WO2008046082A2 - Use of heterocyclic compounds as therapeutic agents - Google Patents

Use of heterocyclic compounds as therapeutic agents Download PDF

Info

Publication number
WO2008046082A2
WO2008046082A2 PCT/US2007/081318 US2007081318W WO2008046082A2 WO 2008046082 A2 WO2008046082 A2 WO 2008046082A2 US 2007081318 W US2007081318 W US 2007081318W WO 2008046082 A2 WO2008046082 A2 WO 2008046082A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
heteroaryl
heterocyclyl
aryl
aralkyl
Prior art date
Application number
PCT/US2007/081318
Other languages
French (fr)
Other versions
WO2008046082A3 (en
Inventor
Mikhail Chafeev
Sultan Chowdhury
Robert Fraser
Jianmin Fu
Rajender Kamboj
Shifeng Liu
Vandna Raina
Mehran Seid Bagherzadeh
Jianyu Sun
Serguei Sviridov
Original Assignee
Xenon Pharmaceuticals Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xenon Pharmaceuticals Inc. filed Critical Xenon Pharmaceuticals Inc.
Publication of WO2008046082A2 publication Critical patent/WO2008046082A2/en
Publication of WO2008046082A3 publication Critical patent/WO2008046082A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems

Definitions

  • the present invention is directed to methods of using certain heterocyclic compounds as therapeutic agents.
  • this invention is directed to the use of certain heterocyclic compounds in treating diseases or conditions such as hypercholesterolemia, benign prostatic hyperplasia, pruritis and cancer.
  • Voltage-gated sodium channels transmembrane proteins that initiate action potentials in nerve, muscle and other electrically excitable cells, are a necessary component of normal sensation, emotions, thoughts and movements (Catterall, W.A., Nature (2001 ), Vol. 409, pp. 988-990).
  • These channels consist of a highly processed alpha subunit that is associated with auxiliary beta subunits.
  • the pore-forming alpha subunit is sufficient for channel function, but the kinetics and voltage dependence of channel gating are in part modified by the beta subunits (Goldin et al., Neuron (2000), Vol. 28, pp. 365-368).
  • Each alpha-subunit contains four homologous domains, I to IV, each with six predicted transmembrane segments.
  • the alpha-subunit of the sodium channel forming the ion-conducting pore and containing the voltage sensors regulating sodium ion conduction has a relative molecular mass of 260,000. Electrophysiological recording, biochemical purification, and molecular cloning have identified ten different sodium channel alpha subunits and four beta subunits (Yu, F.H., et al., Sci. STKE (2004), 253; and Yu, F.H., et al., Neurosci. (2003), 20:7577-85).
  • sodium channels include rapid activation and inactivation when the voltage across the plasma membrane of an excitable cell is depolarized (voltage-dependent gating), and efficient and selective conduction of sodium ions through conducting pores intrinsic to the structure of the protein (Sato, C, et al., Nature (2001 ), 409:1047-1051 ).
  • sodium channels are closed. Following membrane depolarization, sodium channels open rapidly and then inactivate. Channels only conduct currents in the open state and, once inactivated, have to return to the resting state, favoured by membrane hyperpolarization, before they can reopen.
  • Different sodium channel subtypes vary in the voltage range over which they activate and inactivate as well as their activation and inactivation kinetics.
  • Na/1.1 and Na v 1.2 are highly expressed in the brain (Raymond, C. K., et al., J. Biol. Chem. (2004), 279(44):46234-41 ) and are vital to normal brain function. In humans, mutations in Na/1.1 and Na/I .2 result in severe epileptic states and in some cases mental decline (Rhodes, T.H., et al., Proc. Natl.
  • Na/1.3 is broadly expressed throughout the body (Raymond, C. K., et al., op. cit). It has been demonstrated to have its expression upregulated in the dorsal horn sensory neurons of rats after nervous system injury (Hains, B. D., et al., J. Neurosci. (2003), 23(26):8881-92). Many experts in the field have considered Na/I .3 as a suitable target for pain therapeutics (Lai, J., et al., Curr. Opin. Neurobiol. (2003), (3):291 -72003; Wood, J. N., et al., J. Neurobiol.
  • Na/I .4 expression is essentially limited to muscle (Raymond, C.K., et al., op. cit.). Mutations in this gene have been shown to have profound effects on muscle function including paralysis, (Tamaoka A., Intern. Med. (2003), (9):769-70). Thus, this channel can be considered a target for the treatment of abnormal muscle contractility, spasm or paralysis.
  • the cardiac sodium channel, Na/1.5 is expressed mainly in the heart ventricles and atria (Raymond, C.
  • Na/1.5 is central to the genesis of cardiac arrhythmias. Mutations in human Na v 1.5 result in multiple arrhythmic syndromes, including, for example, long QT3 (LQT3), Brugada syndrome (BS), an inherited cardiac conduction defect, sudden unexpected nocturnal death syndrome (SUNDS) and sudden infant death syndrome (SIDS) (Liu, H. et al., Am. J. Pharmacogenomics (2003), 3(3):173-9).
  • Sodium channel blocker therapy has been used extensively in treating cardiac arrhythmias. The first antiarrhythmic drug, quinidine, discovered in 1914, is classified as a sodium channel blocker.
  • Na 1 Zl .6 encodes an abundant, widely distributed voltage-gated sodium channel found throughout the central and peripheral nervous systems, clustered in the nodes of Ranvier of neural axons (Caldwell, J. H., et al., Proc. Natl. Acad. ScL USA (2000), 97(10): 5616-20). Although no mutations in humans have been detected, Nayi .6 is thought to play a role in the manifestation of the symptoms associated with multiple sclerosis and has been considered as a target for the treatment of this disease (Craner, M.J., et al., Proc. Natl. Acad. ScL USA (2004), 101(21):8168-73).
  • Nav1.7 was first cloned from the pheochromocytoma PC12 cell line (Toledo- Aral, J. J., et al., Proc. Natl.Acad. ScL USA (1997), 94:1527-1532). Its presence at high levels in the growth cones of small-diameter neurons suggested that it could play a role in the transmission of nociceptive information. Although this has been challenged by experts in the field as Na v 1.7 is also expressed in neuroendocrine cells associated with the autonomic system (Klugbauer, N., et al., EMBO J. (1995), 14(6): 1084-90) and as such has been implicated in autonomic processes.
  • Na ⁇ I .7 blockers active in a subset of neurons Further support for Na ⁇ I .7 blockers active in a subset of neurons is supported by the finding that two human heritable pain conditions, primary erythermalgia and familial rectal pain, have been shown to map to Na v 1.7 (Yang, Y., et al., J. Med. Genet. (2004), 41 (3): 171 -4).
  • the expression of Na v 1.8 is essentially restricted to the DRG (Raymond, C. K., et al., op. cit.).
  • Na/I .8- null mutant mice were viable, fertile and normal in appearance.
  • WO03/037274A2 describes pyrazole- amides and sulfonamides for the treatment of central or peripheral nervous system conditions, particularly pain and chronic pain by blocking sodium channels associated with the onset or recurrance of the indicated conditions.
  • PCT Published Patent Application No. WO03/037890A2 describes piperidines for the treatment of central or peripheral nervous system conditions, particularly pain and chronic pain by blocking sodium channels associated with the onset or recurrence of the indicated conditions.
  • the compounds, compositions and methods of these inventions are of particular use for treating neuropathic or inflammatory pain by the inhibition of ion flux through a channel that includes a PN3 (Na v 1.8) subunit.
  • Na v 1.9 underlies neurotrophin (BDNF)-evoked depolarization and excitation, and is the only member of the voltage gated sodium channel superfamily to be shown to be ligand mediated (Blum, R., Kafitz, K.W., Konnerth, A., Nature (2002), 419 (6908):687-93).
  • BDNF neurotrophin
  • the limited pattern of expression of this channel has made it a candidate target for the treatment of pain (Lai, J, et al., op. cit.; Wood, J. N., et al., op. cit; Chung, J. M. et al., op. cit).
  • NaX is a putative sodium channel, which has not been shown to be voltage gated.
  • NaX is found in neurons and ependymal cells in restricted areas of the CNS, particularly in the circumventricular organs, which are involved in body-fluid homeostasis (Watanabe, E., et al., J. Neurosci. (2000), 20(20):7743-51 ).
  • NaX-null mice showed abnormal intakes of hypertonic saline under both water- and salt-depleted conditions.
  • TTX sodium channel blocker tetrodotoxin
  • Sodium channels are targeted by a diverse array of pharmacological agents. These include neurotoxins, antiarrhythmics, anticonvulsants and local anesthetics (Clare, J.J., et al., Drug Discovery Today (2000) 5:506-520). All of the current pharmacological agents that act on sodium channels have receptor sites on the alpha subunits. At least six distinct receptor sites for neurotoxins and one receptor site for local anesthetics and related drugs have been identified (Cestele, S. et al., Biochimie (2000), Vol. 82, pp. 883-892).
  • the small molecule sodium channel blockers or the local anesthetics and related antiepileptic and antiarrhythmic drugs interact with overlapping receptor sites located in the inner cavity of the pore of the sodium channel (Catterall, W.A., Neuron (2000), 26:13-25). Amino acid residues in the S6 segments from at least three of the four domains contribute to this complex drug receptor site, with the IVS6 segment playing the dominant role. These regions are highly conserved and as such most sodium channel blockers known to date interact with similar potency with all channel subtypes. Nevertheless, it has been possible to produce sodium channel blockers with therapeutic selectivity and a sufficient therapeutic window for the treatment of epilepsy (e.g.
  • the present invention is directed to the use of certain heterocyclic compounds for the treatment and/or prevention of diseases or conditions, such as hypercholesterolemia, benign prostatic hyperplasia, pruritis, and cancer.
  • the invention provides compounds of formula (I):
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 9 -C(O)R 6 , -R 9 -C(O)OR 6 , -R 9 -C(O)N(R 5 )R 6 , -R 9 -OR 6 , -R 9 -CN, -R 10 -P(O)(OR 6 ) 2 or -R 10 -O-R 10 -OR 6 ; or R 1 is aralkyl substituted by -C(O)N(R 7 )R 8 where: R 7 is hydrogen, alkyl, aryl or aralkyl; and
  • R 8 is hydrogen, alkyl, haloalkyl, -R 10 -CN, -R 10 -OR 6 , -R 10 -N(R 5 )R 6 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 7 and R 8 , together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R 7 and R 8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl,
  • R 1 is aralkyl substituted by one or more substituents selected from the group consisting of -R 9 -OR 6 , -R 9 -C(O)OR 6 , halo, haloalkyl, alkyl, nitro, cyano, aryl (optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R 1 is -R 10 -N(R 11 )R 12 , -R 10 -N(R 13 )C(O)R 12 or -R 10 -N(R 11 )C(O)N(R 11 )R 12 where: each R 11 is hydrogen, alkyl, aryl or aralkyl; each R 12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkylalkylal
  • R 13 is hydrogen, alkyl, aryl, arakyl or -C(O)R 6 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 11 and R 12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 9 -CN, -R 9 -OR 6 , -R 9 -C(O)R 6 , heterocyclyl and heteroaryl; or R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R
  • each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl,
  • the invention provides methods of treating or preventing hypercholesterolemia in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
  • the invention provides methods of treating or preventing benign prostatic hyperplasia in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
  • the invention provides methods of treating or preventing pruritis in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
  • the invention provides methods of treating or preventing cancer in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
  • the invention provides pharmaceutical compositions comprising the compounds of the invention, as set forth above, and pharmaceutically acceptable excipients.
  • the invention provides pharmaceutical therapy in combination with one or more other compounds of the invention or one or more other accepted therapies or as any combination thereof to increase the potency of an existing or future drug therapy or to decrease the adverse events associated with the accepted therapy.
  • the present invention relates to a pharmaceutical composition combining compounds of the present invention with established or future therapies for the indications listed in the invention.
  • this invention is directed to the use of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or the use of a pharmaceutical composition of the invention, comprising a pharmaceutically acceptable excipient and a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in the preparation of a medicament for the treatment and/or prevention of hypercholesterolemia, benign prostatic hyperplasia, pruritis, and/or cancer in a mammal.
  • C 7 -C 12 alkyl describes an alkyl group, as defined below, having a total of 7 to 12 carbon atoms
  • C 4 -Ci 2 cycloalkylalkyl describes a cycloalkylalkyl group, as defined below, having a total of 4 to 12 carbon atoms.
  • the total number of carbons in the shorthand notation does not include carbons that may exist in substituents of the group described. For example, the following terms have the meaning indicated:
  • Ci-Ci O alkyl refers to an alkyl radical as defined below containing one to ten carbon atoms.
  • the C r C 10 alkyl readical may be optionally substituted as defined below for an alkyl group.
  • C 2 -C 12 alkynyl refers to an alknyl radical as defined below containing two to twelve carbon atoms.
  • the C 2 -C 12 alknyl radical may be optionally substituted as defined below for an alkenyl group.
  • CrC ⁇ alkoxy refers to an alkoxy radical as defined below containing one to twelve carbon atoms.
  • the alkyl part of the d-C 12 alkoxy radical may be optionally substituted as defined below for an alkyl group.
  • C 2 -Ci 2 alkoxyalkyl refers to an alkoxyalkyl radical as defined below containing two to twelve carbon atoms. Each alkyl part of the C 2 -C 12 alkoxyalkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 7 -C 12 aralkyl refers to an aralkyl group as defined below containing seven to twelve carbon atoms. The aryl part of the C 7 -C 12 aralkyl radical may be optionally substituted as described below for an aryl group. The alkyl part of the C 7 -C 12 aralkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 7 -C 12 aralkenyl refers to an aralkenyl group as defined below containing seven to twelve carbon atoms.
  • the aryl part of the C 7 -C 12 aralkenyl radical may be optionally substituted as described below for an aryl group.
  • the alkenyl part of the C 7 -C 12 aralkenyl radical may be optionally substituted as defined below for an alkenyl group.
  • C 3 -C 12 cycloalkyl refers to a cycloalkyl radical as defined below having three to twelve carbon atoms.
  • the C 3 -C 12 cycloalkyl radical may be optionally substituted as defined below for a cycloalkyl group.
  • C 4 -C 12 cycloalkylalkyl refers to a cycloalkylalkyl radical as defined below having four to twelve carbon atoms.
  • the C 4 -C 12 cycloalkylalkyl radical may be optionally substituted as defined below for a cycloalkylalkyl group.
  • Amino refers to the -NH 2 radical.
  • Haldroxyl refers to the -OH radical.
  • Niro refers to the -NO 2 radical.
  • Trifluoromethyl refers to the -CF 3 radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, preferably one to eight carbon atoms or one to six carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (/so-propyl), n-butyl, n-pentyl, 1 ,1-dimethylethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, and the like.
  • an alkyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2 , -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2 , -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O),R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 4 R 16 (where t is O to 2), and -S(O) t N(R 14 ,
  • Alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1 ,4-dienyl, and the like.
  • an alkenyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2 , -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2 , -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O),R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 1 R 16 (where t is 0 to 2), and -S(O) t N(R
  • Alkylene or "alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • an alkylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2 , -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2) -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O) t R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 1 R 16 (where t is 0 to 2), and -S(O) t N(R 14
  • alkenylene or “alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like.
  • the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • the points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • an alkenylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2> -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2 , -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O) t R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 1 R 16 (where t is 0 to 2), and -S(O) t N(
  • Alkynylene or “alkynylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one triple bond and having from two to twelve carbon atoms, e.g., propynylene, n-butynylene, and the like.
  • the alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond.
  • the points of attachment of the alkynylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain.
  • an alkynylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2 , -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2 , -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O) t R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 1 R 16 (where t is O to 2), and -S(O) t N
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR 14 , -OC(O)-R 14 , -N(R 14 ) 2 , -C(O)R 14 , -C(O)OR 14 , -C(O)N(R 14 ) 2 , -N(R 14 )C(O)OR 16 , -N(R 14 )C(O)R 16 , -N(R 14 )S(O) t R 16 (where t is 1 to 2), -S(O) 1 OR 16 (where t is 1 to 2), -S(O) 1 OR 16 (
  • Alkoxy refers to a radical of the formula -OR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms.
  • R a is an alkyl radical as defined above containing one to twelve carbon atoms.
  • the alkyl part of the alkoxy radical may be optionally substituted as defined above for an alkyl radical.
  • Alkoxyalkyl refers to a radical of the formula -R 3 -O-R 3 where each R 3 is independently an alkyl radical as defined above.
  • the oxygen atom may be bonded to any carbon in either alkyl radical.
  • Each alkyl part of the alkoxyalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Aryl refers to aromatic monocyclic or multicyclic hydrocarbon ring system consisting only of hydrogen and carbon and containing from 6 to 18 carbon atoms, where the ring system may be partially saturated.
  • Aryl groups include, but are not limited to, groups such as fluorenyl, phenyl and naphthyl.
  • aryl or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals optionally substituted by one or more substituents independently selected from the group consisting of alkyl, akenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, heteroaryl, heteroarylalkyl, -R 15 -OR 14 , -R 15 -OC(O)-R 14 , -R 15 -N(R 14 ) 2 , -R 15 -C(O)R 14 , -R 15 -C(O)OR 14 , -R 15 -C(O)N(R 14 ) 2 , -R 15 -N(R 14 )C(O)OR 16 , -R 15 -N(R 14 )C(O)R 16 , -R 15 -N(R 14 )C(O)R 16 , -R 15 -N
  • Aralkyl refers to a radical of the formula -R a R b where R 3 is an alkyl radical as defined above and R b is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like.
  • the aryl radical(s) may be optionally substituted as described above.
  • Aryloxy refers to a radical of the formula -OR b where R b is an aryl group as defined above.
  • the aryl part of the aryloxy radical may be optionally substituted as defined above.
  • Aralkenyl refers to a radical of the formula -R 0 R b where R c is an alkenyl radical as defined above and R b is one or more aryl radicals as defined above, which may be optionally substituted as described above.
  • the aryl part of the aralkenyl radical may be optionally substituted as described above for an aryl group.
  • the alkenyl part of the aralkenyl radical may be optionally substituted as defined above for an alkenyl group.
  • Aralkyloxy refers to a radical of the formula -OR b where R b is an aralkyl group as defined above.
  • the aralkyl part of the aralkyloxy radical may be optionally substituted as defined above.
  • Cycloalkyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond.
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptly, and cyclooctyl.
  • Polycyclic radicals include, for example, adamantyl, norbomyl, decalinyl, and the like.
  • cycloalkyl is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, oxo, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 15 -OR 14 , -R 15 -OC(O)-R 14 , -R 15 -N(R 14 ) 2 , -R 15 -C(O)R 14 , -R 15 -C(O)OR 14 , -R 15 -C(O)N(R 14 ) 2 , -R 15 -N(R 14 )C(O)OR 16 , -R 15 -
  • Cycloalkylalkyl refers to a radical of the formula -R a Ro where R 3 is an alkyl radical as defined above and R d is a cycloalkyl radical as defined above.
  • the alkyl radical and the cycloalkyl radical may be optionally substituted as defined above.
  • Halo refers to bromo, chloro, fluoro or iodo.
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like.
  • the alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Heterocyclyl refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated.
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1 ,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and
  • heterocyclyl is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 15 -OR 14 , -R 15 -OC(O)-R 14 , -R 15 -N(R 14 ) 2 , -R 15 -C(O)R 14 , -R 15 -C(O)OR 14 , -R 15 -C(O)N(R 14 ) 2 , -R 15 -N(R 14 )C(O)OR 16 , -R 15 -C(O)N(R
  • Heterocyclylalkyl refers to a radical of the formula -R a R e where R a is an alkyl radical as defined above and R e is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom.
  • the alkyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for an alkyl group.
  • the heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
  • Heteroaryl refers to a 5- to 18-membered aromatic ring radical which consists of one to seventeen carbon atoms and from one to ten heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized.
  • Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[jb][1 ,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1 ,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothi
  • heteroaryl is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkoxy, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, oxo, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 15 -OR 14 , -R 15 -OC(O)-R 14 , -R 15 -N(R 14 ) 2 , -R 15 -C(O)R 14 , -R 15 -C(O)OR 14 , -R 15 -C(O)N(R 14 ) 2 , -R 15 -R 15 -
  • Heteroarylalkyl refers to a radical of the formula -R a R f where R 3 is an alkyl radical as defined above and R f is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkyl part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Heteroarylalkenyl refers to a radical of the formula -R b R f where Rb is an alkenyl radical as defined above and R f is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkenyl radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkenyl part of the heteroarylalkenyl radical may be optionally substituted as defined above for an alkenyl group.
  • Trihaloalkyl refers to an alkyl radical, as defined above, that is substituted by three halo radicals, as defined above, e.g., trifluoromethyl.
  • the alkyl part of the trihaloalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Trihaloalkoxy refers to a radical of the formula -OR 9 where R 9 is a trihaloalkyl group as defined above.
  • the trihaloalkyl part of the trihaloalkoxy group may be optionally substituted as defined above for a trihaloalkyl group.
  • Prodrugs is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention.
  • prodrug refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)).
  • a discussion of prodrugs is provided in Higuchi, T., et a/., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, Ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
  • prodrug is also meant to include any covalently bonded carriers, which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention.
  • Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like.
  • the invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of formula (I) being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 O, 31 P, 32 P, 35 S, 18 F, 36 CI, 123 I, and 125 I, respectively.
  • radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action on the sodium channels, or binding affinity to pharmacologically important site of action on the sodium channels.
  • Certain isotopically-labelled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples and Preparations as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • the invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reducation, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically are identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its coversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to human
  • “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.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets, ( e.g. cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildelife and the like.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1 ,2-disulfonic
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, ⁇ /-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are is
  • solvate refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • the compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
  • a “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
  • “Therapeutically effective amount” refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a disease or condition in the mammal, preferably a human.
  • a compound of the invention which constitutes a "therapeutically effective amount” will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • Treating covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:
  • the terms “disease” and “condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
  • the compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centres and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), [R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallisation.
  • stereoisomer refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • the present invention includes tautomers of any said compounds.
  • intermediate compounds of formula (I) and all polymorphs of the aforementioned species and crystal habits thereof are also within the scope of the invention.
  • the chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program, wherein the compounds of the invention are named herein as derivatives of the central core structure, i.e., the 2-oxindole structure.
  • a substituent group is named before the group to which it attaches.
  • cyclopropylethyl comprises an ethyl backbone with cyclopropyl substituent.
  • all bonds are identified, except for some carbon atoms, which are assumed to be bonded to sufficient hydrogen atoms to complete the valency.
  • One embodiment is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is -R 9 -C(O)R 6 , -R 9 -C(O)OR 6 , -R 9 -OR 6 , -R 9 -CN, -R 10 -P(O)(OR 6 ) 2 , -R 10 -O-R 10 -OR 6 , hydrogen, alkyl, haloalkyl, cycloalkylalkyl, heterocyclylalkyl, aryl (optionally substituted by one or more substituents selected from the group consisting of halo and -R 9 -C(O)OR 6 ), aralkyl
  • R 3 is independently selected from the group consisting of hydrogen, halo, haloalkyl,
  • R 4 is independently selected from the group consisting of alkyl, aryl, aralkyl, aralkynyl, heteroaryl, heteroarylalkyl, -R 9 -C(O)R 5 , -N(R 6 )C(O)N(R 5 )R 6 , -R 9 -NO 2 , -R 9 -N(R 5 )R 6 , -R 9 -C(O)OR 6 , -R 9 -N(R 6 )C(O)OR 6 and -Si(R 6 ) 3 , wherein each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R 4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, ary
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R 10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl, aryl or aralkyl, where each of the aryl or aralkyl group for R 1 is optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ; each R 2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R 9 -OR 6 , wherein each of the aryl and heteroaryl groups for R 2 is optionally substituted by one or more substituents
  • R 3 is hydrogen, halo, -R 9 -OR 6 or -R 9 -OC(O)R 6 ;
  • R 4 is independently selected from the group consisting of alkyl, aryl, aralkynyl, heteroaryl, heteroarylalkyl, -R 9 -C(O)R 5 , -N(R 6 )C(O)N(R 5 )R 6 , -R 9 -NO 2 ,
  • each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R 4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloaikyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R 9 -CN, -R 9 -NO 2 , -R 9 -OR 6 , -R 9 -N(R 5 )R 6 , -S(O) m R 5 , -R 9 -C(O)
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl, aryl or aralkyl, where each of the aryl or aralkyl group for R 1 is optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ; each R 2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R 9 -OR 6 , wherein each of the aryl and heteroaryl groups for R 2 is optionally substituted by one or more substituent
  • R 3 is hydrogen, halo, -R 9 -OR 6 or -R 9 -OC(O)R 6 ;
  • R 4 is -R 9 -C(O)R 5 ;
  • each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkeny
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-hydroxy-3-[2-oxo-2-(2-thienyl)ethyl]-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-b]pyridin-2- one; and 3-[2-(2-furyl)-2-oxoethyl]-3-hydroxy-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3-/3]pyridin-2- one.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
  • — J is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
  • R 1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ); each R 2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R 9 -OR 6 , wherein each of the aryl and heteroaryl groups for R 2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -CN, -R 9 -NO 2 , -R 9 -
  • R 3 is hydrogen, halo, -R 9 -OR 6 or -R 9 -OC(O)R 6 ;
  • R 4 is heterocyclylalkyl, heteroaryl or heteroarylalkyl, each optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -CN,
  • each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ); each R 2 is each independently selected from the group consisting of alkyl, halo, phenyl, benzodioxolyl and -R 9 -OR 6 , R 3 is hydrogen, halo, -R 9 -OR 6 or -R 9 -OC(O)R 6 ;
  • R 4 is heterocyclylalkyl, heteroaryl or heteroarylalkyl, each optionally substituted by one or more substituents selected from the group consisting of halo, heterocyclyl, and -R 9 -OR 6 ; each R 6 is independently selected from group consisting of hydrogen, alky!, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
  • R 1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ); R 3 is -R 9 -OR 6 ;
  • R 4 is aryl, aralkyl or aralkynyl, wherein each of the aryl, aralkyl and aralkynyl groups for R 4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R 9 -CN, -R 9 -NO 2 , -R 9 -OR 6 , -R 9 -N(R 5 )R 6 , -S(O) m R 5 , -R 9 -C(O)R 5 , -R 9 -C(O)OR 6 , -R 9 -C
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
  • R 1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R 9 -OR 6 and -R 9 -C(O)OR 6 ); R 3 is -R 9 -OR 6 ;
  • R 4 is aryl, aralkyl or aralkynyl, wherein each of the aryl, aralkyl and aralkynyl groups for R 4 is optionally substituted by one or more substituents selected from the group consisting of halo, oxo and -R 9 -OR 6 ; each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R 9 is
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is hydrogen, alkyl, haloalkyl or cycloalkylalkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OR 6 , -R 9 -N(R 5 )R 6 , -R 9 -C(O)R 5 , -R 9 -
  • each n is independently 1 or 2; or two adjacent R 2 groups, together with the heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R 2 groups, if present, are as described above;
  • R 3 is hydrogen, halo or -R 9 -OR 6 ;
  • R 4 is independently selected from the group consisting of alkyl, aryl, aralkynyl, heteroaryl, heteroarylalkyl, -R 9 -C(O)R 5 , -R 9 -N(R 6 )C(O)OR 6 , -N(R 6 )C(O)N(R 5 )R 6 , -R 9 -NO 2 , -R 9 -N(R 5 )R 6 , -R 9 -C(O)OR 6 , and -Si(R 6 ) 3 , wherein each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R 4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, a
  • each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which they are attached, may form a
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl
  • R 1 is hydrogen, alkyl, haloalkyl or cycloalkylalkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OR 6 , -R 9 -N(R 5 )R 6 , -R 9 -C(O)R 5 , -R 9 -C(O)OR 6 , -R 9 -C(O)N(R 5 )R 6 , -N(R 6 )C(O)R 5 , wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl
  • R 4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R 9 -CN, -R 9 -NO 2 , -R 9 -OR 6 , -R 9 -N(R 5 )R 6 ,
  • each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R 9 -OR 6 ;
  • R 3 is hydrogen or -R 9 -OR 6 ;
  • R 4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of halo, -R 9 -OR 6 and -N(R 6 )C(O)R 5 ; each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkeny
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R 9 -OR 6 ;
  • R 3 is hydrogen or -R 9 -OR 6 ;
  • R 4 is benzodioxolyl optionally substituted by one or more substituents selected from the group consisting of halo and -R 9 -OR 6 ;
  • each R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl;
  • each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1H-pyrrolo[1 ,2-b]pyrazol-2(3H)-one;
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R 9 -OR 6 ;
  • R 3 is hydrogen, halo or -R 9 -OR 6 ;
  • R 4 is independently selected from the group consisting of -R 9 -C(O)R 5 and -R 9 -N(R 6 )C(O)OR 6 ; each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R 9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
  • R 1 is alkyl or aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, -R 9 -OR 6 , heteroaryl and -R 9 -C(O)OR 6 );
  • R 3 is -R 9 -C(O)X, -R 9 -C(O)OR 6 and -R 9 -C(O)N(R 5 )R 6 where X is bromo or chloro;
  • R 4 is independently selected from the group consisting of -R 9 -C(O)R 5 and heteroaryl optionally substituted by one or more substituents selected from the group consisting of halo and R 9 -OR 6 ; each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1, 2, 3 or 4; is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl or aralkyl optionally substituted by one or more substituents selected from the group consisting of halo and -R 9 -C(O)OR 6 ; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OR 6 , -R 9 -N(R 5 )R 6
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4; a fused heteroaryl ring selected from the group consisting of pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R 1 is alkyl; each R 2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R 9 -OR 6 ;
  • R 3 is independently selected from the group consisting of halo, -R 9 -CN, -R 9 -N(R 5 )R 6 and -N(R 6 )C(O)OR 6 ;
  • R 4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, and -R 9 -OR 6 ;
  • each R 5 and R 6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R 5 and R 6 are each attached to the same nitrogen atom, then R 5 and R 6 , together with the nitrogen atom to which they are attached, may form a
  • Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-(1 ,3-benzodioxol-5-yl)-3-fluoro-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3- ⁇ ]pyridin-2-one; 3-(1 ,3-benzodioxol-5-yl)-2-oxo-1-pentyl-2,3-dihydro-1H-pyrrolo[2,3- ⁇ ]pyridine-3- carbonitrile; and
  • One embodiment of the invention is the method of treating or preventing hypercholesterolemia in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
  • Another embodiment of the invention is the method of treating or preventing benign prostatic hyperplasia in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
  • Another embodiment of the invention is the method of treating or preventing pruritis in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
  • Another embodiment of the invention is the method of treating or preventing cancer in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
  • the compounds of the invention modulate, preferably inhibit, ion flux through a voltage-dependent sodium channel in a mammal, especially in a human. Any such modulation, whether it be partial or complete inhibition or prevention of ion flux, is sometimes referred to herein as “blocking” and corresponding compounds as “blockers”.
  • the compounds of the invention modulates the activity of a sodium channel downwards, inhibits the voltage-dependent activity of the sodium channel, and/or reduces or prevents sodium ion flux across a cell membrane by preventing sodium channel activity such as ion flux.
  • the compounds of the invention inhibit the ion flux through a voltage- dependent sodium channel.
  • the compounds are state or frequency dependent modifers of the sodium channels, having a low affinity for the rested/closed state and a high affinity for the inactivated state. These compounds are likely to interact with overlapping sites located in the inner cavity of the sodium conducting pore of the channel similar to that described for other state-dependent sodium channel blockers (Cestele, S., et al., op. cit). These compounds may also be likely to interact with sites outside of the inner cavity and have allosteric effects on sodium ion conduction through the channel pore.
  • the compounds and pharmaceutical compositions of the invention are useful in the treatment and/or prevention of benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and/or pruritis (itch) in a mammal, preferably a human.
  • BPH benign prostatic hyperplasia
  • itch pruritis
  • Benign prostatic hyperplasia also known as benign prostatic hypertrophy, is one of the most common diseases affecting aging men.
  • BPH is a progressive condition which is characterized by a nodular enlargement of prostatic tissue resulting in obstruction of the urethra. Consequences of BPH can include hypertrophy of bladder smooth muscle, a decompensated bladder, acute urinary retention and an increased incidence of urinary tract infection.
  • BPH has a high public health impact and is one of the most common reasons for surgical intervention among elderly men. Attempts have been made to clarify the etiology and pathogenesis and, to that end, experimental models have been developed. Spontaneous animal models are limited to the chimpanzee and the dog. BPH in man and the dog share many common features. In both species, the development of BPH occurs spontaneously with advanced age and can be prevented by early/prepubertal castration. A medical alternative to surgery is very desirable for treating BHP and the consequences.
  • prostatic epithelial hyperplasia in both man and the dog is androgen sensitive, undergoing involution with androgen deprivation and resuming epithelial hyperplasia when androgen is replaced.
  • Cells originating from the prostate gland have been shown to express high levels of voltage gated sodium channels, lmmunostaining studies clearly demonstrated evidence for voltage gated sodium channels in prostatic tissues (Prostate Cancer Prostatic Dis. 2005; 8(3):266-73).
  • Hypercholesterolemia i.e., elevated blood cholesterol
  • CVD cardiovascular diseases
  • lowering the levels of total serum cholesterol in individuals with high levels of cholesterol has been known to reduce the risk of these diseases.
  • the lowering of low density lipoprotein cholesterol in particular is an essential step in the prevention of CVD.
  • hypercholesterolemia therapies Although there are a variety of hypercholesterolemia therapies, there is a continuing need and a continuing search in this field of art for alternative therapies.
  • the invention provides compounds which are useful as antihypercholesterolemia agents and their related conditions.
  • the present compounds may act in a variety of ways. While not wishing to be bound to any particular mechanism of action, the compounds may be direct or indirect inhibitors of the enzyme acyl CoA: cholesterol acyl transferase (ACAT) that results in inhibition of the esterification and transport of cholesterol across the intestinal wall. Another possibility may be that the compounds of the invention may be direct or indirect inhibitors of cholesterol biosynthesis in the liver. It is possible that some compounds of the invention may act as both direct or indirect inhibitors of ACAT and cholesterol biosynthesis.
  • acyl CoA cholesterol acyl transferase
  • Pruritus commonly known as itch
  • itch is a common dermatological condition. While the exact causes of pruritis are complex and poorly understood, there has long been acknowledged to have interactions with pain. In particular, it is believed that sodium channels likely communicate or propagate along the nerve axon the itch signals along the skin. Transmission of the itch impulses results in the unpleasant sensation that elicits the desire or reflex to scratch.
  • the mildly painful stimuli from scratching are effective in abolishing the itch sensation.
  • analgesics such as opioids can generate severe pruritus.
  • the antagonistic interaction between pain and itch can be exploited in pruritus therapy, and current research concentrates on the identification of common targets for future analgesic and antipruritic therapy.
  • itch or skin irritation include, but are not limited to: a) psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritus, and itching caused by skin disorders (e.g., contact dermatitis), systemic disorders, neuropathy, psychogenic factors or a mixture thereof; b) itch caused by allergic reactions, insect bites, hypersensitivity (e.g., dry skin, acne, eczema, psoriasis), inflammatory conditions or injury; c) itch associated with vulvar vestibulitis; and d) skin irritation or inflammatory effect from administration of another therapeutic such as, for example, antibiotics, antivirals and antihistamines.
  • another therapeutic such as, for example, antibiotics, antivirals and antihistamines.
  • the compounds of the invention are also useful in treating or preventing certain hormone sensitive cancers, such as prostate cancer (adenocarcinoma), breast cancer, ovarian cancer, testicular cancer, thyroid neoplasia.
  • hormone sensitive cancers such as prostate cancer (adenocarcinoma), breast cancer, ovarian cancer, testicular cancer, thyroid neoplasia.
  • the voltage gated sodium channels have been demonstrated to be expressed in prostate and breast cancer cells. Up-regulation of neonatal Na(v)1.5 occurs as an integral part of the metastatic process in human breast cancer and could serve both as a novel marker of the metastatic phenotype and a therapeutic target (Clin. Cancer Res.2005, Aug. 1 ; 11(15): 5381-9). Functional expression of voltage-gated sodium channel alpha-subunits, specifically Na/I .7, is associated with strong metastatic potential in prostate cancer (CaP) in vitro.
  • CaP prostate cancer
  • the compounds of the invention are also useful in treating or preventing symptoms associated with BPH such as, but not limited to, acute urinary retention and urinary tract infection.
  • the compounds of the invention are also useful in treating or preventing certain endocrine imbalances or endocrinopathies such as congenital adrenal hyperplasia , hyperthyroidism, hypothyroidism, osteoporosis, osteomalacia, rickets, Cushing's Syndrome, Conn's syndrome, hyperaldosteronism, hypogonadism, hypergonadism, infertility, fertility and diabetes.
  • certain endocrine imbalances or endocrinopathies such as congenital adrenal hyperplasia , hyperthyroidism, hypothyroidism, osteoporosis, osteomalacia, rickets, Cushing's Syndrome, Conn's syndrome, hyperaldosteronism, hypogonadism, hypergonadism, infertility, fertility and diabetes.
  • the present invention readily affords many different means for identification of therapeutic agents, especially as sodium channel modulating agents. Identification of the therapeutic agents can be assessed using a variety of in vitro and in vivo assays, e.g., measuring current, measuring membrane potential, measuring ion flux, (e.g. sodium or guanidinium), measuring sodium concentration, measuring second messengers and transcription levels, and using e.g., voltage-sensitive dyes, radioactive tracers, and patch-clamp electrophysiology.
  • in vitro and in vivo assays e.g., measuring current, measuring membrane potential, measuring ion flux, (e.g. sodium or guanidinium), measuring sodium concentration, measuring second messengers and transcription levels, and using e.g., voltage-sensitive dyes, radioactive tracers, and patch-clamp electrophysiology.
  • One such protocol involves the screening of chemical agents for ability to modulate the activity of a sodium channel thereby identifying it as a modulating agent.
  • a competitive binding assay with known sodium channel toxins such as tetrodotoxin, alpha-scorpion toxins, aconitine, BTX and the like, may be suitable for identifying potential therapeutic agents with high selectivity for a particular sodium channel.
  • the use of BTX in such a binding assay is well known and is described in McNeal, E.T., et al., J. Med. Chem. (1985), 28(3):381-8; and Creveling, C.R., et al., Methods in Neuroscience, Vol.8: Neurotoxins (Conn PM Ed) (1992), pp. 25-37, Academic Press, New York.
  • the assays can be carried out in cells, or cell or tissue extracts expressing the channel of interest in a natural endogenous setting or in a recombinant setting.
  • the assays that can be used include plate assays which measure Na+ influx through surrogate markers such as 14 C-guanidine influx or determine cell depolarization using fluorescent dyes such as the FRET based and other fluorescent assays or a radiolabeled binding assay employing radiolabeled aconitine, BTX, TTX or STX. More direct measurements can be made with manual or automated electrophysiology systems.
  • the guanidine influx assay is explained in more detail below in the Biological Assays section. Throughput of test compounds is an important consideration in the choice of screening assay to be used.
  • Electrophysiological assays using patch clamp techniques is accepted as a gold standard for detailed characterization of sodium channel compound interactions, and as described in Bean et al., op. cit. and Leuwer, M., et al., op. cit.
  • LTS manual low-throughput screening
  • MTS medium-throughput screening
  • HTS high-throughput screening
  • Planar electrodes are capable of achieving high- resistance, cells-attached seals followed by stable, low-noise whole-cell recordings that are comparable to conventional recordings.
  • a suitable instrument is the PatchXpress
  • HEK and CHO stably expressing high levels of the relevant sodium ion channel can be adapted into high-density suspension cultures.
  • assays can be selected which allow the investigator to identify compounds which block specific states of the sodium channel, such as the open state, closed state or the resting state, or which block transition from open to closed, closed to resting or resting to open. Those skilled in the art are generally familiar with such assays.
  • Binding assays are also available, however these are of only limited functional value and information content. Designs include traditional radioactive filter based binding assays or the confocal based fluorescent system available from Evotec OAI group of companies (Hamburg, Germany), both of which are HTS.
  • Radioactive flux assays can also be used.
  • channels are stimulated to open with veratridine or aconitine and held in a stabilized open state with a toxin, and channel blockers are identified by their ability to prevent ion influx.
  • the assay can use radioactive 22 [Na] and 14 [C] guanidinium ions as tracers. FlashPlate &
  • Cytostar-T plates in living cells avoids separation steps and are suitable for HTS.
  • Scintillation plate technology has also advanced this method to HTS suitability.
  • HTS FLIPR system membrane potential kit
  • Molecular Dynamics a division of Amersham Biosciences, Piscataway, NJ. This method is limited to slow membrane potential changes. Some problems may result from the fluorescent background of compounds. Test compounds may also directly influence the fluidity of the cell membrane and lead to an increase in intracellular dye concentrations. Still, because of the functional aspects of the assay, the information content is reasonably good.
  • Sodium dyes can be used to measure the rate or amount of sodium ion influx through a channel. This type of assay provides a very high information content regarding potential channel blockers. The assay is functional and would measure Na+ influx directly. CoroNa Red, SBFI and/or sodium green (Molecular Probes, Inc. Eugene OR) can be used to measure Na influx; all are Na responsive dyes. They can be used in combination with the FLIPR instrument. The use of these dyes in a screen has not been previously described in the literature. Calcium dyes may also have potential in this format.
  • FRET based voltage sensors are used to measure the ability of a test compound to directly block Na influx.
  • HTS systems include the VIPRTM Il FRET system (Aurora Biosciences Corporation, San Diego, CA, a division of Vertex Pharmaceuticals, Inc.) which may be used in conjunction with FRET dyes, also available from Aurora Biosciences.
  • This assay measures sub-second responses to voltage changes. There is no requirement for a modifier of channel function.
  • the assay measures depolarization and hyperpolarizations, and provides ratiometric outputs for quantification.
  • a somewhat less expensive MTS version of this assay employs the FLEXstationTM (Molecular Devices Corporation) in conjunction with FRET dyes from Aurora Biosciences. Other methods of testing the compounds disclosed herein are also readily known and available to those skilled in the art.
  • SAR structure-activity relationship
  • Modulating agents so identified are then tested in a variety of in vivo models so as to determine if they alleviate the diseases or conditions, especially benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and pruritis (itch), with minimal adverse events.
  • BPH benign prostatic hyperplasia
  • itch hypercholesterolemia
  • itch pruritis
  • the assays described below in the Biological Assays Section are useful in assessing the biological activity of the instant compounds.
  • a successful therapeutic agent of the present invention will meet some or all of the following criteria.
  • Oral availability should be at or above 20%.
  • Animal model efficacy is less than about 0.1 ⁇ g to about 100 mg/Kg body weight and the target human dose is between 0.1 ⁇ g to about 100 mg/Kg body weight, although doses outside of this range may be acceptable ("mg/Kg” means milligrams of compound per kilogram of body mass of the subject to whom it is being administered).
  • the therapeutic index or ratio of toxic dose to therapeutic dose
  • the potency (as expressed by IC 50 value) should be less than 10 ⁇ M, preferably below 1 ⁇ M and most preferably below 50 nM.
  • the IC 50 is a measure of the amount of compound required to achieve 50% inhibition of ion flux through a sodium channel, over a specific time period, in an assay of the invention.
  • Compounds of the present invention in the guanidine influx assay have demonstrated IC 50 1 S ranging from less than a nanomolar to less than 10 micromolar.
  • the compounds of the invention can be used in in vitro or in vivo studies as exemplary agents for comparative purposes to find other compounds also useful in treatment of, or protection from, the various diseases disclosed herein.
  • Another aspect of the invention relates to inhibiting Na/1.1 , Na v 1.2, Nav1.3, Na v 1.4, Na v 1.5, Na v 1.6, Na v 1.7, Na/I .8, or Na v 1.9 activity in a biological sample or a patient, which method comprises administering to the patient, or contacting said biological sample with a compound of formula I or a composition comprising said compound.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of Na/1.1 , Na/I .2, Na v 1.3, Na v 1.4, Na v 1.5, Na/I .6, Na v 1.7, Na/I .8, or Na v 1.9 activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, the study of sodium ion channels in biological and pathological phenomena; and the comparative evaluation of new sodium ion channel inhibitors.
  • a compound of the invention as set forth above in the Summary of the Invention, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and/or a pharmaceutical composition of the invention, comprising a pharmaceutically acceptable excipient and one or more compounds of the invention, as set forth above in the Summary of the Invention, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, can also be used in the preparation of a medicament for the treatment and/or prevention of hypercholesterolemia, benign prostatic hyperplasia, pruritis, and/or cancer in a mammal.
  • compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see The
  • composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention.
  • a pharmaceutical composition of the invention may be in the form of a solid or liquid.
  • the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form.
  • the carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
  • the pharmaceutical composition When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
  • the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form.
  • a solid composition will typically contain one or more inert diluents or edible carriers.
  • binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
  • a liquid carrier such as polyethylene glycol or oil.
  • the pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • the liquid pharmaceutical compositions of the invention may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Physiological saline is a preferred adjuvant.
  • a liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of a compound of the invention in the composition. When intended for oral administration, this amount may be varied to be between 0.1 and about 70% of the weight of the composition.
  • Preferred oral pharmaceutical compositions contain between about 4% and about 50% of the compound of the invention.
  • Preferred pharmaceutical compositions and preparations according to the present invention are prepared so that a parenteral dosage unit contains between 0.01 to 10% by weight of the compound prior to dilution of the invention.
  • the pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base.
  • the base for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers.
  • Thickening agents may be present in a pharmaceutical composition for topical administration.
  • the composition may include a transdermal patch or iontophoresis device.
  • Topical formulations may contain a concentration of the compound of the invention from about 0.1 to about 10% w/v (weight per unit volume).
  • the pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug.
  • the composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient.
  • bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
  • the pharmaceutical composition of the invention may include various materials, which modify the physical form of a solid or liquid dosage unit.
  • the composition may include materials that form a coating shell around the active ingredients.
  • the materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents.
  • the active ingredients may be encased in a gelatin capsule.
  • the pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound.
  • Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
  • the pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol.
  • aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols.
  • compositions of the invention may be prepared by methodology well known in the pharmaceutical art.
  • a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution.
  • a surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
  • the compounds of the invention are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • a therapeutically effective daily dose is (for a 70 kg mammal) from about 0.001 mg/kg (i.e., 0.7 mg) to about 100 mg/kg (i.e., 7.0 gm); preferaby a therapeutically effective dose is (for a 70 kg mammal) from about 0.01 mg/kg (i.e., 7 mg) to about 50 mg/kg (i.e., 3.5 gm); more preferably a therapeutically effective dose is (for a 70 kg mammal) from about 1 mg/kg (i.e., 70 mg) to about 25 mg/kg (i.e., 1.75 gm).
  • the total dose required for each treatment can be administered by multiple doses or in a single dose over the course of the day, if desired. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached.
  • the diagnostic pharmaceutical compound or composition can be administered alone or in conjunction with other diagnostics and/or pharmaceuticals directed to the pathology, or directed to other symptoms of the pathology.
  • the recipients of administration of compounds and/or compositions of the invention can be any vertebrate animal, such as mammals.
  • the preferred recipients are mammals of the Orders Primate (including humans, apes and monkeys), Arteriodactyla (including horses, goats, cows, sheep, pigs), Rodenta (including mice, rats, rabbits, and hamsters), and Camivora (including cats, and dogs).
  • the preferred recipients are turkeys, chickens and other members of the same order. The most preferred recipients are humans.
  • a pharmaceutical composition according to the invention for topical applications, it is preferred to administer an effective amount of a pharmaceutical composition according to the invention to target area, e.g., skin surfaces, mucous membranes, and the like, which are adjacent to peripheral neurons which are to be treated.
  • This amount will generally range from about 0.0001 mg to about 1 g of a compound of the invention per application, depending upon the area to be treated, whether the use is diagnostic, prophylactic or therapeutic, the severity of the symptoms, and the nature of the topical vehicle employed.
  • a preferred topical preparation is an ointment, wherein about 0.001 to about 50 mg of active ingredient is used per cc of ointment base.
  • the pharmaceutical composition can be formulated as transdermal compositions or transdermal delivery devices ("patches"). Such compositions include, for example, a backing, active compound reservoir, a control membrane, liner and contact adhesive. Such transdermal patches may be used to provide continuous pulsatile, or on demand delivery of the compounds of the present invention as desired.
  • compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • Controlled release drug delivery systems include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Pat. Nos. 3,845,770 and 4,326,525 and in P. J. Kuzma et al, Regional Anesthesia 22 (6): 543-551 (1997), all of which are incorporated herein by reference.
  • the compositions of the invention can also be delivered through intra-nasal drug delivery systems for local, systemic, and nose-to-brain medical therapies.
  • Controlled Particle Dispersion (CPD)TM technology traditional nasal spray bottles, inhalers or nebulizers are known by those skilled in the art to provide effective local and systemic delivery of drugs by targeting the olfactory region and paranasal sinuses.
  • the invention also relates to an intravaginal shell or core drug delivery device suitable for administration to the human or animal female.
  • the device may be comprised of the active pharmaceutical ingredient in a polymer matrix, surrounded by a sheath, and capable of releasing the compound in a substantially zero order pattern on a daily basis similar to devises used to apply testosterone as desscribed in PCT Patent No. WO 98/50016.
  • kits that contain a pharmaceutical composition which includes one or more compounds of the invention.
  • the kit also includes instructions for the use of the pharmaceutical composition for modulating the activity of ion channels, for the treatment of benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and pruritis (itch), as well as other utilities as disclosed herein.
  • BPH benign prostatic hyperplasia
  • itch hypercholesterolemia
  • cancer cancer and pruritis
  • a commercial package will contain one or more unit doses of the pharmaceutical composition.
  • such a unit dose may be an amount sufficient for the preparation of an intravenous injection.
  • compounds which are light and/or air sensitive may require special packaging and/or formulation.
  • packaging may be used which is opaque to light, and/or sealed from contact with ambient air, and/or formulated with suitable coatings or excipients.
  • R 1 , R 2 , R 3 and R 4 are as defined herein, as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • Suitable protecting groups include hydroxy, amino, mercapto and carboxylic acid.
  • Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., f-butyldimethylsilyl, f-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include benzyl, f-butoxycarbonyl, benzyloxycarbonyl, and the like.
  • Suitable protecting groups for mercapto include -C(O)-R" (where R" is alkyl, aryl or arylalkyl), p-methoxybenzyl, trityl and the like.
  • Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
  • Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein.
  • the protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl- chloride resin.
  • R 1 , R 2 , R 4 and p are defined as in the Summary of the Invention unless specifically defined otherwise, and X is Cl or Br.
  • R 1 group can be introduced to an amino compound of formula (101 ) either by reductive amination, which is well-known to those skilled in the art, or formation of an amide by reacting with a corresponding acyl chloride followed by reduction, which is also well-known to those skilled in the art, to form a high order of amino compound of formula (102).
  • Reaction of the compound of formula (102) with oxalyl chloride gives the compound of formula (103).
  • the compound of formula (103) can be obtained by alkylation of the compound of formula (104) with the chloro or bromo compound of formula (105).
  • alkylation of pyrrole-type compound of formula (106) with the chloro or bromo compound of formula (105) provides the compound of formula (107).
  • the compound of formula (201) can be obtained after the removal of the hydroxyl group of the heterocyclic compound of formula (109) by treating the compound with a silane such as triethylsilane.
  • the compound of formula (201 ) can also be achieved by treating the compound of formula (109) with SOCI 2 /NEt 3 followed by reduction with Zn dust.
  • the compound (201 ) is treated with a silyl compound, such as, but not limited to, trimethylsilyl chloride, to generate the silyl ether intermediate, which is treated with ytterbium (III) trifluoromethanesulfonate and formaldehyde to afford the compound of formula (301).
  • a compound of formula (301 ) can be obtained by treating the compound of formula (201) with a base, such as, but not limited to, LiOH 1 iPr 2 NH, LDA, and subsequently reacting with formaldehyde.
  • the compounds of formula (I) of the invention where R 3 is fluoro can be synthesized following the general procedure as described below in REACTION SCHEME 4.
  • the hydroxyl group of the compound of the formula (109) can be converted to the corresponding chloro group by reacting with a chloride compound such as, but not limited to, thionyl chloride, in the presence of a base such as, but not limited to, diisopropylethylamine or triethylamine, in a solvent such as, but inot limited to, dichloromethane or chloroform.
  • a chloride compound such as, but not limited to, thionyl chloride
  • a base such as, but not limited to, diisopropylethylamine or triethylamine
  • a solvent such as, but inot limited to, dichloromethane or chloroform.
  • Treatment of the generated chloride compound with a nucleophile such as, but not limited to, sodium cyanide or benzylamine, in a solvent such as, but not limited to, tetrahedrofuran or dioxane, provides the compound of formula (I)
  • the reaction was stirred at -78 0 C for 2 h and quenched with saturated ammonium chloride (20.0 mL). After the organic solvent was removed under reduced pressure, the residue was diluted with ethyl acetate (50.0 mL). The organic layer was washed with brine (30.0 mL), dried over anhydrous sodium sulfate and filtered.
  • This example describes an in vitro assay for testing and profiling test agents against human or rat sodium channels stably expressed in cells of either an endogenous or recombinant origin.
  • the assay is also useful for determining the IC-50 of a sodium channel blocking compound.
  • the assay is based on the guanidine flux assay described by Reddy, N. L., et al., J. Med. Chem. (1998), 41(17):3298-302.
  • the guanidine influx assay is a radiotracer flux assay used to determine ion flux activity of sodium channels in a high-throughput microplate-based format.
  • the assay uses 14 C-guanidine hydrochloride in combination with various known sodium channel modulators, to assay the potency of test agents. Potency is determined by an IC-50 calculation. Selectivity is determined by comparing potency of the compound for the channel of interest to its potency against other sodium channels (also called 'selectivity profiling').
  • Each of the test agents is assayed against cells that express the channels of interest.
  • Voltage gated sodium channels are either TTX sensitive or insensitive. This property is useful when evaluating the activities of a channel of interest when it resides in a mixed population with other sodium channels.
  • Table 1 summarizes cell lines useful in screening for a certain channel activity in the presence or absence of TTX.
  • Cells expressing the channel of interest are grown according to the supplier or in the case of a recombinant cell in the presence of selective growth media such as G418 (Gibco/lnvitrogen)
  • the cells are disassociated from the culture dishes with an enzymatic solution (1X) Trypsin/EDTA (Gibco/lnvitrogen) and analyzed for density and viability using haemocytometer (Neubauer).
  • Disassociated cells are washed and resuspended in their culture media then plated into Scintiplates (Beckman Coulter Inc.) (approximately 100,000 cells/ well) and incubated at 37 °C/5 % CO 2 . for 20-24 hours.
  • LNHBSS Low sodium HEPES-buffered saline solution
  • 1mM Choline Chloride, 20 nM HEPES (Sigma), 1mM Calcium Chloride, 5mM Potassium Chloride, 1 mM Magnesium Chloride, 10 mM Glucose agents diluted with LNHBSS are added to each well.
  • concentrations of test agent may be used.
  • the activation/radiolabel mixture contains aconitine (Sigma), and 14 C-guanidine hydrochloride (ARC).
  • the Scintiplates are incubated at ambient temperature. Following the incubation, the Scintplates are extensively washed with LNHBSS supplemented with guanidine (Sigma). The Scintiplates are dried and then counted using a Wallac MicroBeta TriLux (Perkin-Elmer Life Sciences). The ability of the test agent to block sodium channel activity is determined by comparing the amount of 14 C-guanidine present inside the cells expressing the different sodium channels. Based on this data, a variety of calculations, as set out elsewhere in this specification, may be used to determine whether a test agent is selective for a particular sodium channel.
  • IC-50 value of a test agent for a specific sodium channel may be determined using the above general method.
  • IC-50 may be determined using a 3, 8, 10, 12 or 16 point curve in duplicate or triplicate with a starting concentration of 1 , 5 or 10 ⁇ M diluted serially with a final concentration reaching the sub-nanomolar, nanomolar and low micromolar ranges.
  • the mid-point concentration of test agent is set at 1 ⁇ M, and sequential concentrations of half dilutions greater or smaller are applied (e.g. 0.5 ⁇ M; 5 ⁇ M and 0.25 ⁇ M; 10 ⁇ M and 0.125 ⁇ M; 20 ⁇ M etc.).
  • the fold selectivity, factor of selectivity or multiple of selectivity is calculated by dividing the IC-50 value of the test sodium channel by the reference sodium channel, for example, Na v 1.5.
  • Cells expressing the channel of interest were cultured in DMEM growth media (Gibco) with 0.5mg/mL G418, +/-1 % PSG, and 10% heat-inactivated fetal bovine serum at 37C° and 5% CO 2 .
  • DMEM growth media Gibco
  • heat-inactivated fetal bovine serum 37C° and 5% CO 2 .
  • cells were plated on 10mm dishes.
  • the internal (pipette) solution consisted of (in mM): NaCI (5), CaCI 2 (0.1 ) MgCI 2 (2), CsCI (10), CsF (120), HEPES (10), EGTA (10), at pH 7.2.
  • 12.5 ms test pulses to depolarizing voltages from -60 to +90 m V from a holding potential of -110 mV was used to construct current-voltage relationships (I- V curves).
  • I- V curves current-voltage relationships
  • Steady-state inactivation (availability) curves were then constructed by measuring the current activated during a 8.75 ms test pulse following 1 second conditioning pulses to potentials ranging from -110 to -10 m V.
  • a single "diary" protocol with a holding potential of -110 mV was created to record the resting state current (10 ms test pulse), the current after fast inactivation (5 ms pre-pulse of -80 to -50 mV followed by a 10 ms test pulse), and the current during various holding potentials (35 ms ramp to test pulse levels).
  • Compounds were applied during the "diary" protocol and the block was monitored at 15s intervals.
  • the voltage-dependence of the steady-state inactivation in the presence of the compound was determined.
  • Compounds that block the resting state of the channel decreased the current elicited during test pulses from all holding potentials, whereas compounds that primarily blocked the inactivated state decreased the current elicited during test pulses at more depolarized potentials.
  • the currents at the resting state (l rest ) and the currents during the inactivated state (I activated) were used to calculate steady-state affinity of compounds. Based on the Michaelis- Menton model of inhibition, the K r and K, was calculated as the concentration of compound needed to cause 50% inhibition of the l res t or the l, na ct ⁇ vated. respectively.
  • V max is the rate of inhibition
  • h is the Hill coefficient (for interacting sites)
  • K m is Michaelis-Menten constant
  • [Drug] is the concentration of the test compound.
  • the drug concentration is numerically equal to K m and approximates the K r and K 1 , respectively.
  • BPH Benign Prostate Hyperplasia
  • Dogs were closed orally with compounds of the present invention at oral doses of between 0 mg/kg and 100 mg/kg for a period of 4 weeks.
  • a control group received placebo.
  • the animals were sacrificed and the prostate glands dissected out, dabbed dry and then weighed.
  • Compounds of the present invention were shown to be efficacious in a dose dependent manner within a range of 5 mg/kg and 100 mg/kg in significantly reducing the weight of the prostate in dogs when compared to the vehicle treated (0 mg/kg) controls. These compounds had no adverse events, making them ideal candidates for the safe treatment of BPH and the associated symptoms, such as, but not limited to, acute urinary retention and urinary tract infection.
  • Dogs were dosed orally at a range of 0 mg/kg to 100 mg/kg daily with compounds of the present invention for a period of 2- 4 weeks. After 2 and 4 weeks the animals were bled and their serum collected for total cholesterol analysis and compared to the animals dosed with vehicle alone (0 mg/kg).
  • cholesteryl esters in the sample are first hydrolyzed by cholesterol esterase. All cholesterol, whether previously esterified or existing free in the circulation, is then oxidized by cholesterol oxidase to the corresponding ketone and hydrogen peroxide.
  • ADHP (10-acetyl-3,7-dihydroxyphenoxazine) is utilized as a highly sensitive and stable probe for hydrogen peroxide.
  • Horseradish peroxidase catalyzes the reaction of ADHP with hydrogen peroxide to yield the highly fluorescent product resorufin, which can be monitored using excitation wavelengths of 565-580 nm and emission wavelengths of
  • the compounds of the invention exhibited the ability to affect a significant drop in total serum cholesterol when administered in the above assay to dogs in a daily oral dose range of 5-100 mg/kg over a 2- and 4-week period.
  • the compounds of the invention can be evaluated for their activity as antipruritic agents by in vivo test using rodent models.
  • One established model for peripherally elicited pruritus is through the injection of serotonin into the rostral back area (neck) in hairless rats.
  • a dose of a compound of the present invention Prior to serotonin injections (e.g., 2 mg/ml, 50 ⁇ L), a dose of a compound of the present invention can be applied systemically through oral, intravenous or intraperitoneal routes or topically to a circular area fixed diameter (e.g. 18 mm). Following dosing, the serotonin injections are given in the area of the topical dosing. After serotonin injection the animal behaviour is monitored by video recording for 20 min-1.5 h, and the number of scratches in this time compared to vehicle treated animals. Thus, application of a compound of the current invention could suppress serotonin-induced scratching in rats.

Abstract

This invention is directed to the use of heterocyclic compounds for the treatment and/or prevention of diseases or conditions such as hypercholesterolemia, benign prostatic hyperplasia, pruritis and cancer.

Description

USE OF HETEROCYCLIC COMPOUNDS AS THERAPEUTIC AGENTS
FIELD OF THE INVENTION
The present invention is directed to methods of using certain heterocyclic compounds as therapeutic agents. In particular, this invention is directed to the use of certain heterocyclic compounds in treating diseases or conditions such as hypercholesterolemia, benign prostatic hyperplasia, pruritis and cancer.
BACKGROUND OF THE INVENTION
Voltage-gated sodium channels, transmembrane proteins that initiate action potentials in nerve, muscle and other electrically excitable cells, are a necessary component of normal sensation, emotions, thoughts and movements (Catterall, W.A., Nature (2001 ), Vol. 409, pp. 988-990). These channels consist of a highly processed alpha subunit that is associated with auxiliary beta subunits. The pore-forming alpha subunit is sufficient for channel function, but the kinetics and voltage dependence of channel gating are in part modified by the beta subunits (Goldin et al., Neuron (2000), Vol. 28, pp. 365-368). Each alpha-subunit contains four homologous domains, I to IV, each with six predicted transmembrane segments. The alpha-subunit of the sodium channel, forming the ion-conducting pore and containing the voltage sensors regulating sodium ion conduction has a relative molecular mass of 260,000. Electrophysiological recording, biochemical purification, and molecular cloning have identified ten different sodium channel alpha subunits and four beta subunits (Yu, F.H., et al., Sci. STKE (2004), 253; and Yu, F.H., et al., Neurosci. (2003), 20:7577-85).
The hallmarks of sodium channels include rapid activation and inactivation when the voltage across the plasma membrane of an excitable cell is depolarized (voltage-dependent gating), and efficient and selective conduction of sodium ions through conducting pores intrinsic to the structure of the protein (Sato, C, et al., Nature (2001 ), 409:1047-1051 ). At negative or hyperpolarized membrane potentials, sodium channels are closed. Following membrane depolarization, sodium channels open rapidly and then inactivate. Channels only conduct currents in the open state and, once inactivated, have to return to the resting state, favoured by membrane hyperpolarization, before they can reopen. Different sodium channel subtypes vary in the voltage range over which they activate and inactivate as well as their activation and inactivation kinetics.
The sodium channel family of proteins has been extensively studied and shown to be involved in a number of vital body functions. Research in this area has identified variants of the alpha subunits that result in major changes in channel function and activities, which can ultimately lead to major pathophysiological conditions. Implicit with function, this family of proteins are considered prime points of therapeutic intervention. Na/1.1 and Nav1.2 are highly expressed in the brain (Raymond, C. K., et al., J. Biol. Chem. (2004), 279(44):46234-41 ) and are vital to normal brain function. In humans, mutations in Na/1.1 and Na/I .2 result in severe epileptic states and in some cases mental decline (Rhodes, T.H., et al., Proc. Natl. Acad. Sci. USA (2004), 101 (30): 11147-52; Kamiya, K., et al., J. Biol. Chem. (2004), 24(11 ) :2690-8; Pereira, S., et al., Neurology (2004), 63(1 ):191-2). As such both channels have been considered as validated targets for the treatment of epilepsy (see PCT Published Patent Publication No. WO 01/38564).
Na/1.3 is broadly expressed throughout the body (Raymond, C. K., et al., op. cit). It has been demonstrated to have its expression upregulated in the dorsal horn sensory neurons of rats after nervous system injury (Hains, B. D., et al., J. Neurosci. (2003), 23(26):8881-92). Many experts in the field have considered Na/I .3 as a suitable target for pain therapeutics (Lai, J., et al., Curr. Opin. Neurobiol. (2003), (3):291 -72003; Wood, J. N., et al., J. Neurobiol. (2004), 61(1):55-71 ; Chung, J.M., et al., Novartis Found Symp. (2004), 261 :19-27; discussion 27-31 , 47-54). Na/I .4 expression is essentially limited to muscle (Raymond, C.K., et al., op. cit.). Mutations in this gene have been shown to have profound effects on muscle function including paralysis, (Tamaoka A., Intern. Med. (2003), (9):769-70). Thus, this channel can be considered a target for the treatment of abnormal muscle contractility, spasm or paralysis. The cardiac sodium channel, Na/1.5, is expressed mainly in the heart ventricles and atria (Raymond, C. K., et al., op. cit.), and can be found in the sinovial node, ventricular node and possibly Purkinje cells. The rapid upstroke of the cardiac action potential and the rapid impulse conduction through cardiac tissue is due to the opening of Nav1.5. As such, Na/1.5 is central to the genesis of cardiac arrhythmias. Mutations in human Nav1.5 result in multiple arrhythmic syndromes, including, for example, long QT3 (LQT3), Brugada syndrome (BS), an inherited cardiac conduction defect, sudden unexpected nocturnal death syndrome (SUNDS) and sudden infant death syndrome (SIDS) (Liu, H. et al., Am. J. Pharmacogenomics (2003), 3(3):173-9). Sodium channel blocker therapy has been used extensively in treating cardiac arrhythmias. The first antiarrhythmic drug, quinidine, discovered in 1914, is classified as a sodium channel blocker.
Na1Zl .6 encodes an abundant, widely distributed voltage-gated sodium channel found throughout the central and peripheral nervous systems, clustered in the nodes of Ranvier of neural axons (Caldwell, J. H., et al., Proc. Natl. Acad. ScL USA (2000), 97(10): 5616-20). Although no mutations in humans have been detected, Nayi .6 is thought to play a role in the manifestation of the symptoms associated with multiple sclerosis and has been considered as a target for the treatment of this disease (Craner, M.J., et al., Proc. Natl. Acad. ScL USA (2004), 101(21):8168-73).
Nav1.7 was first cloned from the pheochromocytoma PC12 cell line (Toledo- Aral, J. J., et al., Proc. Natl.Acad. ScL USA (1997), 94:1527-1532). Its presence at high levels in the growth cones of small-diameter neurons suggested that it could play a role in the transmission of nociceptive information. Although this has been challenged by experts in the field as Nav1.7 is also expressed in neuroendocrine cells associated with the autonomic system (Klugbauer, N., et al., EMBO J. (1995), 14(6): 1084-90) and as such has been implicated in autonomic processes. The implicit role in autonomic functions was demonstrated with the generation of Nav1.7 null mutants; deleting Na/I .7 in all sensory and sympathetic neurons resulted in a lethal perinatal phenotype. (Nassar, et al., Proc. Natl. Acad. ScL USA (2004), 101 (34): 12706- 11.). In contrast, by deleting the Na/1.7 expression in a subset of sensory neurons that are predominantly nociceptive, a role in pain mechanisms, was demonstrated (Nassar, et al., op. cit). Further support for Na^I .7 blockers active in a subset of neurons is supported by the finding that two human heritable pain conditions, primary erythermalgia and familial rectal pain, have been shown to map to Nav1.7 (Yang, Y., et al., J. Med. Genet. (2004), 41 (3): 171 -4). The expression of Nav1.8 is essentially restricted to the DRG (Raymond, C. K., et al., op. cit.). There are no identified human mutations for Na/I .8. However, Na/I .8- null mutant mice were viable, fertile and normal in appearance. A pronounced analgesia to noxious mechanical stimuli, small deficits in noxious thermoreception and delayed development of inflammatory hyperalgesia suggested to the researchers that Na/I .8 plays a major role in pain signalling (Akopian, A. N., et al., Nat. Neurosci. (1999), 2(6): 541-8). Blocking of this channel is widely accepted as a potential treatment for pain (Lai, J, et al., op. cit; Wood, J. N., et al., op. cit; Chung, J. M., et al., op. cit). PCT Published Patent Application No. WO03/037274A2 describes pyrazole- amides and sulfonamides for the treatment of central or peripheral nervous system conditions, particularly pain and chronic pain by blocking sodium channels associated with the onset or recurrance of the indicated conditions. PCT Published Patent Application No. WO03/037890A2 describes piperidines for the treatment of central or peripheral nervous system conditions, particularly pain and chronic pain by blocking sodium channels associated with the onset or recurrence of the indicated conditions. The compounds, compositions and methods of these inventions are of particular use for treating neuropathic or inflammatory pain by the inhibition of ion flux through a channel that includes a PN3 (Nav1.8) subunit.
The tetrodotoxin insensitive, peripheral sodium channel Na/1.9, disclosed by Dib-Hajj, S. D., et al. (see Dib-Hajj, S. D., et al., Proc. Natl. Acad. Sci. USA (1998), 95(15):8963-8) was shown to reside solely in the dorsal root ganglia. It has been demonstrated that Nav1.9 underlies neurotrophin (BDNF)-evoked depolarization and excitation, and is the only member of the voltage gated sodium channel superfamily to be shown to be ligand mediated (Blum, R., Kafitz, K.W., Konnerth, A., Nature (2002), 419 (6908):687-93). The limited pattern of expression of this channel has made it a candidate target for the treatment of pain (Lai, J, et al., op. cit.; Wood, J. N., et al., op. cit; Chung, J. M. et al., op. cit).
NaX is a putative sodium channel, which has not been shown to be voltage gated. In addition to expression in the lung, heart, dorsal root ganglia, and Schwann cells of the peripheral nervous system, NaX is found in neurons and ependymal cells in restricted areas of the CNS, particularly in the circumventricular organs, which are involved in body-fluid homeostasis (Watanabe, E., et al., J. Neurosci. (2000), 20(20):7743-51 ). NaX-null mice showed abnormal intakes of hypertonic saline under both water- and salt-depleted conditions. These findings suggest that the NaX plays an important role in the central sensing of body-fluid sodium level and regulation of salt intake behaviour. Its pattern of expression and function suggest it as a target for the treatment of cystic fibrosis and other related salt regulating maladies.
Studies with the sodium channel blocker tetrodotoxin (TTX) used to lower neuron activity in certain regions of the brain, indicate its potential use in the treatment of addiction. Drug-paired stimuli elicit drug craving and relapse in addicts and drug- seeking behavior in rats. The functional integrity of the basolateral amygdala (BLA) is necessary for reinstatement of cocaine-seeking behaviour elicited by cocaine- conditioned stimuli, but not by cocaine itself. BLA plays a similar role in reinstatement of heroin-seeking behavior. TTX-induced inactivation of the BLA on conditioned and heroin-primed reinstatement of extinguished heroin-seeking behaviour in a rat model (Fuchs, R.A. and See, R.E., Psychopharmacology (2002) 160(4):425-33). This closely related family of proteins has long been recognised as targets for therapeutic intervention. Sodium channels are targeted by a diverse array of pharmacological agents. These include neurotoxins, antiarrhythmics, anticonvulsants and local anesthetics (Clare, J.J., et al., Drug Discovery Today (2000) 5:506-520). All of the current pharmacological agents that act on sodium channels have receptor sites on the alpha subunits. At least six distinct receptor sites for neurotoxins and one receptor site for local anesthetics and related drugs have been identified (Cestele, S. et al., Biochimie (2000), Vol. 82, pp. 883-892).
The small molecule sodium channel blockers or the local anesthetics and related antiepileptic and antiarrhythmic drugs, interact with overlapping receptor sites located in the inner cavity of the pore of the sodium channel (Catterall, W.A., Neuron (2000), 26:13-25). Amino acid residues in the S6 segments from at least three of the four domains contribute to this complex drug receptor site, with the IVS6 segment playing the dominant role. These regions are highly conserved and as such most sodium channel blockers known to date interact with similar potency with all channel subtypes. Nevertheless, it has been possible to produce sodium channel blockers with therapeutic selectivity and a sufficient therapeutic window for the treatment of epilepsy (e.g. lamotrignine, phenytoin and carbamazepine) and certain cardiac arrhythmias (e.g. lignocaine, tocainide and mexiletine). However, the potency and therapeutic index of these blockers is not optimal and have limited the usefulness of these compounds in a variety of therapeutic areas where a sodium channel blocker would be ideally suited.
SUMMARY OF THE INVENTION
The present invention is directed to the use of certain heterocyclic compounds for the treatment and/or prevention of diseases or conditions, such as hypercholesterolemia, benign prostatic hyperplasia, pruritis, and cancer.
Accordingly, in one aspect, the invention provides compounds of formula (I):
Figure imgf000006_0001
wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000007_0001
is a fused heteroaryl ring or a fused heterocyclyl ring; R1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R9-C(O)R6, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2 or -R10-O-R10-OR6; or R1 is aralkyl substituted by -C(O)N(R7)R8 where: R7 is hydrogen, alkyl, aryl or aralkyl; and
R8 is hydrogen, alkyl, haloalkyl, -R10-CN, -R10-OR6, -R10-N(R5)R6, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R7 and R8, together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R7 and R8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl,
-R9-CN, -R9-OR6, heterocyclyl and heteroaryl; or R1 is aralkyl substituted by one or more substituents selected from the group consisting of -R9-OR6, -R9-C(O)OR6, halo, haloalkyl, alkyl, nitro, cyano, aryl (optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R1 is -R10-N(R11)R12, -R10-N(R13)C(O)R12 or -R10-N(R11)C(O)N(R11)R12 where: each R11 is hydrogen, alkyl, aryl or aralkyl; each R12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R10-OC(O)R6, -R10-C(O)OR6, -R10-C(O)N(R5)R6, -R10-C(O)R6, -R10-OR6, or -R10-CN;
R13 is hydrogen, alkyl, aryl, arakyl or -C(O)R6; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R11 and R12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R9-CN, -R9-OR6, -R9-C(O)R6, heterocyclyl and heteroaryl; or R1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R9-OR6, -R9-C(O)OR6, aryl and aralkyl; each R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -C(S)R5, -C(R5)2C(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6,
-R9-S(O)nN(R5)R6, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(=N-CN)N(R5)R6, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6JS(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring or the fused heterocyclyl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)X, -C(S)R5, -C(R5)2C(O)R6, -R9-OC(O)R6, -R9-C(O)OR6,
-C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -Si(R6)3, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6,, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(N=C(R5)R6)N(R5)R6, wherein X is bromo or chloro, each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl groups for R3 and R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
In another aspect, the invention provides methods of treating or preventing hypercholesterolemia in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above. In another aspect, the invention provides methods of treating or preventing benign prostatic hyperplasia in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
In another aspect, the invention provides methods of treating or preventing pruritis in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
In another aspect, the invention provides methods of treating or preventing cancer in a mammal, preferably a human, wherein the methods comprise administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention as set forth above.
In another aspect, the invention provides pharmaceutical compositions comprising the compounds of the invention, as set forth above, and pharmaceutically acceptable excipients. In another aspect, the invention provides pharmaceutical therapy in combination with one or more other compounds of the invention or one or more other accepted therapies or as any combination thereof to increase the potency of an existing or future drug therapy or to decrease the adverse events associated with the accepted therapy. In one embodiment, the present invention relates to a pharmaceutical composition combining compounds of the present invention with established or future therapies for the indications listed in the invention.
In another aspect, this invention is directed to the use of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or the use of a pharmaceutical composition of the invention, comprising a pharmaceutically acceptable excipient and a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, in the preparation of a medicament for the treatment and/or prevention of hypercholesterolemia, benign prostatic hyperplasia, pruritis, and/or cancer in a mammal.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
Certain chemical groups named herein are preceded by a shorthand notation indicating the total number of carbon atoms that are to be found in the indicated chemical group. For example; C7-C12alkyl describes an alkyl group, as defined below, having a total of 7 to 12 carbon atoms, and C4-Ci2cycloalkylalkyl describes a cycloalkylalkyl group, as defined below, having a total of 4 to 12 carbon atoms. The total number of carbons in the shorthand notation does not include carbons that may exist in substituents of the group described. For example, the following terms have the meaning indicated:
"Ci-CiOalkyl" refers to an alkyl radical as defined below containing one to ten carbon atoms. The CrC10alkyl readical may be optionally substituted as defined below for an alkyl group.
"C2-C12alkynyl" refers to an alknyl radical as defined below containing two to twelve carbon atoms. The C2-C12alknyl radical may be optionally substituted as defined below for an alkenyl group.
"CrC^alkoxy" refers to an alkoxy radical as defined below containing one to twelve carbon atoms. The alkyl part of the d-C12alkoxy radical may be optionally substituted as defined below for an alkyl group.
"C2-Ci2alkoxyalkyl" refers to an alkoxyalkyl radical as defined below containing two to twelve carbon atoms. Each alkyl part of the C2-C12alkoxyalkyl radical may be optionally substituted as defined below for an alkyl group. "C7-C12aralkyl" refers to an aralkyl group as defined below containing seven to twelve carbon atoms. The aryl part of the C7-C12aralkyl radical may be optionally substituted as described below for an aryl group. The alkyl part of the C7-C12aralkyl radical may be optionally substituted as defined below for an alkyl group.
"C7-C12aralkenyl" refers to an aralkenyl group as defined below containing seven to twelve carbon atoms. The aryl part of the C7-C12aralkenyl radical may be optionally substituted as described below for an aryl group. The alkenyl part of the C7-C12aralkenyl radical may be optionally substituted as defined below for an alkenyl group.
"C3-C12cycloalkyl" refers to a cycloalkyl radical as defined below having three to twelve carbon atoms. The C3-C12cycloalkyl radical may be optionally substituted as defined below for a cycloalkyl group.
"C4-C12cycloalkylalkyl" refers to a cycloalkylalkyl radical as defined below having four to twelve carbon atoms. The C4-C12cycloalkylalkyl radical may be optionally substituted as defined below for a cycloalkylalkyl group. In addition to the foregoing, as used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated:
"Amino" refers to the -NH2 radical.
"Cyano" refers to the -CN radical.
"Hydroxyl" refers to the -OH radical. "Imino" refers to the =NH substituent.
"Nitro" refers to the -NO2 radical.
"Oxo" refers to the =0 substituent.
"Thioxo" refers to the =S substituent.
"Trifluoromethyl" refers to the -CF3 radical. "Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to twelve carbon atoms, preferably one to eight carbon atoms or one to six carbon atoms, and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, n-propyl, 1-methylethyl (/so-propyl), n-butyl, n-pentyl, 1 ,1-dimethylethyl (f-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR14, -OC(O)-R14, -N(R14)2, -C(O)R14, -C(O)OR14, -C(O)N(R14)2, -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O),R16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)4R16 (where t is O to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Alkenyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1 ,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR14, -OC(O)-R14, -N(R14)2, -C(O)R14, -C(O)OR14, -C(O)N(R14)2, -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O),R16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)1R16 (where t is 0 to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR14, -OC(O)-R14, -N(R14)2, -C(O)R14, -C(O)OR14, -C(O)N(R14)2) -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O)tR16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)1R16 (where t is 0 to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Alkenylene" or "alkenylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one double bond and having from two to twelve carbon atoms, e.g., ethenylene, propenylene, n-butenylene, and the like. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkenylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkenylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR14, -OC(O)-R14, -N(R14)2> -C(O)R14, -C(O)OR14, -C(O)N(R14)2, -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O)tR16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)1R16 (where t is 0 to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Alkynylene" or "alkynylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one triple bond and having from two to twelve carbon atoms, e.g., propynylene, n-butynylene, and the like. The alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a double bond or a single bond. The points of attachment of the alkynylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkynylene chain may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkenyl, cyano, nitro, aryl, cycloalkyl, heterocyclyl, heteroaryl, oxo, trimethylsilanyl, -OR14, -OC(O)-R14, -N(R14)2, -C(O)R14, -C(O)OR14, -C(O)N(R14)2, -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O)tR16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)1R16 (where t is O to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl. "Alkynyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to twelve carbon atoms, preferably one to eight carbon atoms and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group may be optionally substituted by one of the following groups: alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR14, -OC(O)-R14, -N(R14)2, -C(O)R14, -C(O)OR14, -C(O)N(R14)2, -N(R14)C(O)OR16, -N(R14)C(O)R16, -N(R14)S(O)tR16 (where t is 1 to 2), -S(O)1OR16 (where t is 1 to 2), -S(O)1R16 (where t is O to 2), and -S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl. "Alkoxy" refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. The alkyl part of the alkoxy radical may be optionally substituted as defined above for an alkyl radical.
"Alkoxyalkyl" refers to a radical of the formula -R3-O-R3 where each R3 is independently an alkyl radical as defined above. The oxygen atom may be bonded to any carbon in either alkyl radical. Each alkyl part of the alkoxyalkyl radical may be optionally substituted as defined above for an alkyl group.
"Aryl" refers to aromatic monocyclic or multicyclic hydrocarbon ring system consisting only of hydrogen and carbon and containing from 6 to 18 carbon atoms, where the ring system may be partially saturated. Aryl groups include, but are not limited to, groups such as fluorenyl, phenyl and naphthyl. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals optionally substituted by one or more substituents independently selected from the group consisting of alkyl, akenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, aryl, heteroaryl, heteroarylalkyl, -R15-OR14, -R15-OC(O)-R14, -R15-N(R14)2, -R15-C(O)R14, -R15-C(O)OR14, -R15-C(O)N(R14)2, -R15-N(R14)C(O)OR16, -R15-N(R14)C(O)R16, -R15-N(R14)S(O),R16 (where t is 1 to 2), -R15-S(O)tOR16 (where t is 1 to 2), -R15-S(O)tR16 (where t is 0 to 2), and -R15-S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; each R15 is independently a direct bond or a straight or branched alkylene or alkenylene chain; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Aralkyl" refers to a radical of the formula -RaRb where R3 is an alkyl radical as defined above and Rb is one or more aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like. The aryl radical(s) may be optionally substituted as described above.
"Aryloxy" refers to a radical of the formula -ORb where Rb is an aryl group as defined above. The aryl part of the aryloxy radical may be optionally substituted as defined above. "Aralkenyl" refers to a radical of the formula -R0Rb where Rc is an alkenyl radical as defined above and Rb is one or more aryl radicals as defined above, which may be optionally substituted as described above. The aryl part of the aralkenyl radical may be optionally substituted as described above for an aryl group. The alkenyl part of the aralkenyl radical may be optionally substituted as defined above for an alkenyl group. "Aralkyloxy" refers to a radical of the formula -ORb where Rb is an aralkyl group as defined above. The aralkyl part of the aralkyloxy radical may be optionally substituted as defined above.
"Cycloalkyl" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptly, and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbomyl, decalinyl, and the like. Unless otherwise stated specifically in the specification, the term "cycloalkyl" is meant to include cycloalkyl radicals which are optionally substituted by one or more substituents independently selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, nitro, oxo, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R15-OR14, -R15-OC(O)-R14, -R15-N(R14)2, -R15-C(O)R14, -R15-C(O)OR14, -R15-C(O)N(R14)2, -R15-N(R14)C(O)OR16, -R15-N(R14)C(O)R16, -R15-N(R14)S(O)tR16 (where t is 1 to 2), -R15-S(O)tOR16 (where t is 1 to 2), -R15-S(O)tR16 (where t is 0 to 2), and -R15-S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; each R15 is independently a direct bond or a straight or branched alkylene or alkenylene chain; and each R16 is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Cycloalkylalkyl" refers to a radical of the formula -RaRo where R3 is an alkyl radical as defined above and Rd is a cycloalkyl radical as defined above. The alkyl radical and the cycloalkyl radical may be optionally substituted as defined above. "Halo" refers to bromo, chloro, fluoro or iodo.
"Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, 3-bromo-2-fluoropropyl, 1-bromomethyl-2-bromoethyl, and the like. The alkyl part of the haloalkyl radical may be optionally substituted as defined above for an alkyl group.
"Heterocyclyl" refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1 ,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1 ,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, the term "heterocyclyl" is meant to include heterocyclyl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R15-OR14, -R15-OC(O)-R14, -R15-N(R14)2, -R15-C(O)R14, -R15-C(O)OR14, -R15-C(O)N(R14)2, -R15-N(R14)C(O)OR16, -R15-N(R14)C(O)R16, -R15-N(R14)S(O)tR16 (where t is 1 to 2), -R15-S(O)tOR16 (where t is 1 to 2), -R15-S(O)tR16 (where t is 0 to 2), and -R15-S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; each R15 is independently a direct bond or a straight or branched alkylene or alkenylene chain; and each R16 is alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl. "Heterocyclylalkyl" refers to a radical of the formula -RaRe where Ra is an alkyl radical as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. The alkyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for an alkyl group. The heterocyclyl part of the heterocyclylalkyl radical may be optionally substituted as defined above for a heterocyclyl group.
"Heteroaryl" refers to a 5- to 18-membered aromatic ring radical which consists of one to seventeen carbon atoms and from one to ten heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzthiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[jb][1 ,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1 ,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, the term "heteroaryl" is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkoxy, halo, haloalkyl, haloalkenyl, cyano, oxo, thioxo, nitro, oxo, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R15-OR14, -R15-OC(O)-R14, -R15-N(R14)2, -R15-C(O)R14, -R15-C(O)OR14, -R15-C(O)N(R14)2, -R15-N(R14)C(O)OR16, -R15-N(R14)C(O)R16, -R15-N(R14)S(O)tR16 (where t is 1 to 2), -R15-S(O)tOR16 (where t is 1 to 2), -R15-S(O)tR16 (where t is 0 to 2), and -R15-S(O)tN(R14)2 (where t is 1 to 2) where each R14 is independently hydrogen, alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl; each R15 is independently a direct bond or a straight or branched alkylene or alkenylene chain; and each R16 is alkyl, alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
"Heteroarylalkyl" refers to a radical of the formula -RaRf where R3 is an alkyl radical as defined above and Rf is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkyl radical may be optionally substituted as defined above for a heteroaryl group. The alkyl part of the heteroarylalkyl radical may be optionally substituted as defined above for an alkyl group.
"Heteroarylalkenyl" refers to a radical of the formula -RbRf where Rb is an alkenyl radical as defined above and Rf is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkenyl radical may be optionally substituted as defined above for a heteroaryl group. The alkenyl part of the heteroarylalkenyl radical may be optionally substituted as defined above for an alkenyl group.
"Trihaloalkyl" refers to an alkyl radical, as defined above, that is substituted by three halo radicals, as defined above, e.g., trifluoromethyl. The alkyl part of the trihaloalkyl radical may be optionally substituted as defined above for an alkyl group.
"Trihaloalkoxy" refers to a radical of the formula -OR9 where R9 is a trihaloalkyl group as defined above. The trihaloalkyl part of the trihaloalkoxy group may be optionally substituted as defined above for a trihaloalkyl group. "Prodrugs" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)).A discussion of prodrugs is provided in Higuchi, T., et a/., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, Ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like.
The invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of formula (I) being isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 150, 170, 18O, 31P, 32P, 35S, 18F, 36CI, 123I, and 125I, respectively. These radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action on the sodium channels, or binding affinity to pharmacologically important site of action on the sodium channels. Certain isotopically-labelled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples and Preparations as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reducation, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically are identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its coversion products from the urine, blood or other biological samples.
"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. "Mammal" includes humans and both domestic animals such as laboratory animals and household pets, ( e.g. cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildelife and the like.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution. When a functional group is described as "optionally substituted," and in turn, substitutents on the functional group are also "optionally substituted" and so on, for the purposes of this invention, such iterations are limited to five.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
"Pharmaceutically acceptable salt" includes both acid and base addition salts. "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1 ,5- disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2~naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, Λ/-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
Often crystallizations produce a solvate of the compound of the invention. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor. "Therapeutically effective amount" refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a disease or condition in the mammal, preferably a human. The amount of a compound of the invention which constitutes a "therapeutically effective amount" will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
"Treating" or "treatment" as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:
(i) preventing the disease or condition from occurring in a mammal, in particular, when such mammal is predisposed to the condition but has not yet been diagnosed as having it;
(ii) inhibiting the disease or condition, i.e., arresting its development; (iii) relieving the disease or condition, i.e., causing regression of the disease or condition; or
(iv) relieving the symptoms resulting from the disease or condition.
As used herein, the terms "disease" and "condition" may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centres and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), [R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallisation. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tautomers of any said compounds.
Also within the scope of the invention are intermediate compounds of formula (I) and all polymorphs of the aforementioned species and crystal habits thereof. The chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program, wherein the compounds of the invention are named herein as derivatives of the central core structure, i.e., the 2-oxindole structure. For complex chemical names employed herein, a substituent group is named before the group to which it attaches. For example, cyclopropylethyl comprises an ethyl backbone with cyclopropyl substituent. In chemical structure diagrams, all bonds are identified, except for some carbon atoms, which are assumed to be bonded to sufficient hydrogen atoms to complete the valency.
Thus, for example, a compound of formula (I) wherein p is 0, R1 is pentyl, R3 is hydroxy, R is benzo-1 ,3-dioxolyl; and
Figure imgf000025_0001
is a fused thienyl ring;
Figure imgf000025_0002
is named herein as 4-(1 ,3-benzodioxol-5-yl)-4-hydroxy-6-pentyl-4,6-dihydro-5/-/- thieno[2,3-6]pyrrol-5-one. EMBODIMENTS OF THE INVENTION
Of the various aspects of the invention set forth above in the Summary of the Invention, certain embodiments are preferred.
One embodiment is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1 , 2, 3 or 4;
Figure imgf000025_0003
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is -R9-C(O)R6, -R9-C(O)OR6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2, -R10-O-R10-OR6, hydrogen, alkyl, haloalkyl, cycloalkylalkyl, heterocyclylalkyl, aryl (optionally substituted by one or more substituents selected from the group consisting of halo and -R9-C(O)OR6), aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6and -R9-C(O)OR6), heteroaryl (optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6), or heteroarylalkyl (optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6); each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-OR6, -R9-N(R5)R6, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6,
-R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5 and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above; R3 is independently selected from the group consisting of hydrogen, halo, haloalkyl,
-R9-OR6, -R9-OC(O)R6, -R9-CN, -R9-N(R5)R6, -R9-C(O)R5, -R9-C(O)X, -R9-C(O)OR6 and -N(R6)C(O)OR6, wherein X is chloro or bromo;
R4 is independently selected from the group consisting of alkyl, aryl, aralkyl, aralkynyl, heteroaryl, heteroarylalkyl, -R9-C(O)R5, -N(R6)C(O)N(R5)R6, -R9-NO2, -R9-N(R5)R6, -R9-C(O)OR6, -R9-N(R6)C(O)OR6 and -Si(R6)3, wherein each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a N-heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000027_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl, aryl or aralkyl, where each of the aryl or aralkyl group for R1 is optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6; each R2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R9-OR6, wherein each of the aryl and heteroaryl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and
-N(R6)S(O)nR5, wherein each m is independently 0, 1, or 2 and each n is independently 1 or 2;
R3 is hydrogen, halo, -R9-OR6 or -R9-OC(O)R6;
R4 is independently selected from the group consisting of alkyl, aryl, aralkynyl, heteroaryl, heteroarylalkyl, -R9-C(O)R5, -N(R6)C(O)N(R5)R6, -R9-NO2,
-R9-N(R5)R6, -R9-C(O)OR6 and -Si(R6)3, wherein each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloaikyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloaikyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1 , 2, 3 or 4;
Figure imgf000028_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl, aryl or aralkyl, where each of the aryl or aralkyl group for R1 is optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6; each R2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R9-OR6, wherein each of the aryl and heteroaryl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloaikyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2;
R3 is hydrogen, halo, -R9-OR6 or -R9-OC(O)R6; R4 is -R9-C(O)R5; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-hydroxy-3-[2-oxo-2-(2-thienyl)ethyl]-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-b]pyridin-2- one; and 3-[2-(2-furyl)-2-oxoethyl]-3-hydroxy-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3-/3]pyridin-2- one.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
J is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
R1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6); each R2 is independently selected from the group consisting of alkyl, halo, aryl, heteroaryl and -R9-OR6, wherein each of the aryl and heteroaryl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5 and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2;
R3 is hydrogen, halo, -R9-OR6 or -R9-OC(O)R6;
R4 is heterocyclylalkyl, heteroaryl or heteroarylalkyl, each optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN,
-R9-NO2> -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5 and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000030_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6); each R2 is each independently selected from the group consisting of alkyl, halo, phenyl, benzodioxolyl and -R9-OR6, R3 is hydrogen, halo, -R9-OR6 or -R9-OC(O)R6;
R4 is heterocyclylalkyl, heteroaryl or heteroarylalkyl, each optionally substituted by one or more substituents selected from the group consisting of halo, heterocyclyl, and -R9-OR6; each R6 is independently selected from group consisting of hydrogen, alky!, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0;
Figure imgf000031_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
R1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6); R3 is -R9-OR6;
R4 is aryl, aralkyl or aralkynyl, wherein each of the aryl, aralkyl and aralkynyl groups for R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1, or 2 and each n is independently 1 or 2; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0;
Figure imgf000032_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
R1 is aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, heteroaryl, -R9-OR6 and -R9-C(O)OR6); R3 is -R9-OR6;
R4 is aryl, aralkyl or aralkynyl, wherein each of the aryl, aralkyl and aralkynyl groups for R4 is optionally substituted by one or more substituents selected from the group consisting of halo, oxo and -R9-OR6; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000033_0001
a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is hydrogen, alkyl, haloalkyl or cycloalkylalkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-OR6, -R9-N(R5)R6, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0,
1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 is hydrogen, halo or -R9-OR6;
R4 is independently selected from the group consisting of alkyl, aryl, aralkynyl, heteroaryl, heteroarylalkyl, -R9-C(O)R5, -R9-N(R6)C(O)OR6, -N(R6)C(O)N(R5)R6, -R9-NO2, -R9-N(R5)R6, -R9-C(O)OR6, and -Si(R6)3, wherein each of the aryl, aralkynyl, heteroaryl and heteroarylalkyl groups for R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6,
-R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000034_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl
R1 is hydrogen, alkyl, haloalkyl or cycloalkylalkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-OR6, -R9-N(R5)R6, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above; R3 is hydrogen or -R9-OR6;
R4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6,
-S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000035_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6;
R3 is hydrogen or -R9-OR6;
R4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of halo, -R9-OR6 and -N(R6)C(O)R5; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000036_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6;
R3 is hydrogen or -R9-OR6; R4 is benzodioxolyl optionally substituted by one or more substituents selected from the group consisting of halo and -R9-OR6; each R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1H-pyrrolo[1 ,2-b]pyrazol-2(3H)-one;
4-(1 ,3-benzodioxol-5-yl)-4-hydroxy-6-pentyl-4,6-dihydro-5/-/-thieno[2,3-ό]pyrrol-5-one; 6-(1,3-benzodioxol-5-yl)-6-hydroxy-4-pentyl-4,6-dihydro-5H-thieno[3,2-ιb]pyrrol-5-one; 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[2,3- £>]pyridin-2-one; 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H-pyrrolo[3,2- b]pyridin-2-one;
3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[3,2- c]pyridin-2-one;
6-hydroxy-6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6-dihydro-5/-/-thieno[3,2-
£>]pyrrol-5-one;
3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-b]pyridin-2-one; 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[3,2-/b]pyridin-2-one; 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[3,2-c]pyridin-2-one; 6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6-dihydro-5H-thieno[3,2-b]pyrrol-5-one; 3-(6-hyd roxy- 1 , 3-benzod ioxol-5-yl )-3-( hyd roxymethyl )- 1 -pentyl- 1 , 3-d ihyd ro-2H- pyrrolo[2 , 3-b]pyrid i n-2-one ;
3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-3-(hydroxymethyl)-1 -pentyl-1 ,3-dihydro-2H- pyrrolo[3,2-ib]pyridin-2-one;
3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-3-(hydroxymethyl)-1 -pentyl-1 ,3-dihydro-2/-/- pyrrolo[3,2-c]pyridin-2-one; 6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-6-(hydroxymethyl)-4-pentyl-4,6-dihydro-5H- thieno[3,2-b]pyrrol-5-one; 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-<b]pyridin-2-one; and 3-(1 ,3-benzodioxol-5-yl)-1 -pentyl-1 , 3-dihydro-2H-pyrrolo[2,3-b]pyridin-2-one.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000037_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6;
R3 is hydrogen, halo or -R9-OR6;
R4 is independently selected from the group consisting of -R9-C(O)R5 and -R9-N(R6)C(O)OR6; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O;
Figure imgf000038_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl;
R1 is alkyl or aralkyl (optionally substituted by one or more substituents selected from the group consisting of halo, haloalkyl, -R9-OR6, heteroaryl and -R9-C(O)OR6);
R3 is -R9-C(O)X, -R9-C(O)OR6 and -R9-C(O)N(R5)R6 where X is bromo or chloro; R4 is independently selected from the group consisting of -R9-C(O)R5 and heteroaryl optionally substituted by one or more substituents selected from the group consisting of halo and R9-OR6; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is O, 1, 2, 3 or 4;
Figure imgf000039_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl or aralkyl optionally substituted by one or more substituents selected from the group consisting of halo and -R9-C(O)OR6; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-OR6, -R9-N(R5)R6, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6,
-N(R6)C(O)R5 and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above; R3 and R4 together form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6, where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a N-heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain. Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000040_0001
is a fused heteroaryl ring selected from the group consisting of pyrrolyl, pyrazolyl, pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl or aralkyl optionally substituted by one or more substituents selected from the group consisting of halo and -R9-C(O)OR6; each R2 is independently selected from the group consisting of alkyl, halo and haloalkyl; or two adjacent R2 groups, together with the heteroaryl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above; R3 and R4 together form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a-C(O)R6, where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a N-heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6; each R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000041_0001
a fused heteroaryl ring selected from the group consisting of pyridinyl, pyrimidinyl, thienyl and pyrazinyl; R1 is alkyl; each R2 is independently selected from the group consisting of alkyl, halo, haloalkyl and -R9-OR6;
R3 is independently selected from the group consisting of halo, -R9-CN, -R9-N(R5)R6 and -N(R6)C(O)OR6; R4 is heteroaryl optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, and -R9-OR6; each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; and each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain.
Another embodiment of the invention is a compound of formula (I), as set forth above in the Summary of the Invention, selected from the group consisting of: 3-(1 ,3-benzodioxol-5-yl)-3-fluoro-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-ό]pyridin-2-one; 3-(1 ,3-benzodioxol-5-yl)-2-oxo-1-pentyl-2,3-dihydro-1H-pyrrolo[2,3-ύ]pyridine-3- carbonitrile; and
3-(1 ,3-benzodioxol-5-yl)-3-(benzylamino)-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3-b]pyridin- 2-one.
One embodiment of the invention is the method of treating or preventing hypercholesterolemia in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I). Another embodiment of the invention is the method of treating or preventing benign prostatic hyperplasia in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
Another embodiment of the invention is the method of treating or preventing pruritis in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
Another embodiment of the invention is the method of treating or preventing cancer in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I) as set forth above for the embodiments of the compounds of formula (I).
Specific embodiments of the compounds of formula (I) are described in more detail below in the Preparation of the Compounds of the Invention.
UTILITY AND TESTING OF THE COMPOUNDS QF THE INVENTION
The compounds of the invention modulate, preferably inhibit, ion flux through a voltage-dependent sodium channel in a mammal, especially in a human. Any such modulation, whether it be partial or complete inhibition or prevention of ion flux, is sometimes referred to herein as "blocking" and corresponding compounds as "blockers". In general, the compounds of the invention modulates the activity of a sodium channel downwards, inhibits the voltage-dependent activity of the sodium channel, and/or reduces or prevents sodium ion flux across a cell membrane by preventing sodium channel activity such as ion flux.
The compounds of the invention inhibit the ion flux through a voltage- dependent sodium channel. Preferably, the compounds are state or frequency dependent modifers of the sodium channels, having a low affinity for the rested/closed state and a high affinity for the inactivated state. These compounds are likely to interact with overlapping sites located in the inner cavity of the sodium conducting pore of the channel similar to that described for other state-dependent sodium channel blockers (Cestele, S., et al., op. cit). These compounds may also be likely to interact with sites outside of the inner cavity and have allosteric effects on sodium ion conduction through the channel pore.
Any of these consequences may ultimately be responsible for the overall therapeutic benefit provided by these compounds.
Accordingly, while not wishing to be bound to any particular mechanism of action, the compounds and pharmaceutical compositions of the invention are useful in the treatment and/or prevention of benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and/or pruritis (itch) in a mammal, preferably a human.
Benign prostatic hyperplasia (BPH), also known as benign prostatic hypertrophy, is one of the most common diseases affecting aging men. BPH is a progressive condition which is characterized by a nodular enlargement of prostatic tissue resulting in obstruction of the urethra. Consequences of BPH can include hypertrophy of bladder smooth muscle, a decompensated bladder, acute urinary retention and an increased incidence of urinary tract infection. BPH has a high public health impact and is one of the most common reasons for surgical intervention among elderly men. Attempts have been made to clarify the etiology and pathogenesis and, to that end, experimental models have been developed. Spontaneous animal models are limited to the chimpanzee and the dog. BPH in man and the dog share many common features. In both species, the development of BPH occurs spontaneously with advanced age and can be prevented by early/prepubertal castration. A medical alternative to surgery is very desirable for treating BHP and the consequences.
The prostatic epithelial hyperplasia in both man and the dog is androgen sensitive, undergoing involution with androgen deprivation and resuming epithelial hyperplasia when androgen is replaced. Cells originating from the prostate gland have been shown to express high levels of voltage gated sodium channels, lmmunostaining studies clearly demonstrated evidence for voltage gated sodium channels in prostatic tissues (Prostate Cancer Prostatic Dis. 2005; 8(3):266-73).
Hypercholesterolemia, i.e., elevated blood cholesterol, is an established risk factor in the development of, e.g., atherosclerosis, coronary artery disease, hyperlipidemia, stroke, hyperinsulinemias, hypertension, obesity, diabetes, cardiovascular diseases (CVD), myocardial ischemia, and heart attack. Thus, lowering the levels of total serum cholesterol in individuals with high levels of cholesterol has been known to reduce the risk of these diseases. The lowering of low density lipoprotein cholesterol in particular is an essential step in the prevention of CVD.
Although there are a variety of hypercholesterolemia therapies, there is a continuing need and a continuing search in this field of art for alternative therapies.
The invention provides compounds which are useful as antihypercholesterolemia agents and their related conditions. The present compounds may act in a variety of ways. While not wishing to be bound to any particular mechanism of action, the compounds may be direct or indirect inhibitors of the enzyme acyl CoA: cholesterol acyl transferase (ACAT) that results in inhibition of the esterification and transport of cholesterol across the intestinal wall. Another possibility may be that the compounds of the invention may be direct or indirect inhibitors of cholesterol biosynthesis in the liver. It is possible that some compounds of the invention may act as both direct or indirect inhibitors of ACAT and cholesterol biosynthesis.
Pruritus, commonly known as itch, is a common dermatological condition. While the exact causes of pruritis are complex and poorly understood, there has long been acknowledged to have interactions with pain. In particular, it is believed that sodium channels likely communicate or propagate along the nerve axon the itch signals along the skin. Transmission of the itch impulses results in the unpleasant sensation that elicits the desire or reflex to scratch.
From a neurobiology level, it is believed that there is a shared complexity of specific mediators, related neuronal pathways and the central processes of itch and pain and recent data suggest that there is a broad overlap between pain- and itch- related peripheral mediators and/or receptors (Ikoma et al., Nature Reviews Neuroscience, 7:535-547, 2006). Remarkably, pain and itch have similar mechanisms of neuronal sensitization in the peripheral nervous system and the central nervous system but exhibits intriguing differences as well.
For example, the mildly painful stimuli from scratching are effective in abolishing the itch sensation. In contrast, analgesics such as opioids can generate severe pruritus. The antagonistic interaction between pain and itch can be exploited in pruritus therapy, and current research concentrates on the identification of common targets for future analgesic and antipruritic therapy.
Compounds of the present invention have been shown to have analgesic effects in a number of animal models at oral doses ranging from 1 mg/kg to 100 mg/kg. The compounds of the invention can also be useful for treating pruritus. The types of itch or skin irritation, include, but are not limited to: a) psoriatic pruritis, itch due to hemodyalisis, aguagenic pruritus, and itching caused by skin disorders (e.g., contact dermatitis), systemic disorders, neuropathy, psychogenic factors or a mixture thereof; b) itch caused by allergic reactions, insect bites, hypersensitivity (e.g., dry skin, acne, eczema, psoriasis), inflammatory conditions or injury; c) itch associated with vulvar vestibulitis; and d) skin irritation or inflammatory effect from administration of another therapeutic such as, for example, antibiotics, antivirals and antihistamines.
The compounds of the invention are also useful in treating or preventing certain hormone sensitive cancers, such as prostate cancer (adenocarcinoma), breast cancer, ovarian cancer, testicular cancer, thyroid neoplasia. The voltage gated sodium channels have been demonstrated to be expressed in prostate and breast cancer cells. Up-regulation of neonatal Na(v)1.5 occurs as an integral part of the metastatic process in human breast cancer and could serve both as a novel marker of the metastatic phenotype and a therapeutic target (Clin. Cancer Res.2005, Aug. 1 ; 11(15): 5381-9). Functional expression of voltage-gated sodium channel alpha-subunits, specifically Na/I .7, is associated with strong metastatic potential in prostate cancer (CaP) in vitro. Voltage-gated sodium channel alpha-subunits immunostaining, using antibodies specific to the sodium channel alpha subunit was evident in prostatic tissues and markedly stronger in CaP vs non-CaP patients (Prostate Cancer Prostatic Dis. 2005;8(3):266-73)
The compounds of the invention are also useful in treating or preventing symptoms associated with BPH such as, but not limited to, acute urinary retention and urinary tract infection.
The compounds of the invention are also useful in treating or preventing certain endocrine imbalances or endocrinopathies such as congenital adrenal hyperplasia , hyperthyroidism, hypothyroidism, osteoporosis, osteomalacia, rickets, Cushing's Syndrome, Conn's syndrome, hyperaldosteronism, hypogonadism, hypergonadism, infertility, fertility and diabetes.
The present invention readily affords many different means for identification of therapeutic agents, especially as sodium channel modulating agents. Identification of the therapeutic agents can be assessed using a variety of in vitro and in vivo assays, e.g., measuring current, measuring membrane potential, measuring ion flux, (e.g. sodium or guanidinium), measuring sodium concentration, measuring second messengers and transcription levels, and using e.g., voltage-sensitive dyes, radioactive tracers, and patch-clamp electrophysiology.
One such protocol involves the screening of chemical agents for ability to modulate the activity of a sodium channel thereby identifying it as a modulating agent. A typical assay described in Bean et al., J. General Physiology (1983), 83:613- 642, and Leuwer, M., et al., Sr. J. Pharmacol (2004), 141(1):47-54, uses patch-clamp techniques to study the behaviour of channels. Such techniques are known to those skilled in the art, and may be developed, using current technologies, into low or medium throughput assays for evaluating compounds for their ability to modulate sodium channel behaviour.
A competitive binding assay with known sodium channel toxins such as tetrodotoxin, alpha-scorpion toxins, aconitine, BTX and the like, may be suitable for identifying potential therapeutic agents with high selectivity for a particular sodium channel. The use of BTX in such a binding assay is well known and is described in McNeal, E.T., et al., J. Med. Chem. (1985), 28(3):381-8; and Creveling, C.R., et al., Methods in Neuroscience, Vol.8: Neurotoxins (Conn PM Ed) (1992), pp. 25-37, Academic Press, New York.
These assays can be carried out in cells, or cell or tissue extracts expressing the channel of interest in a natural endogenous setting or in a recombinant setting. The assays that can be used include plate assays which measure Na+ influx through surrogate markers such as 14C-guanidine influx or determine cell depolarization using fluorescent dyes such as the FRET based and other fluorescent assays or a radiolabeled binding assay employing radiolabeled aconitine, BTX, TTX or STX. More direct measurements can be made with manual or automated electrophysiology systems. The guanidine influx assay is explained in more detail below in the Biological Assays section. Throughput of test compounds is an important consideration in the choice of screening assay to be used. In some strategies, where hundreds of thousands of compounds are to be tested, it is not desirable to use low throughput means. In other cases, however, low throughput is satisfactory to identify important differences between a limited number of compounds. Often it will be necessary to combine assay types to identify specific sodium channel modulating compounds.
Electrophysiological assays using patch clamp techniques is accepted as a gold standard for detailed characterization of sodium channel compound interactions, and as described in Bean et al., op. cit. and Leuwer, M., et al., op. cit. There is a manual low-throughput screening (LTS) method which can compare 2-10 compounds per day; a recently developed system for automated medium-throughput screening (MTS) at 20-50 patches (i.e. compounds) per day; and a technology from Molecular Devices Corporation (Sunnyvale, CA) which permits automated high-throughput screening (HTS) at 1000-3000 patches (i.e. compounds) per day.
One automated patch-clamp system utilizes planar electrode technology to accelerate the rate of drug discovery. Planar electrodes are capable of achieving high- resistance, cells-attached seals followed by stable, low-noise whole-cell recordings that are comparable to conventional recordings. A suitable instrument is the PatchXpress
7000A (Axon Instruments Inc, Union City, CA). A variety of cell lines and culture techniques, which include adherent cells as well as cells growing spontaneously in suspension are ranked for seal success rate and stability. Immortalized cells (e.g.
HEK and CHO) stably expressing high levels of the relevant sodium ion channel can be adapted into high-density suspension cultures.
Other assays can be selected which allow the investigator to identify compounds which block specific states of the sodium channel, such as the open state, closed state or the resting state, or which block transition from open to closed, closed to resting or resting to open. Those skilled in the art are generally familiar with such assays.
Binding assays are also available, however these are of only limited functional value and information content. Designs include traditional radioactive filter based binding assays or the confocal based fluorescent system available from Evotec OAI group of companies (Hamburg, Germany), both of which are HTS.
Radioactive flux assays can also be used. In this assay, channels are stimulated to open with veratridine or aconitine and held in a stabilized open state with a toxin, and channel blockers are identified by their ability to prevent ion influx. The assay can use radioactive 22[Na] and 14[C] guanidinium ions as tracers. FlashPlate &
Cytostar-T plates in living cells avoids separation steps and are suitable for HTS.
Scintillation plate technology has also advanced this method to HTS suitability.
Because of the functional aspects of the assay, the information content is reasonably good. Yet another format measures the redistribution of membrane potential using the
FLIPR system membrane potential kit (HTS) available from Molecular Dynamics (a division of Amersham Biosciences, Piscataway, NJ). This method is limited to slow membrane potential changes. Some problems may result from the fluorescent background of compounds. Test compounds may also directly influence the fluidity of the cell membrane and lead to an increase in intracellular dye concentrations. Still, because of the functional aspects of the assay, the information content is reasonably good.
Sodium dyes can be used to measure the rate or amount of sodium ion influx through a channel. This type of assay provides a very high information content regarding potential channel blockers. The assay is functional and would measure Na+ influx directly. CoroNa Red, SBFI and/or sodium green (Molecular Probes, Inc. Eugene OR) can be used to measure Na influx; all are Na responsive dyes. They can be used in combination with the FLIPR instrument. The use of these dyes in a screen has not been previously described in the literature. Calcium dyes may also have potential in this format.
In another assay, FRET based voltage sensors are used to measure the ability of a test compound to directly block Na influx. Commercially available HTS systems include the VIPR™ Il FRET system (Aurora Biosciences Corporation, San Diego, CA, a division of Vertex Pharmaceuticals, Inc.) which may be used in conjunction with FRET dyes, also available from Aurora Biosciences. This assay measures sub-second responses to voltage changes. There is no requirement for a modifier of channel function. The assay measures depolarization and hyperpolarizations, and provides ratiometric outputs for quantification. A somewhat less expensive MTS version of this assay employs the FLEXstation™ (Molecular Devices Corporation) in conjunction with FRET dyes from Aurora Biosciences. Other methods of testing the compounds disclosed herein are also readily known and available to those skilled in the art.
These results provide the basis for analysis of the structure-activity relationship (SAR) between test compounds and the sodium channel. Certain substituents on the core structure of the test compound tend to provide more potent inhibitory compounds. SAR analysis is one of the tools those skilled in the art may now employ to identify preferred embodiments of the compounds of the invention for use as therapeutic agents.
Modulating agents so identified are then tested in a variety of in vivo models so as to determine if they alleviate the diseases or conditions, especially benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and pruritis (itch), with minimal adverse events. The assays described below in the Biological Assays Section are useful in assessing the biological activity of the instant compounds.
Typically, a successful therapeutic agent of the present invention will meet some or all of the following criteria. Oral availability should be at or above 20%. Animal model efficacy is less than about 0.1 μg to about 100 mg/Kg body weight and the target human dose is between 0.1 μg to about 100 mg/Kg body weight, although doses outside of this range may be acceptable ("mg/Kg" means milligrams of compound per kilogram of body mass of the subject to whom it is being administered). The therapeutic index (or ratio of toxic dose to therapeutic dose) should be greater than 100. The potency (as expressed by IC50 value) should be less than 10 μM, preferably below 1 μM and most preferably below 50 nM. The IC50 ("Inhibitory Concentration - 50%") is a measure of the amount of compound required to achieve 50% inhibition of ion flux through a sodium channel, over a specific time period, in an assay of the invention. Compounds of the present invention in the guanidine influx assay have demonstrated IC50 1S ranging from less than a nanomolar to less than 10 micromolar.
In an alternative use of the invention, the compounds of the invention can be used in in vitro or in vivo studies as exemplary agents for comparative purposes to find other compounds also useful in treatment of, or protection from, the various diseases disclosed herein.
Another aspect of the invention relates to inhibiting Na/1.1 , Nav1.2, Nav1.3, Nav1.4, Nav1.5, Nav1.6, Nav1.7, Na/I .8, or Nav1.9 activity in a biological sample or a patient, which method comprises administering to the patient, or contacting said biological sample with a compound of formula I or a composition comprising said compound. The term "biological sample", as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
Inhibition of Na/1.1 , Na/I .2, Nav1.3, Nav1.4, Nav1.5, Na/I .6, Nav1.7, Na/I .8, or Nav1.9 activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, the study of sodium ion channels in biological and pathological phenomena; and the comparative evaluation of new sodium ion channel inhibitors.
A compound of the invention, as set forth above in the Summary of the Invention, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and/or a pharmaceutical composition of the invention, comprising a pharmaceutically acceptable excipient and one or more compounds of the invention, as set forth above in the Summary of the Invention, as a stereoisomer, enantiomer or tautomer or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, can also be used in the preparation of a medicament for the treatment and/or prevention of hypercholesterolemia, benign prostatic hyperplasia, pruritis, and/or cancer in a mammal. PHARMACEUTICAL COMPOSITIONS OF THE INVENTION AND ADMINISTRATION
Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrastemal injection or infusion techniques. Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see The
Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention.
A pharmaceutical composition of the invention may be in the form of a solid or liquid. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.
When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may be present: binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent. When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension. The liquid may be for oral administration or for delivery by injection, as two examples. When intended for oral administration, preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
The liquid pharmaceutical compositions of the invention, whether they be solutions, suspensions or other like form, may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. An injectable pharmaceutical composition is preferably sterile.
A liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained. Typically, this amount is at least 0.01% of a compound of the invention in the composition. When intended for oral administration, this amount may be varied to be between 0.1 and about 70% of the weight of the composition. Preferred oral pharmaceutical compositions contain between about 4% and about 50% of the compound of the invention. Preferred pharmaceutical compositions and preparations according to the present invention are prepared so that a parenteral dosage unit contains between 0.01 to 10% by weight of the compound prior to dilution of the invention.
The pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers. Thickening agents may be present in a pharmaceutical composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device. Topical formulations may contain a concentration of the compound of the invention from about 0.1 to about 10% w/v (weight per unit volume).
The pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug. The composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient. Such bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
The pharmaceutical composition of the invention may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients may be encased in a gelatin capsule.
The pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
The pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols.
The pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system. The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy. Generally, a therapeutically effective daily dose is (for a 70 kg mammal) from about 0.001 mg/kg (i.e., 0.7 mg) to about 100 mg/kg (i.e., 7.0 gm); preferaby a therapeutically effective dose is (for a 70 kg mammal) from about 0.01 mg/kg (i.e., 7 mg) to about 50 mg/kg (i.e., 3.5 gm); more preferably a therapeutically effective dose is (for a 70 kg mammal) from about 1 mg/kg (i.e., 70 mg) to about 25 mg/kg (i.e., 1.75 gm).
The ranges of effective doses provided herein are not intended to be limiting and represent preferred dose ranges. However, the most preferred dosage will be tailored to the individual subject, as is understood and determinable by one skilled in the relevant arts, (see, e.g., Berkowet al., eds., The Merck Manual, 16th edition, Merck and Co., Rahway, N. J., 1992; Goodmanetna., eds..Goodman and Oilman's The Pharmacological Basis of Therapeutics, 10th edition, Pergamon Press, Inc., Elmsford, N.Y., (2001); Avery's Drug Treatment: Principles and Practice of Clinical Pharmacology and Therapeutics, 3rd edition, ADIS Press, LTD., Williams and Wilkins, Baltimore, MD. (1987), Ebadi, Pharmacology, Little, Brown and Co., Boston, (1985); Osolci al., eds., Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Co., Easton, PA (1990); Katzung, Basic and Clinical Pharmacology, Appleton and Lange, Norwalk, CT (1992)).
The total dose required for each treatment can be administered by multiple doses or in a single dose over the course of the day, if desired. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. The diagnostic pharmaceutical compound or composition can be administered alone or in conjunction with other diagnostics and/or pharmaceuticals directed to the pathology, or directed to other symptoms of the pathology. The recipients of administration of compounds and/or compositions of the invention can be any vertebrate animal, such as mammals. Among mammals, the preferred recipients are mammals of the Orders Primate (including humans, apes and monkeys), Arteriodactyla (including horses, goats, cows, sheep, pigs), Rodenta (including mice, rats, rabbits, and hamsters), and Camivora (including cats, and dogs). Among birds, the preferred recipients are turkeys, chickens and other members of the same order. The most preferred recipients are humans.
For topical applications, it is preferred to administer an effective amount of a pharmaceutical composition according to the invention to target area, e.g., skin surfaces, mucous membranes, and the like, which are adjacent to peripheral neurons which are to be treated. This amount will generally range from about 0.0001 mg to about 1 g of a compound of the invention per application, depending upon the area to be treated, whether the use is diagnostic, prophylactic or therapeutic, the severity of the symptoms, and the nature of the topical vehicle employed. A preferred topical preparation is an ointment, wherein about 0.001 to about 50 mg of active ingredient is used per cc of ointment base. The pharmaceutical composition can be formulated as transdermal compositions or transdermal delivery devices ("patches"). Such compositions include, for example, a backing, active compound reservoir, a control membrane, liner and contact adhesive. Such transdermal patches may be used to provide continuous pulsatile, or on demand delivery of the compounds of the present invention as desired.
The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art. Controlled release drug delivery systems include osmotic pump systems and dissolutional systems containing polymer-coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U.S. Pat. Nos. 3,845,770 and 4,326,525 and in P. J. Kuzma et al, Regional Anesthesia 22 (6): 543-551 (1997), all of which are incorporated herein by reference. The compositions of the invention can also be delivered through intra-nasal drug delivery systems for local, systemic, and nose-to-brain medical therapies. Controlled Particle Dispersion (CPD)™ technology, traditional nasal spray bottles, inhalers or nebulizers are known by those skilled in the art to provide effective local and systemic delivery of drugs by targeting the olfactory region and paranasal sinuses. The invention also relates to an intravaginal shell or core drug delivery device suitable for administration to the human or animal female. The device may be comprised of the active pharmaceutical ingredient in a polymer matrix, surrounded by a sheath, and capable of releasing the compound in a substantially zero order pattern on a daily basis similar to devises used to apply testosterone as desscribed in PCT Patent No. WO 98/50016.
Current methods for ocular delivery include topical administration (eye drops), subconjunctival injections, periocular injections, intravitreal injections, surgical implants and iontophoresis (uses a small electrical current to transport ionized drugs into and through body tissues). Those skilled in the art would combine the best suited excipients with the compound for safe and effective intra-occular administration.
The most suitable route will depend on the nature and severity of the condition being treated. Those skilled in the art are also familiar with determining administration methods (oral, intravenous, inhalation, sub-cutaneous, rectal etc.), dosage forms, suitable pharmaceutical excipients and other matters relevant to the delivery of the compounds to a subject in need thereof. KITS-OF-PARTS
The present invention also provides kits that contain a pharmaceutical composition which includes one or more compounds of the invention. The kit also includes instructions for the use of the pharmaceutical composition for modulating the activity of ion channels, for the treatment of benign prostatic hyperplasia (BPH), hypercholesterolemia, cancer and pruritis (itch), as well as other utilities as disclosed herein. Preferably, a commercial package will contain one or more unit doses of the pharmaceutical composition. For example, such a unit dose may be an amount sufficient for the preparation of an intravenous injection. It will be evident to those of ordinary skill in the art that compounds which are light and/or air sensitive may require special packaging and/or formulation. For example, packaging may be used which is opaque to light, and/or sealed from contact with ambient air, and/or formulated with suitable coatings or excipients.
PREPARATION OF THE COMPOUNDS OF THE INVENTION
The following Reaction Schemes illustrate methods to make compounds of this invention, i.e., compounds of formula (I):
wherein
Figure imgf000056_0001
, p, R1, R2, R3 and R4 are as defined herein, as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
It is understood that in the following description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.
It will also be appreciated by those skilled in the art that in the process described below the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., f-butyldimethylsilyl, f-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include benzyl, f-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(O)-R" (where R" is alkyl, aryl or arylalkyl), p-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters.
Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein.
The use of protecting groups is described in detail in Greene, T.W. and P. G. M. Wuts, Greene's Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. The protecting group may also be a polymer resin such as a Wang resin or a 2-chlorotrityl- chloride resin.
It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as "prodrugs". All prodrugs of compounds of this invention are included within the scope of the invention.
The following Reaction Schemes illustrate methods to make compounds of this invention. It is understood that one skilled in the art would be able to make these compounds by similar methods or by methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make in a similar manner as described below other compounds of formula (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, e.g., Smith, M. B. and J. March, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in this invention.
In the following Reaction Schemes, R1, R2, R4 and p are defined as in the Summary of the Invention unless specifically defined otherwise, and X is Cl or Br.
In general, the compounds of formula (I) of the invention where R3 is -OH can be synthesized following the general procedure as described in REACTION SCHEME 1 .
REACTION SCHEME 1
Figure imgf000058_0001
(101) (102) (106)
R1 X
(105)
Figure imgf000058_0002
R4M (108)
Figure imgf000058_0003
R1
(109) Formula (I)
R1 group can be introduced to an amino compound of formula (101 ) either by reductive amination, which is well-known to those skilled in the art, or formation of an amide by reacting with a corresponding acyl chloride followed by reduction, which is also well-known to those skilled in the art, to form a high order of amino compound of formula (102). Reaction of the compound of formula (102) with oxalyl chloride gives the compound of formula (103). Alternatively, the compound of formula (103) can be obtained by alkylation of the compound of formula (104) with the chloro or bromo compound of formula (105). Alternatively, alkylation of pyrrole-type compound of formula (106) with the chloro or bromo compound of formula (105) provides the compound of formula (107). Treatment of the compound of formula (107) with N- bromosuccinimide in a solvent such as, but not limited to, dimethylsulfoxide, affords the product of formula (103). The treatment of the compound of formula (103) with a nucleophile such as, but not limited to, a Grignard reagent or enolate of formula (108), affords the compound of formula (I) (109) of the invention where R3 is -OH.
In general, the compounds of formula (I) of the invention where R3 is -H can be synthesized following the general procedure as described below in REACTION SCHEME 2.
REACTION SCHEME 2
Figure imgf000059_0001
(109) Formula (I) (201) Formula (I)
The compound of formula (201) can be obtained after the removal of the hydroxyl group of the heterocyclic compound of formula (109) by treating the compound with a silane such as triethylsilane. The compound of formula (201 ) can also be achieved by treating the compound of formula (109) with SOCI2/NEt3 followed by reduction with Zn dust.
In general, the compounds of formula (I) of the invention where R3 is -CH2OH can be synthesized following the general procedure as described below in REACTION SCHEME 3.
REACTION SCHEME 3
Figure imgf000059_0002
(201) Formula (I) (301) Formula (I)
The compound (201 ) is treated with a silyl compound, such as, but not limited to, trimethylsilyl chloride, to generate the silyl ether intermediate, which is treated with ytterbium (III) trifluoromethanesulfonate and formaldehyde to afford the compound of formula (301). Alternatively, a compound of formula (301 ) can be obtained by treating the compound of formula (201) with a base, such as, but not limited to, LiOH1 iPr2NH, LDA, and subsequently reacting with formaldehyde. In general, the compounds of formula (I) of the invention where R3 is fluoro can be synthesized following the general procedure as described below in REACTION SCHEME 4.
REACTION SCHEME 4
Figure imgf000060_0001
(109) Formula (I) (401) Formula (I)
Treatment of the compound of formula (109) with a fluorinating reagent such as, but not limited to, diethylaminosulfur trifluoride (DAST), in a solvent such as, but not limited to, chloroform, provides the fluoro compound of the formula (I) (401 ).
In general, the compounds of formula (I) of the invention where R3 is -CN or -N(R5)R6 can be synthesized following the general procedure as described below in REACTION SCHEME 5.
REACTION SCHEME 5
Figure imgf000060_0002
(109) Formula (I) (501 ) Formula (I)
The hydroxyl group of the compound of the formula (109) can be converted to the corresponding chloro group by reacting with a chloride compound such as, but not limited to, thionyl chloride, in the presence of a base such as, but not limited to, diisopropylethylamine or triethylamine, in a solvent such as, but inot limited to, dichloromethane or chloroform. Treatment of the generated chloride compound with a nucleophile such as, but not limited to, sodium cyanide or benzylamine, in a solvent such as, but not limited to, tetrahedrofuran or dioxane, provides the compound of formula (I) (501).
The following specific Preparations (for the preparation of starting materials and intermediates) and Examples (for the preparation of the compounds of the invention) and the Biological Examples (for the assays used to demonstrate the utility of the compounds of the invention) are provided as a guide to assist in the practice of the invention, and are not intended as a limitation on the scope of the invention.
PREPARATION 1 Synthesis of 1-pentyl-1H-pyrrolo[1 ,2-b]pyrazole-2,3-dione A. Synthesis of Λ/-f(1E)-pentylidenel-1/-/-pyrrol-1 -amine
A mixture of 1H-pyrrol-1 -amine (4.0 g, 49.0 mmol), valeraldehyde (4.10 g, 49.0 mmol) and molecular sieves (4 A) in ethanol (30.0 ml.) was stirred at ambient temperature overnight. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to dryness to give the title compound (unstable): MS (ES+) m/z 151.2 (M + 1 ).
B. Synthesis of Λ/-pentyl-1 H-pyrrol-1 -amine
To a solution of Λ/-[(1E)-pentylidene]-1H-pyrrol-1 -amine in THF (100 ml.) was added LiAIH4 (3.80 g, 100 mmol) in small portions. The reaction mixture was stirred at ambient temperature for 20 h and quenched with the addition of saturated sodium sulfate solution dropwise. The mixture was filtered through celite, and the filtrate was concentrated under reduced pressure. The residue was subjected to column chromatography to afford the title compound (3.65 g, 51%): MS (ES+) m/z 153.2 (M + 1 )-
C. Synthesis of 1 -pentyl-1 H-pyrrolof 1 ,2-iblpyrazole-2,3-dione To a solution of Λ/-pentyl-1/-/-pyrrol-1 -amine (7.0O g, 46.0 mmol) in dichloroethane (200 ml.) was added oxalyl chloride (7.50 g, 60.0 mmol) at -78 0C. The reaction mixture was stirred at ambient temperature overnight and quenched with water. The organic layer was separated, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was subjected to column chromatography to afford the title compound (0.70 g, 7%): MS (ES+) m/z 229.3 (M + 23).
PREPARATION 2 Synthesis of 6-pentyl-4H-thieno[2,3-b]pyrrole-4,5(6H)-dione
A. Synthesis of Λ/-pentylthiophen-2-amine A mixture of 2-iodothiophene (21.0 g, 100 mmol), n-pentylamine (13.5 g, 150 mmol), Cu metal (0.64 g), K3PO4 (42.4 g, 200 mmol) and water (3.60 g) in 2- (dimethylamino)ethanol (100 mL) was heated at 60 0C for 16 hours. The reaction mixture was poured into water and extracted with ether. The ether layer was separated, washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to give the title compound (8.90 g, 53%): MS (ES+) m/z 170.3 (M + 1 ).
B. Synthesis of 6-pentyl-4H-thienor2.3-fclpyrrole-4.5(6/-/Vdione
A mixture of Λ/-pentylthiophen-2-amine (8.90 g, 53.0 mmol) and oxalyl chloride (11.0 g, 87.0 mmol) in chloroform (200 mL) was heated at 60 0C for 5 hours. The reaction mixture was washed with water, brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (0.90 g, 8%): MS (ES+) m/z 246.3 (M + 23).
PREPARATION 3 Synthesis of 4-pentyl-4H-thieno[3,2-6]pyrrole-5,6-dione A. Synthesis of Λ/-3-thienylpentanamide
To a solution of thiophen-3-amine (Galvez, C, et al, J. Heterocycl. Chem. (1984), 21 :393-5) (5.70 g, 57.0 mmol) and triethylamine (5.82 g, 58.0 mmol) in dichloromethane (100 mL) was added pentanoyl chloride (6.93 g, 57.0 mmol) dropwise at 0 0C. The reaction mixture was stirred at ambient temperature overnight and quenched with water (50.0 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to afford the title compound: MS (ES+) m/z 184.3 (M + 1 ).
B. Synthesis of Λ/-pentylthiophen-3-amine
To a solution of Λ/-3-thienylpentanamide (13.4 g, 73.0 mmol) in THF (200 mL) was added LiAIH4 (3.50 g, 100 mmol) at ambient temperature. The resulting mixture was stirred at ambient temperature for 16 h and at 60 0C for 1 h. After cooling down to ambient temperature, the reaction was quenched by the addition of saturated sodium sulfate dropwise until the color changed from green to white and diluted with THF (200 mL). The reaction mixture was filtered through celite and the filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to yield the title compound (9.70 g, 79%): MS (ES+) m/z 170.3 (M + 1 ). C. Synthesis of 4-pentyl-4H-thieno[3,2-/?1pyrrole-5,6-dione
To a solution of Λ/-pentylthiophen-3-amine (7.30 g, 4.30 mmol) in ether (50.0 mL) was added a solution of oxalyl chloride (6.00 ml_, 42.0 mmol) in ether (50.0 mL) slowly at -10 0C. The reaction mixture was stirred at ambient temperature for 3 h and quenched with cold water. The organic layer was separated, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (5.10 g, 53%): MS (ES+) m/z 246.3 (M + 23).
PREPARATION 4 Synthesis of 1 -pentyl-1 H-pyrrolo[2,3-b]pyridine-2,3-dione
A. Synthesis of 1-pentyl-1/-/-pyrrolof2,3-ib1pyridine
To a suspension of sodium hydride in anhydrous Λ/,/V-dimethylformamide (40.0 mL) was added 1H-pyrrolo[2,3-jb]pyridine (5.00 g, 42.4 mmol) at 0 0C. The reaction mixture was stirred for 0.5 h, followed by the addition of 1-bromopentane (9.25 g, 61.2 mmol). The reaction mixture was stirred at ambient temperature for 3.5 h , quenched with water (20.0 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic layers was washed with water (3 x 50.0 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to give the title compound (8.00 g, 100%) as a pale yellow oil: 1H NMR (300 MHz, CDCI3) δ 8.29 (dd, 1 H), 7.86 (d, 1 H), 7.19 (d, 1H), 7.02-6.98 (m, 1 H), 6.41 (d, 1 H), 4.25 (t, 2H), 1.89-1.79 (m, 2H), 1.35-1.25 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 147.4, 142.6, 128.6, 127.9, 120.6, 115.5, 99.2, 44.6, 30.1 , 29.0, 22.4, 13.9.
B. Synthesis of 1-pentyl-1/-/-pyrrolof2,3-blpyridine-2,3-dione
A 2-neck round bottom flask (1 L) was charged with 1 -pentyl-1 H-pyrrolo[2,3- b]pyridine (17.4 g, 92.6 mmol) in anhydrous dimethylsulfoxide (300 mL) and bubbled with nitrogen. To the reaction solution was added Λ/-bromosuccinimide (34.3 g, 193 mmol) in portion over 15 min at 0 0C. The reaction mixture was heated at 60 0C for 6 h followed by at ambient temperature for 16 h. The reaction mixture was diluted with water (200 mL) and stirred for 0.5 h followed by extraction with ethyl acetate (3 x 200 mL). The combined organic layers was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to give the title compound as a yellow solid, which was crystallized from ether as an orange solid (14.6 g, 72%): 1H NMR (300 MHz, CDCI3) δ 8.41 (dd, 1H), 7.78 (dd, 1H), 7.03 (dd, 1 H), 3.79 (t, 2H), 1.77-1.66 (m, 2H), 1.34-1.29 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 219.1 , 182.2, 164.0, 158.2, 155.8, 132.8, 119.4, 112.0, 39.3, 28.9, 27.2, 22.3, 13.9.
PREPARATION 5 Synthesis of 1 -pentyl-1 /-/-pyrrolo[3,2-/b]pyridine-2,3-dione
A. Synthesis of 1-pentyl-1/-/-pyrrolor3,2-άlpyridine
Following the procedure as described in PREPARATION 4A, and making non- critical variations using 1/-/-pyrrolo[3,2-ό]pyridine to replace 1/-/-pyrrolo[2,3-b]pyridine, the title compound was obtained (75%) as a yellow oil: 1H NMR (300 MHz, CDCI3) δ 8.39 (d, 1 H), 7.56 (d, 1 H), 7.25 (d, 1 H), 7.05-7.01 (m, 1 H), 6.63 (d, 1 H), 4.05-3.99 (m, 2H), 1.79-1.72 (m, 2H), 1.31-1.45 (m, 4H), 0.81 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 146.8, 142.9, 131.0, 128.9, 116.5, 116.1 , 102.0, 46.6, 30.0, 29.0, 22.2, 14.0; MS (ES+) m/z 189.3 (M + 1).
B. Synthesis of 1-pentyl-1/-/-pyrrolo[3.2-ιb1pyridine-2,3-dione Following the procedure as described in PREPARATION 4B, and making non- critical variations using 1-pentyl-7/-/-pyrrolo[3,2-b]pyridine to replace 1 -pentyl-1 H- pyrrolo[2,3-t»]pyridine, the title compound was obtained (44%) as a yellow solid: Rf = 0.22 (ethyl acetate/hexane, 30%).
PREPARATION 6 Synthesis of 1-pentyl-1/-/-pyrrolo[3,2-c]pyridine-2,3-dione
Following the procedure as described in PREPARATION 4A, and making non- critical variations using 1/-/-pyrrolo[3,2-c]pyridine-2,3-dione (Rivalle, C, et al, J. Heterocycl. Chem. (1997), 34:441 ) to replace 1H-pyrrolo[2,3-6]pyridine, the title compound was obtained (36%): 1H NMR (300 MHz, CDCI3) δ 8.71-8.64 (m, 2H), 6.90 (d, 1 H), 3.71 (t, 2H), 1.74-1.62 (m, 2H), 1.41-1.27 (m, 4H), 0.89 (t, 3H); MS (ES+) m/z 219.3 (M + 1 ).
EXAMPLE 1 Synthesis of 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1 -pentyl-1 /-/-pyrrolo[1 ,2-b]pyrazol-
2(3H)-one To a solution of 1 -pentyl-1 H-pyrrolo[1 ,2-b]pyrazole-2,3-dione (0.7O g, 3.40 mmol) in THF was added 3,4-(methylenedioxy)phenylmagnesium bromide (4.00 mL, 1.0 M solution in THF/toluene, 4.00 mmol) at 10 0C. The reaction mixture was stirred at ambient temperature for two hours and quenched with saturated ammonium chloride solution. The organic layer was separated, dried over sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (0.09 g, 8%): 1H NMR (300 MHz, CDCI3)δ 7.00 (d, 1 H) , 6.89 (dd, 1H), 6.78 (dd, 1 H), 6.73 (d, 1 H), 6.28-6.18 (m, 2H), 5.93 (s, 2H), 3.89 (dt, 2H), 3.05 (br, 1 H), 1.80-1.68 (m, 2H), 1.32 (dt, 4H), 0.86 (t, 3H); MS (ES+) m/z 351.3 (M + 23).
EXAMPLE 2
Synthesis of 4-(1 ,3-benzodioxol-5-yl)-4-hydroxy-6-pentyl-4,6-dihydro-5H-thieno[2,3- b]pyrrol-5-one
Following the procedure as described in Example 1 , and making non-critical variations using 6-pentyl-4H-thieno[2,3-ό]pyrrole-4,5(6H)-dione to replace 1-pentyl-1H- pyrrolo[1 ,2-b]pyrazole-2,3-dione, the title compound was obtained (32%): 1H NMR (300 MHz, CDCI3) δδ.92-6.71 (m, 5H), 5.92 (s, 2H), 3.75-3.52 (m, 2H), 2.99 (br, 1H), 1.78-1.67 (m, 2H), 1.38-1.28 (m, 4H), 0.87 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 179.5, 147.9, 147.6, 146.9, 133.5, 128.1 , 121.8, 119.1 , 117.1 , 108.1 , 106.6, 101.2, 78.5, 42.7,
28.7, 27.3, 22.2, 13.9; MS (ES+) m/z 368.3 (M + 23).
EXAMPLE 3
Synthesis of 6-(1 ,3-benzodioxol-5-yl)-6-hydroxy-4-pentyl-4,6-dihydro-5H-thieno[3,2- b]pyrrol-5-one
Following the procedure as described in Example 1 , and making non-critical variations using 4-pentyl-4H-thieno[3,2-b]pyrrole-5,6-dione to replace 1-pentyl-1H- pyrrolo[1 ,2-jb]pyrazole-2,3-dione, the title compound was obtained (21%): 1H NMR (300 MHz, CDCI3) δ 7.39 (d, 1 H), 6.91-6.71 (m, 4H), 5.92 (s, 2H), 3.65 (m, 2H), 3.16 (br, 1 H), 1.68 (m, 2H), 1.31 (m, 4H), 0.87 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 180.0, 147.8, 147.5, 146.1, 134.2, 129.5, 122.9, 119.1, 111.8, 108.0, 106.5, 101.2, 79.2, 41.9, 28.8,
27.8, 22.3, 14.0; MS (ES+) m/z 368.3 (M + 23).
EXAMPLE 4
Synthesis of 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H- pyrrolo[2,3-b]pyridin-2-one
To a solution of 1 ,3-benzodioxol-5-ol in THF (40.0 mL) was added a solution of iso-propyl magnesium chloride (7.90 mL, 15.9 mmol, 2.0 M in THF) dropwise at 0 0C over 5 min. The reaction mixture was stirred for 30 min upon which time colorless precipitate formed. After the solvent was removed under reduced pressure, the residue was dissolved in anhydrous dichloromethane (40.0 ml.) and cooled to 0 0C followed by the addition of a solution of 1-pentyl-1H-pyrrolo[2,3-b]pyridine-2,3-dione (1.84 g, 8.44 mmol) in dichloromethane (10.0 ml_). The reaction mixture was stirred at ambient temperature for 16 h and quenched with saturated ammonium chloride solution (30.0 ml_). The organic layer was separated and washed with water (3 x 25.0 ml_), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was crystallized from ethyl acetate and ether to afford the title compound (2.20 g, 73%) as a beige solid: 1H NMR (300 MHz, CDCI3) δ 8.29 (dd, 1 H), 7.74 (dd, 1 H), 7.08 (dd, 1 H), 6.60 (s, 1H), 6.24 (s, 1 H), 5.87 (dd, 2H), 3.78 (d, 2H), 1.77-1.67 (m, 2H), 1.33-1.28 (m, 4H), 0.85 (d, 3H); 13C NMR (75 MHz, DMSO-Gf6) δ 176.9, 157.7, 148.9, 147.3, 147.2, 139.7, 131.1 , 127.7, 119.3, 118.3, 107.1 , 101.1 , 97.8, 74.6, 40.7, 29.0, 27.0, 22.3, 14.4; MS (ES+) m/z 357 (M + 1).
EXAMPLE 5
Synthesis of 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H- pyrrolo[3,2-<b]pyridin-2-one
Following the procedure as described in EXAMPLE 4, and making non-critical variations using 1-pentyl-1/-/-pyrrolo[3,2-b]pyridine-2,3-dione to replace 1-pentyl-1H- pyrrolo[2,3-ib]pyridine-2,3-dione, the title compound was obtained (71%) as a pale yellow solid: 1H NMR (300 MHz, CDCI3) δ 8.17 (d, 1H), 7.29-7.26 (m, 1 H), 7.16 (d, 1 H), 6.52 (s, 1 H), 6.43 (s, 1 H), 5.82 (d, 2H), 3.86-3.76 (m, 1 H), 3.70-3.57 (m, 1H), 1.68-1.63 (m, 2H), 1.33-1.31 (m, 4H), 0.86 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 174.8, 153.3, 151.0, 149.0, 141.8, 141.0, 137.0, 124.8, 116.3, 115.3, 106.8, 101.9, 101.4, 77.5, 40.3, 28.9, 26.8, 22.2, 13.9; MS (ES+) m/z 357.5 (M + 1 ), 339.5 (M - 17).
EXAMPLE 6 Synthesis of 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2/-/- pyrrolo[3,2-c]pyridin-2-one
To a solution of 1 ,3-benzodioxol-5-ol (0.27 g, 1.90 mmol) in THF (10.0 mL) was added iso-propylmagnesium chloride (0.97 mL, 2 M solution in THF, 1.90 mmol) slowly at 0 0C. The mixture was allowed to stir at ambient temperature for 1 hour followed by the addition of 1-pentyl-1H-pyrrolo[3,2-c]pyridine-2,3-dione (0.21 g, 0.96 mmol). The resulting mixture was stirred at ambient temperature overnight, quenched with saturated ammonium chloride (20.0 mL). The mixture was extracted with ethyl acetate (3 x 50.0 mL). The combined organic layers was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo. The residue was subjected to column chromatography (ethyl acetate/hexane, 1/2) to give the title compound (0.52 g, 40%) as a white solid: mp 193-195 0C; 1H NMR (300 MHz, DMSO-J6) δ 9.12 (s, 1 H), 8.30 (d, 1 H), 7.88 (s, 1 H)1 7.22 (s, 1 H), 7.04 (d, 1 H), 6.64 (s, 1H), 6.21 (s, 1 H), 5.93- 5.87 (m, 2H), 3.70-3.50 (m, 2H), 1.63-1.48 (m, 2H), 1.36-1.23 (m, 4H), 0.84 (t, 3H); 13C NMR (75 MHz, DMSO-J6) δ 177.0, 151.4, 150.6, 148.5, 147.3, 143.4, 140.0, 128.6, 119.6, 107.1 , 104.6, 101.2, 97.8, 73.9, 28.9, 26.8, 22.4, 14.4; MS (ES+) m/z 357.2 (M + 1 ).
EXAMPLE 7 Synthesis of 6-hydroxy-6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6-dihydro-5H- thieno[3,2-b]pyrrol-5-one
Following the procedure as described in EXAMPLE 4, and making non-critical variations using 4-pentyl-4H-thieno[3,2-jb]pyrrole-5,6-dione to replace 1-pentyl-1H- pyrrolo[2,3-jb]pyridine-2,3-dione, the title compound was obtained (26%) as a green solid: MS (ES+) m/z 384.4 (M + 23).
EXAMPLE 8 Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3- b]pyridin-2-one
To a solution of 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3- dihydro-2/-/-pyrrolo[2,3-ib]pyridin-2-one (4.00 g, 11.2 mmol) in anhydrous dichloromethane (80.0 mL) was added diisopropyl ethylamine (6.10 mL) and thionyl chloride (2.77 g, 23.5 mmol) under nitrogen at 0 0C. The reaction mixture was stirred at 0 0C for 1 h and concentrated in vacuo to dryness. The residue was dissolved in THF/acetic acid (7:3, 100 mL) followed by the addition of Zn dust (3.08 g, 47.1 mmol) in one portion. The reaction mixture was stirred at ambient temperature for 16 h, filtered and the residue was washed with ethyl acetate (30.0 mL). The filtrate was concentrated in vacuo to dryness. The residue was dissolved in ethyl acetate (200 mL), washed with saturated ammonium chloride (3 x 50.0 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to give the title compound (2.92 g, 76%) as a solid: 1H NMR (300 MHz, CDCI3) δ 8.64 (br, 1 H), 8.26 (d, 1 H), 7.52 (d, 1 H), 7.05 (dd, 1 H), 6.53 (s, 1 H), 6.25 (s, 1 H), 5.84 (d, 2H), 5.02 (s, 1H), 3.86-3.75 (m, 2H), 1.76-1.67 (m, 2H), 1.33-1.28 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 178.5, 157.4, 150.9, 147.8, 147.5, 141.6, 133.2, 121.7, 118.7, 114.1 , 106.4, 101.2, 101.1 , 46.5, 39.8, 28.9, 27.3, 22.3, 13.9; MS (ES+) m/z 341 (M + 1). EXAMPLE 9
Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H-pyrrolo[3,2- b]pyridin-2-one
Following the procedure as described in EXAMPLE 8, and making non-critical variations using 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H- pyrrolo[3,2-<b]pyridin-2-one to replace 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 - pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3-b]pyridin-2-one, the title compound was obtained (50%): MS (ES+) m/z 341.1 (M+1 ).
EXAMPLE 10
Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[3,2- c]pyridin-2-one
A mixture of 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 , 3-dihydro- 2/-/-pyrrolo[3,2-c]pyridin-2-one (0.15 g, 0.42 mmol), triethylsilane (1.60 mL, 10.0 mmol) and trifluroacetic acid (0.74 mL, 10.0 mmol) was stirred at ambient temperature overnight. The mixture was diluted with ethyl acetate (100 mL), washed with water, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo. The residue was triturated with diethyl ether to give the title compound as a white solid (not stable, turned to red in the air): MS (ES+) m/z 341.4 (M + 1 ).
EXAMPLE 11
Synthesis of 6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6-dihydro-5H-thieno[3,2- b]pyrrol-5-one
To a solution of 6-hydroxy-6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6- dihydro-5H- thieno[3,2-ύ]pyrrol-5-one (1.71 g, 4.70 mmol) in CH2CI2 (30.0 mL) were added trifluoroacetic acid (6.00 g, 52.6 mmol) and triethylsilane (5.00 g, 43.0 mmol) at 0 0C. The reaction mixture was stirred at ambient temperature for 16 hours and diluted with CH2CI2 (50.0 mL). The mixture was washed with water (2 x 50.0 mL), dried over Na2SO4 and filtered. The filtrate was evaporated under reduced pressure. The residue was subjected to column chromatography to give the title compound (0.80 g, 49%) as a green solid: MS (ES+) m/z 346.4 (M + 1 ). EXAMPLE 12 Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-3-(hydroxymethyl)-1-pentyl-1 ,3- dihydro-2H-pyrrolo[2,3-b]pyridin-2-one
To a solution of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2/-/- pyrrolo[2,3-6]pyridin-2-one (2.75 g, 8.08 mmol) in anhydrous dichloromethane (40.0 ml.) were added triethylamine (4.91 g, 48.5 mmol) and chlorotrimethylsilane (3.51 g, 32.3 mmol) under nitrogen at 0 0C. The reaction mixture was stirred at 0 0C for 2 h and diluted with anhydrous dichloromethane (50.0 mL). The organic layer was washed with water (2 x 25.0 mL), dried over magnesium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The gummy brown residue was dissolved in THF (40.0 mL) followed by the addition of formaldehyde solution (2.20 mL, 80.8 mmol, 37 wt% in water) and ytterbium (III) trifluoromethanesulfonate (1.25 g, 2.02 mmol). The reaction mixture was stirred at ambient temperature for 36 h and diluted with dichloromethane (100 mL). The organic layer was washed with saturated NaHCO3 (50.0 mL), saturated ammonium chloride (50.0 mL) and water (50.0 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to afford the title compound (2.85 g, 98%): 1H NMR (300 MHz, CDCI3) δ 10.02 (s, 1H), 8.29 (dd, 1 H), 7.72 (dd, 1 H), 7.13 (dd, 1 H), 6.55 (s, 1 H), 6.46 (s, 1H), 5.86 (dd, 2H), 4.37 (dd, 2H), 3.77-3.84 (m, 2H), 3.25 (br, 1 H), 1.63-1.77 (m, 2H), 1.36-1.22 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 179.9, 156.6, 152.3, 148.4, 147.5, 141.5, 133.8, 124.3, 118,7, 111.3, 107.9, 101.9, 101.4, 64.3, 59.1, 39.9, 31.6, 27.2, 22.3, 13.9; MS (ES+) m/z 371.1 (M + 1 ).
EXAMPLE 13
Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-3-(hydroxymethyl)-1-pentyl-1 ,3- dihydro-2/-/-pyrrolo[3,2-b]pyridin-2-one
To a solution of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H- pyrrolo[3,2-/b]pyridin-2-one (1.60 g, 4.70 mmol) in anhydrous tetrahydrofuran (30.0 mL) was added a solution of pre-prepared lithium diisopropylamide (10.3 mmol) in anhydrous tetrahydrofuran (30.0 mL) at -78 0C. The reaction mixture was stirred at -78 0C for 0.5 h followed by the addiotion of para-formaldehyde (0.85 g, 28.2 mmol) in one portion. The reaction was stirred at -78 0C for 2 h and quenched with saturated ammonium chloride (20.0 mL). After the organic solvent was removed under reduced pressure, the residue was diluted with ethyl acetate (50.0 mL). The organic layer was washed with brine (30.0 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness to give the title compound (1.95 g, 100%): 1H NMR (300 MHz, CDCI3) δ 8.22 (dd, 1 H), 7.22-7.12 (m, 2H), 6.51 (s, 1 H)1 6.06 (s, 1 H), 5.83 (d, 2H), 4.89 (s, 2H), 3.83-3.61 (m, 2H), 1.75-1.61 (m, 2H), 1.39-1.29 (m, 4H), 0.89 (t, 3H).
EXAMPLE 14
Synthesis of 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-3-(hydroxymethyl)-1 -pentyl-1 ,3- dihydro-2H-pyrrolo[3,2-c]pyridin-2-one
Following the procedure described in EXAMPLE 13, and making non-critical variations using 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1 ,3-dihydro-2/-/- pyrrolo[3,2-c]pyridin-2-one to replace 3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1 -pentyl-1, 3- dihydro-2H-pyrrolo[3,2-b]pyridin-2-one, the title compound was obtained: MS (ES+) A77/Z 371.4 (M + 1 ).
EXAMPLE 15
Synthesis of 6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-6-(hydroxymethyl)-4-pentyl-4,6- dihydro-5H-thieno[3,2-b]pyrrol-5-one
Following the procedure as described in EXAMPLE 12, and making non-critical variations using 6-(6-hydroxy-1 ,3-benzodioxol-5-yl)-4-pentyl-4,6-dihydro-5H- thieno[3,2-ύ]pyrrol-5-one to replace 3-(6-hydroxy-1 , 3-benzodioxol-5-yl)-1 -pentyl-1 ,3- dihydro-2/-/-pyrrolo[2,3-ib]pyridin-2-one, the title compound was obtained (10%): MS (ES+) /77/z 376.1(M + 1 ), 398.5 (M + 23).
EXAMPLE 16 Synthesis of 3-(1,3-benzodioxol-5-yl)-3-hydroxy-1 -pentyl-1 ,3-dihydro-2/-/-pyrrolo[2,3- b]pyridin-2-one
To a solution of 1-pentyl-7H-pyrrolo[2,3-b]pyridine-2,3-dione (0.32 g, 1.45 mmol) in anhydrous THF (20.0 mL) was added dropwise a solution of (3,4- methylenedioxy)phenyl bromide (2.20 mL, 1.0 M solution in THF/toluene, 2.17 mmol) at -78 0C under nitrogen. The reaction mixture was stirred at ambient temperature overnight and quenched with saturated NH4CI solution (15.0 mL). The mixture was concentrated in vacuo. The aqueous residue was extracted with ethyl acetate, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo to dryness The residue was subjected to column chromatography to give the title compound (0.46 g, 93%): mp 104-105 0C; 1H NMR (300 MHz, CDCI3) δ 8.18 (dd, 1H), 7.48 (dd, 1H), 6.93 (dd, 1 H), 6.89 (d, 1 H), 6.77 (dd, 1 H), 6.71 (d, 1 H), 5.92 (s, 2H), 4.04 (br, 1 H), 3.78 (dt, 2H), 1.77-1.67 (m, 2H), 1.36-1.27 (m, 4H), 0.86 (t, 3H); 13C NMR (75 MHz, CDCI3) δ
177.3, 156.7, 148.6, 148.1 , 147.9, 133.3, 132.3, 126.3, 118.8, 118.8, 108.3, 106.1 , 101.3, 77.2, 39.5, 29.0, 27.3, 22.3, 14.0; MS (ES+) m/z 341 (M + 1 ).
EXAMPLE 17 Synthesis of 3-(1 ,3-benzodioxol-5-yl)-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3-jb]pyridin-2- one
Following the procedure as described in EXAMPLE 8, and making non-critical variations using 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1 ,3-dihydro-2H- pyrrolo[2,3-ib]pyridin-2-one to replace 3-hydroxy-3-(6-hydroxy-1 ,3-benzodioxol-5-yl)-1- pentyl-1 ,3-dihydro-2H-pyrrolo[2,3-jb]pyridin-2-one, the title compound was obtained (75%): mp 75-77 0C; 1H NMR (300 MHz, CDCI3) δ 8.19 (d, 1 H), 7.36 (d, 1H), 6.92 (dd, 1 H), 6.75 (d, 1 H), 6.64 (dd, 1 H), 6.57 (d, 1 H), 5.90 (s, 2H), 4.48 (s, 1 H), 3.85-3.76 (m, 2H), 1.77-1.67 (m, 2H), 1.36-1.27 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ
175.4, 157.6, 148.2, 147.4, 132.2, 129.1 , 123.6, 121.8, 118.2, 108.7, 108.5, 101.2, 50.9 39.5, 29.0, 27.4, 22.4, 14.0; MS (ES+) m/z 326 (M + 1 ).
EXAMPLE 18 Synthesis of 3-hydroxy-3-[2-oxo-2-(2-thienyl)ethyl]-1 -pentyl-1 ,3-dihydro-2H-pyrrolo[2,3- ό]pyridin-2-one
To a mixture of 1 -pentyl-1 /-/-pyrrolo[2,3-b]pyridine-2,3-dione (0.62 g, 2.82 immol) in ethanol (12.0 mL) was added diisopropylethylamine (0.10 mL) and 1- thiophen-2-ylethanone (0.53 g, 4.23 mmol) at ambient temperature. The yellow reaction mixture was heated to reflux for 2 h, cooled to ambient temperature and kept stirring for 17 h upon which time precipitate was formed. The solid was collected by filtration, washed with methanol and ether to afford the title compound (0.63 g, 64%) as a colorless solid: 1H NMR (300 MHz, CDCI3) δ 8.18 (dd, 1 H), 7.68-7.61 (m, 3H), 7.08 (dd, 1 H), 6.89 (dd, 1 H), 4.89 (s, 1H), 3.78-3.71 (m, 3H), 3.43 (d, 1H), 1.75-1.65 (m, 2H), 1.36-1.28 (m, 4H), 0.85 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 190.2, 176.0, 156.9,
148.5, 143.3, 135.1 , 133.2, 131.9, 128.4, 124.5, 118.5, 73.9, 44.9, 39.5, 29.0, 27.2. 22.4, 14.0; MS (ES+) m/z 345 (M + 1 ). EXAMPLE 19 Synthesis of 3-[2-(2-furyl)-2-oxoethyl]-3-hydroxy-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3- b]pyridin-2-one
Following the procedure as described in EXAMPLE 18, making non-critical variations using 1-furan-2-ylethanone to replace 1-thiophen-2-ylethanone, the title compound was obtained (71%) as a colorless solid: 1H NMR (300 MHz, CDCI3) δ 8.14 (dd, 1 H), 7.60 (dd, 1 H), 7.54 (d, 1 H), 7.18 (d, 1 H), 6.89 (dd, 1 H), 6.50 (dd, 1 H), 4.82 (s, 1 H), 3.74 (t, 2H)1 3.49 (ABq, 2H), 1.75-1.65 (m, 2H), 1.34-1.28 (m, 4H), 0.84 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 186.1 , 176.1 , 156.9, 152.0, 148.5, 147.3, 131.9, 124.4, 118.5, 112.7, 73.9, 44.0, 39.4, 29.0, 27.1 , 22.3, 14.0; MS (ES+) m/z 330 (M + 2).
EXAMPLE 20 Synthesis of 3-(1 ,3-benzodioxol-5-yl)-3-fluoro-1-pentyl-1 ,3-dihydro-2H-pyrrolo[2,3- b]pyridin-2-one
A solution of 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1 ,3-dihydro-2H- pyrrolo[2,3-ib]pyridin-2-one (0.26 g, 0.76 mmol) in anhydrous chloroform (2.00 mL) was added dropwise to a solution diethylaminosulfur trifluoride (DAST) (0.18 g, 1.14 mmol) in anhydrous chloroform (7.00 mL) over 45 min under nitrogen at 0 0C. The yellow reaction mixture was stirred at 0 0C for 4 h and diluted with ether (10.0 mL). The mixture was washed with water (2 x 5.00 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (0.17 g, 64%): \1H NMR (300 MHz, CDCI3) δ 8.31 (dt, 1 H), 7.67 (dt, 1H), 7.09 (dd, 1 H), 6.96 (d, 1 H), 6.82 (d, 1 H), 6.77-6.73 (m, 1 H), 5.98 (d, 2H), 3.73 (t, 2H), 1.73-1.63 (m, 2H), 1.35-1.19 (m, 4H), 0.83 (t, 3H); 13C NMR (75 MHz, CDCI3) δ 171.9, 171.6, 157.7, 157.6, 150.3, 150.3, 148.9 148.8, 148.3, 133.7 128.9 128.5, 121.2, 120.9, 119.8, 119.8, 119.1 , 119.1 ,
117.3, 108.1 , 106.3, 106.2, 102.0, 39.1 , 28.6, 26.8, 22.0, 13.2; MS (ES+) m/z 343 (M + 1 ), 323 (M - F).
EXAMPLE 21
Synthesis of 3-(1 ,3-benzodioxol-5-yl)-2-oxo-1 -pentyl-2,3-dihydro-1 /-/-pyrrolo[2, 3- 6]pyridine-3-carbonitrile
To a solution of 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1 ,3-dihydro-2H- pyrrolo[2,3-b]pyridin-2-one (0.68 g, 2.00 mmol) in anhydrous dichloromethane (20.0 mL) was added diisopropylethylamine (0.78 g, 6.00 mmol) followed by the addition of thionyl chloride (0.47 g, 4.00 mmol) at 0 0C. The reaction mixture was stirred for 0.5 h and concentrated under reduced pressure. The gummy residue was dissolved in anhydrous tetrahydrofuran (20.0 mL) followed by the addition of sodium cyanide (0.20 g, 4.00 mmol). The reaction mixture was stirred at ambient temperature for 16 h, diluted with water (20.0 mL) and extracted with ethyl acetate (3 x 50.0 mL). The combined organic layers was washed with water (20.0 mL), saturated ammonium chloride (30.0 mL), and brine (20.0 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (0.46 g, 65%): MS (ES+) m/z 350 (M + 1).
EXAMPLE 22
Synthesis of 3-(1 ,3-benzodioxol-5-yl)-3-(benzylamino)-1-pentyl-1 ,3-dihydro-2/-/- pyrrolo[2,3-jfc>]pyridin-2-one To a solution of 3-(1 ,3-benzodioxol-5-yl)-3-hydroxy-1-pentyl-1 ,3-dihydro-2H- pyrrolo[2,3-jb]pyridin-2-one (0.68 g, 2.00 mmol) in anhydrous dichloromethane (20.0 mL) was added diisopropylethylamine (0.78 g, 6.00 mmol) and thionyl chloride (0.47 g, 4.00 mmol) at 0 °C. The reaction mixture was stirred for 0.5 h, and concentrated under reduced pressure. The gummy residue was dissolved in anhydrous dioxane (20.0 mL) followed by the addition of benzylamine (0.43 g, 4.00 mmol). The reaction mixture was heated at reflux for 16 h, cooled to ambient temperature, diluted with water (20.0 mL) and extracted with ethyl acetate (3 x 50.0 mL). The combined organic layers was washed with water (20.0 mL), saturated ammonium chloride (30.0 mL), and brine (20.0 mL). The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to dryness. The residue was subjected to column chromatography to afford the title compound (0.63 g, 73%) as a gummy material: MS (ES+) m/z 430 (M + 1 ).
BIOLOGICAL ASSAYS
Various techniques are known in the art for testing the activity of compounds of the invention. In order that the invention described herein may be more fully understood, the following biological assays are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner. BIOLOGICAL EXAMPLE 1 Guanidine Influx Assay (in vitro assay)
This example describes an in vitro assay for testing and profiling test agents against human or rat sodium channels stably expressed in cells of either an endogenous or recombinant origin. The assay is also useful for determining the IC-50 of a sodium channel blocking compound. The assay is based on the guanidine flux assay described by Reddy, N. L., et al., J. Med. Chem. (1998), 41(17):3298-302.
The guanidine influx assay is a radiotracer flux assay used to determine ion flux activity of sodium channels in a high-throughput microplate-based format. The assay uses 14C-guanidine hydrochloride in combination with various known sodium channel modulators, to assay the potency of test agents. Potency is determined by an IC-50 calculation. Selectivity is determined by comparing potency of the compound for the channel of interest to its potency against other sodium channels (also called 'selectivity profiling'). Each of the test agents is assayed against cells that express the channels of interest. Voltage gated sodium channels are either TTX sensitive or insensitive. This property is useful when evaluating the activities of a channel of interest when it resides in a mixed population with other sodium channels. The following Table 1 summarizes cell lines useful in screening for a certain channel activity in the presence or absence of TTX.
TABLE 1
Figure imgf000074_0001
Figure imgf000075_0001
It is also possible to employ recombinant cells expressing these sodium channels. Cloning and propagation of recombinant cells are known to those skilled in the art (see, for example, Klugbauer, N, et al., EMBO J. (1995), 14(6): 1084-90; and Lossin, C, et al., Neuron (2002), 34:877-884).
Cells expressing the channel of interest are grown according to the supplier or in the case of a recombinant cell in the presence of selective growth media such as G418 (Gibco/lnvitrogen) The cells are disassociated from the culture dishes with an enzymatic solution (1X) Trypsin/EDTA (Gibco/lnvitrogen) and analyzed for density and viability using haemocytometer (Neubauer). Disassociated cells are washed and resuspended in their culture media then plated into Scintiplates (Beckman Coulter Inc.) (approximately 100,000 cells/ well) and incubated at 37 °C/5 % CO2. for 20-24 hours. After an extensive wash with Low sodium HEPES-buffered saline solution (LNHBSS) (150 mM Choline Chloride, 20 nM HEPES (Sigma), 1mM Calcium Chloride, 5mM Potassium Chloride, 1 mM Magnesium Chloride, 10 mM Glucose) agents diluted with LNHBSS are added to each well. (Varying concentrations of test agent may be used). The activation/radiolabel mixture contains aconitine (Sigma), and 14C-guanidine hydrochloride (ARC).
After loading the cells with test agent and activation/radiolabel mixture, the Scintiplates are incubated at ambient temperature. Following the incubation, the Scintplates are extensively washed with LNHBSS supplemented with guanidine (Sigma). The Scintiplates are dried and then counted using a Wallac MicroBeta TriLux (Perkin-Elmer Life Sciences). The ability of the test agent to block sodium channel activity is determined by comparing the amount of 14C-guanidine present inside the cells expressing the different sodium channels. Based on this data, a variety of calculations, as set out elsewhere in this specification, may be used to determine whether a test agent is selective for a particular sodium channel. IC-50 value of a test agent for a specific sodium channel may be determined using the above general method. IC-50 may be determined using a 3, 8, 10, 12 or 16 point curve in duplicate or triplicate with a starting concentration of 1 , 5 or 10μM diluted serially with a final concentration reaching the sub-nanomolar, nanomolar and low micromolar ranges. Typically the mid-point concentration of test agent is set at 1 μM, and sequential concentrations of half dilutions greater or smaller are applied (e.g. 0.5 μM; 5 μM and 0.25 μM; 10 μM and 0.125 μM; 20 μM etc.). The IC-50 curve is calculated using the 4 Parameter Logistic Model or Sigmoidal Dose-Response Model formula (fit = (A+((B-A)/(1+((C/x)ΛD)))). The fold selectivity, factor of selectivity or multiple of selectivity, is calculated by dividing the IC-50 value of the test sodium channel by the reference sodium channel, for example, Nav1.5.
Representative compounds of the invention, when tested in the above assay using a known cell line that expresses a sodium channel, demonstrated an IC50 (nM) activity level as set forth below in Table 2 wherein "A" refers to an IC50 activity level of from 1 nM to 10 nM, "B" refers to an IC50 activity level from 10 nM to 100 nM, "C" refers to an IC50 activity level from 100 nM to 1000 nM, and "D" refers to an IC50 activity level equal to or greater than 1000 nM. The Synthetic Example numbers provided in Table 2 correspond to the Synthetic Examples herein: TABLE 2
Figure imgf000076_0001
Figure imgf000077_0001
BIOLOGICAL EXAMPLE 2 Electrophysiological Assay (In vitro assay)
Cells expressing the channel of interest were cultured in DMEM growth media (Gibco) with 0.5mg/mL G418, +/-1 % PSG, and 10% heat-inactivated fetal bovine serum at 37C° and 5% CO2. For electrophysiological recordings, cells were plated on 10mm dishes.
Whole cell recordings were examined by established methods of whole cell voltage clamp (Bean et al., op. cit.) using an Axopatch 200B amplifier and Clampex software (Axon Instruments, Union City, CA). All experiments were performed at ambient temperature. Electrodes were fire-polished to resistances of 2-4 Mohm.s Voltage errors and capacitance artifacts were minimized by series resistance compensation and capacitance compensation, respectively. Data were acquired at 40 kHz and filtered at 5 kHz. The external (bath) solution consisted of: NaCI (140 mM), KCI (5 mM), CaCI2 (2 mM), MgCI2 (1 mM), HEPES (10 mM) at pH 7.4. The internal (pipette) solution consisted of (in mM): NaCI (5), CaCI2 (0.1 ) MgCI2 (2), CsCI (10), CsF (120), HEPES (10), EGTA (10), at pH 7.2. To estimate the steady-state affinity of compounds for the resting and inactivated state of the channel (Kr and K1, respectively), 12.5 ms test pulses to depolarizing voltages from -60 to +90 m V from a holding potential of -110 mVwas used to construct current-voltage relationships (I- V curves). A voltage near the peak of the IV-curve (-30 to 0 m V) was used as the test pulse throughout the remainder of the experiment. Steady-state inactivation (availability) curves were then constructed by measuring the current activated during a 8.75 ms test pulse following 1 second conditioning pulses to potentials ranging from -110 to -10 m V. To monitor channels at steady-state, a single "diary" protocol with a holding potential of -110 mVwas created to record the resting state current (10 ms test pulse), the current after fast inactivation (5 ms pre-pulse of -80 to -50 mV followed by a 10 ms test pulse), and the current during various holding potentials (35 ms ramp to test pulse levels). Compounds were applied during the "diary" protocol and the block was monitored at 15s intervals.
After the compounds equilibrated, the voltage-dependence of the steady-state inactivation in the presence of the compound was determined. Compounds that block the resting state of the channel decreased the current elicited during test pulses from all holding potentials, whereas compounds that primarily blocked the inactivated state decreased the current elicited during test pulses at more depolarized potentials. The currents at the resting state (lrest) and the currents during the inactivated state (I activated) were used to calculate steady-state affinity of compounds. Based on the Michaelis- Menton model of inhibition, the Kr and K, was calculated as the concentration of compound needed to cause 50% inhibition of the lrest or the l,nactιvated. respectively.
% inhibition = Vmay*rDruqlh [Drug]h + K x nm h
Vmax is the rate of inhibition, h is the Hill coefficient (for interacting sites), Km is Michaelis-Menten constant, and [Drug] is the concentration of the test compound. At 50% inhibition (1/2Vmax) of the lrest or Inactivated, the drug concentration is numerically equal to Km and approximates the Kr and K1, respectively.
BIOLOGICAL EXAMPLE 3
In Vivo Assay for Benign Prostate Hyperplasia (BPH) The effectiveness of the compounds of the present invention for treating BPH was demonstrated by the following in vivo assay. Dogs were closed orally with compounds of the present invention at oral doses of between 0 mg/kg and 100 mg/kg for a period of 4 weeks. A control group received placebo. The animals were sacrificed and the prostate glands dissected out, dabbed dry and then weighed. Compounds of the present invention were shown to be efficacious in a dose dependent manner within a range of 5 mg/kg and 100 mg/kg in significantly reducing the weight of the prostate in dogs when compared to the vehicle treated (0 mg/kg) controls. These compounds had no adverse events, making them ideal candidates for the safe treatment of BPH and the associated symptoms, such as, but not limited to, acute urinary retention and urinary tract infection.
BIOLOGICAL EXAMPLE 4
In Vivo Assay for Antihypercholesterlemia Efficacy and Antiatherosclerotic Efficacy The compounds of this invention possess antihypercholesterolemia efficacy and antiatherosclerotic efficacy, as evidenced by their activity in the assays described below. Dogs have cardiovascular systems similar to that of humans, making them ideal for studying the effects of medicinal compounds designed to treat cardiovascular disorders.
Dogs were dosed orally at a range of 0 mg/kg to 100 mg/kg daily with compounds of the present invention for a period of 2- 4 weeks. After 2 and 4 weeks the animals were bled and their serum collected for total cholesterol analysis and compared to the animals dosed with vehicle alone (0 mg/kg).
The measurement of cholesterol is one of the most common tests performed in the clinical laboratory setting. Simple fluorometric methods for the sensitive quantitation of total cholesterol in plasma or serum are commonly used. In one assay, cholesteryl esters in the sample are first hydrolyzed by cholesterol esterase. All cholesterol, whether previously esterified or existing free in the circulation, is then oxidized by cholesterol oxidase to the corresponding ketone and hydrogen peroxide.
ADHP (10-acetyl-3,7-dihydroxyphenoxazine) is utilized as a highly sensitive and stable probe for hydrogen peroxide. Horseradish peroxidase catalyzes the reaction of ADHP with hydrogen peroxide to yield the highly fluorescent product resorufin, which can be monitored using excitation wavelengths of 565-580 nm and emission wavelengths of
585-595 nm.
The compounds of the invention exhibited the ability to affect a significant drop in total serum cholesterol when administered in the above assay to dogs in a daily oral dose range of 5-100 mg/kg over a 2- and 4-week period.
BIOLOGICAL EXAMPLE 5 In Vivo Assay for Treatment of Pruritis
The compounds of the invention can be evaluated for their activity as antipruritic agents by in vivo test using rodent models. One established model for peripherally elicited pruritus is through the injection of serotonin into the rostral back area (neck) in hairless rats. Prior to serotonin injections (e.g., 2 mg/ml, 50 μL), a dose of a compound of the present invention can be applied systemically through oral, intravenous or intraperitoneal routes or topically to a circular area fixed diameter (e.g. 18 mm). Following dosing, the serotonin injections are given in the area of the topical dosing. After serotonin injection the animal behaviour is monitored by video recording for 20 min-1.5 h, and the number of scratches in this time compared to vehicle treated animals. Thus, application of a compound of the current invention could suppress serotonin-induced scratching in rats.
* * * * *
All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

WHAT IS CLAIMED IS
1. A method of treating or preventing hypercholesterolemia in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I):
Figure imgf000081_0001
wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000081_0002
is a fused heteroaryl ring or a fused heterocyclyl ring; R1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R9-C(O)R6, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2 or -R10-O-R10-OR6; or R1 is aralkyl substituted by -C(O)N(R7)R8 where: R7 is hydrogen, alkyl, aryl or aralkyl; and R8 is hydrogen, alkyl, haloalkyl, -R10-CN, -R10-OR6, -R10-N(R5)R6, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R7 and R8, together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R7 and R8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl,
-R9-CN, -R9-OR6, heterocyclyl and heteroaryl; or R1 is aralkyl substituted by one or more substituents selected from the group consisting of -R9-OR6, -R9-C(O)OR6, halo, haloalkyl, alkyl, nitro, cyano, aryl (optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R1 is -R10-N(R11)R12, -R10-N(R13)C(O)R12 or -R10-N(R11)C(O)N(R11)R12 where: each R11 is hydrogen, alkyl, aryl or aralkyl; each R12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R10-OC(O)R6, -R10-C(O)OR6, -R10-C(O)N(R5)R6, -R10-C(O)R6, -R10-OR6, or -R10-CN;
R13 is hydrogen, alkyl, aryl, arakyl or -C(O)R6; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R11 and R12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R9-CN, -R9-OR6, -R9-C(O)R6, heterocyclyl and heteroaryl; or R1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R9-OR6, -R9-C(O)OR6, aryl and aralkyl; each R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -C(S)R5, -C(R5)2C(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(=N-CN)N(R5)R6, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring or the fused heterocyclyl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)X, -C(S)R5, -C(R5)2C(O)R6, -R9-OC(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -Si(R6)3, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6,, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(N=C(R5)R6)N(R5)R6, wherein X is bromo or chloro, each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl groups for R3 and R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
2. A method of treating or preventing benign prostatic hyperplasia in a mammal, wherein the methods comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I):
Figure imgf000084_0001
wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000084_0002
is a fused heteroaryl ring or a fused heterocyclyl ring; R1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R9-C(O)R6, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2 or -R10-O-R10-OR6; or R1 is aralkyl substituted by -C(O)N(R7)R8 where: R7 is hydrogen, alkyl, aryl or aralkyl; and
R8 is hydrogen, alkyl, haloalkyl, -R10-CN, -R10-OR6, -R10-N(R5)R6, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R7 and R8, together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R7 and R8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, -R9-CN, -R9-OR6, heterocyclyl and heteroaryl; or R1 is aralkyl substituted by one or more substituents selected from the group consisting of -R9-OR6, -R9-C(O)OR6, halo, haloalkyl, alkyl, nitro, cyano, aryl (optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R1 is -R10-N(R11)R12, -R10-N(R13)C(O)R12 or -R10-N(R11)C(O)N(R11)R12 where: each R11 is hydrogen, alkyl, aryl or aralkyl; each R12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R10-OC(O)R6, -R10-C(O)OR6, -R10-C(O)N(R5)R6, -R10-C(O)R6, -R10-OR6, or -R10-CN;
R13 is hydrogen, alkyl, aryl, arakyl or -C(O)R6; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R11 and R12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R9-CN, -R9-OR6, -R9-C(O)R6, heterocyclyl and heteroaryl; or R1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R9-OR6, -R9-C(O)OR6, aryl and aralkyl; each R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -C(S)R5, -C(R5)2C(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(=N-CN)N(R5)R6, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2) -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring or the fused heterocyclyl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)X, -C(S)R5, -C(R5)2C(O)R6, -R9-OC(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -Si(R6)3, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6,, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(N=C(R5)R6)N(R5)R6, wherein X is bromo or chloro, each m is independently 0, 1, or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl groups for R3 and R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a
N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
3. A method of treating or preventing pruritis in a mammal, wherein the methods comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I):
Figure imgf000087_0001
wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000087_0002
is a fused heteroaryl ring or a fused heterocyclyl ring;
R is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R9-C(O)R6, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2 or -R10-O-R10-OR6; or R1 is aralkyl substituted by -C(O)N(R7)R8 where: R7 is hydrogen, alkyl, aryl or aralkyl; and R8 is hydrogen, alkyl, haloalkyl, -R10-CN, -R10-OR6, -R10-N(R5)R6, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R7 and R8, together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R7 and R8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, -R9-CN, -R9-OR6, heterocyclyl and heteroaryl; or R1 is aralkyl substituted by one or more substituents selected from the group consisting of -R9-OR6, -R9-C(O)OR6, halo, haloalkyl, alkyl, nitro, cyano, aryl
(optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R1 is -R10-N(R11)R12, -R10-N(R13)C(O)R12 or -R10-N(R11)C(O)N(R11)R12 where: each R11 is hydrogen, alkyl, aryl or aralkyl; each R12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R10-OC(O)R6, -R10-C(O)OR6, -R10-C(O)N(R5)R6, -R10-C(O)R6, -R10-OR6, or -R10-CN;
R13 is hydrogen, alkyl, aryl, arakyl or -C(O)R6; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R11 and R12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R9-CN, -R9-OR6, -R9-C(O)R6, heterocyclyl and heteroaryl; or R1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R9-OR6, -R9-C(O)OR6, aryl and aralkyl; each R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2,
-R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -C(S)R5,
-C(R5)2C(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(=N-CN)N(R5)R6, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring or the fused heterocyclyl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)X, -C(S)R5, -C(R5)2C(O)R6, -R9-OC(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -Si(R6)3> -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6,, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(N=C(R5)R6)N(R5)R6, wherein X is bromo or chloro, each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl groups for R3 and R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
4. A method of treating or preventing cancer in a mammal, wherein the methods comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I):
Figure imgf000090_0001
wherein: p is 0, 1 , 2, 3 or 4;
Figure imgf000091_0001
a fused heteroaryl ring or a fused heterocyclyl ring; R1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, aralkyl, aralkenyl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R9-C(O)R6, -R9-C(O)OR6,
-R9-C(O)N(R5)R6, -R9-OR6, -R9-CN, -R10-P(O)(OR6)2 or -R10-O-R10-OR6; or R1 is aralkyl substituted by -C(O)N(R7)R8 where:
R7 is hydrogen, alkyl, aryl or aralkyl; and
R8 is hydrogen, alkyl, haloalkyl, -R10-CN, -R10-OR6, -R10-N(R5)R6, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R7 and R8, together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl group for R7 and R8 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, -R9-CN, -R9-OR6, heterocyclyl and heteroaryl; or R1 is aralkyl substituted by one or more substituents selected from the group consisting of -R9-OR6, -R9-C(O)OR6, halo, haloalkyl, alkyl, nitro, cyano, aryl
(optionally substituted by cyano), aralkyl (optionally substituted by one or more alkyl groups), heterocyclyl and heteroaryl; or R1 is -R10-N(R11)R12, -R10-N(R13)C(O)R12 or -R10-N(R11)C(O)N(R11)R12 where: each R11 is hydrogen, alkyl, aryl or aralkyl; each R12 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R10-OC(O)R6, -R10-C(O)OR6, -R10-C(O)N(R5)R6, -R10-C(O)R6, -R10-OR6, or -R10-CN;
R13 is hydrogen, alkyl, aryl, arakyl or -C(O)R6; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R11 and R12 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R9-CN, -R9-OR6, -R9-C(O)R6, heterocyclyl and heteroaryl; or R1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of alkyl, halo, haloalkyl, -R9-OR6, -R9-C(O)OR6, aryl and aralkyl; each R2 is independently selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -C(S)R5, -C(R5)2C(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(=N-CN)N(R5)R6, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R2 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or two adjacent R2 groups, together with the fused heteroaryl ring or the fused heterocyclyl ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, aryl, heterocyclyl and heteroaryl, and the remaining R2 groups, if present, are as described above;
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -N=C(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)X, -C(S)R5, -C(R5)2C(O)R6, -R9-OC(O)R6, -R9-C(O)OR6, -C(S)OR5, -R9-C(O)N(R5)R6, -C(S)N(R5)R6, -Si(R6)3, -N(R6)C(O)R5, -N(R6)C(S)R5, -N(R6)C(O)OR6, -N(R6)C(S)OR5, -N(R6)C(O)N(R5)R6, -N(R6)C(S)N(R5)R6, -N(R6)S(O)nR5, -N(R6)S(O)nN(R5)R6, -R9-S(O)nN(R5)R6,, -N(R6)C(=NR6)N(R5)R6, and -N(R6)C(N=C(R5)R6)N(R5)R6, wherein X is bromo or chloro, each m is independently 0, 1 , or 2 and each n is independently 1 or 2; and wherein each of the cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, aralkynyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl groups for R3 and R4 is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, halo, haloalkyl, haloalkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, oxo, -R9-CN, -R9-NO2, -R9-OR6, -R9-N(R5)R6, -S(O)mR5, -R9-C(O)R5, -R9-C(O)OR6, -R9-C(O)N(R5)R6, -N(R6)C(O)R5, and -N(R6)S(O)nR5, wherein each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R3 and R4 together may form =NS(O)2R6, =N-R15, =N-O-R6 or =R9a -C(O)R6 (where R9a is a straight or branched alkenylene chain wherein the alkenylene chain is attached to the carbon to which R3 and R4 is attached through a double bond and R15 is a heterocyclyl optionally substituted by alkyl, haloalkyl or -R9-OR6); each R5 and R6 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl and optionally substituted heteroaryl; or when R5 and R6 are each attached to the same nitrogen atom, then R5 and R6, together with the nitrogen atom to which they are attached, may form a N-heterocyclyl or N-heteroaryl; each R9 is a direct bond or an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; and each R10 is an optionally substituted straight or branched alkylene chain, an optionally substituted straight or branched alkenylene chain or an optionally substituted straight or branched alkynylene chain; as a stereoisomer, enantiomer, tautomer thereof or mixtures thereof; or a pharmaceutically acceptable salt, solvate or prodrug thereof.
PCT/US2007/081318 2006-10-12 2007-10-12 Use of heterocyclic compounds as therapeutic agents WO2008046082A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85123606P 2006-10-12 2006-10-12
US60/851,236 2006-10-12

Publications (2)

Publication Number Publication Date
WO2008046082A2 true WO2008046082A2 (en) 2008-04-17
WO2008046082A3 WO2008046082A3 (en) 2008-06-19

Family

ID=39283677

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/081318 WO2008046082A2 (en) 2006-10-12 2007-10-12 Use of heterocyclic compounds as therapeutic agents

Country Status (1)

Country Link
WO (1) WO2008046082A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7700641B2 (en) 2005-04-11 2010-04-20 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
US7799798B2 (en) 2005-04-11 2010-09-21 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8263606B2 (en) 2008-10-17 2012-09-11 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8445696B2 (en) 2009-10-14 2013-05-21 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8450358B2 (en) 2009-06-29 2013-05-28 Xenon Pharmaceuticals Inc. Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
US8466188B2 (en) 2006-10-12 2013-06-18 Xenon Pharmaceuticals Inc. Use of spiro-oxindole compounds as therapeutic agents
US8993574B2 (en) 2008-04-24 2015-03-31 F2G Ltd Pyrrole antifungal agents
US9504671B2 (en) 2010-02-26 2016-11-29 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
US9682033B2 (en) 2015-02-05 2017-06-20 Teva Pharmaceuticals International Gmbh Methods of treating postherpetic neuralgia with a topical formulation of a spiro-oxindole compound
US10201524B2 (en) 2014-11-21 2019-02-12 F2G Limited Antifungal agents
US10709714B2 (en) 2013-11-22 2020-07-14 Clifton Life Sciences LLC Gastrin antagonists for treatment and prevention of osteoporosis
US11819503B2 (en) 2019-04-23 2023-11-21 F2G Ltd Method of treating coccidioides infection

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006110654A1 (en) * 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006110654A1 (en) * 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ANGER T ET AL: "MEDICINAL CHEMISTRY OF NEURONAL VOLTAGE-GATED SODIUM CHANNEL BLOCKERS" JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, US, vol. 44, no. 2, 18 January 2001 (2001-01-18), pages 115-137, XP009049196 ISSN: 0022-2623 *
FRASER SCOTT P ET AL: "Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis" CLINICAL CANCER RESEARCH, THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 11, no. 15, 1 August 2005 (2005-08-01), pages 5381-5389, XP002415107 ISSN: 1078-0432 *
SMITH P ET AL: "Sodium channel protein expression enhances the invasiveness of rat and human prostate cancer cells" FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 423, no. 1, 13 February 1998 (1998-02-13), pages 19-24, XP004261853 ISSN: 0014-5793 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7799798B2 (en) 2005-04-11 2010-09-21 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents
US7935721B2 (en) 2005-04-11 2011-05-03 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
US8106087B2 (en) 2005-04-11 2012-01-31 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
US7700641B2 (en) 2005-04-11 2010-04-20 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
US8466188B2 (en) 2006-10-12 2013-06-18 Xenon Pharmaceuticals Inc. Use of spiro-oxindole compounds as therapeutic agents
US9452168B2 (en) 2008-04-24 2016-09-27 F2G Ltd Pyrrole antifungal agents
US8993574B2 (en) 2008-04-24 2015-03-31 F2G Ltd Pyrrole antifungal agents
US8916580B2 (en) 2008-10-17 2014-12-23 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US9458178B2 (en) 2008-10-17 2016-10-04 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8263606B2 (en) 2008-10-17 2012-09-11 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8415370B2 (en) 2008-10-17 2013-04-09 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
US8450358B2 (en) 2009-06-29 2013-05-28 Xenon Pharmaceuticals Inc. Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
US9480677B2 (en) 2009-06-29 2016-11-01 Xenon Pharmaceuticals Inc. Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
US8883840B2 (en) 2009-06-29 2014-11-11 Xenon Pharmaceuticals Inc. Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
US9695185B2 (en) 2009-10-14 2017-07-04 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8742109B2 (en) 2009-10-14 2014-06-03 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US9260446B2 (en) 2009-10-14 2016-02-16 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US8445696B2 (en) 2009-10-14 2013-05-21 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
US9504671B2 (en) 2010-02-26 2016-11-29 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
US10709714B2 (en) 2013-11-22 2020-07-14 Clifton Life Sciences LLC Gastrin antagonists for treatment and prevention of osteoporosis
US10201524B2 (en) 2014-11-21 2019-02-12 F2G Limited Antifungal agents
US10596150B2 (en) 2014-11-21 2020-03-24 F2G Limited Antifungal agents
US11065228B2 (en) 2014-11-21 2021-07-20 F2G Limited Antifungal agents
US9682033B2 (en) 2015-02-05 2017-06-20 Teva Pharmaceuticals International Gmbh Methods of treating postherpetic neuralgia with a topical formulation of a spiro-oxindole compound
US11819503B2 (en) 2019-04-23 2023-11-21 F2G Ltd Method of treating coccidioides infection

Also Published As

Publication number Publication date
WO2008046082A3 (en) 2008-06-19

Similar Documents

Publication Publication Date Title
WO2008046082A2 (en) Use of heterocyclic compounds as therapeutic agents
EP1888592B1 (en) Spiroheterocyclic compounds and their uses as therapeutic agents
US8466188B2 (en) Use of spiro-oxindole compounds as therapeutic agents
WO2008046065A1 (en) Spiroheterocyclic compounds and their uses as therapeutic agents
EP2350091B1 (en) Spiro-oxindole compounds and their use as therapeutic agents
US20110237567A9 (en) Tricyclic spiro-oxindole derivatives and their uses as therapeutic agents
US20080103151A9 (en) Heterocyclic compounds and their uses as therapeutic agents
WO2008134553A1 (en) Methods of using bicyclic compounds in treating sodium channel-mediated diseases
US20110294842A9 (en) Spiro (furo [3, 2-c] pyridine-3-3&#39; -indol) -2&#39; (1&#39;h)-one derivatives and related compounds for the treatment of sodium-channel mediated diseases, such as pain
WO2010053998A1 (en) Spiro-condensed indole derivatives as sodium channel inhibitors
WO2008046087A2 (en) Spiro compounds and their uses as therapeutic agents
EP2079737A1 (en) Pyridopyrimidinone compounds useful in treating sodium channel-mediated diseases or conditions
MX2012000028A (en) Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents.
WO2008109856A2 (en) Methods of using diazepinone compounds in treating sodium channel-mediated diseases or conditions
WO2008121859A1 (en) Methods of using tricyclic compounds in treating sodium channel-mediated diseases or conditions
WO2008134547A1 (en) Methods of using hydroxycyclohexane and hydroxypiperidine compounds in treating sodium channel-mediated diseases or conditions
WO2008106633A1 (en) Methods of using benzodioxine compounds in treating sodium channel-mediated diseases or conditions
CN101636398A (en) Pyridopyrimidinone compounds useful in treating sodium channel-mediated diseases or conditions

Legal Events

Date Code Title Description
NENP Non-entry into the national phase in:

Ref country code: DE

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

Ref document number: 07854024

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 07854024

Country of ref document: EP

Kind code of ref document: A2