WO2008060789A2 - Use of spiro-oxindole compounds as therapeutic agents - Google Patents

Use of spiro-oxindole compounds as therapeutic agents Download PDF

Info

Publication number
WO2008060789A2
WO2008060789A2 PCT/US2007/081247 US2007081247W WO2008060789A2 WO 2008060789 A2 WO2008060789 A2 WO 2008060789A2 US 2007081247 W US2007081247 W US 2007081247W WO 2008060789 A2 WO2008060789 A2 WO 2008060789A2
Authority
WO
WIPO (PCT)
Prior art keywords
indol
heteroaryl
furo
benzodioxole
aryl
Prior art date
Application number
PCT/US2007/081247
Other languages
English (en)
French (fr)
Other versions
WO2008060789A3 (en
Inventor
Mikhail Chafeev
Sultan Chowdhury
Robert Fraser
Jianmin Fu
Rajender Kamboj
Duanjie Hou
Shifeng Liu
Mehran Seid Bagherzadeh
Serguei Sviridov
Shaoyi Sun
Jianyu Sun
Nagasree Chakka
Tom Hsieh
Vandna Raina
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39339863&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2008060789(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Xenon Pharmaceuticals Inc. filed Critical Xenon Pharmaceuticals Inc.
Priority to AU2007319580A priority Critical patent/AU2007319580B2/en
Priority to JP2009532606A priority patent/JP5460324B2/ja
Priority to BRPI0719210 priority patent/BRPI0719210A2/pt
Priority to CA002665698A priority patent/CA2665698A1/en
Priority to US12/445,264 priority patent/US8466188B2/en
Priority to EP07868434A priority patent/EP2073806B1/en
Priority to AT07868434T priority patent/ATE545416T1/de
Priority to MX2009003876A priority patent/MX2009003876A/es
Publication of WO2008060789A2 publication Critical patent/WO2008060789A2/en
Publication of WO2008060789A3 publication Critical patent/WO2008060789A3/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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention is directed to methods of using spiro-oxindole compounds as therapeutic agents.
  • this invention is directed to the use of certain spiro-oxindole 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, WA, 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/I .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 Nay1.1 and Na/I .2 result in severe epileptic states and in some cases mental decline (Rhodes, T.H., et al., Proc.
  • Na/I .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/1.3 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 v 1.5 is expressed mainly in the heart ventricles and atria (Raymond, C.
  • Na v 1.5 is central to the genesis of cardiac arrhythmias. Mutations in human Na ⁇ I .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.
  • Nav1.6 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, Na/I .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).
  • Na v 1.7 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/I .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/1.7 blockers active in a subset of neurons Further support for Na/1.7 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 Nay1.7 (Yang, Y., et al., J. Med. Genet. (2004), 41 (3): 171 -4).
  • the expression of Na/1.8 is essentially restricted to the DRG (Raymond, C. K., et al., op. cit.). There are no identified human mutations for Nav/1.8. However, Na/1.8- 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/1.8) subunit.
  • Na/I .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, JJ. , 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 spiro-oxindole 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):
  • j and k are each independently 0, 1 , 2 or 3;
  • Q is -C(R 1a )H-, -C(O)-, -O-, -S(O) n ,- (where m is 0, 1 or 2), -CF 2 -, -C(O)O-, -C(O)N(R 5 )- or -N(R 5 )C(O)-;
  • R 1a is hydrogen or -OR 5 ;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where:
  • R 6 is hydrogen, alkyl, aryl or aralkyl
  • R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 , -R 9 -N(R 4 )R 5 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 6 and R 7 , 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 groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo,
  • R 12 is hydrogen, alkyl, aryl, arakyl or -C(O)R 5 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 10 and R 11 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 8 -CN, -R 8 -OR 5 , -R 8 -C(O)R 5 , heterocyclyl and heteroaryl; or R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo,
  • each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R 3a and R 3b , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and
  • R 3c and R 3d are as defined above; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R M are as defined above; or R 30 and R 3d , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R 3b are as defined above; each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each
  • 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 -C 12 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:
  • C-Ci O alkyl refers to an alkyl radical as defined below containing one to ten carbon atoms.
  • the C-i-C 10 alkyl readical may be optionally substituted as defined below for an alkyl group.
  • C 2 -Ci 2 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 -C 12 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 aryi 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 -Ci 2 aralkyl radical may be optionally substituted as defined below for an alkyl group.
  • C 7 -Ci 2 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) 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
  • 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 O to 2), and -S(O) t N(R
  • 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, /7-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(0) t 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) 4 R 16 (where t is O to 2), and -S(O),N(R
  • 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 3 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 c 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, norbornyl, 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 R d 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[b][1,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazoiyl, benzo[4,6]imidazo[1 ,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophen
  • 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 a 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 R b 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 al., "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 0, 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:
  • 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 of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: at least one of j and k is 1 and the other is 0 or 1 ; Q is -O-; R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 , -R 9 -S(O) m -R 5 (where m is 0, 1 or 2), -R 8 -OR 5 , -R 8 -CN, -R 9 -P(O)(OR 5 ) 2 , or -R 9 -O-R 9 -OR 5 ;
  • each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R 3a and R 3b , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3c and R ⁇ are as defined above; or R 3b and R 30 , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R 3d are as defined above; or R 3c and R 3d , together with the carbon ring
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ; Q is -O-;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, cycloalkylalkyl, -R 8 -OR 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen.halo or alkyl; R 3a and R 3d are both hydrogen;
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is a straight or branched alkylene chain, a straight or
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1'-(2-cyclopropylethyl)spiro[furo[2,3-/][1 ,3]benzodioxole-7,3 l -indol]-2 l (1 l /-/)-one; 1 '-pentylspiro[furo[2,3-f][1 ,3]benzodioxole-7,3'-indol]-2'(1 '/-/)-one; 4'-bromo-1 '-pentylspiro[furo[2,3-/][1 ,3]benzodioxole-7,3'-indol]-2'(1 'H)-one; ethyl (2'-oxospiro[furo[2,3-f][1 ,3]benzodioxole-7
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, cycloalkylalkyl, -R 8 -OR 5 , -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 , -R 8 -CN, -R 9 -P(O)(OR 5 ) 2 , or -R 9 -O-R 9 -OR 5 ;
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, halo or alkyl;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, cycloalkyl, cycloalkylalkyl, -R 8 -OR 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, halo or alkyl;
  • R 3a and R 3d are both hydrogen
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: ⁇ . ⁇ -dimethyl- ⁇ . ⁇ -dihydrospirotbenzoti ⁇ -ib ⁇ -jbldifuran-S.S'-indo ⁇ XI ⁇ Vone; ethyl ( ⁇ -bromo-e. ⁇ -dimethyl ⁇ '-oxo- ⁇ .e-dihydrospirotbenzoti ⁇ -b ⁇ . ⁇ b'ldifuran-S.S 1 - indol]-1'(2'H)-yl)acetate; ethyl (2 l -oxo-6,7-dihydro-5/-/-spiro[indeno[5,6-b]furan-3,3 l -indol]-1'(2'H)-yl)acetate; ethyl (2-oxo-5',6',7',8'-
  • R 1 is aryl, heteroaryl or heterocyclyl
  • R 2a , R 2b , R 2c and R 2d are each hydrogen
  • R 3b and R 3c together with the carbon ring atoms to which they are directly attached, form a fused dioxolyl ring.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: at least one of j and k is 1 and the other is 0 or 1 ; Q is -O-;
  • R 1 is hydrogen, alkyl, -R 8 -C(O)OR 5 or -R 8 -C(O)N(R 4 )R 5 ;
  • 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
  • R 2a is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R 8 -NO 2 , -R 8 -OR 5 , -R 8 -N(R 4 )R 5 , -S(O) m R 4 , -R 8 -S(O) n N(R 4 )R 5 ,
  • each m is independently O 1 1 , or 2 and each n is independently 1 or 2;
  • R 2b , R 2c and R 2d are each hydrogen;
  • R 3a , R 3b , R 30 and R 3d are each independently selected from hydrogen or halo; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl and R 3a and R 3d are as defined above;
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl,
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or j is 1 and k is 0; Q is -O-;
  • R 1 is hydrogen or alkyl
  • R 2a is selected from the group consisting of alkyl, haloalkenyl, aryl, aralkyl, aralkenyl, heterocyclyl, heteroaryl, -R 8 -C(O)N(R 4 )R 5 , and -R 8 -N(R 4 )R 5 ; wherein each of the aryl, aralkyl, aralkenyl, heterocyclyl and heteroaryl groups for R 2a is optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R
  • R 3a , R 3b , R 30 and R 3d are each independently selected from hydrogen or halo; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused dioxolyl ring or a fused optionally substituted tetrahydrofuranyl ring, and R 3a and R 3d are as defined above; each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; and each R 8 is a
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 4'-[6-(dimethylamino)pyridin-3-yl]-1 '-pentylspiro[furo[2,3-f][1 ,3]benzodioxole-7,3'-indol]-
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where: R 6 is hydrogen, alkyl, aryl or aralkyl; and
  • R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 , -R 9 -N(R 4 )R 5 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 6 and R 7 , 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 groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, -R 8 -CN, -R 8
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ; Q is -O-;
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where: R 6 is hydrogen, alkyl, aryl or aralkyl; and R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 , -R 9 -N(R 4 )R 5 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 6 and R 7 , 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 groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consist
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ; Q is -O-;
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where: R 6 is hydrogen, alkyl, aryl or aralkyl; and R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 or -R 9 -N(R 4 )R 5 ; or R 6 and R 7 , together with the nitrogen to which they are attached, form a heterocyclyl or heteroaryl; wherein each aryl, aralkyl, heterocyclyl and heteroaryl groups for R 6 and R 7 is optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl,
  • R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen;
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is a straight or branched alkylene chain, a straight
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: /V-CS-methylbutyO ⁇ -KZ-oxospirotfurop.S- ⁇ I .Slbenzodioxole-Z.S'-indoO-i ⁇ 'H)- yl)methyl]benzamide; ⁇ /, ⁇ /-diisopropyl-2-[(2'-oxospiro[furo[2,3-f][1 ,3]benzodioxole-7,3'-indol]-r(2 l H)- yl)methyl]benzamide; ⁇ /-butyl-2-[(2 l -oxospiro[furo[2,3-/][1 > 3]benzodioxole-7 > 3'-indol]-1 l (2 l H)- yl)methyl]benzamide;
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is O and k is i ; Q is -O-;
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where: R 6 is hydrogen, alkyl, aryl or aralkyl; and
  • R 7 is aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroaryl groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, -R 8 -CN, -R 8 -OR 5 , heterocyclyl and heteroaryl;
  • R 2a , R 2b , R 2c and R 2d are each hydrogen;
  • R 3a and R 36 are each hydrogen;
  • R 3b and R 3c together with the carbon ring atoms to which they are directly attached, form a fused dioxolyl ring;
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: ⁇ /-[2-(4-chlorophenyl)ethyl]-2-[(2'-oxospiro[furo[2,3-/][1 > 3]benzodioxole-7,3'-inclol]-
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: at least one of j and k is 1 and the other is 0 or 1 ;
  • R 1 is aralkyl optionally substituted by one or more substituents selected from the group consisting of -R 8 -OR 5 , -C(O)OR 5 , halo, haloalkyl, alkyl, nitro, cyano, aryl, aralkyl, heterocyclyl and heteroaryl;
  • R 2a , R 2b , R 2c and R 2d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R 8 -NO 2 , -R 8 -OR 5 , -R 8
  • R 3c and R 3d are as defined above; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R M are as defined above; or R 3c and R 3d , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 33 and R 3b are as defined above; each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each
  • R 1 is aralkyl optionally substituted by one or more substituents selected from the group consisting of -R 8 -OR 5 , -C(O)OR 5 , halo, haloalkyl, alkyl, nitro, cyano, aryl, aralkyl, heterocyclyl and heteroaryl;
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, alkyl or halo;
  • R 3a , R 3b , R 3c and R 3d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl,
  • each m is independently 0, 1 , or 2 and each n is independently 1 or 2; or R 3a and R 3b , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and
  • R 3c and R 3d are as defined above; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R 3d are as defined above; or R 30 and R 3d , together with the carbon ring atoms to which they are directly attached, may form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and R 3a and R 3b are as defined above; each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or
  • R 1 is aralkyl optionally substituted by one or more substituents selected from the group consisting of -R 8 -OR 5 , -C(O)OR 5 , halo, haloalkyl, alkyl, nitro, cyano, aryl, aralkyl, heterocyclyl and heteroaryl;
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, alkyl or halo;
  • R 3a , R 3b , R 3c and R 3d are each independently selected from the group consisting of hydrogen, halo, and -R 8 -OR 5 ; or R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, dioxolyl, tetrahydrofuranyl, and heteroaryl, and R 3a and R 3d are each hydrogen;
  • each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl;
  • each R 8 is a direct bond or a straight or branched alky
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: methyl 2-[(2'-oxospiro[furo[2,3-/][1 ,3]benzodioxole-7,3"-indol]-1 '(2 1 H)- yl)methyl]benzoate; methyl 3-[(2'-oxospiro[furo[2,3-f][1 ,3]benzodioxole-7,3'-indol]-1 '(2'H)- yl)methyl]benzoate; methyl 4-[(2'-oxospiro[furo[2,3-f][1 ,3]benzodioxole-7,3'-indol]-1 '(2 1 H)- yl)methyl]benzoate; r-(diphenylmethyl)spiro[furo[2,3-f][1
  • R 1 is -R 9 -N(R 10 )R 11 , -R 9 -N(R 12 )C(O)R 11 or -R 9 -N(R 10 )C(O)N(R 10 )R 11 where: each R 10 is hydrogen, alkyl, aryl, aralkyl or heteroaryl; each R 11 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OC(O)R 5 , -R 9 -C(O)OR 5 , -R 9 -C(O)N(R 4 )R 5 , -R 9 -C(O)R 5 , -R 9 -N(R 4 )R 5 , -R 9 -OR 5 , or -R 9 -CN; R
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ; Q is -O-; R 1 is -R 9 -N(R 10 )R 11 , -R 9 -N(R 12 )C(O)R 11 or -R 9 -N(R 10 )C(O)N(R 10 )R 11 where: each R 10 is hydrogen, alkyl, aryl, aralkyl or heteroaryl; each R 11 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OC(O)R 5 , -R 9 -C(O)OR 5 , -R 9 -C(O)N(R 4 )R 5 , -R 9
  • R 12 is hydrogen, alkyl, aryl, arakyl or -C(O)R 5 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 10 and R 11 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 8 -CN, -R 8 -OR 5 , -R 8 -C(O)R 5 , heterocyclyl and heteroaryl; R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen; R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cyclo
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ;
  • R 1 is -R 9 -N(R 10 )R 11 where: each R 10 is hydrogen, alkyl, aryl, aralkyl or heteroaryl; each R 11 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OC(O)R 5 , -R 9 -C(O)OR 5 , -R 9 -C(O)N(R 4 )R 5 , -R 9 -C(O)R 5 , -R 9 -N(R 4 )R 5 , -R 9 -OR 5 , or -R 9 -CN; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroary
  • each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is a straight or branched alkylene chain, a straight or
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1 l -[2-(diethylamino)ethyl]spiro[furo[2,3-/][1 ,3]benzodioxole-7,3 l -indol]-2'(1 ⁇ )-one; r-[2-(pyridin-2-ylamino)ethyl]spiro[furo[2,3-/][1 ,3]benzodioxole-7,3 l -indol]-2'(1'H)-one;
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 ;
  • R 1 is -R 9 -N(R 12 )C(O)R 11 where: each R 10 is hydrogen, alkyl, aryl, aralkyl or heteroaryl; each R 11 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 -OC(O)R 5 , -R 9 -C(O)OR 5 , -R 9 -C(O)N(R 4 )R 5 , -R 9 -C(O)R 5 , -R 9 -N(R 4 )R 5 , -R 9 -OR 5 , or -R 9 -CN;
  • R 12 is hydrogen, alkyl, aryl, arakyl or -C(O)R 5 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 10 and R 11 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 8 -CN, -R 8 -OR 5 , -R 8 -C(O)R 5 , heterocyclyl and heteroaryl; R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen; R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused dioxolyl
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1'-(3-aminopropyl)spiro[furo[2,3-/][1 ,3]benzodioxole-7,3'-indol]-2'(r/-/)-one; 3-chloro- ⁇ /-[3-(2'-oxospiro[furo[2,3-f][1 ,3]benzodioxole-7,3 l -indol]-r(2 ⁇ )- yl)propyl]thiophene-2-carboxamide;
  • R 1 is -R 9 -N(R 10 )C(O)N(R 10 )R 11 where: each R 10 is hydrogen, alkyl, aryl, aralkyl or heteroaryl; each R 11 is hydrogen, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 9 OC(O)R 5 , -R 9 -C(O)OR 5 , -R 9 -C(O)N(R 4 )R 5 , -R 9 -C(O)R 5 , -R 9 -N(R 4 )R 5 , -R 9 -OR 5 , or -R 9 -CN; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, hetero
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1-(4-fluorophenyl)-3-[2-(2'-oxospiro[furo[2,3-/][1 ,3]benzodioxole-7,3'-indol]-1'(2 1 /-/)- yl)ethyl]urea;
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: at least one of j and k is 1 and the other is 0 or 1 ; Q is -O-;
  • R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -OR 5 , -R 8 -C(O)OR 5 , -R 8 -N(R 4 )R 5 , -R 8 -C(O)N(R 4 )R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R 8 -NO 2 , -R 8 -OR 5 ,
  • R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -OR 5 , -R 8 -C(O)OR 5 , -R 8 -N(R 4 )R 5 , -R 8 -C(O)N(R 4 )R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, halo, alkyl or
  • R 3a , R 3b , R 3c and R 3d are each independently selected from hydrogen, halo, alkyl or
  • R 3b and R 3c together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, and
  • R 3a and R 3d are each hydrogen; each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain.
  • R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -OR 5 , -R 8 -C(O)OR 5 , -R 8 -N(R 4 )R 5 , -R 8 -C(O)N(R 4 )R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from hydrogen, halo, alkyl or
  • R 33 , R 3b , R 30 and R 3d are each independently selected from hydrogen, halo, alkyl or -R 8 -OR 5 ; or R 3b and R 30 , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from dioxolyl or tetrahydrofuranyl, and R 3a and R 3d are each hydrogen; each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of:
  • R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -OR 5 , -R 8 -C(O)OR 5 , -R 8 -N(R 4 )R 5 , -R 8 -C(O)N(R 4 )R 5 , -R 8 -N(R 5 )C(O)R 4 , -R 8 -S(O) m R 4 (where m is 0, 1 or 2), -R 8 -CN, or -R 8 -NO 2 ; R 2a , R
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 4'-(6-methoxypyridin-3-yl)-1 '- ⁇ [5-(trifluoromethyl)-2-furyl]methyl ⁇ spiro[furo[2,3- fl[1 ⁇ ]ben. ⁇ dioxole-7 l 3 l -indol]-2 l (1 l H)-one; 4'-[6-(dimethylamino)pyridin-3-yl]-1 l - ⁇ [5-(trifluoromethyl)-2-furyl]methyl ⁇ spiro[furo[2,3-
  • R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -OR 5 , -R 8 -C(O)OR 5 , -R 8 -N(R 4 )R 5 , -R 8 -C(O)N(R 4 )R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R 8 -NO 2 , -R 8 -OR 5 ,
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: ⁇ -bromo-i'- ⁇ -chloro ⁇ -thienyOmethyljspiroCI-benzofuran-S.S'-indolJ ⁇ XI'HJ-one; 1'-(pyridin-3-ylmethyl)-6-(trifluoromethoxy)spiro[1-benzofuran-3,3'-indol]-2'(1'/-/)-one hydrochloride; 6-(trifluoromethoxy)-1'- ⁇ [5-(trifluoromethyl)-2-furyl]methyl ⁇ spiro[1-benzofuran-3,3'-indol]-
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2; Q is -O-;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen;
  • R 3b and R 3c together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl; each R 4 and R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, and heteroarylalkyl; or when R 4 and R 5 are each attached to the same nitrogen atom, then R 4 and R 5 , together with the nitrogen atom to which they are attached, may form a heterocyclyl or heteroaryl; and each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2;
  • Q is -C(R 1a )H-, -C(O)-, -CF 2 -, -C(O)O- or -N(R 5 )C(O)-;
  • R 1a is hydrogen or -OR 5 ;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where:
  • R 6 is hydrogen, alkyl, aryl or aralkyl
  • R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 , -R 9 -N(R 4 )R 5 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 6 and R 7 , 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 groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo,
  • R 12 is hydrogen, alkyl, aryl, arakyl or -C(O)R 5 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 10 and R 11 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 8 -CN, -R 8 -OR 5 , -R 8 -C(O)R 5 , heterocyclyl and heteroaryl; or R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo,
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2; Q is -C(R 1a )H-; R 1a is hydrogen or -OR 5 ;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN, -R 8 -NO 2 , -R 8 -OR 5 , -R 8 -N(R 4 )R 5 ,
  • 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 R 2a , R 2b , R 2c and R 2d may be optionally substituted by one or more substituents selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalky
  • R 3a , R 3b , R 30 and R 3d are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, alkoxy, halo, haloalkyl, haloalkenyl, haloalkoxy, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, aralkenyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -R 8 -CN-R 8 -NO 2 , -R 8 -OR 5 ,
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2; Q is -C(R 1a )H-; R 1a is hydrogen or -OR 5 ;
  • R 1 is hydrogen, alky!, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 2a , R 2b , R 2c and R 2d are each hydrogen;
  • R 2a , R 2b , R 2c and R 2d are each hydrogen; R 38 and R 3d are each hydrogen; R 3b and R 30 , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl; each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl; each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2; Q is -C(R 1a )H-;
  • R 1a is hydrogen or -OR 5 ;
  • R 1 is pentyl;
  • R 2a , R 2b , R 2c and R 2d are each hydrogen;
  • R 3a and R 36 are each hydrogen;
  • R 3b and R 3c together with the carbon ring atoms to which they are directly attached, form a fused dioxolyl ring;
  • each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1'-pentyl-6,7-dihydrospiro[indeno[5,6-c/][1 ,3]dioxole-5,3'-indol]-2'(1'/-/)-one; i-pentyl-T'. ⁇ '-dihydro-e ⁇ -spirotindole-S. ⁇ '-naphthop.S-cOti .SjdioxolJ ⁇ CIHVonei and Z-methoxy-r-pentyl- ⁇ J-dihydrospirofindenof ⁇ -cfUI .Sldioxole-S.S'-indolJ ⁇ XI ⁇ J-one.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and
  • Q is -C(O)-, -CF 2 -, -C(O)O- or -N(R 5 )C(O)-;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 ,
  • R 1 is aralkyl substituted by -C(O)N(R 6 )R 7 where:
  • R 6 is hydrogen, alkyl, aryl or aralkyl
  • R 7 is hydrogen, alkyl, haloalkyl, -R 9 -CN, -R 9 -OR 5 , -R 9 -N(R 4 )R 5 , aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl; or R 6 and R 7 , 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 groups for R 6 and R 7 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo,
  • R 12 is hydrogen, alkyl, aryl, arakyl or -C(O)R 5 ; and wherein each aryl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl and heteroarylalkyl groups for R 10 and R 11 may be optionally substituted by one or more substituents selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, halo, haloalkyl, nitro, -R 8 -CN, -R 8 -OR 5 , -R 8 -C(O)R 5 , heterocyclyl and heteroaryl; or R 1 is heterocyclylalkyl or heteroarylalkyl where the heterocyclylalkyl or the heteroarylalkyl group is optionally substituted by one or more substituents selected from the group consisting of oxo, alkyl, halo,
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2;
  • Q is -C(O)-, -CF 2 -, -C(O)O- or -N(R 5 )C(O)-;
  • R 1 is hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, aryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, -R 8 -C(O)R 5 , -R 8 -C(O)OR 5 , -R 8 -C(O)N(R 4 )R 5 , -S(O) 2 -R 5 , -R 9 -S(O) m -R 5 (where m is 0, 1 or 2), -R 8 -OR 5 , -R 8 -CN, -R 9 -P(O)(OR 5 ) 2> or
  • R 2a , R 2b , R 2c and R 2d are each hydrogen; R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen; R 3b and R 3c , together with the carbon ring atoms to which they are directly attached, form a fused ring selected from cycloalkyl, heterocyclyl, aryl or heteroaryl; each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl; each R 8 is a direct bond or a straight or branched alkylene chain, a straight or branched alkenylene chain or a straight or branched alkynylene chain; and each R 9 is a straight or branched alkylene chain, a straight or
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention wherein: j is 0 and k is 1 or 2;
  • Q is -C(O)-, -CF 2 -, -C(O)O- or -N(R 5 )C(O)-;
  • R 1 is pentyl;
  • R 2a , R 2b , R 2c and R 2d are each hydrogen; R 3a and R 3d are each hydrogen;
  • each R 5 is independently selected from group consisting of hydrogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxyalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl and heteroaryl.
  • Another embodiment of the invention are the compounds of formula (I) as set forth above in the Summary of the invention, selected from the group consisting of: 1'-pentylspiro[indeno[5,6-c/][1 ,3]dioxole-5,3 l -indole]-2',7(1'H,6/-/)-dione; 1-pentyl-6'H-spiro[indole-3 > 5'-naphtho[2,3-d][1 ,3]dioxole]-2,8'(1H,7 l H)-dione; 8 ⁇ 8'-difluoro-1 -pentyl-Z ⁇ '-dihydro- ⁇ -spiropndole-S. ⁇ '-naphthop.S-c/KI ,3]dioxol]-
  • 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
  • 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.
  • 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.
  • skin disorders e.g., contact dermatitis
  • hypersensitivity e.g., dry skin, acne, eczema, psoriasis
  • itch associated with vulvar vestibulitis e.g., itch associated with vulvar vestibulitis
  • 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 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.
  • 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. 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
  • 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 v 1.1 , Na/I .2, Nav1.3, Na v 1.4, Na/1.5, Na v 1.6, Ua v 1.7, Na/1.8, or Na ⁇ I .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 v 1.2, Na v 1.3, Na v 1.4, Na v 1.5, Na/I .6, Na/I .7, Na 1 Zi .8, or Nay"] .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.
  • compositions 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 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.
  • 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 sal
  • the pharmaceutical composition when 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.
  • 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.
  • the pharmaceutical 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., 0.07 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., 0.7 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.
  • 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, 16 th edition, Merck and Co., Rahway, NJ., 1992; Goodmanetna., eds..Goodman and Oilman's The Pharmacological Basis of Therapeutics, 10 th 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.
  • 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.
  • Effective amounts of a diagnostic pharmaceutical compound or composition of the invention are from about 0.1 ⁇ g to about 100 mg/Kg body weight, administered at intervals of 4-72 hours, for a period of 2 hours to 1 year, and/or any range or value therein, such as 0.0001-0.001 , 0.001-0.01 , 0.01-0.1 , 0.1-1.0,1.0-10, 5- 10, 10-20, 20-50 and 50-100 mg/Kg, at intervals of 1-4, 4-10, 10-16, 16-24, 24-36, 24- 36, 36-48, 48-72 hours, for a period of 1-14, 14-28, or 30-44 days, or 1-24 weeks, or any range or value therein.
  • 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 Carnivora (including cats, and dogs).
  • the preferred recipients are turkeys, chickens and other members of the same order.
  • the most preferred recipients are humans.
  • 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.
  • compositions 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.
  • 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.
  • 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.
  • kits that contain a pharmaceutical composition which includes one or more compounds of the above formulae.
  • 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 2a , R 2b , R 2c , R 2d , R 3a , R 3b , R 3c , and R 3d 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.
  • Suitable protecting groups include hydroxy, amino, mercapto and carboxylic acid.
  • Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (e.g., f-butyldimethylsilyl, ?-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like.
  • Suitable protecting groups for amino, amidino and guanidino include 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.
  • protecting groups are described in detail in Greene, T.W. and P. G. M. Wuts, 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.
  • 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.
  • R 1 , R 2a , R 2b , R 2c , R 2d , R 3a , R 3b , R 3c and R 30 are defined as in the Specification unless specifically defined otherwise.
  • X is Cl or Br.
  • R 11 is an alkyl group.
  • the compounds of formula (I) of the invention where Q is -O-, j is 0 and k is 1 can be synthesized following the general procedure as described below in REACTION SCHEME 1.
  • an isatin compound of formula (101 ) is alkylated with the chloro or bromo compound of formula (102) to afford the product of formula (103).
  • the phenol compound of formula (104) is treated with a Grignard reagent of formula (105) at low temperature (0 0 C) to form the phenoxymagnesium halide intermediate which reacts with the keto-carbonyl group of the isatin compound of formula (103) in a solvent, such as, but not limited to, methylene chloride or toluene, to afford the oxindole of formula (106).
  • the compound of formula (107) is obtained after the removal of the hydroxyl group at C-3 position of the oxindole by treating the compound of formula (106) with silane such as triethylsilane.
  • the compound of formula (107) can also be achieved by treating the compound of formula (106) with SOCI 2 /NEt 3 then reduction with Zn dust.
  • Compound of formula (107) 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 (108).
  • a silyl compound such as, but not limited to, trimethylsilyl chloride
  • compound of formula (108) can be obtained by treating the compound of formula (107) with a base, such as, but not limited to, LiOH, JPr 2 NH, LDA, and subsequently reacting with formaldehyde.
  • Intramolecular cyclization via Mitsunobu reaction affords the compound of formula (I) of the invention where Q is -O-, j is 0 and k is 1.
  • REACTION SCHEME 1.1 illustrates a schematic synthesis of amide and heterocyclic compounds as compounds of formula (I).
  • R 1 consists of an ester group
  • a compound such as a compound of formula (109) (in which A is alkyl or aralkyl) can be converted to the corresponding carboxylic acid compound of formula (110) by treatment of a compound of formula (109) with a base such as, but not limited to, lithium hydroxide, sodium hydroxide or potassium hydroxide, in a mixed solvent such as, but not limited to, tetrahydrofuran or methanol with water.
  • a base such as, but not limited to, lithium hydroxide, sodium hydroxide or potassium hydroxide
  • a mixed solvent such as, but not limited to, tetrahydrofuran or methanol with water.
  • the acid compound of formula (110) can be converted to a mixed anhydride, by treatment with iso-butyl chloroformate in the presence of a base such as, but not limited to, N- methylmorpholine, or to the corresponding acid chloride, by treament with oxalyl chloride in the presence of catalytic amount of ⁇ /, ⁇ /-dimethylformamide in a solvent such as, but not limited to, toluene, dichloromethane or chloroform.
  • a base such as, but not limited to, N- methylmorpholine
  • treament with oxalyl chloride in the presence of catalytic amount of ⁇ /, ⁇ /-dimethylformamide in a solvent such as, but not limited to, toluene, dichloromethane or chloroform.
  • the mixed anhydride can react directly with, or the acid chloride can react with, in the presence of a base such as, but not limited to, triethylamine or diisopropyl ethylamine, a primary or secondary amine to form the amide compound of formula (111) as a compound of formula (I).
  • a base such as, but not limited to, triethylamine or diisopropyl ethylamine, a primary or secondary amine
  • the acid compound of formula (110) can react with an aromatic diamine compound in a solvent such as, but not limited to, toluene to form the benzimidazole compound of formula (111.1 ) as a compound of formula (I).
  • REACTION SCHEME 1.2 illustrates a schematic synthesis of amine compounds as compounds of formula (I).
  • the protecting group such as, but not limited to, phthalimido or terf-butyloxycarbonyl
  • either the primary or secondary amino compound of formula (113) can be formed.
  • Reaction of the amino compound of formula (113) with an acyl chloride in the presence of a base such as, but not limited to, triethylamine or diisopropyl ethylamine, in a solvent such as, but not limited to, toluene, dichloromethane or chloroform provides the amide compound of formula (114) as a compound of formula (I).
  • REACTION SCHEME 1.3 illustrates a schematic synthesis of amine compounds as compounds of formula (I).
  • the alcohol compound of formula (118) upon removal of the protecting group in compound of formula (117), can be oxidized to the aldehyde compound of (119) by using an oxidant such as, but not limited to, pyridinium dichromate or Dess-Martin's reagent.
  • an oxidant such as, but not limited to, pyridinium dichromate or Dess-Martin's reagent.
  • the amine compound of formula (120) can be obtained as a compound of formula (I) through the reductive amination of the aldehyde compound of formula (119) with a primary or secondary amine.
  • REACTION SCHEME 1.4 illustrates an alternative synthesis of compounds of formula (I) with the introduction of a variety of R 1 groups.
  • Compound of formula (121) where PG is a protecting group such as, but not limited to, diphenylmethyl, can be synthesized through the sequence as shown in REACTION SCHEME 1 above. The protecting group can be removed under a high pressure of hydrogen such as 60 psi to form the oxindole compound of formula (122).
  • a compound of formula (I) can be achieved by alkylation of the compound of formula (122) with a halide reagent XR 1 (where X is chloro, bromo or iodo) in the presence of a base such as, but not limited to, sodium hydride, sodium bis(trimethylsilyl)amide, and lithium hydroxide, in a solvent such as, but not limited to, ⁇ /, ⁇ /-dimethylformamide, tetrahydrofuran, acetone or acetonitrile.
  • a base such as, but not limited to, sodium hydride, sodium bis(trimethylsilyl)amide, and lithium hydroxide
  • a solvent such as, but not limited to, ⁇ /, ⁇ /-dimethylformamide, tetrahydrofuran, acetone or acetonitrile.
  • reaction of compound of formula (122) with an alcohol under Mitsunobu reaction conditions in the presence of a phosphine reagent such as, but not limited to, triphenylphosphine, tributylphosphine or trimethyl phosphine, and azadicarboxylate of diethyl, diisopropyl or di-tert-butyl in a solvent such as, but not limited to, tetrahydrofuran, ethyl acetate or dichloromethane, provides the compound of formula (I).
  • a phosphine reagent such as, but not limited to, triphenylphosphine, tributylphosphine or trimethyl phosphine, and azadicarboxylate of diethyl, diisopropyl or di-tert-butyl in a solvent such as, but not limited to, tetrahydrofuran, ethyl acetate or dichloromethane
  • R 2a , R 2b , R 2c , R 2d , R 3a , R 3b , R 3c or R 3d of the compound of formula (I) is a bromo or trifluoromethylsulfonyloxy group
  • further derivatives can be synthesized as shown in REACTION SCHEME 1.5 and REACTION SCHEME 1.6 below.
  • the triflate compound can be obtained by treating the bromo compound with diborane in the presence of a palladium catalyst followed by sequential oxidation with hydrogen peroxide/sodium hydroxide and reaction with trifluoromethanesulfonyl anhydride.
  • Compounds of formula (123) or (129) (with either a bromo or a trifluoromethylsulfonyloxy group for R 2a , R 2b , R 2c , R 2d , R 3a , R 3b , R 3c or R 3d ) can react with zinc cyanide or tributyltin cyanide and potassium cyanide in the presence of a palladium catalyst such as, but not limited to, palladium acetate or tris(dibenzylideneacetone)dipalladium(0), and a ligand such as, but not limited to, tri(o- tolyl)phosphine, 1 ,1'-bis(diphenylphosphino)ferrocene or 2-(di-tert- butylphosphino)biphenyl in a solvent such as, but not limited to, N, N- dimethylformamide or acetonitrile to provide the cyano compounds of formula (124) or formula (130)
  • Compound of formula (123) or formula (129) (with either a bromo or a trifluoromethylsulfonyloxy group for R 2a , R 2b , R 2c , R 2d , R 3a , R 3b , R 3c or R 3d ) can react with a primary or secondary amine in the presence of a palladium catalyst such as, but not limited to, tetrakis(triphenylphosphine)palladium(0) or tris(dibenzylideneacetone)dipalladium(0), with or without a ligand such as, but not limited to, triphenylphosphine, tri(o-tolyl)phosphine, 1 ,1'- bis(diphenylphosphino)ferrocene or 2-(di-tert-butylphosphino)biphenyl, a base such as, but not limited to, sodium carbonate, cesium carbonate or sodium
  • the compound of formula (I) of the invention where Q is -O and k is 1 can be synthesized following the general procedure as described below in REACTION SCHEME 2.
  • a compound of formula (201 ) is treated with a lithium reagent of formula (202), such as, but not limited to, n-BuLi at low temperature followed by the reaction with keto-carbonyl group of the isatin compound of formula (103) in a solvent, such as, but not limited to, THF to afford the oxindole of formula (203).
  • the compound of formula (204) is obtained after the removal of the hydroxyl group at C-3 position of the oxindole by treating the compound of formula (203) with silane such as triethylsilane.
  • the compound of formula (204) can also be achieved by treating the compound of formula (203) with SOCI 2 /NEt 3 then reduction with Zn dust.
  • Compound of formula (204) 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 (205).
  • a compound of formula (205) can be obtained by treating the compound of formula (204) with a base, such as, but not limited to, LiOH, iPr 2 NH, or LDA, and subsequently reacting with formaldehyde. Intramolecular cyclization via Mitsunobu reaction affords the compound of formula (I) of the invention where Q is -O- and k is 1.
  • the compound of formula (I) of the invention where Q is -O-or -S- and k is 0 can be synthesized following the general procedure as described below in REACTION SCHEME 3 wherein intramolecular cyclization of the compound of formula (203) via Mitsunobu reaction affords the compound of formula (I) of the invention where Q is -O- or -S-and k is 0.
  • the compound of formula (I) of the invention where Q is -C(O)-, -(CH 2 )- or -(CF 2 )- and j is 0 can be synthesized following the general procedure as described in REACTION SCHEME 4.
  • the Grignard reagent of formula (401) reacts with keto-carbonyl group of the isatin compound of formula (103) in a solvent, such as, but not limited to, methylene chloride or toluene to afford the oxindole of formula (402).
  • the compound of formula (403) is obtained after the removal of the hydroxyl group at C-3 position of the oxindole by treating the compound of formula (402) with silane such as triethylsilane.
  • the compound of formula (403) can also be achieved by treating the compound of formula (402) with SOCI 2 /NEt 3 , followed by reduction with Zn dust.
  • Compound of formula (403) is alkylated at C-3 position of oxindole ring with a compound of formula (404) to afford the compound of formula (405) which is subjected to saponification to generate the carboxylic acid of formula (406).
  • This carboxylic acid is then converted to an acid chloride of formula (407) following procedures known to one skilled in the art.
  • Intramolecular cyclization in the presence of an Lewis acid, such as, but not limited to, tin(IV) chloride yields the compound of formula (I) of the invention where Q is -C(O)- and j is 0.
  • the compound of formula (I) of the invention where Q is -O-, j is 0 and k is 1 can be synthesized following the general procedure as described below in REACTION SCHEME 5.
  • the phenol compound of formula (104) is treated with a Grignard reagent of formula (105) at low temperature (0 0 C) to form the phenoxymagnesium halide intermediate which reacts with the keto-carbonyl group of the isatin compound of formula (101 ) in a solvent, such as, but not limited to, tetrahedrofuran, methylene chloride or toluene, to afford the heterocyclic compound of formula (501).
  • a solvent such as, but not limited to, tetrahedrofuran, methylene chloride or toluene

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
PCT/US2007/081247 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents WO2008060789A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU2007319580A AU2007319580B2 (en) 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents
JP2009532606A JP5460324B2 (ja) 2006-10-12 2007-10-12 治療剤としてのスピロ−オキシインドール化合物の使用
BRPI0719210 BRPI0719210A2 (pt) 2006-10-12 2007-10-12 Uso de compostos espiro-oxindol como agentes terapêuticos
CA002665698A CA2665698A1 (en) 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents
US12/445,264 US8466188B2 (en) 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents
EP07868434A EP2073806B1 (en) 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents
AT07868434T ATE545416T1 (de) 2006-10-12 2007-10-12 Verwendung von spiro-oxindol-verbindungen als therapeutika
MX2009003876A MX2009003876A (es) 2006-10-12 2007-10-12 Uso de compuestos de espiro-oxindol como agentes terapeuticos.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85178706P 2006-10-12 2006-10-12
US60/851,787 2006-10-12

Publications (2)

Publication Number Publication Date
WO2008060789A2 true WO2008060789A2 (en) 2008-05-22
WO2008060789A3 WO2008060789A3 (en) 2008-07-24

Family

ID=39339863

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/081247 WO2008060789A2 (en) 2006-10-12 2007-10-12 Use of spiro-oxindole compounds as therapeutic agents

Country Status (14)

Country Link
US (1) US8466188B2 (pt-PT)
EP (1) EP2073806B1 (pt-PT)
JP (2) JP5460324B2 (pt-PT)
CN (2) CN103271906A (pt-PT)
AR (1) AR063280A1 (pt-PT)
AT (1) ATE545416T1 (pt-PT)
AU (1) AU2007319580B2 (pt-PT)
BR (1) BRPI0719210A2 (pt-PT)
CA (1) CA2665698A1 (pt-PT)
CL (1) CL2007002950A1 (pt-PT)
MX (1) MX2009003876A (pt-PT)
RU (1) RU2009117642A (pt-PT)
TW (1) TW200833695A (pt-PT)
WO (1) WO2008060789A2 (pt-PT)

Cited By (25)

* 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
WO2011047173A3 (en) * 2009-10-14 2012-01-12 Xenon Pharmaceuticals Inc. Pharmaceutical compositions for oral administration
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
JP2012505895A (ja) * 2008-10-17 2012-03-08 ゼノン・ファーマシューティカルズ・インコーポレイテッド 治療剤としてのスピロオキシインドール化合物およびそれらの使用
WO2012049555A1 (en) 2010-10-13 2012-04-19 Lupin Limited Spirocyclic compounds as voltage-gated sodium channel modulators
WO2011106729A3 (en) * 2010-02-26 2012-11-15 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
JP2012532107A (ja) * 2009-06-29 2012-12-13 ゼノン・ファーマシューティカルズ・インコーポレイテッド スピロ−オキシインドール化合物のエナンチオマーおよび治療剤としてのその使用
JP2013508290A (ja) * 2009-10-14 2013-03-07 ゼノン・ファーマシューティカルズ・インコーポレイテッド スピロ−オキシインドール化合物のための合成方法
US8466188B2 (en) 2006-10-12 2013-06-18 Xenon Pharmaceuticals Inc. Use of spiro-oxindole compounds as therapeutic agents
WO2013130013A1 (en) * 2012-02-27 2013-09-06 Nanyang Polytechnic Novel compounds and uses thereof
US8598156B2 (en) 2010-03-25 2013-12-03 Glaxosmithkline Llc Chemical compounds
CN103554121A (zh) * 2013-10-16 2014-02-05 华东师范大学 3,3-螺(2-四氢呋喃)氧化吲哚多环状化合物及其应用
CN103554120A (zh) * 2013-10-16 2014-02-05 华东师范大学 3,3-螺(2-四氢呋喃)氧化吲哚多环状化合物的制备方法
WO2014143601A1 (en) * 2013-03-14 2014-09-18 Eli Lilly And Company CDC7 Inhibitors
WO2015151001A1 (en) 2014-03-29 2015-10-08 Lupin Limited Sulfonamide compounds as voltage gated sodium channel modulators
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
US9487535B2 (en) 2012-04-12 2016-11-08 Xenon Pharmaceuticals Inc. Asymmetric syntheses for spiro-oxindole compounds useful as therapeutic agents
WO2017037682A1 (en) 2015-09-04 2017-03-09 Lupin Limited Sulfonamide compounds as voltage-gated sodium channel modulators
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
WO2018163077A1 (en) 2017-03-08 2018-09-13 Lupin Limited Indanyl compounds as voltage gated sodium channel modulators
US10100060B2 (en) 2016-06-16 2018-10-16 Xenon Pharmaceuticals Inc. Asymmetric synthesis of funapide
CN108976243A (zh) * 2018-08-23 2018-12-11 青岛农业大学 通过二甲基呋喃与含氧化吲哚邻羟基苄醇合成螺-色满-4,3′-氧化吲哚的合成方法
WO2020011731A1 (en) * 2018-07-12 2020-01-16 UCB Biopharma SRL Spirocyclic indane analogues as il-17 modulators
US11434230B2 (en) 2016-01-20 2022-09-06 Chemocentryx, Inc. 2-oxindole compounds

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110294842A9 (en) * 2006-10-12 2011-12-01 Xenon Pharmaceuticals Inc. Spiro (furo [3, 2-c] pyridine-3-3' -indol) -2' (1'h)-one derivatives and related compounds for the treatment of sodium-channel mediated diseases, such as pain
US8217051B2 (en) * 2009-02-17 2012-07-10 Hoffmann-La Roche Inc. Spiroindolinone derivatives
EP2407469A1 (en) * 2010-07-13 2012-01-18 Chemo Ibérica, S.A. Salt of sitagliptin
US9108972B2 (en) * 2011-03-03 2015-08-18 Cadila Healthcare Limited Salts of DPP-IV inhibitor
CN102321095B (zh) * 2011-05-26 2014-09-10 兰州大学 手性螺环吲哚-吡喃嘧啶碱类化合物及其制备和应用
DK3080134T3 (en) 2013-12-13 2018-10-22 Vertex Pharma PRODRUGS OF PYRIDONAMIDS USED AS MODULATORS OF SODIUM CHANNELS
JP6532474B2 (ja) * 2014-02-06 2019-06-19 アッヴィ・インコーポレイテッド 6−ヘテロアリールオキシ−および6−アリールオキシ−キノリン−2−カルボキシアミドならびにその使用
TW201726637A (zh) 2015-12-18 2017-08-01 默沙東藥廠 對電位閘控鈉通道具選擇活性之羥基烷基胺-及羥基環烷基胺-取代之二胺-芳基磺胺化合物
KR20200006128A (ko) 2017-05-16 2020-01-17 버텍스 파마슈티칼스 인코포레이티드 나트륨 채널의 조절제로서의 중수소화 피리돈 아미드 및 이의 프로드럭
JOP20200001A1 (ar) 2017-07-11 2022-10-30 Vertex Pharma كاربوكسأميدات بوصفها معدلات لقنوات الصوديوم
US11529337B2 (en) 2018-02-12 2022-12-20 Vertex Pharmaceuticals Incorporated Method of treating pain
JP2021531256A (ja) 2018-07-09 2021-11-18 リーバー インスティチュート インコーポレイテッドLieber Institute, Inc. NaV1.8を阻害するピリダジン化合物
WO2020146612A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Esters and carbamates as modulators of sodium channels
WO2020146682A1 (en) 2019-01-10 2020-07-16 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
WO2020176763A1 (en) 2019-02-27 2020-09-03 Vertex Pharmaceuticals Incorporated Dosage form comprising prodrug of na 1.8 sodium channel inhibitor
WO2020219867A1 (en) 2019-04-25 2020-10-29 Vertex Pharmaceuticals Incorporated Pyridone amide co-crystal compositions for the treatment of pain
CN112442042B (zh) * 2019-08-30 2023-10-20 广东东阳光药业股份有限公司 一种螺环吲哚化合物的制备方法
AU2020397059A1 (en) 2019-12-06 2022-07-21 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
AU2022286511A1 (en) 2021-06-04 2023-11-30 Vertex Pharmaceuticals Incorporated Hydroxy and (halo)alkoxy substituted tetrahydrofurans as modulators of sodium channels
CA3221788A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran-2-carboxamides as modulators of sodium channels
IL308953A (en) 2021-06-04 2024-01-01 Vertex Pharma N-Hydroalkyl(Hetero)Aryl)Tetrahydrofuran Carboxamides as Natrene Channel Modulators
CN117794918A (zh) 2021-06-04 2024-03-29 沃泰克斯药物股份有限公司 经取代的四氢呋喃类似物作为钠通道调节剂
CA3221939A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamide analogs as modulators of sodium channels
JP2024520649A (ja) 2021-06-04 2024-05-24 バーテックス ファーマシューティカルズ インコーポレイテッド (2r,3s,4s,5r)-4-[[3-(3,4-ジフルオロ-2-メトキシ-フェニル)-4,5-ジメチル-5-(トリフルオロメチル)テトラヒドロフラン-2-カルボニル]アミノ]ピリジン-2-カルボキサミドを含む固体剤形及び投与レジメン
WO2023205463A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205465A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205468A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
TW202408501A (zh) 2022-04-22 2024-03-01 美商維泰克斯製藥公司 用於治療疼痛之雜芳基化合物
CN115160331B (zh) * 2022-08-04 2023-06-02 青岛农业大学 一种氧化吲哚螺烯丙基取代色满骨架及其制备方法
WO2024123815A1 (en) 2022-12-06 2024-06-13 Vertex Pharmaceuticals Incorporated Process for the synthesis of substituted tetrahydrofuran modulators of sodium channels

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005035498A1 (ja) * 2003-10-08 2005-04-21 Dainippon Sumitomo Pharma Co., Ltd. 含窒素二環性化合物の摂食調節剤としての用途
WO2005099689A1 (en) * 2004-04-01 2005-10-27 Case Western Reserve University Topical delivery of phthalocyanines
WO2006110917A2 (en) * 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents

Family Cites Families (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3189617A (en) * 1961-02-03 1965-06-15 Sterling Drug Inc 1-aryloxindoles and their preparation
DE1956237A1 (de) 1969-11-08 1971-05-13 Basf Ag Spiro-pyrrolizidon-oxindole
DE2113343A1 (de) 1971-03-19 1972-09-21 Thiemann Chem Pharm Fab Indolo[2,3-b] chinolone und Verfahren zu ihrer Herstellung
US3723459A (en) * 1971-04-23 1973-03-27 Mc Neil Labor Inc 2-oxospiro (indoline -3,4{40 -thiochroman) derivatives
US3845770A (en) * 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
SE400966B (sv) * 1975-08-13 1978-04-17 Robins Co Inc A H Forfarande for framstellning av 2-amino-3-(eller 5-)bensoyl-fenylettiksyror
US4670566A (en) * 1979-07-12 1987-06-02 A. H. Robins Company, Incorporated 3-methyl-hio-4-(5-, 6-, or 7-)phenylindolindolin-2-ones
US4326525A (en) * 1980-10-14 1982-04-27 Alza Corporation Osmotic device that improves delivery properties of agent in situ
US4440785A (en) * 1980-10-30 1984-04-03 A. H. Robins Company, Inc. Methods of using 2-aminobiphenylacetic acids, esters, and metal salts thereof to treat inflammation
US4438130A (en) * 1981-11-12 1984-03-20 The Upjohn Company Analgesic 1-oxa-, aza- and thia-spirocyclic compounds
JPS60142984A (ja) 1983-12-28 1985-07-29 Kyorin Pharmaceut Co Ltd 新規なスピロピロリジン−2,5−ジオン誘導体およびその製造法
US4569942A (en) 1984-05-04 1986-02-11 Pfizer Inc. N,3-Disubstituted 2-oxindole-1-carboxamides as analgesic and antiinflammatory agents
JPS6130554A (ja) 1984-07-23 1986-02-12 Ono Pharmaceut Co Ltd プロスタグランジン類似化合物のある特定の立体配置を有する異性体及びそれらを有効成分として含有する治療剤
US4690943A (en) 1984-09-19 1987-09-01 Pfizer Inc. Analgesic and antiinflammatory 1,3-diacyl-2-oxindole compounds
US4721721A (en) 1984-12-18 1988-01-26 Rorer Pharmaceutical Corporation 6-(4-thiazole) compounds, cardiotonic compositions including the same, and their uses
DE3608088C2 (de) 1986-03-07 1995-11-16 Schering Ag Pharmazeutische Präparate, enthaltend Cyclodextrinclathrate von Carbacyclinderivaten
WO1993012786A1 (en) 1986-07-10 1993-07-08 Howard Harry R Jr Indolinone derivatives
CA1322197C (en) 1987-07-17 1993-09-14 Bernd Buchmann 9-halogen-(z) prostaglandin derivatives, process for their production and their use as pharmaceutical agents
US5182289A (en) * 1988-06-14 1993-01-26 Schering Corporation Heterobicyclic compounds having antiinflammatory activity
US5206261A (en) 1989-07-25 1993-04-27 Taiho Pharmaceutical Company, Limited Oxindole derivative
DE3932953A1 (de) 1989-10-03 1991-04-11 Boehringer Mannheim Gmbh Neue 2-bicyclo-benzimidazole, verfahren zu ihrer herstellung und diese verbindungen enthaltende arzneimittel
DE3935514A1 (de) 1989-10-25 1991-05-02 Boehringer Mannheim Gmbh Neue bicyclo-imidazole, verfahren zu ihrer herstellung und diese verbindungen enthaltende arzneimittel
US5304121A (en) * 1990-12-28 1994-04-19 Boston Scientific Corporation Drug delivery system making use of a hydrogel polymer coating
US5023265A (en) * 1990-06-01 1991-06-11 Schering Corporation Substituted 1-H-pyrrolopyridine-3-carboxamides
US5484778C1 (en) 1990-07-17 2001-05-08 Univ Cleveland Hospitals Phthalocynine photosensitizers for photodynamic therapy and methods for their use
CA2095718A1 (en) 1990-11-22 1992-05-23 Hans-Rudolf Waespe Isonicotinic acid derivatives and related spiro compounds with herbicidal action
US5116854A (en) * 1991-06-28 1992-05-26 Pfizer Inc. Anti-inflammatory 1-heteroaryl-3-acyl-2-oxindoles
US5686624A (en) * 1992-01-30 1997-11-11 Sanofi 1-benzenesulfonyl-1,3-dihydro-indol-2-one derivatives, their preparation and pharmaceutical compositions in which they are present
FR2708605A1 (fr) 1993-07-30 1995-02-10 Sanofi Sa Dérivés du N-sulfonylindol-2-one, leur préparation, les compositions pharmaceutiques en contenant.
US5663431A (en) * 1992-01-30 1997-09-02 Sanofi 1-benzenesulfonyl-1,3-dihydro-indol-2-one derivatives, their preparation and pharmaceutical compositions in which they are present
FR2686878B1 (fr) 1992-01-30 1995-06-30 Sanofi Elf Derives du n-sulfonyl oxo-2 indole, leur preparation, les compositions pharmaceutiques en contenant.
US5849780A (en) * 1992-01-30 1998-12-15 Sanofi 1-benzenesulfonyl-1-1,3-dihydroindol-2-one derivatives, their preparation and pharmaceutical compositions in which they are present
JPH08503450A (ja) 1992-08-06 1996-04-16 ワーナー−ランバート・コンパニー 蛋白チロシンキナーゼを阻害し、かつ抗腫瘍特性を有する2−チオインドール(セレノインドール)および関連ジスルフィド(セレニド)
US5278162A (en) * 1992-09-18 1994-01-11 The Du Pont Merck Pharmaceutical Company 3,3'-disubstituted-1,3-dihydro-2H-pyrrolo[2,3-b]heterocyclic-2-one useful in the treatment of cognitive disorders of man
US5296478A (en) * 1992-10-07 1994-03-22 The Dupont Merck Pharmaceutical Co. 1-substituted oxindoles as cognition enhancers
US5776936A (en) * 1992-11-13 1998-07-07 Pharmacia & Upjohn Company Marcfortine/paraherquamide derivatives useful as antiparasitic agents
DE4242451A1 (de) * 1992-12-16 1994-06-23 Basf Ag Verfahren zur Herstellung von 5-Ringheterocyclen
US5298522A (en) 1993-01-22 1994-03-29 Pfizer Inc. 6-chloro-5-fluoro-3-(2-thenoyl)-2-oxindole-1-carboxamide as an analgesic and anti-inflammatory agent while maintaining a normal urine protein/creatinine ratio
US5716981A (en) * 1993-07-19 1998-02-10 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
FR2708606B1 (fr) * 1993-07-30 1995-10-27 Sanofi Sa Dérivés du N-phénylalkylindol-2-one, leur préparation, les compositions pharmaceutiques en contenant.
US5502072A (en) 1993-11-26 1996-03-26 Pfizer Inc. Substituted oxindoles
AT400950B (de) * 1994-02-04 1996-04-25 Immodal Pharmaka Gmbh Verfahren zur technischen herstellung definierter isomerengemische aus verbindungen mit spirozyklischen - aminocarboxyl- und/oder spirozyklischen - aminocarbonyl-systemen
EP0754183A1 (fr) * 1994-04-07 1997-01-22 Cemaf Nouveaux derives de spiro[indole-pyrrolidine] agonistes melatoninergiques, leur procede de preparation et leur utilisation a titre de medicament
US5618819A (en) * 1994-07-07 1997-04-08 Adir Et Compagnie 1,3-dihydro-2H-pyrrolo[2,3-b]pyridin-2-one and oxazolo[4,5-b]pyridin-2-(3H)-one compounds
FR2722195B1 (fr) 1994-07-07 1996-08-23 Adir Nouveaux derives de 1,3-dihydro-2h-pyrrolo(2,3-b) pyridin-2-ones et oxazolo(4,5-b) pyridin-2(3h)-ones, leur procede de preparation et les compositions pharmaceutiques qui les contiennent
US6099562A (en) * 1996-06-13 2000-08-08 Schneider (Usa) Inc. Drug coating with topcoat
CA2235686C (en) 1995-10-24 2007-06-26 Sanofi Indolin-2-one derivatives, process for their production and the pharmaceutical compositions containing them
FR2740136B1 (fr) * 1995-10-24 1998-01-09 Sanofi Sa Derives d'indolin-2-one, procede pour leur preparation et les compositions pharmaceutiques les contenant
HUP9600855A3 (en) 1996-04-03 1998-04-28 Egyt Gyogyszervegyeszeti Gyar Process for producing tenidap
JPH1095766A (ja) 1996-09-19 1998-04-14 Sanwa Kagaku Kenkyusho Co Ltd アセトアミド誘導体、及びその用途
FR2757157B1 (fr) 1996-12-13 1999-12-31 Sanofi Sa Derives d'indolin-2-one, procede pour leur preparation et compositions pharmaceutiques les contenant
ATE467418T1 (de) * 1997-01-20 2010-05-15 Immodal Pharmaka Gmbh Verfahren und stoffe zur freisetzung eines wachstumsfaktors aus endothelzellen, und nach dem verfahren freigesetzter wachstumsfaktor sowie seine verwendung
NO317155B1 (no) 1997-02-04 2004-08-30 Ono Pharmaceutical Co <omega>-cykloalkyl-prostagladin-E<N>2</N>-derivater
PT1008588E (pt) 1997-02-10 2003-09-30 Ono Pharmaceutical Co Derivados da 11,15-o-diaklquil-prostaglandina e processo de producao dos mesmos e farmaco que os contem como ingrediente activo
AU741482B2 (en) 1997-05-07 2001-11-29 Allergan Pharmaceuticals International Limited Intravaginal drug delivery devices for the administration of testosterone and testosterone precursors
US6262293B1 (en) 1997-12-25 2001-07-17 Ono Pharmaceutical Co., Ltd. ω-Cycloalkly-prostaglandin e2 derivatives
JP4087938B2 (ja) 1998-02-04 2008-05-21 高砂香料工業株式会社 ヒノキチオ−ル類の分岐サイクロデキストリン包接化合物からなる抗菌剤およびそれを含有する組成物
ATE260240T1 (de) 1998-04-01 2004-03-15 Nortran Pharmaceuticals Inc Aminocyclohexylverbindungen und deren verwendung
US20040038970A1 (en) * 1998-06-12 2004-02-26 Societe De Conseils De Recherches Etd' Application Scientifiques, S.A.S. A Paris, France Corp. Beta-carboline compounds
US6235780B1 (en) 1998-07-21 2001-05-22 Ono Pharmaceutical Co., Ltd. ω-cycloalkyl-prostaglandin E1 derivatives
AU5227999A (en) 1998-07-27 2000-02-21 Abbott Laboratories Substituted oxazolines as antiproliferative agents
US6407101B1 (en) * 1999-05-04 2002-06-18 American Home Products Corporation Cyanopyrroles
US6355648B1 (en) * 1999-05-04 2002-03-12 American Home Products Corporation Thio-oxindole derivatives
CA2373990C (en) 1999-05-21 2007-05-08 Bristol-Myers Squibb Company Pyrrolotriazine inhibitors of kinases
ATE396722T1 (de) * 1999-07-21 2008-06-15 Boehringer Ingelheim Pharma Kleine moleküle zur behandlung von endzündlichen erkrankungen
NZ516452A (en) 1999-07-21 2004-06-25 Astrazeneca Ab Spirooxindole Derivatives
US6566372B1 (en) * 1999-08-27 2003-05-20 Ligand Pharmaceuticals Incorporated Bicyclic androgen and progesterone receptor modulator compounds and methods
CA2394229C (en) 1999-11-26 2010-04-13 Mcgill University Loci for idiopathic generalized epilepsy, mutations thereof and method using same to assess, diagnose, prognose or treat epilepsy
FR2807038B1 (fr) 2000-04-03 2002-08-16 Sanofi Synthelabo Nouveaux derives d'indolin-2-one, leur preparation et les compositions pharmaceutiques les contenant
AU2002213204A1 (en) 2000-10-13 2002-04-22 Bristol-Myers Squibb Company Selective maxi-K- potassium channel openers functional under conditions of high intracellular calcium concentration, methods and uses thereof
DE60120748T2 (de) 2000-11-10 2007-05-16 Eli Lilly And Co., Indianapolis 3-substituierte oxindolderivate als beta-3-agonisten
US6670357B2 (en) * 2000-11-17 2003-12-30 Bristol-Myers Squibb Company Methods of treating p38 kinase-associated conditions and pyrrolotriazine compounds useful as kinase inhibitors
US20030078278A1 (en) 2001-06-26 2003-04-24 Pfizer Inc. Spiropiperidine compounds as ligands for ORL-1 receptor
ATE325119T1 (de) * 2001-08-14 2006-06-15 Lilly Co Eli 3-substituierte oxindol-beta-3-agonisten
EP1451173A4 (en) 2001-11-01 2005-10-26 Icagen Inc PIPERIDINE
ATE455104T1 (de) 2001-11-01 2010-01-15 Icagen Inc Pyrazolamide zur anwendung in der behandlung von schmerz
WO2003044016A1 (en) * 2001-11-20 2003-05-30 Eli Lilly And Company 3-SUBSTITUTED OXINDOLE β3 AGONISTS
SE0104341D0 (sv) * 2001-12-20 2001-12-20 Astrazeneca Ab New use
WO2003064425A1 (en) 2002-01-28 2003-08-07 Pfizer Japan Inc. N-substituted spiropiperidine compounds as ligands for orl-1 receptor
US6995144B2 (en) * 2002-03-14 2006-02-07 Eisai Co., Ltd. Nitrogen containing heterocyclic compounds and medicines containing the same
WO2003078394A1 (en) 2002-03-15 2003-09-25 Eli Lilly And Company Dihydroindol-2-one derivatives as steroid hormone nuclear receptor modulators
GB0213715D0 (en) 2002-06-14 2002-07-24 Syngenta Ltd Chemical compounds
IL165395A0 (en) 2002-06-25 2006-01-15 Wyeth Corp Use of thio-oxubdile derivatives in treatment of hormone-related conditions
US7115649B2 (en) 2002-06-25 2006-10-03 Wyeth Methods of treating skin disorders using thio-oxindole derivatives
US7256218B2 (en) 2002-11-22 2007-08-14 Jacobus Pharmaceutical Company, Inc. Biguanide and dihydrotriazine derivatives
WO2004100954A1 (en) * 2003-05-16 2004-11-25 Pfizer Products Inc. Treatment of psychotic and depressive disorders
MXPA05012325A (es) * 2003-05-16 2006-01-30 Pfizer Prod Inc Procedimiento para potenciar la cognicion usando zipirasidona.
CA2525868A1 (en) * 2003-05-16 2004-11-25 Pfizer Products Inc. Anxiety treatments with ziprasidone
MXPA05012320A (es) * 2003-05-16 2006-01-30 Pfizer Prod Inc Tratamientos de trastornos bipolares y sintomas asociados.
CN101693697A (zh) 2003-07-30 2010-04-14 泽农医药公司 哌嗪衍生物和它们作为治疗剂的用途
DE10337184A1 (de) 2003-08-13 2005-03-10 Gruenenthal Gmbh Substituierte 3-Pyrrolidin-Indol-Derivate
WO2005016913A1 (en) * 2003-08-19 2005-02-24 Pfizer Japan, Inc. Tetrahydroisoquinoline or isochroman compounds as orl-1 receptor ligands for the treatment of pain and cns disorders
EP1557166A1 (en) 2004-01-21 2005-07-27 Newron Pharmaceuticals S.p.A. Alpha-aminoamide derivatives useful in the treatment of lower urinary tract disorders
GB0406867D0 (en) 2004-03-26 2004-04-28 F2G Ltd Antifungal agents
US7354925B2 (en) 2004-03-29 2008-04-08 Pfizer Inc. Alpha aryl or heteroaryl methyl beta piperidino propanamide compounds as ORL1-receptor antagonists
GT200500063A (es) 2004-04-01 2005-10-14 Metodo para tratar la esquizofrenia y/o anormalidades glucoregulatorias
KR20060130781A (ko) 2004-04-08 2006-12-19 토포타겟 에이/에스 다이페닐 인돌-2-온 화합물 및 암 치료에 있어서의 그의용도
BRPI0510260A (pt) 2004-04-27 2007-10-23 Wyeth Corp método para purificar um composto, e, kit farmacêutico
MXPA06012660A (es) 2004-05-05 2007-03-23 Unibioscreen Sa Derivados de naftalimida para el tratamiento de cancer.
TWI350168B (en) 2004-05-07 2011-10-11 Incyte Corp Amido compounds and their use as pharmaceuticals
CA2566477A1 (en) 2004-05-14 2005-11-24 Pfizer Products Inc. Pyrimidine derivatives for the treatment of abnormal cell growth
CN101001850A (zh) 2004-06-24 2007-07-18 因塞特公司 酰胺基化合物及其作为药物的应用
GT200500186A (es) 2004-07-07 2006-03-02 Regimenes anticonceptivos con antagonistas del receptor de progesterona y kits
GT200500183A (es) 2004-08-09 2006-04-10 Moduladores del receptor de progesterona que comprenden derivados de pirrol-oxindol y sus usos
GT200500185A (es) 2004-08-09 2006-04-10 Moduladores del receptor de progesterona que comprenden derivados de pirrol-oxindol y sus usos
JP4677323B2 (ja) 2004-11-01 2011-04-27 キヤノン株式会社 画像処理装置及び画像処理方法
AU2005306476A1 (en) 2004-11-18 2006-05-26 Incyte Corporation Inhibitors of 11-beta hydroxyl steroid dehydrogenase type 1 and methods of using the same
DE102005007694A1 (de) 2005-02-18 2006-09-21 Henkel Kgaa Mittel zum Färben von keratinhaltigen Fasern
CN101160314B (zh) 2005-02-22 2012-05-23 密执安州立大学董事会 Mdm2的小分子抑制剂以及其应用
AR053710A1 (es) 2005-04-11 2007-05-16 Xenon Pharmaceuticals Inc Compuestos espiroheterociclicos y sus usos como agentes terapeuticos
AR056317A1 (es) 2005-04-20 2007-10-03 Xenon Pharmaceuticals Inc Compuestos de oxindol y composicion farmaceutica
AR053713A1 (es) 2005-04-20 2007-05-16 Xenon Pharmaceuticals Inc Compuestos heterociclicos y sus usos como agentes terapeuticos
MX2007013450A (es) * 2005-04-29 2008-01-21 Wyeth Corp Procedimniento de preparacion de oxindoles y tio-oxindoles 3,3-disustituidos.
EP1888581A2 (en) * 2005-05-16 2008-02-20 Gilead Sciences, Inc. Hiv-integrase inhibitor compounds
EP1924264B9 (en) 2005-09-01 2014-02-19 F.Hoffmann-La Roche Ag Diaminopyrimidines as p2x3 and p2x2/3 modulators
US20110294842A9 (en) * 2006-10-12 2011-12-01 Xenon Pharmaceuticals Inc. Spiro (furo [3, 2-c] pyridine-3-3' -indol) -2' (1'h)-one derivatives and related compounds for the treatment of sodium-channel mediated diseases, such as pain
WO2008046083A2 (en) 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Use of oxindole compounds as therapeutic agents
CL2007002951A1 (es) 2006-10-12 2008-02-01 Xenon Pharmaceuticals Inc Uso de compuestos derivados de espiroheterociclicos en el tratamiento de hipercolesterolemia, hiperplasia benigna de prostata, pruritis, cancer.
CL2007002950A1 (es) 2006-10-12 2008-02-01 Xenon Pharmaceuticals Inc Uso de compuestos derivados de espiro-oxindol en el tratamiento de hipercolesterolemia, hiperplasia benigna de prostata, pruritis, cancer
WO2008046084A2 (en) 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents
WO2008046087A2 (en) 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Spiro compounds and their uses as therapeutic agents
CA2666136A1 (en) 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Tricyclic spiro-oxindole derivatives and their uses as therapeutic agents
WO2008046082A2 (en) 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Use of heterocyclic compounds as therapeutic agents
WO2008117050A1 (en) 2007-03-27 2008-10-02 Astrazeneca Ab Pyrazolyl-amino-substituted pyrazines and their use for the treatment of cancer
US8263606B2 (en) * 2008-10-17 2012-09-11 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
EP2350091B1 (en) 2008-10-17 2015-06-03 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
WO2010053998A1 (en) 2008-11-05 2010-05-14 Xenon Pharmaceuticals, Inc. Spiro-condensed indole derivatives as sodium channel inhibitors
US20110269788A1 (en) * 2008-12-29 2011-11-03 Xenon Pharmaceuticals Inc. Spiro-oxindole-derivatives as sodium channel blockers
WO2010132352A2 (en) 2009-05-11 2010-11-18 Xenon Pharmaceuticals Inc. Spiro compounds and their use as therapeutic agents
AR077252A1 (es) 2009-06-29 2011-08-10 Xenon Pharmaceuticals Inc Enantiomeros de compuestos de espirooxindol y sus usos como agentes terapeuticos
EP2733145A1 (en) 2009-10-14 2014-05-21 Xenon Pharmaceuticals Inc. Synthetic methods for spiro-oxindole compounds
WO2011047173A2 (en) 2009-10-14 2011-04-21 Xenon Pharmaceuticals Inc. Pharmaceutical compositions for oral administration
PE20121699A1 (es) 2010-02-26 2012-12-22 Xenon Pharmaceuticals Inc Composiciones farmaceuticas del compuesto espiro-oxindol para administracion topica

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005035498A1 (ja) * 2003-10-08 2005-04-21 Dainippon Sumitomo Pharma Co., Ltd. 含窒素二環性化合物の摂食調節剤としての用途
WO2005099689A1 (en) * 2004-04-01 2005-10-27 Case Western Reserve University Topical delivery of phthalocyanines
WO2006110917A2 (en) * 2005-04-11 2006-10-19 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents

Non-Patent Citations (4)

* 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 cited in the application *
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 *
VILLAMIL ET AL: "Efficacy of lidocaine in the treatment of pruritus in patients with chronic cholestatic liver diseases" AMERICAN JOURNAL OF MEDICINE, XX, XX, vol. 118, no. 10, 27 September 2005 (2005-09-27), pages 1160-1163, XP005783681 ISSN: 0002-9343 *

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8106087B2 (en) 2005-04-11 2012-01-31 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
US7935721B2 (en) 2005-04-11 2011-05-03 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
JP2012505895A (ja) * 2008-10-17 2012-03-08 ゼノン・ファーマシューティカルズ・インコーポレイテッド 治療剤としてのスピロオキシインドール化合物およびそれらの使用
US8101647B2 (en) 2008-10-17 2012-01-24 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
US8916580B2 (en) 2008-10-17 2014-12-23 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
JP2015010090A (ja) * 2008-10-17 2015-01-19 ゼノン・ファーマシューティカルズ・インコーポレイテッドXenon Pharmaceuticals Inc. 治療剤としてのスピロオキシインドール化合物およびそれらの使用
CN103664973B (zh) * 2008-10-17 2017-04-19 泽农医药公司 螺羟吲哚化合物及其作为治疗剂的用途
US9458178B2 (en) 2008-10-17 2016-10-04 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
CN105175433A (zh) * 2008-10-17 2015-12-23 泽农医药公司 螺羟吲哚化合物及其作为治疗剂的用途
US8415370B2 (en) 2008-10-17 2013-04-09 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their uses as therapeutic agents
EP2350090B1 (en) * 2008-10-17 2015-06-03 Xenon Pharmaceuticals Inc. Spiro-oxindole compounds and their use as therapeutic agents
JP2012532107A (ja) * 2009-06-29 2012-12-13 ゼノン・ファーマシューティカルズ・インコーポレイテッド スピロ−オキシインドール化合物のエナンチオマーおよび治療剤としてのその使用
US8450358B2 (en) 2009-06-29 2013-05-28 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
US9480677B2 (en) 2009-06-29 2016-11-01 Xenon Pharmaceuticals Inc. Enantiomers of spiro-oxindole compounds and their uses as therapeutic agents
JP2017002085A (ja) * 2009-06-29 2017-01-05 ゼノン・ファーマシューティカルズ・インコーポレイテッドXenon Pharmaceuticals Inc. スピロ−オキシインドール化合物のエナンチオマーおよび治療剤としてのその使用
RU2544852C2 (ru) * 2009-10-14 2015-03-20 Ксенон Фармасьютикалз Инк. Способы синтеза спиро-оксиндольных соединений
US8445696B2 (en) 2009-10-14 2013-05-21 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
WO2011047173A3 (en) * 2009-10-14 2012-01-12 Xenon Pharmaceuticals Inc. Pharmaceutical compositions for oral administration
US9695185B2 (en) 2009-10-14 2017-07-04 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
JP2013508290A (ja) * 2009-10-14 2013-03-07 ゼノン・ファーマシューティカルズ・インコーポレイテッド スピロ−オキシインドール化合物のための合成方法
JP2013521232A (ja) * 2010-02-26 2013-06-10 ゼノン・ファーマシューティカルズ・インコーポレイテッド 局所投与のためのスピロオキシインドール化合物の医薬組成物および治療剤としてのその使用
CN102946859A (zh) * 2010-02-26 2013-02-27 泽农医药公司 用于局部给药的螺-羟吲哚化合物的药物组合物及其作为治疗剂的用途
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
WO2011106729A3 (en) * 2010-02-26 2012-11-15 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
AU2011220396B2 (en) * 2010-02-26 2016-10-13 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
EP3266444A1 (en) * 2010-02-26 2018-01-10 Xenon Pharmaceuticals Inc. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
CN105726531A (zh) * 2010-02-26 2016-07-06 泽农医药公司 用于局部给药的螺-羟吲哚化合物的药物组合物及其作为治疗剂的用途
US8598156B2 (en) 2010-03-25 2013-12-03 Glaxosmithkline Llc Chemical compounds
WO2012049555A1 (en) 2010-10-13 2012-04-19 Lupin Limited Spirocyclic compounds as voltage-gated sodium channel modulators
WO2013130013A1 (en) * 2012-02-27 2013-09-06 Nanyang Polytechnic Novel compounds and uses thereof
US9487535B2 (en) 2012-04-12 2016-11-08 Xenon Pharmaceuticals Inc. Asymmetric syntheses for spiro-oxindole compounds useful as therapeutic agents
EA027012B1 (ru) * 2013-03-14 2017-06-30 Эли Лилли Энд Компани Ингибиторы cdc7
KR20150119101A (ko) * 2013-03-14 2015-10-23 일라이 릴리 앤드 캄파니 Cdc7 억제제
US9156824B2 (en) 2013-03-14 2015-10-13 Eli Lilly And Company CDC7 inhibitors
KR101718645B1 (ko) 2013-03-14 2017-03-21 일라이 릴리 앤드 캄파니 Cdc7 억제제
WO2014143601A1 (en) * 2013-03-14 2014-09-18 Eli Lilly And Company CDC7 Inhibitors
CN103554120A (zh) * 2013-10-16 2014-02-05 华东师范大学 3,3-螺(2-四氢呋喃)氧化吲哚多环状化合物的制备方法
CN103554121A (zh) * 2013-10-16 2014-02-05 华东师范大学 3,3-螺(2-四氢呋喃)氧化吲哚多环状化合物及其应用
WO2015151001A1 (en) 2014-03-29 2015-10-08 Lupin Limited Sulfonamide compounds as voltage gated sodium channel modulators
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
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
WO2017037682A1 (en) 2015-09-04 2017-03-09 Lupin Limited Sulfonamide compounds as voltage-gated sodium channel modulators
US10239869B2 (en) 2015-09-04 2019-03-26 Lupin Limited Sulfonamide compounds as voltage-gated sodium channel modulators
US11434230B2 (en) 2016-01-20 2022-09-06 Chemocentryx, Inc. 2-oxindole compounds
US10100060B2 (en) 2016-06-16 2018-10-16 Xenon Pharmaceuticals Inc. Asymmetric synthesis of funapide
WO2018163077A1 (en) 2017-03-08 2018-09-13 Lupin Limited Indanyl compounds as voltage gated sodium channel modulators
WO2020011731A1 (en) * 2018-07-12 2020-01-16 UCB Biopharma SRL Spirocyclic indane analogues as il-17 modulators
US11458124B2 (en) 2018-07-12 2022-10-04 UCBBiopharma Srl Spirocyclic indane analogues as IL-17 modulators
CN108976243A (zh) * 2018-08-23 2018-12-11 青岛农业大学 通过二甲基呋喃与含氧化吲哚邻羟基苄醇合成螺-色满-4,3′-氧化吲哚的合成方法

Also Published As

Publication number Publication date
AR063280A1 (es) 2009-01-21
US8466188B2 (en) 2013-06-18
EP2073806B1 (en) 2012-02-15
MX2009003876A (es) 2009-05-11
TW200833695A (en) 2008-08-16
ATE545416T1 (de) 2012-03-15
CN101631546A (zh) 2010-01-20
AU2007319580B2 (en) 2012-11-29
BRPI0719210A2 (pt) 2015-05-05
JP2013256539A (ja) 2013-12-26
CN103271906A (zh) 2013-09-04
WO2008060789A3 (en) 2008-07-24
US20100173967A1 (en) 2010-07-08
US20110172282A9 (en) 2011-07-14
RU2009117642A (ru) 2010-11-20
JP2010506854A (ja) 2010-03-04
CA2665698A1 (en) 2008-05-22
AU2007319580A1 (en) 2008-05-22
JP5460324B2 (ja) 2014-04-02
EP2073806A2 (en) 2009-07-01
CL2007002950A1 (es) 2008-02-01

Similar Documents

Publication Publication Date Title
EP2073806B1 (en) Use of spiro-oxindole compounds as therapeutic agents
US7700641B2 (en) Spiro-oxindole compounds and their uses as therapeutic agents
JP5834104B2 (ja) 治療剤としてのスピロオキシインドール化合物およびそれらの使用
WO2008046083A2 (en) Use of oxindole compounds as therapeutic agents

Legal Events

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

Ref document number: 200780038111.X

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2007868434

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007319580

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2665698

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2009/003876

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2009532606

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007319580

Country of ref document: AU

Date of ref document: 20071012

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2009117642

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12445264

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0719210

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090409