow crystalline solid (111.0 mg, 56%). 1H NMR (300.132 MHz, CDCl3) δ 8.49 (t, J=5.3 Hz, 1H), 7.91 (dd, J=9.3, 5.2 Hz, 1H), 7.52 (t, J=8.8 Hz, 1H), 7.16-7.07 (m, 2H), 6.98 (dd, J=9.1, 4.5 Hz, 1H), 3.70 (s, 3H), 3.44 (q, J=6.7 Hz, 2H), 1.64 (sextet, J=7.3 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H). MS APCI, m/z=373 (M+H). HPLC 2.66 min.
  • Example 92 4-Amino-8-(2-chloro-5-methoxyphenyl)-7-fluoro-cinnoline-3-carboxylic acid propylamide
  • The title compound was prepared from 4-amino-7-fluoro-8-iodo-N-propylcinnoline-3-carboxamide (250 mg, 0.67 mmol) and 2-chloro-5-methoxyphenyl boronic acid (279 mg, 1.50 mmol) according to Method A to afford a solid (181 mg, 72%). 1H NMR (500.333 MHz, CDCl3) δ 8.41 (s, 1H), 7.90 (dd, J=9.1, 5.1 Hz, 1H), 7.49 (t, J=8.7 Hz, 1H), 7.41 (dd, J=6.9, 2.7 Hz, 1H), 6.94-6.92 (m, 2H), 3.79 (s, 3H), 3.44 (q, J=6.7 Hz, 2H), 1.65 (sextet, J=7.2 Hz, 2H), 0.99 (t, J=7.3 Hz, 3H). MS APCI, m/z=389/391 (M+H). HPLC 2.13 min.
  • Example 93 4-amino-N-cyclopropyl-8-(2,6-dimethoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2,6-dimethoxypyridine-3-boronic acid (170 mg, 0.94 mmol) were reacted. After purification the title compound (132 mg, 77% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.03 (d, J=4.9 Hz, 1H), 8.38 (d, J=8.0 Hz, 1H), 7.76 (m, 2H), 7.67 (d, J=8.0 Hz, 1H), 6.48 (d, J=8.0 Hz, 1H), 3.93 (s, 3H), 3.77 (s, 3H), 2.96 (m, 1H), 0.70 (m, 4H). MS APCI, m/z=366.
  • Example 94 4-amino-N-cyclopropyl-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2-methoxy-5-methylphenylboronic acid (155 mg, 0.94 mmol) were reacted. After purification the title compound (120 mg, 74% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.00 (d, J=4.9 Hz, 1H), 8.38 (d, J=7.1 Hz, 1H), 7.74 (t, J=7.7 Hz, 1H), 7.68 (d, J=7.1 Hz, 1H), 7.19 (d, J=8.5 Hz, 1H), 7.06 (s, 1H), 6.98 (d, J=8.5 Hz, 1H), 3.57 (s, 3H), 2.96 (m, 1H), 2.28 (s, 3H), 0.70 (m, 4H). MS APCI, m/z=349.
  • Example 95 4-amino-N-cyclopropyl-8-(2,4-dimethoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2,4-dimethoxyphenylboronic acid (171 mg, 0.94 mmol) were reacted. After purification the title compound (136 mg, 80% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.00 (d, J=4.8 Hz, 1H), 8.35 (d, J=8.4 Hz, 1H), 7.72 (t, J=7.7 Hz, 1H), 7.68 (ad, J=7.1 Hz, 1H), 7.18 (d, J=8.3 Hz, 1H), 6.66 (s, 1H), 6.59 (ad, J=8.3 Hz, 1H), 3.83 (s, 3H), 3.61 (s, 3H), 2.96 (m, 1H), 0.70 (m, 4H). MS APCI, m/z=365.
  • Example 96 4-amino-N-cyclopropyl-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2,4-dimethoxypyrimidine-5-boronic acid (171 mg, 0.94 mmol) were reacted. After purification the title compound (133 mg, 78% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.51 (bm, 1H), 8.31 (s, 1H), 7.89 (a dd, J=2.3, 7.4 Hz, 1H), 7.75 (m, 2H), 4.06 (s, 3H), 3.92 (s, 3H), 2.96 (m, 1H), 0.88 (m, 2H), 0.65 (m, 2H).
  • MS APCI, m/z=367, HPLC 2.07 min
  • Example 97 4-amino-N-cyclopropyl-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (100 mg, 0.33 mmol) and 2,5-dimethoxyphenylboronic acid (118 mg, 0.65 mmol) were reacted. After purification the title compound (101 mg, 85% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (bm, 1H), 7.85 (a dd, J=2.2, 7.6 Hz, 1H), 7.73 (m, 2H), 6.95 (m, 2H), 6.91 (m, 1H), 3.77 (s, 3H), 3.63 (s, 3H), 2.96 (m, 1H), 0.88 (m, 2H), 0.65 (m, 2H). MS APCI, m/z=365, HPLC 2.42 min.
  • Example 98 4-amino-N-ethyl-8-(2-fluoro-6-methoxy-phenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-ethyl-cinnoline-3-carboxamide and 2-fluoro-6-methoxy-phenyl boronic acid were reacted to afford the title compound as a off-white solid. 1H NMR (500 MHz, CDCl3) δ 8.50 (br, 1H), 7.90 (m, 1H), 7.74 (m, 2H), 7.36 (m, 1H), 6.84 (m, 2H), 3.70 (s, 3H), 3.52 (m, 2H), 1.25 (t, J=7.0 Hz, 3H). MS APCI, m/z=341 (M+H).
  • Example 99 4-amino-7-fluoro-8-(2-fluoro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using Method G, 4-amino-8-bromo-7-fluoro-N-propyl-cinnoline-3-carboxamide and 2-fluoro-6-methoxy-phenyl boronic acid were reacted to afford the title compound as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.16 (br, 1H), 9.06 (m, 1H), 8.58 (m, 1H), 8.32 (br, 1H), 7.76 (m, 1H), 7.50 (m, 1H), 7.02 (m, 1H), 6.94 (m, 1H), 3.68 (s, 3H), 3.31-3.25 (m, 2H), 1.57 (m, 2H), 0.89 (t, J=7.0 Hz, 3H). MS APCI, m/z=373 (M+H).
  • Example 100 4-amino-7-cyano-8-(2,4-dimethoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using Method G, 4-amino-8-bromo-7-cyano-N-propyl-cinnoline-3-carboxamide and 2,4-dimethoxyphenyl boronic acid were reacted to afford the title compound as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.51 (br, 1H), 7.87 (m, 2H), 7.27 (m, 1H), 6.66 (m, 2H), 3.88 (s, 3H), 3.74 (s, 3H), 3.45 (m, 2H), 1.64 (apparent sextet, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=392 (M+H).
  • Example 101 4-amino-N-cyclobutyl-8-(2-fluoro-6-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclobutyl-cinnoline-3-carboxamide (145 mg) and 2-fluoro-6-methoxy-phenyl boronic acid (191 mg) were reacted to afford the title compound (32 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.20 (d, 1H), 8.44 (m, 1H), 7.76 (m, 2H), 7.44 (m, 1H), 6.99 (d, 1H), 6.90 (t, 1H), 4.49 (m, 1H), 3.65 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=367 (M+H).
  • Example 102 4-amino-N-cyclopropyl-8-(2-fluoro-6-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method H, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (120 mg, 0.39 mmol) and 2-fluoro-6-methoxyphenylboronic acid (250 mg, 1.47 mmol) were reacted (refluxed 2 hours). After purification the title compound (82 mg, 60% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (bm, 1H), 7.93 (m, 1H), 7.75 (m, 1H), 7.36 (apparent q, J=7.0 Hz, 1H), 6.84 (m, 2H), 3.69 (s, 3H), 2.96 (m, 1H), 0.85 (m, 2H), 0.63 (m, 2H). MS APCI, m/z=353 (M+H) HPLC 1.74 min.
  • Example 103 4-amino-8-(2-chloro-6-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using Method G, 4-amino-8-bromo-N-propyl-cinnoline-3-carboxamide (600 mg) and 2-chloro-6-methoxy-phenylboronic acid (1051 mg) were reacted to afford the title compound (263 mg) as a off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.55 (br, 1H), 7.94-7.88 (m, 1H), 7.79-7.65 (m, 2H), 7.37-7.29 (m, 1H), 7.15 (m, 1H), 6.94 (m, 1H), 3.65 (s, 3H), 3.44 (apparent quartet, J=7.0 Hz, 2H), 1.64 (apparent sextet, J=7.0 Hz, 2H), 0.99 (t, J=7.0 Hz, 3H). MS APCI, m/z=371 (M+H) HPLC 1.86 min.
  • Example 104 4-amino-7-fluoro-8-(2-fluoro-3-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (250 mg, 0.67 mmol) and (2-fluoro-3-methoxyphenyl)boronic acid (193.4 mg, 1.136 mmol) were reacted to afford the title compound (72.0 mg, 29% yield) as an off-white solid. 1H NMR (500 MHz, CDCl3) δ 8.49 (bs, 1H), 7.94 (d×d, J=4.9 Hz, J=9.2 Hz, 1H), 7.55 (t, J=8.5 Hz, 1H), 7.21 (m, 1H), 7.03-7.10 (m, 2H), 3.94 (s, 3H), 3.44 (q, J=6.7 Hz, J=13.4 Hz, 2H), 1.64 (apparent sextet, J=7.3 Hz, 2H), 0.98 (t, J=7.3 Hz, 3H). MS APCI, m/z=373 (M+H) HPLC 2.72 min.
  • Example 107 4-amino-7-fluoro-8-(4-fluoro-2-methoxy-phenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (250 mg, 0.67 mmol) and (2-methoxy-4-fluorophenyl)boronic acid (227.1 mg, 1.336 mmol) were reacted to afford the title compound (131.7 mg, 53% yield) as an off-white solid. 1H NMR (500 MHz, CDCl3) δ 8.49 (bs, 1H), 7.89 (d×d, J=5.5 Hz, J=9.2 Hz, 1H), 7.52 (apparent triplet, J=8.9 Hz, 1H), 7.30 (m, 1H), 6.82 (d×d, J=2.4 Hz, J=8.5 Hz, 1H), 6.79 (d×d, J=2.4 Hz, J=12.8 Hz, 1H), 3.72 (s, 3H), 3.44 (q, J=6.7 Hz, J=13.4 Hz, 2H), 1.64 (apparent sextet, J=7.3 Hz, 2H), 0.99 (t, J=7.6 Hz, 3H). MS APCI, m/z=373 (M+H) HPLC 2.70 min.
  • Example 108 4-amino-7-fluoro-8-(2-fluoro-4-methoxyphenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (250.0 mg, 0.67 mmol) and (2-fluoro-4-methoxyphenyl)boronic acid (227.1 mg, 1.336 mmol) were reacted to afford the title compound (165.4 mg, 67% yield) as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.51 (bs, 1H), 7.90 (d×d, J=5.5 Hz, J=9.2 Hz, 1H), 7.53 (apparent triplet, J=8.9 Hz, 1H), 7.41 (t, J=8.5 Hz, 1H), 6.86 (d×d, J=2.4 Hz, J=8.5 Hz, 1H), 6.80 (d×d, J=2.4 Hz, J=11.6 Hz, 1H), 3.87 (s, 3H), 3.44 (q, J=6.7 Hz, J=12.8 Hz, 2H), 1.65 (apparent sextet, J=7.3 Hz, 2H), 1.00 (t, J=7.3 Hz, 3H). MS APCI, m/z=373 (M+H) HPLC 2.78 min.
  • Example 110 4-amino-7-fluoro-8-(2-methyl-5-fluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method A, 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (300 mg, 0.80 mmol) and (2-methyl-5-fluorophenyl)boronic acid (246.8 mg, 1.60 mmol) were reacted to afford the title compound (164.7 mg, 58% yield) as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.48 (bs, 1H), 7.93 (d×d, J=5.3 Hz, J=9.4 Hz, 1H), 7.54 (apparent t, J=8.6 Hz, 1H), 7.30 (d×d, J=5.6 Hz, J=8.3 Hz, 1H), 6.99-7.09 (m, 2H), 3.44 (apparent q, J=7.0 Hz, 2H), 2.03 (s, 3H), 1.64 (apparent sextet, J=7.4 Hz, 2H), 0.99 (t, J=7.4 Hz, 3H). MS APCI, m/z=357 (M+H) HPLC 2.78 min.
  • Example 113 4-amino-7-fluoro-8-(2,5-difluorophenyl)-N-propyl-cinnoline-3-carboxamide
  • Using method B, 4-amino-7-fluoro-8-iodo-N-propyl-cinnoline-3-carboxamide (150 mg, 0.40 mmol) and (2,5-difluorophenyl)boronic acid (519.0 mg, 3.29 mmol) were reacted to afford the title compound (50.5 mg, 35% yield) as an off-white solid. 1H NMR (300 MHz, CDCl3) δ 8.48 (bs, 1H), 7.96 (d×d, J=5.3 Hz, J=9.3 Hz, 1H), 7.55 (t, J=9.1 Hz, 1H), 7.13-7.23 (m, 3H), 3.45 (apparent q, J=6.9 Hz, J=13.1 Hz, 2H), 1.65 (apparent sextet, J=7.3 Hz, 2H), 1.00 (t, J=7.4 Hz, 3H). MS APCI, m/z=361 (M+H) HPLC 2.98 min.
  • Example 124 4-amino-N-cyclobutyl-7-fluoro-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-7-fluoro-N-cyclobutyl-cinnoline-3-carboxamide (175 mg) and 2-methoxy-5-methyl-phenyl boronic acid (187 mg) were reacted to afford the title compound (128 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.18 (d, 1H), 8.50 (m, 1H), 7.71 (m, 1H), 7.24 (m, 1H), 7.07 (m, 1H), 7.03 (m, 1H), 4.49 (m, 1H), 3.61 (s, 3H), 2.29 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=381 (M+H).
  • Example 125 4-amino-N-cyclobutyl-7-fluoro-8-(5-fluoro-2-methoxy-phenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-7-fluoro-N-cyclobutyl-cinnoline-3-carboxamide (175 mg) and 5-fluoro-2-methoxy-phenyl boronic acid (191 mg) were reacted to afford the title compound (141 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.21 (d, 1H), 8.53 (m, 1H), 7.74 (m, 1H), 7.28 (m, 1H), 7.17 (m, 2H), 4.49 (m, 1H), 3.64 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=385 (M+H).
  • Example 128 4-amino-N-cyclobutyl-8-(2,4-dimethoxyphenyl)-7-fluoro-cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-7-fluoro-N-cyclobutyl-cinnoline-3-carboxamide (175 mg) and 2,4-dimethoxy-phenyl boronic acid (205 mg) were reacted to afford the title compound (133 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.20 (d, 1H), 8.47 (m, 1H), 7.70 (m, 1H), 7.18 (m, 1H), 6.70 (m, 1H), 6.64 (m, 1H), 4.49 (m, 1H), 3.85 (s, 3H), 3.65 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=397 (M+H).
  • Example 130 4-amino-N-cyclobutyl-8-(2,4-dimethoxypyrimidin-5-yl)-7-fluoro-cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-7-fluoro-N-cyclobutyl-cinnoline-3-carboxamide (175 mg) and 2,4-dimethoxypyrimidin-5-yl boronic acid (207 mg) were reacted to afford the title compound (88 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.24 (d, 1H), 8.57 (m, 1H), 8.38 (s, 1H), 7.77 (m, 1H), 4.49 (m, 1H), 4.00 (s, 3H), 3.84 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=399 (M+H).
  • Example 131 4-amino-N-cyclobutyl-8-(2,6-dimethoxypyridin-3-yl)-7-fluoro-cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-7-fluoro-N-cyclobutyl-cinnoline-3-carboxamide (175 mg) and 2,6-dimethoxypyridin-3-yl boronic acid (206 mg) were reacted to afford the title compound (122 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.22 (d, 1H), 8.50 (m, 1H), 7.73 (t, 1H), 7.67 (d, 1H), 6.53 (d, 1H), 4.49 (m, 1H), 3.95 (s, 3H), 3.80 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=398 (M+H).
  • Example 139 4-amino-N-cyclobutyl-8-(2-methoxy-5-methyl-phenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclobutyl-cinnoline-3-carboxamide (145 mg) and 2-methoxy-5-methyl-phenyl boronic acid (186 mg) were reacted to afford the title compound (94 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.20 (d, 1H), 8.38 (d, 1H), 7.76-7.66 (m, 2H), 7.20 (m, 1H), 7.07 (m, 1H), 7.00 (m, 1H), 4.50 (m, 1H), 3.58 (s, 3H), 2.29 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=363 (M+H).
  • Example 142 4-amino-N-cyclobutyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclobutyl-cinnoline-3-carboxamide (145 mg) and 4-methoxypyridin-3-yl boronic acid (172 mg) were reacted to afford the title compound (31 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.24 (d, 1H), 8.52 (m, 1H), 8.44 (m, 1H), 8.33 (s, 1H), 7.78 (m, 2H), 7.18 (d, 1H), 4.49 (m, 1H), 3.72 (s, 3H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=350 (M+H).
  • Example 147 4-amino-N-cyclobutyl-8-(3,5-dimethylphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclobutyl-cinnoline-3-carboxamide (145 mg) and 3,5-dimethylphenyl boronic acid (169 mg) were reacted to afford the title compound (59 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.31 (d, 1H), 8.38 (d, 1H), 7.78 (m, 2H), 7.28 (s, 2H), 7.06 (s, 1H), 4.52 (m, 1H), 2.35 (s, 6H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=347 (M+H).
  • Example 154 4-amino-8-(4-chlorophenyl)-N-cyclobutyl-cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclobutyl-cinnoline-3-carboxamide (145 mg) and 4-chlorophenyl boronic acid (176 mg) were reacted to afford the title compound (112 mg) as white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.31 (d, 1H), 8.43 (d, 1H), 7.86 (m, 1H), 7.79 (m, 1H), 7.73 (m, 2H), 7.54 (m, 2H), 4.52 (m, 1H), 2.26-2.10 (m, 4H), 1.72-1.62 (m, 2H). MS APCI, m/z=353 (M+H).
  • Example 156 4-amino-N-cyclopropyl-8-(2-fluoro-6-methylpyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (61 mg, 0.20 mmol) and 2-fluoro-6-methylpyridine-3-boronic acid (62 mg, 0.36 mmol) were reacted. After purification the title compound (41 mg, 60% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.52 (bm, 1H), 7.92 (a t, J=7.5 Hz, 2H), 7.85 (m, 1H), 7.75 (at, J=7.7 Hz, 1H), 7.18 (a d, J=7.5 Hz, 1H), 2.96 (m, 1H), 2.58 (s, 3H), 0.88 (m, 2H), 0.65 (m, 2H). MS APCI, m/z=338, HPLC 2.17 min.
  • Example 157 4-amino-N-cyclopropyl-7-fluoro-8-(5-fluoro-6-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (175 mg, 0.48 mmol) and 5-fluoro-6-methoxypyridine-3-boronic acid (162 mg, 0.96 mmol) were reacted. After purification the title compound (106 mg, 61% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.48 (bm, 1H), 8.15 (m, 1H), 7.92 (a dd, J=5.2, 9.0 Hz, 1H), 7.65 (a d, J=10.7 Hz, 1H), 7.56 (a t, J=9.0 Hz, 1H), 4.10 (s, 3H), 2.96 (m, 1H), 0.88 (m, 2H), 0.65 (m, 2H). MS APCI, m/z=372, HPLC 2.59 min.
  • Example 158 4-amino-N-cyclopropyl-7-fluoro-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (188 mg, 0.50 mmol) and 2-methoxypyridine-3-boronic acid (155 mg, 1.01 mmol) were reacted. After purification the title compound (110 mg, 62% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.48 (bm, 1H), 8.29 (ad, J=5.0 Hz, 1H), 7.91 (a dd, J=5.2, 9.2 Hz, 1H), 7.67 (a d, J=7.3 Hz, 1H), 7.56 (a t, J=8.9 Hz, 1H), 7.05 (a dd, J=5.0, 7.3 Hz, 1H), 3.88 (s, 3H), 2.96 (m, 1H), 0.88 (m, 2H), 0.65 (m, 2H). MS APCI, m/z=354, HPLC 2.23 min.
  • Example 159 4-amino-N-cyclopropyl-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 4-methylpyridine-3-boronic acid (128 mg, 0.94 mmol) were reacted. After purification the title compound (96 mg, 64% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.53 (d, J=5.0 Hz, 1H), 8.50 (bm, 1H), 8.47 (s, 1H), 7.95 (a d, J=8.3 Hz, 1H), 7.77 (t, J=7.7 Hz, 1H), 7.69 (apparent d, J=7.0 Hz, 1H), 2.96 (m, 1H), 2.10 (s, 3H), 0.88 (m, 2H), 0.62 (m, 2H). MS APCI, m/z=320, HPLC 1.55 min.
  • Example 160 4-amino-N-cyclopropyl-7-fluoro-8-(4-methylpyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 4-methylpyridine-3-boronic acid (150 mg, 0.96 mmol) were reacted. After purification the title compound (100 mg, 62% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.56 (d, J=5.1 Hz, 1H), 8.47 (s, 1H), 8.46 (bm, 1H), 7.99 (a dd, J=5.2, 9.2 Hz, 1H), 7.58 (t, J=8.7 Hz, 1H), 7.30 (d, J=5.0 Hz, 1H), 2.96 (m, 1H), 2.11 (s, 3H), 0.88 (m, 2H), 0.65 (m, 2H). MS APCI, m/z=338, HPLC 1.68 min.
  • Example 161 4-amino-N-cyclopropyl-7-fluoro-8-(2,6-dimethoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2,6-dimethoxypyridine-3-boronic acid (176 mg, 0.96 mmol) were reacted. After purification the title compound (124 mg, 67% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.02 (d, J=4.8 Hz, 1H), 8.51 (m, 1H), 7.73 (t, J=9.1 Hz, 1H), 7.66 (d, J=8.0 Hz, 1H), 6.51 (d, J=8.0 Hz, 1H), 3.94 (s, 3H), 3.78 (s, 3H), 2.93 (m, 1H), 0.70 (m, 4H). MS APCI, m/z=384.
  • Example 162 4-amino-N-cyclopropyl-7-fluoro-8-(6-dimethylpyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 6-methylpyridine-3-boronic acid (150 mg, 0.96 mmol) were reacted. After purification the title compound (18 mg, 11% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.05 (d, J=4.8 Hz, 1H), 8.58 (s, 1H), 8.56 (dd, J=5.6, 9.3 Hz, 1H), 7.86 (ad, J=8.1 Hz, 1H), 7.80 (t, J=9.3 Hz, 1H), 7.39 (d, J=8.0 Hz, 1H), 2.96 (m, 1H), 2.56 (s, 3H), 0.70 (m, 4H). MS APCI, m/z=338.
  • Example 163 4-amino-N-cyclopropyl-7-fluoro-8-(2,4-dimethoxypyrimidin-5-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2,4-dimethoxypyrimidin-5-boronic acid (176 mg, 0.96 mmol) were reacted. After purification the title compound (73 mg, 39% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.04 (d, J=4.9 Hz, 1H), 8.57 (dd, J=5.6, 9.3 Hz, 1H), 8.37 (s, 1H) 7.77 (t, J=9.1 Hz, 1H), 3.99 (s, 3H), 3.84 (s, 3H), 2.93 (m, 1H), 0.70 (m, 4H). MS APCI, m/z=385.
  • Example 164 4-amino-N-cyclopropyl-7-fluoro-8-(2,5-dimethoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2,5-dimethoxyphenylboronic acid (174 mg, 0.96 mmol) were reacted. After purification the title compound (142 mg, 77% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.99 (d, J=4.8 Hz, 1H), 8.51 (a dd, J=5.5, 9.3 Hz, 1H), 7.72 (t, J=9.0 Hz, 1H), 7.06 (a d, J=9.0 Hz, 1H), 7.00 (m, 1H), 6.86 (a d, J=3.0 Hz, 1H), 3.72 (s, 3H), 3.58 (s, 3H), 2.93 (m, 1H), 0.68 (m, 4H). MS APCI, m/z=383.
  • Example 165 4-amino-N-cyclopropyl-7-fluoro-8-(5-fluoro-2-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2,5-dimethoxyphenylboronic acid (162 mg, 0.96 mmol) were reacted. After purification the title compound (150 mg, 84% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.02 (d, J=4.8 Hz, 1H), 8.54 (a dd, J=5.5, 9.3 Hz, 1H), 7.74 (t, J=9.1 Hz, 1H), 7.27 (at, J=8.7 Hz, 1H), 7.15 (m, 2H), 3.64 (s, 3H), 2.95 (m, 1H), 0.67 (m, 4H). MS APCI, m/z=371.
  • Example 166 4-amino-N-cyclopropyl-7-fluoro-8-(2-fluoro-6-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method G, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (372 mg, 1.00 mmol) and 2-fluoro-6-methoxyphenylboronic acid (1.40 g, 8.24 mmol) were reacted. After purification the title compound (117 mg, 33% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.50 (bm, 1H), 7.94 (dd, J=5.2, 9.2 Hz, 1H), 7.53 (a t, J=8.7 Hz, 1H), 7.40 (apparent q, J=7.8 Hz, 1H), 6.86 (m, 2H), 3.72 (s, 3H), 2.96 (m, 1H), 0.88 (m, 2H), 0.62 (m, 2H). MS APCI, m/z=371.
  • Example 167 4-amino-N-cyclopropyl-7-fluoro-8-(2-methoxy-5-methylphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2-methoxy-5-methylphenylboronic acid (160 mg, 0.96 mmol) were reacted. After purification the title compound (134 mg, 76% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.98 (d, J=4.8 Hz, 1H), 8.50 (a dd, J=5.5, 9.3 Hz, 1H), 7.71 (t, J=9.1 Hz, 1H), 7.23 (m, 1H), 7.106 (s, 1H), 7.02 (d, J=8.4 Hz, 1H), 3.60 (s, 3H), 2.95 (m, 1H), 2.28 (s, 3H), 0.67 (m, 4H). MS APCI, m/z=367.
  • Example 168 4-amino-N-cyclopropyl-7-fluoro-8-(2,4-dimethoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2,4-dimethoxyphenylboronic acid (175 mg, 0.96 mmol) were reacted. After purification the title compound (110 mg, 60% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 8.98 (d, J=4.8 Hz, 1H), 8.47 (a dd, J=5.6, 9.3 Hz, 1H), 7.70 (t, J=9.0 Hz, 1H), 7.17 (d, J=8.0 Hz, 1H), 6.70 (s, 1H), 6.63 (a d, J=8.3 Hz, 1H), 3.84 (s, 3H), 3.64 (s, 3H), 2.95 (m, 1H), 0.67 (m, 4H). MS APCI, m/z=383.
  • Example 174 4-amino-N-cyclopropyl-8-(5-fluoro-2-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 5-fluoro-2-methoxyphenylboronic acid (158 mg, 0.94 mmol) were reacted. After purification the title compound (125 mg, 76% yield) was obtained as a white solid. 1H NMR (300 MHz, CDCl3) δ 8.54 (bm, 1H), 7.87 (a dd, J=3.1, 6.6 Hz, 1H), 7.73 (m, 2H), 7.10 (m, 2H), 6.95 (m, 1H), 3.66 (s, 3H), 2.96 (m, 1H), 0.86 (m, 2H), 0.64 (m, 2H). MS APCI, m/z=353.
  • Example 175 4-amino-N-cyclopropyl-8-(4-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 4-methoxypyridine-3-boronic acid (640 mg, 4.20 mmol) were reacted. After purification the title compound (52 mg, 33% yield) was obtained as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.58 (d, J=5.8 Hz, 1H), 8.57 (bm, 1H), 8.44 (s, 1H), 7.91 (m, 1H), 7.74 (m, 2H), 6.94 (d, J=5.8 Hz, 1H), 3.77 (s, 3H), 2.96 (m, 1H), 0.86 (m, 2H), 0.64 (m, 2H). MS APCI, m/z=336.
  • Example 176 4-amino-N-cyclopropyl-8-(2-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2-methoxypyridine-3-boronic acid (142 mg, 0.94 mmol) were reacted. After purification the title compound (118 mg, 76% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.04 (d, J=4.9 Hz, 1H), 8.43 (m, 1H), 8.24 (d, J=5.0 Hz, 1H), 7.77 (m, 2H), 7.71 (a d, J=7.2 Hz, 1H), 7.71 (a dd, J=5.1, 7.2 Hz, 1H), 3.73 (s, 3H), 2.95 (m, 1H), 0.67 (m, 4H). MS APCI, m/z=336.
  • Example 180 4-amino-N-cyclopropyl-8-(5-fluoro-6-methoxypyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 5-fluoro-6-methoxypyridine-3-boronic acid (159 mg, 0.94 mmol) were reacted. After purification the title compound (146 mg, 88% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.13 (d, J=4.9 Hz, 1H), 8.45 (d, J=7.4 Hz, 1H), 8.29 (s, 1H), 8.11 (a d, J=13.8 Hz, 1H), 7.95 (a d, J=7.2 Hz, 1H), 7.81 (a t, J=7.8 Hz, 1H), 4.03 (s, 3H), 2.95 (m, 1H), 0.67 (m, 4H). MS APCI, m/z=354.
  • Example 181 4-amino-N-cyclopropyl-8-(2-fluoro-3-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 2-fluoro-3-methoxyphenylboronic acid (158 mg, 0.94 mmol) were reacted. After purification the title compound (128 mg, 78% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.04 (d, J=4.9 Hz, 1H), 8.48 (dd, J=2.6, 7.2 Hz, 1H), 7.79 (m, 2H), 7.23 (m, 2H), 7.02 (m, 1H), 3.90 (s, 3H), 2.95 (m, 1H), 0.67 (m, 4H). MS APCI, m/z=353.
  • Example 182 4-amino-N-cyclopropyl-8-(6-methylpyridin-3-yl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-8-bromo-N-cyclopropyl-cinnoline-3-carboxamide (143 mg, 0.47 mmol) and 6-methylpyridine-3-boronic acid (128 mg, 0.94 mmol) were reacted. After purification the title compound (119 mg, 80% yield) was obtained as a white solid. MS APCI, m/z=320.
  • Example 185 4-amino-N-cyclopropyl-7-fluoro-8-(2-fluoro-3-methoxyphenyl)cinnoline-3-carboxamide
  • Using Method A, 4-amino-7-fluoro-8-iodo-N-cyclopropyl-cinnoline-3-carboxamide (178 mg, 0.48 mmol) and 2-fluoro-3-methoxyphenylboronic acid (162 mg, 0.96 mmol) were reacted. After purification the title compound (100 mg, 56% yield) was obtained as a white solid. 1H NMR (500 MHz, DMSO-d6) δ 9.04 (d, J=4.8 Hz, 1H), 8.59 (m, 1H), 7.26 (m, 2H), 7.02 (m, 1H), 3.90 (s, 3H), 2.93 (m, 1H), 0.69 (m, 4H).
  • MS APCI, m/z=371.
  • Method AA
  • Preparation of Xenopus oocytes
  • Xenopus laevis frogs (Xenopus I, Kalamazoo, Mich.) were anesthetized using 0.15% tricaine. Surgically removed ovarian lobes were teased out in OR2 solution (82 NaCl, 2.5 KCl, 5 HEPES, 1.5 NaH2PO4, 1 MgCl2, 0.1 EDTA, in mM, pH 7.4). The oocytes were defolliculated by incubation in 25 mL OR2 containing 0.2% collagenase 1A (SIGMA) two times for about 60 minutes on a platform shaker and stored in Leibovitz's L-15 medium. Oocytes were injected the following day in 0.5× Leibovitz's L-15 medium containing 50 mg/ml gentamycin, 10 units/ml penicillin, and 10 mg/ml streptomycin.
  • Method BB
  • Preparation and Injection of cRNA
  • Capped cRNAs from the linearized vectors containing human (α1, β2 and γ2 subunits of the GABAA receptor genes were mixed in ratio of 1:1:30. Oocytes were injected with 25-50 mL of mixed RNA with an appx molar ratio for (α1, β2, and γ2 as 1:1:10. Oocyte recordings were done 2-10 days after injection. The same methods apply to subtypes derived from α2β3γ2, α5β3γ2, and α5β3γ2, except for 1:1:1 ratio was used for α, β, and γ subunits.
  • Method CC Two-Electrode Voltage-Clamping Measurements
  • All measurements were done in a medium containing ND-96 (96 NaCl, 2 KCl, 1.8 CaCl2.2H2O, 1 MgCl2.6H2O, 5 HEPES, in mM, pH 7.5). Two-electrode voltage-clamp recording was carried out using OpusXpress amplifier (Axon Instruments, Foster City, Calif.), which allows simultaneous recording from 8 oocytes. Oocytes were impaled with two electrodes of 1-2 MΩ tip resistance when filled with 3M KCl. Recordings were begun when membrane potential became stable at potentials negative to −50-60 mV. Membrane potential was held at −60 mV. Typical leak currents were between 0-40 nA, and rarely if a few cells did have a relatively high leak (>100 nA) they were not used. For the determination of the GABA EC10, a series of 30 s pulses with increasing concentrations of GABA were applied to the cells every 5 minutes. After calculating EC10 for GABA for each oocyte, a series of 30 s GABA pulses were applied at 5 minutes interval, with increasing doses of the modulator. The concentration of GABA corresponded to the EC10 value calculated for each oocyte. The modulator pulses started 30 s before the GABA pulse so as to allow preincubation with the modulator. A set of 3 pulses with just GABA without modulator was given prior to the modulator-containing pulses to define the baseline GABA response. Two oocytes per each experiment were dedicated to observe the effect of diazepam on GABA response to ensure the presence of 72 subunit in the GABAA pentameric complex, which imparts diazepam sensitivity to the complex.
  • Method DD Calculation of Current Amplitude and Curve Fitting
  • Current amplitude (i) was measured from baseline to peak using Clampfit (Axon Inst., Foster City, Calif.). Potentiation was calculated as percent change from the baseline GABA current flux 100×(imod/icontrol)−1) where imod=current mediated by modulator+GABA and icontrol=current mediated by GABA alone. A value of 100% potentiation means that modulator has caused the control current to double. Similarly, a value of −50% potentiation means the presence of modulator caused a 50% decrease in the control current. Various other data shown here were fitted and plotted using GraphPad Prism (GraphPad Software, Inc. San Diego, Calif.). The percentage potentiation was converted to relative potentiation by dividing it with percentage potentiation value obtained from the same assay with diazepam as a control.
  • Method EE GABAA1 Binding Method Reagents
  • Assay and Wash Buffer: 50 mM Tris-Citrate, 200 mM NaCl, pH 7.8
  • Compounds at 10 mM in DMSO: Put 75 μl in column 1 of compound plate.
  • Flumazenil, 10 mM (for NSB)
  • Membranes (α1, β2, γ2 receptor subunits transfected into Sf9 cells and harvested; prepared by Cell Trends, stored at −80° C.) Sonicate thawed membranes for about 5-10 seconds at setting 3 on Brinkman sonicator, then dilute membranes 1:71 in assay buffer (working conc.=100 ug/ml protein). Keep on ice.
  • [3H]-Flunitrazepam (Cat #NET567): Prepare 10× stock=30 nM, [F] in assay=˜3 nM
  • Assay (See below for Automation Programs.)
  • 1. On PlateMate, prepare 1:3 serial dilutions (30 μl+60 μl) in DMSO for final assay concentrations of 10 μM to 170 pM (Automation Programs 1 and 2). Add 5 ul of 30 uM flumazenil to wells 12 D-E for 50% control wells.
  • 2. Spot 2 μl of compound dilutions into dry plate (Automation Program 3). Manually spot 2 μl 10 mM flumazenil into wells 12 F-H for nonspecific control.
  • 3. Make 1:100 dilution in assay buffer (2 μl into 200 μl) and dispense 25 μl compound into assay plates (Automation Program 4).
  • 4. Dispense 2001 membranes into assay plate (Automation Program 5).
  • 5. Add 25 μl [3H]-Flunitrazepam (Automation Program 6). Incubate for 1 hr at 4° C.
  • 6. Collect membranes on a cell harvester onto GF/B filter plates (pre-wet with dH2O and wash 5×400 μl/well, with cold assay buffer. (First 3 washes are considered hot; last two are cold.)
  • 7. Dry plates for 2-3 hours at RT.
  • 8. Add 40 μl Microscint 40/well (Automation Program 7); seal plates. Count on a TopCount.
  • Automation Programs
  • 1. PlateMate add 60 ul DMSO for dilutions 96w: 96/300 ul head, 5516 tips in columns 2-12, compound plate in left stacker A, DMSO reservoir on stage 2
  • 2. PlateMate 11pt-dilut one-third GABAA: 96/300 ul head, 5516 tips in column 1 of serial dilution magazine, compound plate in left stacker A
  • 3. PlateMate 2 ul addition of cmpd dry new wash: 96/30 ul head, 5506 tips, compound plate in left stacker A, dilution plate in right stacker A, 100% DMSO in reservoir on stage 2, must change to fresh DMSO every 4-6 plates.
  • 4. PlateMate tip chg mix and disp 25 ul to assay plate 96w: 96/300 ul head, 5516 tips, dilution plate in left stacker A, assay plates in right stacker A, auto fill assay buffer reservoir on stage 2, need to change tips after every plate.
  • 5. PlateMate add 200 ul membranes 96w: 96/300 ul head, 5516 tips, assay plates in left stacker A, membrane reservoir on stage 2.
  • 6. RapidPlate add 25 ul hot (number of plates): 100 μl (yellow box) tips in position 1, hot reservoir in position 2, plates beginning in position 3
  • 7. RapidPlate add microscint 40 ul (number of plates): 200 μl (burgundy box) tips in position 1, Microscint 40 reservoir in position 2, plates beginning in position 3.
  • Data Analysis
  • Data is analyzed by calculating percent of control, IC50, and Ki in an XLfit template. The following formula is used in the templates:
  • Ki = IC 50 1 + [ ligand ] / K D
  • Method FF GABAA2 Binding Method Reagents
  • Assay and Wash Buffer: 50 mM Tris-Citrate, 200 mM NaCl, pH 7.8
  • Compounds at 10 mM in DMSO: Put 75 μl in column 1 of compound plate.
  • Flumazenil, 10 mM (for NSB)
  • Membranes (α2, β3, γ2 receptor subunits transfected into Sf9 cells and harvested; prepared by Paragon at 12.5 mg/ml, stored at −80° C.) Sonicate thawed membranes for about 5-10 seconds at setting 3 on Brinkman sonicator, then dilute membranes 1:50 in assay buffer (working conc.=250 ug/ml protein). Keep on ice.
  • [3H]-Flunitrazepam (Cat #NET567): Prepare 10× stock=20 nM, [F} in assay=˜2 nM
  • Assay (See Below for Automation Programs.)
  • 1. On PlateMate, prepare 1:3 serial dilutions (30 μl+60 μl) in DMSO for final assay concentrations of 10 μM to 170 pM (Automation Programs 1 and 2). Add 5 ul of 30 uM flumazenil to wells 12 D-E for 50% control wells.
  • 2. Spot 2 μl of compound dilutions into dry plate (Automation Program 3). Manually spot 2 μl 10 mM flumazenil into wells 12 F-H for nonspecific control.
  • 3. Make 1:100 dilution in assay buffer (2 μl into 200 μl) and dispense 25 μl compound into assay plates (Automation Program 4).
  • 4. Dispense 2001 membranes into assay plate (Automation Program 5).
  • 5. Add 25 μl [3H]-Flunitrazepam (Automation Program 6). Incubate for 1 hr at 4° C.
  • 6. Collect membranes on a cell harvester onto GF/B filter plates (pre-wet with dH2O and wash 5×400 μl/well, with cold assay buffer. (First 3 washes are considered hot; last two are cold.)
  • 7. Dry plates for 2-3 hours at RT.
  • 8. Add 40 μl Microscint 40/well (Automation Program 7); seal plates. Count on a TopCount.
  • Automation Programs
  • 1. PlateMate add 60 ul DMSO for dilutions 96w: 96/300 ul head, 5516 tips in columns 2-12, compound plate in left stacker A, DMSO reservoir on stage 2.
  • 2. PlateMate 11pt-dilut one-third GABAA: 96/300 ul head, 5516 tips in column 1 of serial dilution magazine, compound plate in left stacker A.
  • 3. PlateMate 2 ul addition of cmpd dry new wash: 96/30 ul head, 5506 tips, compound plate in left stacker A, dilution plate in right stacker A, 100% DMSO in reservoir on stage 2, must change to fresh DMSO every 4-6 plates.
  • 4. PlateMate tip chg mix and disp 25 ul to assay plate 96w: 96/300 ul head, 5516 tips, dilution plate in left stacker A, assay plates in right stacker A, auto fill assay buffer reservoir on stage 2, need to change tips after every plate.
  • 5. PlateMate add 200 ul membranes 96w: 96/300 ul head, 5516 tips, assay plates in left stacker A, membrane reservoir on stage 2.
  • 6. RapidPlate add 25 ul hot (number of plates): 100 μl (yellow box) tips in position 1, hot reservoir in position 2, plates beginning in position 3.
  • 7. RapidPlate add microscint 40 ul (number of plates): 2001 (burgundy box) tips in position 1, Microscint 40 reservoir in position 2, plates beginning in position 3.
  • Data Analysis
  • Data is analyzed by calculating percent of control, IC50, and Ki in an XLfit template. The following formula is used in the templates:
  • Ki = IC 50 1 + [ ligand ] / K D
  • Method GG GABAA3 Binding Method Reagents
  • Assay and Wash Buffer: 50 mM Tris-Citrate, 200 mM NaCl, pH 7.8
  • Compounds at 10 mM in DMSO: Put 75 ul in column 1 of compound plate.
  • Flumazenil, 10 mM (for NSB)
  • Membranes (α3, β3, γ2 receptor subunits transfected into Sf9 cells and harvested; prepared by Cell Trends, stored at −80° C.) Sonicate thawed membranes for about 5-10 seconds at setting 3 on Brinkman sonicator, then dilute membranes 1:125 to make a solution of 200 ug/mL in assay buffer. Keep on ice.
  • [3H]-Flunitrazepam (Cat #NET567): Prepare 10× stock=30 nM, [F} in assay=˜3 nM
  • Assay (See below for Automation Programs.)
  • 1. On PlateMate, prepare 1:3 serial dilutions (30 μl+60 μl) in DMSO for final assay concentrations of 10 μM to 170 μM (Automation Programs 1 and 2). Add 5 μl of 30 μM flumazenil to wells 12 D-E for 50% control wells.
  • 2. Spot 2 μl of compound dilutions into dry plate (Automation Program 3). Manually spot 2 μl 10 mM flumazenil into wells 12 F-H for nonspecific control.
  • 3. Make 1:100 dilution in assay buffer (2 μl into 200 μl) and dispense 25 μl compound into assay plates (Automation Program 4).
  • 4. Dispense 200 μl membranes into assay plate (Automation Program 5).
  • 5. Add 25 μl [3H]-Flunitrazepam (Automation Program 6). Incubate for 1 hr at 4° C.
  • 6. Collect membranes on a cell harvester onto GF/B filter plates (pre-wet with dH2O and wash 5×400 μl/well, with cold assay buffer. (First 3 washes are considered hot; last two are cold.)
  • 7. Dry plates for 2-3 hours at RT.
  • 8. Add 40 μl Microscint 40/well (Automation Program 7); seal plates. Count on a TopCount.
  • Automation Programs
  • 1. PlateMate add 60 μl DMSO for dilutions 96w: 96/300 μl head, 5516 tips in columns 2-12, compound plate in left stacker A, DMSO reservoir on stage 2.
  • 2. PlateMate 11pt-dilut one-third GABAA: 96/300 μl head, 5516 tips in column 1 of serial dilution magazine, compound plate in left stacker A.
  • 3. PlateMate 2 μl addition of cmpd dry new wash: 96/30 μl head, 5506 tips, compound plate in left stacker A, dilution plate in right stacker A, 100% DMSO in reservoir on stage 2, must change to fresh DMSO every 4-6 plates.
  • 4. PlateMate tip chg mix and disp 25 μl to assay plate 96w: 96/300 μl head, 5516 tips, dilution plate in left stacker A, assay plates in right stacker A, auto fill assay buffer reservoir on stage 2, need to change tips after every plate.
  • 5. PlateMate add 200 μl membranes 96w: 96/300 μl head, 5516 tips, assay plates in left stacker A, membrane reservoir on stage 2.
  • 6. RapidPlate add 25 μl hot (number of plates): 100 μl (yellow box) tips in position 1, hot reservoir in position 2, plates beginning in position 3.
  • 7. RapidPlate add microscint 40 μl (number of plates): 200 μl (burgundy box) tips in position 1, Microscint 40 reservoir in position 2, plates beginning in position 3.
  • Data Analysis
  • Data is analyzed by calculating percent of control, IC50, and Ki in an XLfit template. The following formula is used in the templates:
  • Ki = IC 50 1 + [ ligand ] / K D
  • Method HH GABAA5 Binding Method Reagents
  • Assay and Wash Buffer: 50 mM Tris-Citrate, 200 mM NaCl, pH 7.8
  • Compounds at 10 mM in DMSO: Put 75 μl in column 1 of compound plate.
  • Flumazenil, 10 mM (for NSB)
  • Membranes (α5, β3, γ2 receptor subunits transfected into Sf9 cells and harvested; prepared by Cell Trends, stored at −80° C.) Sonicate thawed membranes for about 5-10 seconds at setting 3 on Brinkman sonicator, then dilute membranes 1:31 in assay buffer (working conc.=500 ug/ml protein). Keep on ice.
  • [3H]-Flunitrazepam (Cat #NET567): Prepare 10× stock=20 nM, [F] in assay=˜2 nM
  • Assay (See below for Automation Programs.)
  • 1. On PlateMate, prepare 1:3 serial dilutions (30 μl+60 μl) in DMSO for final assay concentrations of 10 μM to 170 pM (Automation Programs 1 and 2). Add 5 ul of 30 uM flumazenil to wells 12 D-E for 50% control wells.
  • 2. Spot 2 μl of compound dilutions into dry plate (Automation Program 3). Manually spot 2 μl 10 mM flumazenil into wells 12 F-H for nonspecific control.
  • 3. Make 1:100 dilution in assay buffer (2 μl into 200 μl) and dispense 25 μl compound into assay plates (Automation Program 4).
  • 4. Dispense 2001 membranes into assay plate (Automation Program 5).
  • 5. Add 25 μl [3H]-Flunitrazepam (Automation Program 6). Incubate for 1 hr at 4° C.
  • 6. Collect membranes on a cell harvester onto GF/B filter plates (pre-wet with dH2O and wash 5×400 μl/well, with cold assay buffer. (First 3 washes are considered hot; last two are cold.)
  • 7. Dry plates for 2-3 hours at RT.
  • 8. Add 40 μl Microscint 40/well (Automation Program 7); seal plates. Count on a TopCount.
  • Automation Programs
  • 1. PlateMate add 60 ul DMSO for dilutions 96w: 96/300 ul head, 5516 tips in columns 2-12, compound plate in left stacker A, DMSO reservoir on stage 2.
  • 2. PlateMate 11pt-dilut one-third GABAA: 96/300 ul head, 5516 tips in column 1 of serial dilution magazine, compound plate in left stacker A.
  • 3. PlateMate 2 ul addition of cmpd dry new wash: 96/30 ul head, 5506 tips, compound plate in left stacker A, dilution plate in right stacker A, 100% DMSO in reservoir on stage 2, must change to fresh DMSO every 4-6 plates.
  • 4. PlateMate tip chg mix and disp 25 ul to assay plate 96w: 96/300 ul head, 5516 tips, dilution plate in left stacker A, assay plates in right stacker A, auto fill assay buffer reservoir on stage 2, need to change tips after every plate.
  • 5. PlateMate add 200 ul membranes 96w: 96/300 ul head, 5516 tips, assay plates in left stacker A, membrane reservoir on stage 2.
  • 6. RapidPlate add 25 ul hot (number of plates): 100 μl (yellow box) tips in position 1, hot reservoir in position 2, plates beginning in position 3.
  • 7. RapidPlate add microscint 40 ul (number of plates): 2001 (burgundy box) tips in position 1, Microscint 40 reservoir in position 2, plates beginning in position 3.
  • Data Analysis
  • Data is analyzed by calculating percent of control, IC50, and Ki in an XLfit template. The following formula is used in the templates:
  • Ki = IC 50 1 + [ ligand ] / K D
    GABAA2
    Binding Ki Relative Potentiation for Relative Potentiation for
    Compound (M) GABAA1 GABAA2
    Figure US20090018112A1-20090115-C00248
    1.44E-10 −0.015 0.15
    Figure US20090018112A1-20090115-C00249
    2.66E-10 −0.008 0.18
    Figure US20090018112A1-20090115-C00250
    3.01E-10
    Figure US20090018112A1-20090115-C00251
    3.27E-10 0.25
    Figure US20090018112A1-20090115-C00252
    3.41E-10
    Figure US20090018112A1-20090115-C00253
    3.64E-10 0.12 0.063
    Figure US20090018112A1-20090115-C00254
    3.87E-10 0.0017 0.12
    Figure US20090018112A1-20090115-C00255
    4.03E-10 0.063 0.13
    Figure US20090018112A1-20090115-C00256
    5.05E-10 0.15 0.14
    Figure US20090018112A1-20090115-C00257
    5.52E-10 0.38
    Figure US20090018112A1-20090115-C00258
    5.58E-10 −0.11 0.17
    Figure US20090018112A1-20090115-C00259
    5.60E-10 0.17 0.25
    Figure US20090018112A1-20090115-C00260
    5.65E-10 −0.028 0.18
    Figure US20090018112A1-20090115-C00261
    6.00E-10 0.51
    Figure US20090018112A1-20090115-C00262
    6.13E-10 0.053 0.18
    Figure US20090018112A1-20090115-C00263
    6.15E-10 0.11 0.13
    Figure US20090018112A1-20090115-C00264
    6.24E-10 0.28
    Figure US20090018112A1-20090115-C00265
    6.98E-10 −0.048 0.16
    Figure US20090018112A1-20090115-C00266
    7.10E-10 0.12 0.21
    Figure US20090018112A1-20090115-C00267
    7.35E-10 0.032 0.12
    Figure US20090018112A1-20090115-C00268
    8.52E-10 0.095 0.37
    Figure US20090018112A1-20090115-C00269
    9.18E-10 0.12 0.39
    Figure US20090018112A1-20090115-C00270
    9.37E-10 0.23 0.37
    Figure US20090018112A1-20090115-C00271
    9.41E-10 0.05 0.23
    Figure US20090018112A1-20090115-C00272
    9.80E-10 0.085 0.22
    Figure US20090018112A1-20090115-C00273
    9.96E-10 −0.078 0.29
    Figure US20090018112A1-20090115-C00274
    1.00E-09 −0.093 0.02
    Figure US20090018112A1-20090115-C00275
    1.00E-09
    Figure US20090018112A1-20090115-C00276
    1.03E-09 0.27
    Figure US20090018112A1-20090115-C00277
    1.11E-09 0.38 0.4
    Figure US20090018112A1-20090115-C00278
    1.13E-09 −0.083 0.17
    Figure US20090018112A1-20090115-C00279
    1.18E-09 0.088 0.31
    Figure US20090018112A1-20090115-C00280
    1.25E-09 0.14 0.19
    Figure US20090018112A1-20090115-C00281
    1.31E-09 0.21 0.28
    Figure US20090018112A1-20090115-C00282
    1.34E-09 0.21
    Figure US20090018112A1-20090115-C00283
    1.36E-09 0.47 0.59
    Figure US20090018112A1-20090115-C00284
    1.49E-09 0.016 0.21
    Figure US20090018112A1-20090115-C00285
    1.62E-09 0.014 0.23
    Figure US20090018112A1-20090115-C00286
    1.67E-09 −0.038 0.1
    Figure US20090018112A1-20090115-C00287
    1.67E-09 0.053 0.27
    Figure US20090018112A1-20090115-C00288
    1.68E-09 0.083 0.11
    Figure US20090018112A1-20090115-C00289
    1.72E-09 −0.088 0.12
    Figure US20090018112A1-20090115-C00290
    1.73E-09 0.26 0.32
    Figure US20090018112A1-20090115-C00291
    1.82E-09 0.067 0.053
    Figure US20090018112A1-20090115-C00292
    1.84E-09 0.2 0.52
    Figure US20090018112A1-20090115-C00293
    1.93E-09 0.06 0.033
    Figure US20090018112A1-20090115-C00294
    2.07E-09
    Figure US20090018112A1-20090115-C00295
    2.10E-09 0.2 0.097
    Figure US20090018112A1-20090115-C00296
    2.17E-09 0.41 0.59
    Figure US20090018112A1-20090115-C00297
    2.23E-09 0.18 0.29
    Figure US20090018112A1-20090115-C00298
    2.30E-09 0.004 0.063
    Figure US20090018112A1-20090115-C00299
    2.34E-09 −0.11 −0.033
    Figure US20090018112A1-20090115-C00300
    2.42E-09 0.12 0.16
    Figure US20090018112A1-20090115-C00301
    2.50E-09 0.18 0.15
    Figure US20090018112A1-20090115-C00302
    2.60E-09 −0.11 0.054
    Figure US20090018112A1-20090115-C00303
    2.69E-09
    Figure US20090018112A1-20090115-C00304
    2.73E-09 0.19 0.29
    Figure US20090018112A1-20090115-C00305
    2.88E-09 0.1 0.16
    Figure US20090018112A1-20090115-C00306
    2.89E-09 0.47
    Figure US20090018112A1-20090115-C00307
    2.89E-09 0.16 0.45
    Figure US20090018112A1-20090115-C00308
    3.22E-09 0.28 0.11
    Figure US20090018112A1-20090115-C00309
    3.22E-09 0.0058 0.18
    Figure US20090018112A1-20090115-C00310
    3.41E-09 0.045 0.26
    Figure US20090018112A1-20090115-C00311
    3.44E-09 0.19 −0.0075
    Figure US20090018112A1-20090115-C00312
    3.88E-09 0.33 0.28
    Figure US20090018112A1-20090115-C00313
    4.11E-09 −0.16 0.14
    Figure US20090018112A1-20090115-C00314
    4.17E-09 0 0.12
    Figure US20090018112A1-20090115-C00315
    4.21E-09 0.07 0.34
    Figure US20090018112A1-20090115-C00316
    4.34E-09 0.13 0.22
    Figure US20090018112A1-20090115-C00317
    4.34E-09 0.12 0.38
    Figure US20090018112A1-20090115-C00318
    4.36E-09 −0.02 0.28
    Figure US20090018112A1-20090115-C00319
    5.00E-09 0.28 0.57
    Figure US20090018112A1-20090115-C00320
    5.06E-09 0.17 0.19
    Figure US20090018112A1-20090115-C00321
    5.11E-09 0.09 0.16
    Figure US20090018112A1-20090115-C00322
    5.51E-09 0.28 0.38
    Figure US20090018112A1-20090115-C00323
    5.56E-09 0.02 0.22
    Figure US20090018112A1-20090115-C00324
    5.71E-09 0.87
    Figure US20090018112A1-20090115-C00325
    5.78E-09 0.1 0.14
    Figure US20090018112A1-20090115-C00326
    5.82E-09 0.25 0.16
    Figure US20090018112A1-20090115-C00327
    5.98E-09
    Figure US20090018112A1-20090115-C00328
    6.13E-09 0.13 0.52
    Figure US20090018112A1-20090115-C00329
    6.40E-09 0.1 0.23
    Figure US20090018112A1-20090115-C00330
    6.75E-09 0.47 0.33
    Figure US20090018112A1-20090115-C00331
    7.24E-09 0.23 0.18
    Figure US20090018112A1-20090115-C00332
    7.24E-09 0.17 0.23
    Figure US20090018112A1-20090115-C00333
    7.38E-09 −0.063 0.1
    Figure US20090018112A1-20090115-C00334
    7.41E-09 0.1 0.22
    Figure US20090018112A1-20090115-C00335
    7.45E-09 0.098 0.54
    Figure US20090018112A1-20090115-C00336
    7.49E-09 −0.065 0.013
    Figure US20090018112A1-20090115-C00337
    7.51E-09 0.34
    Figure US20090018112A1-20090115-C00338
    8.14E-09 −0.16 0.16
    Figure US20090018112A1-20090115-C00339
    8.49E-09 −0.26 −0.004
    Figure US20090018112A1-20090115-C00340
    8.60E-09 −0.018 0.26
    Figure US20090018112A1-20090115-C00341
    8.72E-09 0.02 0.19
    Figure US20090018112A1-20090115-C00342
    8.77E-09 −0.08 0.16
    Figure US20090018112A1-20090115-C00343
    8.84E-09 0.4 0.46
    Figure US20090018112A1-20090115-C00344
    8.91E-09 0.0075 0.33
    Figure US20090018112A1-20090115-C00345
    9.58E-09 −0.046 0.08
    Figure US20090018112A1-20090115-C00346
    1.04E-08 0.34 0.26
    Figure US20090018112A1-20090115-C00347
    1.05E-08 0.026 0.4
    Figure US20090018112A1-20090115-C00348
    1.05E-08 0.23
    Figure US20090018112A1-20090115-C00349
    1.09E-08 0.11 0.11
    Figure US20090018112A1-20090115-C00350
    1.12E-08 0.24 0.22
    Figure US20090018112A1-20090115-C00351
    1.23E-08 0.18 0.53
    Figure US20090018112A1-20090115-C00352
    1.23E-08 −0.15 0.11
    Figure US20090018112A1-20090115-C00353
    1.33E-08 0.32
    Figure US20090018112A1-20090115-C00354
    1.39E-08 0.32
    Figure US20090018112A1-20090115-C00355
    1.44E-08 0.005 0.35
    Figure US20090018112A1-20090115-C00356
    1.51E-08 0.14 0.33
    Figure US20090018112A1-20090115-C00357
    1.52E-08
    Figure US20090018112A1-20090115-C00358
    1.54E-08 −0.11 0.06
    Figure US20090018112A1-20090115-C00359
    1.61E-08 0.36
    Figure US20090018112A1-20090115-C00360
    1.68E-08 0.23 0.35
    Figure US20090018112A1-20090115-C00361
    1.76E-08 0.21 0.27
    Figure US20090018112A1-20090115-C00362
    1.84E-08 0.48
    Figure US20090018112A1-20090115-C00363
    1.86E-08 −0.26 0.038
    Figure US20090018112A1-20090115-C00364
    1.86E-08 0.11 0.24
    Figure US20090018112A1-20090115-C00365
    1.94E-08 0.027 −0.032
    Figure US20090018112A1-20090115-C00366
    1.95E-08 −0.066 0.11
    Figure US20090018112A1-20090115-C00367
    1.98E-08 0.087 0.45
    Figure US20090018112A1-20090115-C00368
    2.00E-08 −0.14 0.12
    Figure US20090018112A1-20090115-C00369
    2.02E-08 0.03 0.19
    Figure US20090018112A1-20090115-C00370
    2.04E-08 −0.067 −0.01
    Figure US20090018112A1-20090115-C00371
    2.20E-08 0.28 0.48
    Figure US20090018112A1-20090115-C00372
    2.21E-08 −0.012 0.14
    Figure US20090018112A1-20090115-C00373
    2.54E-08 0.086 0.04
    Figure US20090018112A1-20090115-C00374
    2.66E-08 0.07 0.18
    Figure US20090018112A1-20090115-C00375
    2.90E-08 0.013 0.11
    Figure US20090018112A1-20090115-C00376
    2.98E-08 0.013 0.25
    Figure US20090018112A1-20090115-C00377
    3.50E-08 0.15 0.16
    Figure US20090018112A1-20090115-C00378
    3.58E-08 0.19 0.43
    Figure US20090018112A1-20090115-C00379
    3.58E-08 0.79 0.32
    Figure US20090018112A1-20090115-C00380
    3.72E-08 −0.14 0.12
    Figure US20090018112A1-20090115-C00381
    4.33E-08 0.22 0.09
    Figure US20090018112A1-20090115-C00382
    4.58E-08 0.013 0.098
    Figure US20090018112A1-20090115-C00383
    4.78E-08 0.87 0.48
    Figure US20090018112A1-20090115-C00384
    5.38E-08 −0.038 0.08
    Figure US20090018112A1-20090115-C00385
    5.65E-08 0.4 0.67
    Figure US20090018112A1-20090115-C00386
    9.14E-08 0.12 0.08
    Figure US20090018112A1-20090115-C00387
    9.15E-08 0.13 0.26
    Figure US20090018112A1-20090115-C00388
    1.09E-07
    Figure US20090018112A1-20090115-C00389
    1.15E-07 0.32 0.04
    Figure US20090018112A1-20090115-C00390
    1.29E-07 0.15 0.33
    Figure US20090018112A1-20090115-C00391
    1.50E-07 0.09 0.023
    Figure US20090018112A1-20090115-C00392
    3.56E-07 0.44 0.13
    Figure US20090018112A1-20090115-C00393
    5.14E-07
    Figure US20090018112A1-20090115-C00394
    6.24E-07
    Figure US20090018112A1-20090115-C00395
    5.50E-06
    Figure US20090018112A1-20090115-C00396
    5.50E-06
    Figure US20090018112A1-20090115-C00397
    5.50E-06
    Figure US20090018112A1-20090115-C00398
    5.50E-06
    Figure US20090018112A1-20090115-C00399
    5.50E-06
    Figure US20090018112A1-20090115-C00400
    5.50E-06
    Figure US20090018112A1-20090115-C00401
    5.50E-06
    Figure US20090018112A1-20090115-C00402
    5.50E-06
    Figure US20090018112A1-20090115-C00403
    5.50E-06
    Figure US20090018112A1-20090115-C00404
    5.50E-06
    Figure US20090018112A1-20090115-C00405
    5.50E-06
    Figure US20090018112A1-20090115-C00406
    5.50E-06
    Figure US20090018112A1-20090115-C00407
    5.50E-06
    Figure US20090018112A1-20090115-C00408
    5.50E-06
    Figure US20090018112A1-20090115-C00409
    9.00E-09 −0.010 0.223
    Figure US20090018112A1-20090115-C00410
    3.00E-10 −0.040 0.119
    Figure US20090018112A1-20090115-C00411
    8.22E-09 −0.028 0.357
    Figure US20090018112A1-20090115-C00412
    7.85E-10 0.077 0.337
    Figure US20090018112A1-20090115-C00413
    5.01E-10 −0.054 0.247
    Figure US20090018112A1-20090115-C00414
    9.27E-10 0.091 0.243
    Figure US20090018112A1-20090115-C00415
    4.02E-09 −0.166 0.074
    Figure US20090018112A1-20090115-C00416
    4.54E-09 −0.006 0.104
  • Method II MT1 GTPγ35S-SPA Assay Test Validation Standards
  • 2-Iodomelatonin and 6-Chloromelatonin with known activities were used as validation standard during the assay development. The EC50 of 2-Iodomelatonin and 6-Chloromelatonin were ˜3E-11 M and ˜1.5E-10 M respectively in GTPγS assay of hMT1 recombinant cell membranes.
  • Cells and/or Microorganisms
  • HEK293F (human embryonic kidney 293 floating cell line) was suspension cultured in Free Style 293 Expression Medium, and expanded in house and stored in liquid nitrogen in cell freezing medium.
  • Buffers, Solutions, Cell Media
    Make 2 Liters
    Lazareno GTPγS Assay Buffer: Buffer
     20 mM HEPES Sigma H-4034, FW 238.3 9.532 g
    100 mM NaCl Sigma S-9625, FW 58.44 11.688 g 
     10 mM MgCl2.6H2O Sigma M-2670, FW 203.3 4.066 g
    pH 7.4 Adjust with NaOH
    Make 2 Liters
    Membrane Prepration Buffer: Buffer
     20 mM HEPES Sigma H-4034, FW 238.3 9.532 g
     3 mM MgCl2.6H2O Sigma M-2670, FW 203.3 1.220 g
     1 mM EGTA Sigma E-3889, FW 380.4 0.761 g
    pH 7.4 Adjust with NaOH
  • Test Compound Preparation
  • Test compounds were synthesized in house. Solid compounds were solubilized at 10 mM in DMSO; then 1:3 further diluted in DMSO in 96-well U-bottom plates using PlateMate on the assay day. 2 μl of diluted compounds were transferred to Opti-assay-plates.
  • Reference Compounds Preparation
  • Reference compound, 2-Iodomelatonin, was prepared the way same as test compound.
  • Compounds Used to Normalize Experimental Results
  • 2-Iodomelatonin for normalization was diluted in DMSO at concentration 50×3 nM (its EC100 concentration=3 nM). 2 μl of 150 nM 2-Iodomelatonin was then transferred to Opti-assay-plates.
  • Cell Lines and Microorganisms
  • HEK293F (human embryonic kidney 293 floating cell line) cells transiently expressed human Melatonin receptor 1 (MT 1) were harvested 48 hours post-transfection. The cell pellets were homogenized using Polytron; and the cell membranes were prepared from GTPγS assay.
  • Preparation of Protein/Membranes Containing Target
  • The cell pellets were homogenized with Polytron in ice-cold buffer: 20 mM HEPES, 3 mM MgCl2, 1 mM EGTA, pH7.4. (Freshly add protease inhibitor cocktail tables from Roche). The samples were centrifuged at 18,500 rpm for 30 mins at 4° C. in Sorvall SS-34 rotor. The membrane pellets were collected and washed with the ice-cold buffer. The samples were centrifuged at 18,500 rpm for 30 mins at 4° C. again. The membranes were resuspended in the ice-cold buffer with protease inhibitors. The protein concentration of the membrane was determined. The membranes were aliquoted and stored at −80° C.
  • Test Method
  • Plate format (if plates are used, as shown in the following table)
  • *Numbers denote “Compound #, Dilution #, Replicate #”
    *Plate direction moves from highest concentration to lowest concentration
    Number of compounds per plate: 8
    Number of replicates per compound: 1
    Number of dilutions per compound: 11
    Test Plate: DR96_02_C12[LR.1]
    1 2 3 4 5 6 7 8 9 10 11 12
    A 1, 1, 1 1, 2, 1 1, 3, 1 1, 4, 1 1, 5, 1 1, 6, 1 1, 7, 1 1, 8, 1 1, 9, 1 1, 10, 1 1, 11, 1 MAX
    B 2, 1, 1 2, 2, 1 2, 3, 1 2, 4, 1 2, 5, 1 2, 6, 1 2, 7, 1 2, 8, 1 2, 9, 1 2, 10, 1 2, 11, 1 MAX
    C 3, 1, 1 3, 2, 1 3, 3, 1 3, 4, 1 3, 5, 1 3, 6, 1 3, 7, 1 3, 8, 1 3, 9, 1 3, 10, 1 3, 11, 1 MAX
    D 4, 1, 1 4, 2, 1 4, 3, 1 4, 4, 1 4, 5, 1 4, 6, 1 4, 7, 1 4, 8, 1 4, 9, 1 4, 10, 1 4, 11, 1 MAX
    E 5, 1, 1 5, 2, 1 5, 3, 1 5, 4, 1 5, 5, 1 5, 6, 1 5, 7, 1 5, 8, 1 5, 9, 1 5, 10, 1 5, 11, 1 MIN
    F 6, 1, 1 6, 2, 1 6, 3, 1 6, 4, 1 6, 5, 1 6, 6, 1 6, 7, 1 6, 8, 1 6, 9, 1 6, 10, 1 6, 11, 1 MIN
    G 7, 1, 1 7, 2, 1 7, 3, 1 7, 4, 1 7, 5, 1 7, 6, 1 7, 7, 1 7, 8, 1 7, 9, 1 7, 10, 1 7, 11, 1 MIN
    H 8, 1, 1 8, 2, 1 8, 3, 1 8, 4, 1 8, 5, 1 8, 6, 1 8, 7, 1 8, 8, 1 8, 9, 1 8, 10, 1 8, 11, 1 MIN
    MAX response (100% effect) was determined as the effect of 3 nM of 2-Iodomelatonin.
    MIN response (0% effect) was determined as the effect of vehicle control.
  • Description of Experimental Procedure
  • Human MT1/HEK293F membrane (10 μg/well) was mixed with WGA-SPA beads (300 μg/well) and GDP (10 μM) in certain volume of Lazareno assay buffer (20 mM HEPES, 100 mM NaCl, 10 mM MgCl2, pH7.4). The membrane combo was kept on ice for 30-60 mins. Test compounds were 1:3 diluted in DMSO from 10 mM stock, and transferred 2 μl of diluted compounds to Opti assay plates-96 using PlateMate. GTPγ35S was added to the membrane mixture prior to dispensing 100 μl the membrane combo to the assay plates-96. The final concentration of GTPγ35S was 200 pM. The assay plates were shaking on a plate shaker for 1.5 hours at room temperature. The assay plates were spun at 2000 rpm for 5 mins in bench top centrifuge. The assay plates were measured in TopCount to capture the data within 4 hours.
  • Summary of the different experimental conditions (the role of various results types)
  • Final concentrations of the constituents
  •  10 μg/well hMT1/HEK293F membranes
    300 μg/well WGA-SPA beads
     10 μM GDP
    200 pM GTPγ35S
     10 μM Start concentration of test compound
    2% DMSO
     20 mM HEPES
    100 mM NaCl
     10 mM MgCl2
    pH7.4
  • Treatments Used in Different Experimental Conditions
  • The test compounds would be heated to 65° C. if they were not soluble at 10 mM in DMSO. The start concentration in general was 10 μM, but could be adjusted based on its potency. Every single batch of membranes had to be validated for its optimal assay conditions, such as, define the optimal GDP concentration, SPA beads amount and EC100 concentration of normalization compound.
  • Calculation of Results
  • Compounds were evaluated for their agonist potency (EC50) and efficacy (Emax). Concentration-response curves were analyzed to determine the EC50 by ActivityBase using equation model #205. Compound's % activity was calculated according to the 100% and 0% activities defined on the same plate as the sample data. Wells A12-C12 were used to define 100% activity, and D12-G12 for 0% activity. More details could be found from the Plate Format above.
  • Results (Dependent Variables, Dependent Measurements) and their Calculation Method
  • The raw values for the replicates in the Minimum Control experimental condition were averaged. The raw values for the replicates in the Maximum Control experimental condition were averaged. The average Minimum Control was subtracted from the average Maximum Control resulting in the Data Window. The average for the Minimum Control was subtracted from each raw value in the Compound Data experimental condition resulting in the Specific Response for each data value in the Compound Data condition. Each Specific Response in the Compound Data condition was divided by the Data Window then multiplied by 100 resulting in the Percent Response. The EC50 and SlopeFactor were determined by fitting the Percent Inhibition and the concentrations of test compound to model 205 in XLfit—y=A+((B−A)/(1+((C/x)̂D))—with parameter A constrained to 0 and parameter B constrained to 100.
  • Certain compounds of the invention have been tested using the above-identified assay (Method II). The results are shown in the following table.
  • MT1 Receptor
    Binding
    %
    Inhibition Ki GTPgS
    at 1 μM (M) EC50 (M) Emax
    Figure US20090018112A1-20090115-C00417
    86 6.00E−08 3.75E−08 100
    Figure US20090018112A1-20090115-C00418
    96 5.60E−08 5.60E−08 61
    Figure US20090018112A1-20090115-C00419
    82 9.20E−08 9.20E−08 64
    Figure US20090018112A1-20090115-C00420
    93 2.76E−07 2.76E−07 70
    Figure US20090018112A1-20090115-C00421
    91  3.6E−08 2.67E−08 61
    Figure US20090018112A1-20090115-C00422
    93  3.1E−08 2.36E−08 64
    Figure US20090018112A1-20090115-C00423
    89  4.9E−08 3.43E−08 60
    Figure US20090018112A1-20090115-C00424
    88  9.0E−08 1.55E−08 54
  • Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference (including, but not limited to, journal articles, U.S. and non-U.S. patents, patent application publications, international patent application publications, and the like) cited in the present application is incorporated herein by reference in its entirety.

Claims (22)

1. A compound selected from 4-amino-8-(2-fluoro-6-methoxy-phenyl)-N-propylcinnoline-3-carboxamide and pharmaceutically acceptable salts thereof.
2. A method of treating or preventing an anxiety disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1.
3. The method of claim 2 wherein the anxiety disorder is panic disorder, panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, social anxiety disorder, obsessive-compulsive disorder, posttraumatic stress disorder, acute stress disorder, generalized anxiety disorder, or generalized anxiety disorder due to a general medical condition.
4. The method of claim 2 wherein the mammal is a human.
5. A method of treating or preventing an anxiety disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1 and at least one cognitive enhancing agent, memory enhancing agent, or choline esterase inhibitor.
6. The method of claim 5 wherein the anxiety disorder is panic disorder, panic disorder without agoraphobia, panic disorder with agoraphobia, agoraphobia without history of panic disorder, specific phobia, social phobia, social anxiety disorder, obsessive-compulsive disorder, posttraumatic stress disorder, acute stress disorder, generalized anxiety disorder, or generalized anxiety disorder due to a general medical condition.
7. The method of claim 5 wherein the mammal is a human.
8. A method of treating or preventing a cognitive disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1.
9. The method of claim 8 wherein the cognitive disorder is Alzheimer's disease, dementia, dementia due to Alzheimer's disease, or dementia due to Parkinson's disease.
10. The method of claim 8 wherein the mammal is a human.
11. A method of treating or preventing a cognitive disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1 and at least one cognitive enhancing agent, memory enhancing agent, or choline esterase inhibitor.
12. The method of 11 wherein the cognitive disorder is Alzheimer's disease, dementia, dementia due to Alzheimer's disease, or dementia due to Parkinson's disease.
13. The method of claim 11 wherein the mammal is a human.
14. A method of treating or preventing a mood disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1.
15. The method of claim 14 wherein the mood disorder is a depressive disorder.
16. The method of claim 15 wherein the depressive disorder is major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, or mixed episodes associated with bipolar disorder.
17. The method of claim 14 wherein the mammal is a human.
18. A method of treating or preventing a mood disorder in a mammal, comprising administering to the mammal a therapeutically effective amount of a compound of claim 1 and at least one cognitive enhancing agent, memory enhancing agent, or choline esterase inhibitor.
19. The method of claim 18 wherein the mood disorder is a depressive disorder.
20. The method of claim 19 wherein the depressive disorder is major depressive disorder, dysthymic disorder, bipolar depression and/or bipolar mania, bipolar I with or without manic, depressive or mixed episodes, bipolar II, cyclothymic disorder, mood disorder due to a general medical condition, manic episodes associated with bipolar disorder, or mixed episodes associated with bipolar disorder.
21. The method of claim 19 wherein the mammal is a human.
22. An atropisomer of a compound according to claim 1.
US12/199,034 2005-12-20 2008-08-27 Compounds and Uses Thereof Abandoned US20090018112A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/199,034 US20090018112A1 (en) 2005-12-20 2008-08-27 Compounds and Uses Thereof
US13/031,763 US20110144331A1 (en) 2005-12-20 2011-02-22 Cinnoline Compounds for the Treatment of Anxiety, Cognitive and Mood Disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US75213705P 2005-12-20 2005-12-20
US82369306P 2006-08-28 2006-08-28
US11/611,943 US7465795B2 (en) 2005-12-20 2006-12-18 Compounds and uses thereof
US12/199,034 US20090018112A1 (en) 2005-12-20 2008-08-27 Compounds and Uses Thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/611,943 Division US7465795B2 (en) 2005-12-20 2006-12-18 Compounds and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/031,763 Continuation US20110144331A1 (en) 2005-12-20 2011-02-22 Cinnoline Compounds for the Treatment of Anxiety, Cognitive and Mood Disorders

Publications (1)

Publication Number Publication Date
US20090018112A1 true US20090018112A1 (en) 2009-01-15

Family

ID=38174480

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/611,943 Active US7465795B2 (en) 2005-12-20 2006-12-18 Compounds and uses thereof
US12/199,034 Abandoned US20090018112A1 (en) 2005-12-20 2008-08-27 Compounds and Uses Thereof
US13/031,763 Abandoned US20110144331A1 (en) 2005-12-20 2011-02-22 Cinnoline Compounds for the Treatment of Anxiety, Cognitive and Mood Disorders

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/611,943 Active US7465795B2 (en) 2005-12-20 2006-12-18 Compounds and uses thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/031,763 Abandoned US20110144331A1 (en) 2005-12-20 2011-02-22 Cinnoline Compounds for the Treatment of Anxiety, Cognitive and Mood Disorders

Country Status (2)

Country Link
US (3) US7465795B2 (en)
TW (1) TW200730506A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080318925A1 (en) * 2007-06-19 2008-12-25 Astrazeneca Ab Compounds and Uses Thereof - 849
US8435988B2 (en) 2010-10-06 2013-05-07 Glaxosmithkline Llc Benzimidazole derivatives as P13 kinase inhibitors
DE212011100209U1 (en) 2011-09-13 2014-04-22 U.S. Nutraceuticals Llc Dba Valensa International Plant-derived seed extract rich in essential fatty acids derived from perilla seeds
US10683293B2 (en) 2014-08-04 2020-06-16 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US11447479B2 (en) 2019-12-20 2022-09-20 Nuevolution A/S Compounds active towards nuclear receptors
US11613532B2 (en) 2020-03-31 2023-03-28 Nuevolution A/S Compounds active towards nuclear receptors
US11780843B2 (en) 2020-03-31 2023-10-10 Nuevolution A/S Compounds active towards nuclear receptors

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2391472T3 (en) * 2005-12-20 2012-11-27 Astrazeneca Ab Substituted cinnoline derivatives as GABAA receptor modulators and method for their synthesis
PE20071068A1 (en) * 2005-12-20 2007-12-13 Glaxo Group Ltd 3- (4 - {[4- (4 - {[3- (3,3-DIMETHYL-1-PIPERIDINYL) PROPYL] OXY} PHENYL) -1-PIPERIDINYL] CARBONYL} -1-NAPHTHALENYL) PROPANOIC OR PROPENOIC ACID, SALTS OF THE SAME, AS ANTAGONISTS OF THE H1 AND H3 RECEPTORS
US7465795B2 (en) * 2005-12-20 2008-12-16 Astrazeneca Ab Compounds and uses thereof
US20100105614A1 (en) 2006-10-25 2010-04-29 Somaxon Pharmaceuticals, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
WO2008070795A2 (en) * 2006-12-06 2008-06-12 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
WO2010123440A1 (en) * 2009-04-21 2010-10-28 Astrazeneca Ab Pharmaceutical composition comprising 4-amino-8-(2-fluoro-6-methoxy-phenyl)-n- propylcinnoline-3-carboxamide hydrogen sulphate and rate-controlling polymer
WO2010123443A1 (en) * 2009-04-21 2010-10-28 Astrazeneca Ab Pharmaceutical composition comprising 4-amino-8-(2-fluoro-6-methoxy-phenyl)-n- propylcinnoline-3-carboxamide hydrogen sulphate.
US20110040208A1 (en) * 2009-08-11 2011-02-17 Abbott Diabetes Care Inc. Integrated lancet and test strip and methods of making and using same
WO2011021979A1 (en) * 2009-08-18 2011-02-24 Astrazeneca Ab Cinnoline compounds, their preparation, and their use
EP3615527B1 (en) * 2017-06-30 2024-03-06 Elanco Animal Health GmbH New azaquinoline derivatives

Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4230713A (en) * 1979-01-19 1980-10-28 Ici Americas Inc. Heterocyclic tetrahydro-1-alkyl-4-oxo-1H-imidazol-2-ylidene urea and phenyl esters of tetrahydro-1-alkyl-4-oxo-1H-imidazol-2-ylidene carbamic acid compounds
US4511568A (en) * 1982-05-12 1985-04-16 Ici Americas Inc. CNS-Depressant pyrazolopyridines
US4546104A (en) * 1983-11-04 1985-10-08 Ici Americas Inc. Pyrazolopyridine cycloalkanones and process for their preparation
US4552883A (en) * 1982-06-15 1985-11-12 Ici Americas Inc. Pyrazolo[3,4-b]pyridine carboxylic acid esters and their pharmaceutical use
US4563525A (en) * 1983-05-31 1986-01-07 Ici Americas Inc. Process for preparing pyrazolopyridine compounds
US4705793A (en) * 1984-08-20 1987-11-10 Ici Americas Inc. Pyrazolo[3,4-b]pyrrolo[3,4-e]pyridine-5(1H)-one and 1-H-pyrazolo[3,4-b][1,6]naphthyridine-5(6H)-one derivations, useful as anti-anxiety agents
US4745121A (en) * 1984-10-04 1988-05-17 Ici Americas Inc. Pyrazolo[3,4-b]pyridine-5-carboxamides and their use as anxiolytic agents
US4886800A (en) * 1985-05-30 1989-12-12 Ici Americas Inc. Substituted cinnoline derivatives as CNS depressants
US4925844A (en) * 1988-02-09 1990-05-15 Ici Americas Inc. Antagonizing the pharmacological effects of a benzodiazepine receptor agonist
US4975435A (en) * 1986-05-06 1990-12-04 Ici Americas Inc. Certain 1H-pyrrold[3,4-b]quinolin-1-one-9-amino-2,3-dihydro derivatives useful for treating anxiety
US5190951A (en) * 1990-10-19 1993-03-02 Ss Pharmaceutical Co., Ltd. Quinoline derivatives
US5240934A (en) * 1990-10-19 1993-08-31 Ss Pharmaceutical Co., Ltd. Quinoline derivatives
US5646153A (en) * 1991-05-10 1997-07-08 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5756804A (en) * 1995-07-25 1998-05-26 Hoechst Aktiengesellschaft Homogeneous process for carrying out cross-coupling reactions
US5801263A (en) * 1996-05-17 1998-09-01 Hoechst Aktiengesellschaft Process for preparing tertiary phosphines containing phosphinate or phosphonate groups, and novel tertiary phosphines containing phosphinate groups
US6015904A (en) * 1993-03-30 2000-01-18 Sworin; Michael Stable reagents for the preparation of radio pharmaceuticals
US6362216B1 (en) * 1998-10-27 2002-03-26 Array Biopharma Inc. Compounds which inhibit tryptase activity
US6417357B1 (en) * 1998-03-18 2002-07-09 Ciba Specialty Chemicals Corporation Coupling reactions with palladium catalysts
US6566571B1 (en) * 1999-04-10 2003-05-20 Degussa Ag Method of producing biaryls
US20040097485A1 (en) * 2002-10-31 2004-05-20 Tularik Inc. Antiinflammation agents
US20040102360A1 (en) * 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
US20040167165A1 (en) * 2003-01-16 2004-08-26 Geetha Shankar Methods of treating conditions associated with an Edg-7 receptor
US20040186148A1 (en) * 2003-03-20 2004-09-23 Schering Corporation Cannabinoid receptor ligands
US6800784B1 (en) * 1999-11-26 2004-10-05 Rhodia Chimie Process for preparing a polyaromatic compound
US20050113283A1 (en) * 2002-01-18 2005-05-26 David Solow-Cordero Methods of treating conditions associated with an EDG-4 receptor
US6984756B2 (en) * 2000-05-19 2006-01-10 Eli Lilly And Company Process for preparing biphenyl compounds
US20060264439A1 (en) * 2005-05-17 2006-11-23 Supergen, Inc. Inhibitors of polo-like kinase-1
US7425556B2 (en) * 2005-12-20 2008-09-16 Astrazeneca Ab Compounds and uses thereof
US7465795B2 (en) * 2005-12-20 2008-12-16 Astrazeneca Ab Compounds and uses thereof
US20080318925A1 (en) * 2007-06-19 2008-12-25 Astrazeneca Ab Compounds and Uses Thereof - 849
US20090012084A1 (en) * 2007-01-25 2009-01-08 Astrazeneca Ab 3-cinnolinecarboxamide derivatives and their use for treating cancer

Family Cites Families (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DD249011A5 (en) 1986-06-20 1987-08-26 Ici Americas Inc,Us PROCESS FOR THE PRODUCTION OF CINNOLIN COMPOUNDS
ATE79540T1 (en) 1988-02-09 1992-09-15 Ici America Inc PHARMACEUTICAL COMPOSITION.
WO1992016497A1 (en) 1991-03-22 1992-10-01 Japan Tobacco Inc. Amino acid derivative having renin inhibitory activity
US5710158A (en) 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
ATE159009T1 (en) 1991-05-10 1997-10-15 Rhone Poulenc Rorer Int BIS MONO- AND BICYCLIC ARYL AND HETEROARYL DERIVATIVES WITH AN INHIBITING EFFECT ON THE EGF AND/OR PDGF RECEPTOR TYROSINKINASE
GB9607219D0 (en) 1996-04-04 1996-06-12 Smithkline Beecham Plc Novel compounds
JP2002506073A (en) 1998-03-12 2002-02-26 ノボ ノルディスク アクティーゼルスカブ Modulators of protein tyrosine phosphatase (PTPases)
GB9919957D0 (en) 1999-08-23 1999-10-27 Merck Sharp & Dohme Therapeutic agents
GB9921150D0 (en) 1999-09-07 1999-11-10 Merck Sharp & Dohme Therapeutic agents
GB9921351D0 (en) 1999-09-09 1999-11-10 Merck Sharp & Dohme Therapeutic agents
GB9927687D0 (en) 1999-11-23 2000-01-19 Merck Sharp & Dohme Therapeutic agents
GB9929685D0 (en) 1999-12-15 2000-02-09 Merck Sharp & Dohme Therapeutic agents
GB9929687D0 (en) 1999-12-15 2000-02-09 Merck Sharp & Dohme Therapeutic agents
CN1398258A (en) 2000-03-21 2003-02-19 法玛西雅厄普约翰美国公司 4-hydroxycinnoline-3-carboxyamides as antiviral agents
ES2249433T3 (en) 2000-05-19 2006-04-01 Eli Lilly And Company BIFENOL COMPOUND PREPARATION PROCEDURE.
EP1294723A1 (en) 2000-05-24 2003-03-26 MERCK SHARP & DOHME LTD. 3-phenyl-imidazo-pyrimidine derivatives as ligands for gaba receptors
AUPQ841300A0 (en) 2000-06-27 2000-07-20 Fujisawa Pharmaceutical Co., Ltd. New aminoalcohol derivatives
GB0018473D0 (en) 2000-07-27 2000-09-13 Merck Sharp & Dohme Therapeutic agents
AU2002212530B2 (en) 2000-11-10 2006-08-17 Merck Sharp & Dohme Limited Imidazo-triazine derivatives as ligands for GABA receptors
GB0027561D0 (en) 2000-11-10 2000-12-27 Merck Sharp & Dohme Therapeutic agents
GB0111191D0 (en) 2001-05-08 2001-06-27 Merck Sharp & Dohme Therapeutic agents
WO2003003009A1 (en) 2001-06-29 2003-01-09 7Tm Pharma A/S Use of metal-ion chelates in validating biological molecules as drug targets in test animal models
GB0117060D0 (en) 2001-07-12 2001-09-05 Merck Sharp & Dohme Therapeutic agents
GB0119803D0 (en) 2001-08-14 2001-10-10 Merck Sharp & Dohme Therapeutic agents
GB0119828D0 (en) 2001-08-14 2001-10-10 Merck Sharp & Dohme Therapeutic agents
GB0120345D0 (en) 2001-08-21 2001-10-17 Merck Sharp & Dohme Therapeutic agents
AUPR738301A0 (en) 2001-08-30 2001-09-20 Starpharma Limited Chemotherapeutic agents
GB0122696D0 (en) 2001-09-20 2001-11-14 Merck Sharp & Dohme Therapeutic agents
AU2003214873A1 (en) 2002-01-18 2003-09-02 Ceretek Llc Methods of treating conditions associated with an edg receptor
GB0208394D0 (en) 2002-04-11 2002-05-22 Merck Sharp & Dohme Therapeutic agents
PL189894B1 (en) 2002-04-15 2005-10-31 Univ Medyczny W Lodzi Method of manufacture of 6,7,8-substituted 4-hydroxycynnoline-3-carboxyl acids
GB0210127D0 (en) 2002-05-02 2002-06-12 Merck Sharp & Dohme Therapeutic agents
GB0210124D0 (en) 2002-05-02 2002-06-12 Merck Sharp & Dohme Therapeutic agents
GB0212048D0 (en) 2002-05-24 2002-07-03 Merck Sharp & Dohme Therapeutic agents
GB0212049D0 (en) 2002-05-24 2002-07-03 Merck Sharp & Dohme Therapeutic agents
CA2491820A1 (en) 2002-07-17 2004-01-22 Warner-Lambert Company Llc Combination of an allosteric alkyne inhibitor of matrix metalloproteinase-13 with a selective inhibitor of cyclooxygenase-2 that is not celecoxib or valdecoxib
WO2004014865A1 (en) 2002-08-13 2004-02-19 Merck Sharp & Dohme Limited Phenylpyridazine derivatives as ligands for gaba receptors
GB0218876D0 (en) 2002-08-13 2002-09-25 Merck Sharp & Dohme Therapeutic agents
SE0202461D0 (en) 2002-08-14 2002-08-14 Astrazeneca Ab Novel compounds
EP1558585B1 (en) 2002-10-04 2013-09-25 Prana Biotechnology Limited Neurologically-active compounds
GB0225501D0 (en) 2002-11-01 2002-12-11 Merck Sharp & Dohme Therapeutic agents
GB0226462D0 (en) 2002-11-13 2002-12-18 Merck Sharp & Dohme Therapeutic agents
GB0301350D0 (en) 2003-01-21 2003-02-19 Merck Sharp & Dohme Therapeutic agents
WO2004076452A1 (en) 2003-02-26 2004-09-10 Merck Sharp & Dohme Limited 5,8-DIFLUOROIMIDAZO[1,2-a]PYRIDINES AS GABA-A α2/α3 LIGANDS FOR TREATING ANXIETY AND/OR DEPRESSION
US7767817B2 (en) 2003-09-05 2010-08-03 Binghe Wang Water soluble boronic acid fluorescent reporter compounds and methods of use thereof
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
CA2546353A1 (en) 2003-11-19 2005-06-09 Array Biopharma Inc. Bicyclic inhibitors of mek and methods of use thereof
PT1699800E (en) 2003-12-23 2010-04-12 Novartis Ag Bicyclic heterocyclic p-38 kinase inhibitors

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4230713A (en) * 1979-01-19 1980-10-28 Ici Americas Inc. Heterocyclic tetrahydro-1-alkyl-4-oxo-1H-imidazol-2-ylidene urea and phenyl esters of tetrahydro-1-alkyl-4-oxo-1H-imidazol-2-ylidene carbamic acid compounds
US4511568A (en) * 1982-05-12 1985-04-16 Ici Americas Inc. CNS-Depressant pyrazolopyridines
US4645838A (en) * 1982-05-12 1987-02-24 Ici Americas Inc. Pyrazolopyridine compounds, and intermediates, useful as anxiolytic agents
US4552883A (en) * 1982-06-15 1985-11-12 Ici Americas Inc. Pyrazolo[3,4-b]pyridine carboxylic acid esters and their pharmaceutical use
US4563525A (en) * 1983-05-31 1986-01-07 Ici Americas Inc. Process for preparing pyrazolopyridine compounds
US4546104A (en) * 1983-11-04 1985-10-08 Ici Americas Inc. Pyrazolopyridine cycloalkanones and process for their preparation
US4705793A (en) * 1984-08-20 1987-11-10 Ici Americas Inc. Pyrazolo[3,4-b]pyrrolo[3,4-e]pyridine-5(1H)-one and 1-H-pyrazolo[3,4-b][1,6]naphthyridine-5(6H)-one derivations, useful as anti-anxiety agents
US4745121A (en) * 1984-10-04 1988-05-17 Ici Americas Inc. Pyrazolo[3,4-b]pyridine-5-carboxamides and their use as anxiolytic agents
US4886800A (en) * 1985-05-30 1989-12-12 Ici Americas Inc. Substituted cinnoline derivatives as CNS depressants
US4975435A (en) * 1986-05-06 1990-12-04 Ici Americas Inc. Certain 1H-pyrrold[3,4-b]quinolin-1-one-9-amino-2,3-dihydro derivatives useful for treating anxiety
US5118688A (en) * 1986-05-06 1992-06-02 Ici Americas Inc. Tetrahydropyridoquinolone derivatives useful as anxiolytic agents
US4925844A (en) * 1988-02-09 1990-05-15 Ici Americas Inc. Antagonizing the pharmacological effects of a benzodiazepine receptor agonist
US5190951A (en) * 1990-10-19 1993-03-02 Ss Pharmaceutical Co., Ltd. Quinoline derivatives
US5240934A (en) * 1990-10-19 1993-08-31 Ss Pharmaceutical Co., Ltd. Quinoline derivatives
US5300517A (en) * 1990-10-19 1994-04-05 Ss Pharmaceutical Co., Ltd. Piperidine compounds having anti-acetylcholinesterase activity
US5646153A (en) * 1991-05-10 1997-07-08 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US6057320A (en) * 1991-05-10 2000-05-02 Aventis Pharmaceuticals Products Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US6015904A (en) * 1993-03-30 2000-01-18 Sworin; Michael Stable reagents for the preparation of radio pharmaceuticals
US5756804A (en) * 1995-07-25 1998-05-26 Hoechst Aktiengesellschaft Homogeneous process for carrying out cross-coupling reactions
US6140265A (en) * 1995-07-25 2000-10-31 Clariant Gmbh Catalyst for cross-coupling reactions
US5801263A (en) * 1996-05-17 1998-09-01 Hoechst Aktiengesellschaft Process for preparing tertiary phosphines containing phosphinate or phosphonate groups, and novel tertiary phosphines containing phosphinate groups
US6417357B1 (en) * 1998-03-18 2002-07-09 Ciba Specialty Chemicals Corporation Coupling reactions with palladium catalysts
US6362216B1 (en) * 1998-10-27 2002-03-26 Array Biopharma Inc. Compounds which inhibit tryptase activity
US6566571B1 (en) * 1999-04-10 2003-05-20 Degussa Ag Method of producing biaryls
US6800784B1 (en) * 1999-11-26 2004-10-05 Rhodia Chimie Process for preparing a polyaromatic compound
US6984756B2 (en) * 2000-05-19 2006-01-10 Eli Lilly And Company Process for preparing biphenyl compounds
US20050113283A1 (en) * 2002-01-18 2005-05-26 David Solow-Cordero Methods of treating conditions associated with an EDG-4 receptor
US20040102360A1 (en) * 2002-10-30 2004-05-27 Barnett Stanley F. Combination therapy
US20040097485A1 (en) * 2002-10-31 2004-05-20 Tularik Inc. Antiinflammation agents
US20040167165A1 (en) * 2003-01-16 2004-08-26 Geetha Shankar Methods of treating conditions associated with an Edg-7 receptor
US20040186148A1 (en) * 2003-03-20 2004-09-23 Schering Corporation Cannabinoid receptor ligands
US20060264439A1 (en) * 2005-05-17 2006-11-23 Supergen, Inc. Inhibitors of polo-like kinase-1
US7425556B2 (en) * 2005-12-20 2008-09-16 Astrazeneca Ab Compounds and uses thereof
US7465795B2 (en) * 2005-12-20 2008-12-16 Astrazeneca Ab Compounds and uses thereof
US20090036454A1 (en) * 2005-12-20 2009-02-05 Astrazeneca Ab Compounds and Uses Thereof
US20090012084A1 (en) * 2007-01-25 2009-01-08 Astrazeneca Ab 3-cinnolinecarboxamide derivatives and their use for treating cancer
US20080318925A1 (en) * 2007-06-19 2008-12-25 Astrazeneca Ab Compounds and Uses Thereof - 849

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080318925A1 (en) * 2007-06-19 2008-12-25 Astrazeneca Ab Compounds and Uses Thereof - 849
US10660898B2 (en) 2010-10-06 2020-05-26 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US8435988B2 (en) 2010-10-06 2013-05-07 Glaxosmithkline Llc Benzimidazole derivatives as P13 kinase inhibitors
US8541411B2 (en) 2010-10-06 2013-09-24 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US8674090B2 (en) 2010-10-06 2014-03-18 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US8865912B2 (en) 2010-10-06 2014-10-21 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US9062003B2 (en) 2010-10-06 2015-06-23 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US9156797B2 (en) 2010-10-06 2015-10-13 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US9872860B2 (en) 2010-10-06 2018-01-23 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US10314845B2 (en) 2010-10-06 2019-06-11 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
DE212011100209U1 (en) 2011-09-13 2014-04-22 U.S. Nutraceuticals Llc Dba Valensa International Plant-derived seed extract rich in essential fatty acids derived from perilla seeds
US10683293B2 (en) 2014-08-04 2020-06-16 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US10689383B2 (en) 2014-08-04 2020-06-23 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US11254681B2 (en) 2014-08-04 2022-02-22 Nuevolution A/S Optionally fused heterocyclyl-substituted derivatives of pyrimidine useful for the treatment of inflammatory, metabolic, oncologic and autoimmune diseases
US11447479B2 (en) 2019-12-20 2022-09-20 Nuevolution A/S Compounds active towards nuclear receptors
US11613532B2 (en) 2020-03-31 2023-03-28 Nuevolution A/S Compounds active towards nuclear receptors
US11780843B2 (en) 2020-03-31 2023-10-10 Nuevolution A/S Compounds active towards nuclear receptors

Also Published As

Publication number Publication date
TW200730506A (en) 2007-08-16
US20110144331A1 (en) 2011-06-16
US20070142382A1 (en) 2007-06-21
US7465795B2 (en) 2008-12-16

Similar Documents

Publication Publication Date Title
US7425556B2 (en) Compounds and uses thereof
US7465795B2 (en) Compounds and uses thereof
TWI412525B (en) Quinoline amide m1 receptor positive allosteric modulators
US8895580B2 (en) Quinolinone-pyrazolone M1 receptor positive allosteric modulators
US20080318943A1 (en) Compounds and Uses Thereof - 848
JP5658255B2 (en) Pyranylarylmethylbenzoquinazolinone M1 receptor positive allosteric modulator
CA2743562A1 (en) Aryl methyl benzoquinazolinone m1 receptor positive allosteric modulators
AU2010216263A1 (en) Pyrazolo [4,3-c] cinnolin-3-one M1 receptor positive allosteric modulators
US20100184738A1 (en) Uses of cinnoline compounds to treat schizophrenia
AU2014282977B2 (en) 1-sulfonyl piperidine derivatives as modulators of prokineticin receptors
CN101379042A (en) Substituted cinnoline derivatives as GABAA-receptor modulators and method for their synthesis

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION