US20140187533A1 - Benzimidazole inhibitors of the sodium channel - Google Patents

Benzimidazole inhibitors of the sodium channel Download PDF

Info

Publication number
US20140187533A1
US20140187533A1 US14/002,943 US201214002943A US2014187533A1 US 20140187533 A1 US20140187533 A1 US 20140187533A1 US 201214002943 A US201214002943 A US 201214002943A US 2014187533 A1 US2014187533 A1 US 2014187533A1
Authority
US
United States
Prior art keywords
optionally substituted
trifluoromethyl
bis
benzo
imidazol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/002,943
Other languages
English (en)
Inventor
Hassan Pajouhesh
Richard Holland
Lingyun Zhang
Hossein Pajouhesh
Jason Lamontagne
Brendan Whelan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Epirus Biopharmaceuticals Inc
Original Assignee
Zalicus Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zalicus Pharmaceuticals Ltd filed Critical Zalicus Pharmaceuticals Ltd
Priority to US14/002,943 priority Critical patent/US20140187533A1/en
Assigned to ZALICUS PHARMACEUTICALS LTD. reassignment ZALICUS PHARMACEUTICALS LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOLLAND, RICHARD, LAMONTAGNE, Jason, WHELAN, BRENDAN, ZHANG, LINGYUN, PAJOUHESH, HASSAN, PAJOUHESH, HOSSEIN
Publication of US20140187533A1 publication Critical patent/US20140187533A1/en
Assigned to EPIRUS BIOPHARMACEUTICALS, INC. reassignment EPIRUS BIOPHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZALICUS PHARMACEUTICALS LTD.
Assigned to DEGIACOMO, INTERIM TRUSTEE, MARK G. reassignment DEGIACOMO, INTERIM TRUSTEE, MARK G. COURT APPOINTMENT (SEE DOCUMENT FOR DETAILS). Assignors: EPIRUS BIOPHARMACEUTICALS, INC.
Assigned to DEGIACOMO, INTERIM TRUSTEE, MARK G. reassignment DEGIACOMO, INTERIM TRUSTEE, MARK G. COURT APPOINTMENT (SEE DOCUMENT FOR DETAILS). Assignors: EPIRUS BIOPHARMACEUTICALS, INC.
Assigned to AnaBios Corporation reassignment AnaBios Corporation ASSET PURCHASE AGREEMENT Assignors: DEGIACOMO, INTERIM TRUSTEE, MARK G.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C237/06Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • 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
    • A61K31/401Proline; Derivatives thereof, e.g. captopril
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D205/00Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
    • C07D205/02Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D205/04Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/60Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/14Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/06Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members
    • C07D241/08Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • the invention relates to compounds useful in treating conditions associated with voltage-gated ion channel function, particularly conditions associated with sodium channel activity. More specifically, the invention concerns benzimdiazole compounds that are useful in treatment numerous diseases and conditions.
  • Voltage-gated sodium (Na v ) channels are present in neurons and excitable tissues where they contribute to processes such as membrane excitability and muscle contraction (Ogata et al., Jpn. J. Pharmacol . (2002) 88(4) 365-77).
  • Na v 1.1-1.9 transmembrane ⁇ -subunits from a single Na v 1 family combine with auxiliary ⁇ -subunits that modify channel function to form functional Na v channels.
  • Na v 1 ⁇ -subunit isoforms Of the nine Na v 1 ⁇ -subunit isoforms, five are expressed in the dorsal root ganglion where they are involved in setting the resting membrane potential and the threshold for generating action potentials, and also contribute to the upstroke as well as firing of action potentials during sustained depolarization.
  • TTX tetrodotoxin
  • the tetrodotoxin (TTX) sensitive Na v 1.7 and TTX-insensitive Na v 1.8 channel subtypes act as major contributors to both inflammatory and neuropathic pain (Momin et al., Curr Opin Neurobiol. 18(4):383-8, 2008; Rush et al., J. Physiol. 579(Pt 1):1-14, 2007).
  • Novel allosteric modulators of voltage-gated ion channels are thus desired.
  • Modulators may affect the kinetics and/or the voltage potentials of, e.g., Na v 1.7 and/or Na v 1.8 channels.
  • the invention relates to compounds useful in conditions modulated by voltage-gated ion channels (e.g., voltage gated sodium channels).
  • voltage-gated ion channels e.g., voltage gated sodium channels.
  • the invention features a compound having a structure according to the following formula,
  • each of R 1 , R 2 , R 3 , and R 4 is selected, independently, from H, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 haloalkyl, optionally substituted C6-C10 aryl, or optionally substituted 5 to 6-membered heteroaryl, where at least one of R 1 , R 2 , R 3 , and R 4 is halogen or optionally substituted C1-C6 haloalkyl;
  • R 5 is H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C10 heteroalkyl;
  • R 6 is —R 6A or —CH 2 R 6B ;
  • R 6A is NH 2 , optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, optionally substituted pyrimidine, optionally substituted C1-C10 aminoalkyl, optionally substituted C1-C10 hydroxyalkyl, optionally substituted C1-C10 alkoxyalkyl, optionally substituted C1-C10 haloalkyl, or optionally substituted C1-C10 alkylsulfonyl; or R 6A has a structure according to
  • n is an integer between 0-4;
  • Z 1 is CH 2 , NH, NCH 3 , or O;
  • L 1 is —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , or —CH 2 NHC( ⁇ O)CH 2 ;
  • each R 2A and R 2C when present, is selected from OH, N(R 2B ) 2 , halogen, and unsubstituted C1-C3 alkyl, or two R 2A combine to form an oxo ( ⁇ O) group, and wherein no more than two R 2A combine to form an oxo group; and
  • each R 2B is, independently, H or unsubstituted C1-C6 alkyl
  • R 2D is H, OH, or NH 2 ;
  • R 6B is optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, or optionally substituted pyrimidine.
  • R 6A is NH 2 , optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, optionally substituted pyrimidine, optionally substituted C1-C10 aminoalkyl, optionally substituted C1-C10 hydroxyalkyl, optionally substituted C1-C10 alkoxyalkyl, optionally substituted C1-C10 haloalkyl, or optionally substituted C1-C10 alkylsulfonyl.
  • R 6 has a structure according to
  • R 5 is H.
  • R 5 is optionally substituted C1-C10 heteroalkyl.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • R 6 is —CH 2 R 6B , and R 6B is optionally substituted azetidine.
  • R 6 is optionally substituted C1-C10 aminoalkyl.
  • the C1-C10 aminoalkyl includes an oxo ( ⁇ O) substituent, an alkoxy substituent, an N-sulfonyl group, or any combination thereof.
  • the compound has a structure according to the following formula,
  • n 0 or 1
  • R 7 is H or —C( ⁇ O)R 7A , where R 7A is unsubstituted C1-C6 alkyl or optionally substituted C1-C10 aminoalkyl.
  • n 0.
  • n 1
  • R 7 is H or C( ⁇ O)R 7A , where R 7A is unsubstituted C1-C3 alkyl or an optionally substituted C1-C10 aminoalkyl including a terminal —NH 2 group.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • R 6 is optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, optionally substituted pyrimidine.
  • R 6 includes a —NH 2 substituent.
  • the compound has a structure according to the following formula,
  • R 6B is optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, or optionally substituted pyrimidine.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • R 5 is H.
  • R 6 is an optionally substituted C1-C10 aminoalkyl group.
  • the C1-C10 aminoalkyl includes a terminal —NH 2 group.
  • the C1-C10 aminoalkyl includes an oxo ( ⁇ O) substituent.
  • R 6 is —(CH 2 ) m1 (NR 6C ) m2 (C ⁇ O) m3 (CH 2 ) m4 NR 6D R 6E or —(CH 2 ) m1 (C(CH 3 ) 2 ) m2 (CH 2 ) m4 NR 6C R 6D where each of m1 and m4 is, independently, an integer between 1-6; each of m2 and m3 is, independently, 0 or 1; each of R 6C and R 6E is, independently, H or unsubstituted C1-C6 alkyl; and R 6D is H, unsubstituted C1-C6 alkyl, or an N-protecting group.
  • R 6 is —(CH 2 ) m1 NH 2 , —CH 2 NHC( ⁇ O)CH 2 NH 2 , —C(CH 3 ) 2 CH 2 NH 2 , —C(CH 3 ) 2 NH 2 , and where m1 is 1, 2, or 3.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • one and only one of R 2 or R 3 is optionally substituted phenyl.
  • R 5 is H.
  • R 6 is optionally substituted C1-C3 haloalkyl, optionally substituted C1-C10 alkoxyalkyl, optionally substituted C1-C10 hydroxyalkyl, or optionally substituted C1-C10 alkylsulfonyl.
  • R 6 is —(CH 2 ) m1 CF 3 , —(CH 2 ) m1 OR 6F , —(CH 2 ) m1 SO 2 R 6G , where m1 is an integer between 1-6, R 68 is H or CH 3 , and R 6G is unsubstituted C1-C3 alkyl.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • R 5 is H.
  • the invention features a compound having a structure according to the following formula,
  • each of X 1 , X 2 , and X 3 is N or CR 4 , and where one and only one of X 1 , X 2 , and X 3 is N;
  • L 1 is a covalent bond, —CH 2 , —CHR 5A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , —CH 2 NHC( ⁇ O)CH 2 , or —CH 2 CH 2 ;
  • each of R 1 , R 2 , and R 4 is, independently, H, unsubstituted C1-C3 alkyl, optionally substituted C1-C3 haloalkyl, or halogen;
  • R 3 is H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C10 heteroalkyl;
  • R 5A is selected from optionally substituted C1-C3 alkyl
  • R 1 , R 2 , R 3 , and R 5 is halogen or optionally substituted C1-C3 haloalkyl.
  • L 1 is CH 2 or CHCF 3 .
  • R 3 is H.
  • the invention features a compound having a structure according to the following formula,
  • n is an integer between 0-4;
  • R 1 is selected from —CH 2 R 3A , —CHR 4A R 3A , —CH 2 C( ⁇ O)R 3A , —C( ⁇ O)CH 2 R 3A , —CH 2 C( ⁇ O)NR 4B R 3A , —CH 2 C( ⁇ O)NR 4B CH 2 R 3A , —CH 2 NR 4B C( ⁇ O)CH 2 R 3A , —R 3B , —CH 2 CH 2 R 3B , and —CH 2 C( ⁇ O)NR 4B CHR 4C R 3C ;
  • each R 2 when present, is selected from OH, N(R 2A ) 2 , halogen, and unsubstituted C1-C3 alkyl, or two R 2 combine to form an oxo ( ⁇ O) group, and where no more than two R 2 combine to form an oxo group;
  • each R 2A is, independently, H or unsubstituted C1-C6 alkyl
  • R 3A is selected from
  • R 3B is selected from
  • R 3C is optionally substituted pyridine
  • R 4A is optionally substituted C1-C3 alkyl
  • R 4B is H or optionally substituted C1-C3 alkyl
  • R 4C is C1-C3 haloalkyl
  • Z 1 is selected from CH 2 , O, and NR 5 , where R 5 is H or unsubstituted C1-C6 alkyl;
  • Z 2 is NH, NR 6 , CHR 2 , CR 6 R 2 , where R 6 is a covalent bond to R 1 .
  • n 0.
  • n is 2 or 4.
  • two R 2 combine to form an oxo group.
  • R 2 is CH 3 .
  • Z 1 is O, NH, CH 2 , or NCH 3 .
  • Z 2 is N, CH, or CNH 2 .
  • Z 1 is CH 2 , NH, NCH 3 , or O;
  • L 1 is —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , or —CH 2 NHC( ⁇ O)CH 2 ;
  • R 5 is H or C1-C10 heteroalkyl
  • each of R 1 , R 2 , R 3 , and R 4 is, independently, H, unsubstituted C1-C3 alkyl, optionally substituted C1-C3 haloalkyl, or halogen, and
  • R 1 , R 2 , R 3 , R 4 , and R R5 is not H.
  • Z 1 is NH
  • n 2 or 4.
  • two R 2A combine to form an oxo group.
  • R 2A is CH 3 .
  • R 1 and R 4 are both H.
  • R 1 and R 4 are, independently, F, CF 3 , or Cl.
  • R 3 is F, Cl, or CF 3 .
  • R 2 is F, Cl, or CF 3 .
  • R 5 is H.
  • R 5 is optionally substituted C1-C10 hydroxyalkyl or C1-C10 aminoalkyl.
  • L 1 is CH 2 .
  • the compound has a structure according to the following formula,
  • Z 1 is CH 2 , NH, NCH 3 , or O;
  • each of X 1 , X 2 , and X 3 is N or CR 8C , and where one and only one of X 1 , X 2 , and X 3 is N;
  • L 1 is a covalent bond, —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , —CH 2 NHC( ⁇ O)CH 2 , or —CH 2 CH 2 ;
  • each of R 8A , R 8B , and R 8C is, independently, H, unsubstituted C1-C3 alkyl, optionally substituted C1-C3 haloalkyl, or halogen, and
  • Z 1 is NH
  • n is 2 or 4. In certain embodiments, two R 2 combine to form an oxo group.
  • R 2 is CH 3 .
  • X 2 is N.
  • At least one of R 8A , R 8B , and R 8C is F, Cl, or CF 3 .
  • L 1 is —CH 2 C( ⁇ O)NHCH 2 or —CH 2 NHC( ⁇ O)CH 2 .
  • the compound has a structure according to the following formula,
  • Z 1 is CH 2 or NH
  • L 1 is a covalent bond, —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , —CH 2 C( ⁇ O)NHCHCF 3 —, or —CH 2 NHC( ⁇ O)CH 2 ;
  • n is an integer between 0-4;
  • each R 2C when present, is independently, OH, NH 2 , NHCH 3 , N(CH 3 ) 2 , or unsubstituted C1-C3 alkyl, or two R 2C groups combine to form an oxo ( ⁇ O) group, and wherein no more than one R 2B or R 2C group can be OH NH 2 , NHCH 3 , or N(CH 3 ) 2 ;
  • R 2D is H, OH, or NH 2 ,
  • R 5 is H or C1-C10 heteroalkyl
  • each of R 1 , R 2 , R 3 , and R 4 is, independently, H, unsubstituted C1-C3 alkyl, optionally substituted C1-C3 haloalkyl or halogen, and
  • R 1 , R 2 , R 3 , R 4 , and R 5 is not H.
  • Z 1 is CH 2 .
  • n is 0 or 1.
  • one of R 2 and R 3 is NH 2 , NHCH 3 , or N(CH 3 ) 2 .
  • R 1 and R 4 are both H.
  • R 1 and R 4 are, independently, F, CF 3 , or Cl.
  • R 3 is F, Cl, or CF 3 .
  • R 2 is F, Cl, or CF 3 .
  • R 5 is H.
  • the compound has a structure according to the following formula,
  • Z 1 is CH 2 , NH, NCH 3 , or O;
  • L 1 is a covalent bond, —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , —CH 2 NHC( ⁇ O)CH 2 , or —CH 2 CH 2 ;
  • R 7 is selected from H, optionally substituted C1-C6 alkyl, and optionally substituted C1-C10 heteroalkyl;
  • each of R 8A and R 8B is selected, independently, from H, halogen, unsubstituted C1-C3 alkyl, and C1-C3 haloalkyl, and
  • R 7 , R 8A and R 8B is not H.
  • Z 1 is N.
  • n 2 or 4.
  • two R 2 combine to form an oxo group.
  • R 2 is CH 3 .
  • R 7 is unsubstituted C1-C3 alkyl.
  • At least one of R 8A and R 8B is F, Cl, or CF 3 .
  • L 1 is CH 2 .
  • the invention features a compound having a structure according to the following formula,
  • each of R 1 , R 2 , and R 3 is, independently, H, unsubstituted C1-C6 alkyl, optionally substituted C1-C6 haloalkyl, or halogen;
  • n 1 or 2;
  • each R 4 and R 5 is, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 haloalkyl, or R 4 and R 5 combine to form an optionally substituted C3-C6 cycloalkyl, or R 4 and R 5 combine to form an oxo (C ⁇ O) group;
  • each of R 6 and R 8 is, independently, H or optionally substituted C1-C6 alkyl; or R 6 and R 8 combine to form an optionally substituted three-to-nine membered heterocyclyl, or R 6 and R 7A combine to form an optionally substituted three-to-nine membered heterocyclyl;
  • n 1 or 2;
  • each R 7A and R 7B is, independently H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 haloalkyl; or R 6 combines with R 7A to form an optionally substituted three-to-nine heterocyclyl; or an R 7A and R 7B group on the same carbon combine to form an optionally substituted C3-C6 cycloalkyl; or, when n is 2, both R 7A groups combine to form an optionally substituted C3-C6 cycloalkyl.
  • each of R 1 , R 2 , and R 3 is, independently, H, C1-C3 haloalkyl, or halogen.
  • one of R 1 , R 2 , and R 3 is H.
  • two of R 1 , R 2 , and R 3 are, independently, CF 3 , Cl, or F.
  • R 4 and R 5 are both H; or R 4 and R 5 are both CH 3 ; or R 4 and R 5 combine to form an optionally substituted C3-C6 cycloalkyl; or R 4 and R 5 combine to form an oxo (C ⁇ O) group.
  • R 6 combines with R 7A to form a three-to-six membered heterocyclyl ring, or wherein R 6 and R 8 combine to form an optionally substituted three-to-six membered heterocyclyl.
  • R 6 is H.
  • R 7A is H and R 7B is optionally substituted C1-C6 alkyl.
  • the compound has a structure according to a formula that is
  • n is 1, and R 7A and R 7B are both H, or R 7A is H and R 7B is optionally substituted C1-C6 alkyl.
  • R 6 is H and R 8 is optionally substituted C1-C6 alkyl, or wherein R 6 and R 8 combine to form an optionally substituted five- to six-membered heterocyclyl (e.g., an unsubstituted five- to six-membered heterocyclyl or a five- to six-membered heterocyclyl that includes a phenyl substituent).
  • the invention features a compound having a structure selected from the group consisting of any of Compounds (1)-(236) of Table 1, or a pharmaceutically acceptable salt or solvate thereof.
  • the invention features a pharmaceutical composition that includes any of the compounds described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is formulated in unit dosage form (e.g., a tablet, caplet, capsule, lozenge, film, strip, gelcap, or syrup).
  • unit dosage form e.g., a tablet, caplet, capsule, lozenge, film, strip, gelcap, or syrup.
  • the invention features method to treat a disease or condition by administering to a subject in need of such treatment an effective amount of any of the compounds described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1), or a pharmaceutical composition thereof.
  • an effective amount of any of the compounds described herein e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1
  • a pharmaceutical composition thereof e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1
  • the disease or condition is pain, epilepsy, Parkinson's disease, a mood disorder (e.g., a major depressive disorder (e.g., atypical depression, melancholic depression, psychotic major depression, catatonic depression, postpartum depression, seasonal affective disorder, dysthymia, and depressive disorder not otherwise specified (DD-NOS)), recurrent brief depression, minor depressive disorder, or a bipolar disorder), psychosis (e.g., schizophrenia), tinnitus, amyotropic lateral sclerosis, glaucoma, ischaemia, spasticity disorders, obsessive compulsive disorder, restless leg syndrome, and Tourette syndrome.
  • a mood disorder e.g., a major depressive disorder (e.g., atypical depression, melancholic depression, psychotic major depression, catatonic depression, postpartum depression, seasonal affective disorder, dysthymia, and depressive disorder not otherwise specified (DD-NOS)
  • DD-NOS
  • the subject is a fasted subject.
  • the subject is a fed subject.
  • the condition is pain or epilepsy.
  • the pain is inflammatory pain (e.g., inflammatory pain caused by rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, inflammatory bowel disease, primary dysmenorrhea, or endometriosis) or neuropathic pain.
  • inflammatory pain e.g., inflammatory pain caused by rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, inflammatory bowel disease, primary dysmenorrhea, or endometriosis
  • neuropathic pain e.g., inflammatory pain caused by rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, inflammatory bowel disease, primary dysmenorrhea, or endometriosis
  • neuropathic pain e.g., inflammatory pain caused by rheumatoid arthritis
  • the pain is chronic pain.
  • the chronic pain is peripheral neuropathic pain; central neuropathic pain, musculoskeletal pain, headache, visceral pain, or mixed pain.
  • the peripheral neuropathic pain is post-herpetic neuralgia, diabetic neuropathic pain, neuropathic cancer pain, HIV-associated neuropathy, erythromelalgia, failed back-surgery syndrome, trigeminal neuralgia, or phantom limb pain; said central neuropathic pain is multiple sclerosis related pain, Parkinson disease related pain, post-stroke pain, post-traumatic spinal cord injury pain, lumbosacral radiculopathy, cervical radiculopathy, brachial radiculopathy, or pain in dementia; the musculoskeletal pain is osteoarthritic pain and fibromyalgia syndrome; inflammatory pain such as rheumatoid arthritis, or endometriosis; the headache is migraine, cluster headache, tension headache syndrome, facial pain, or headache caused by other diseases; the visceral pain is interstitial cystitis, irritable bowel syndrome, or chronic pelvic pain syndrome; or the mixed pain is lower back pain, neck and shoulder pain, burning mouth
  • the headache is migraine.
  • the pain is acute pain.
  • the acute pain is nociceptive pain or post-operative pain.
  • the invention features a method of modulating a voltage-gated sodium channel, the method including contacting a cell with any of the compounds described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1).
  • any of the compounds described herein e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1).
  • alkoxy represents a chemical substituent of formula —OR, where R is an optionally substituted C1-C6 alkyl group, unless otherwise specified.
  • the alkyl group can be substituted, e.g., the alkoxy group can have 1, 2, 3, 4, 5 or 6 substituent groups as defined herein.
  • alkoxyalkyl represents a heteroalkyl group, as defined herein, that is described as an alkyl group that is substituted with an alkoxy group.
  • exemplary unsubstituted alkoxyalkyl groups include between 2 to 12 carbons.
  • the alkyl and the alkoxy each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group.
  • alkyl As used herein, the term “alkyl,” “alkenyl” and “alkynyl” include straight-chain, branched-chain and cyclic monovalent substituents, as well as combinations of these, containing only C and H when unsubstituted. Examples include methyl, ethyl, isobutyl, cyclohexyl, cyclopentylethyl, 2-propenyl, 3-butynyl, and the like.
  • cycloalkyl represents a monovalent saturated or unsaturated non-aromatic cyclic alkyl group having between three to nine carbons (e.g., a C3-C9 cycloalkyl), unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.2.1]heptyl, and the like.
  • the cycloalkyl group includes one carbon-carbon double bond
  • the cycloalkyl group can be referred to as a “cycloalkenyl” group.
  • Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, and the like.
  • the alkyl, alkenyl and alkynyl groups contain 1-12 carbons (e.g., C1-C12 alkyl) or 2-12 carbons (e.g., C2-C12 alkenyl or C2-C12 alkynyl).
  • the alkyl groups are C1-C8, C1-C6, C1-C4, C1-C3, or C1-C2 alkyl groups; or C2-C8, C2-C6, C2-C4, or C2-C3 alkenyl or alkynyl groups.
  • any hydrogen atom on one of these groups can be replaced with a substituent as described herein.
  • Heteroalkyl, heteroalkenyl and heteroalkynyl are similarly defined and contain at least one carbon atom but also contain one or more O, S or N heteroatoms or combinations thereof within the backbone residue whereby each heteroatom in the heteroalkyl, heteroalkenyl or heteroalkynyl group replaces one carbon atom of the alkyl, alkenyl or alkynyl group to which the heteroform corresponds.
  • the heteroalkyl, heteroalkenyl and heteroalkynyl groups have C at each terminus to which the group is attached to other groups, and the heteroatom(s) present are not located at a terminal position. As is understood in the art, these heteroforms do not contain more than three contiguous heteroatoms.
  • heteroatom is O or N.
  • heterocyclyl represents cyclic heteroalkyl or heteroalkenyl that is, e.g., a 3-, 4-, 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • the 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds.
  • heterocyclyl also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group.
  • heterocyclyl includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like.
  • heteroalkyl is defined as C1-C6, it will contain 1-6 C, N, O, or S atoms such that the heteroalkyl contains at least one C atom and at least one heteroatom, for example 1-5 carbons and 1 N atom, or 1-4 carbons and 2 N atoms.
  • heteroalkyl is defined as C1-C6 or C1-C4, it would contain 1-5 carbons or 1-3 carbons respectively, i.e., at least one C is replaced by O, N or S.
  • heteroalkenyl or heteroalkynyl when defined as C2-C6 (or C2-C4), it would contain 2-6 or 2-4 C, N, O, or S atoms, since the heteroalkenyl or heteroalkynyl contains at least one carbon atom and at least one heteroatom, e.g. 2-5 carbons and 1 N atom, or 2-4 carbons, and 2 O atoms. Further, heteroalkyl, heteroalkenyl or heteroalkynyl substituents may also contain one or more carbonyl groups.
  • heteroalkyl, heteroalkenyl and heteroalkynyl groups include CH 2 OCH 3 , CH 2 N(CH 3 ) 2 , CH 2 OH, (CH 2 ) n NR 2 , OR, COOR, CONR 2 , (CH 2 ) n OR,(CH 2 ) n COR, (CH 2 ) n COOR, (CH 2 ) n SR, (CH 2 ) n SOR, (CH 2 ) n SO 2 R 2 , (CH 2 ) n CONR 2 , NRCOR, NRCOOR, OCONR 2 , OCOR and the like wherein the R group contains at least one C and the size of the substituent is consistent with the definition of e.g., alkyl, alkenyl, and alkynyl, as described herein (e.g., n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12).
  • alkylene alkenylene
  • alkynylene alkynylene
  • alk divalent or trivalent groups having a specified size, typically C1-C2, C1-C3, C1-C4, C1-C6, or C1-C8 for the saturated groups (e.g., alkylene or alk) and C2-C3, C2-C4, C2-C6, or C2-C8 for the unsaturated groups (e.g., alkenylene or alkynylene).
  • saturated groups e.g., alkylene or alk
  • C2-C3, C2-C4, C2-C6, or C2-C8 unsaturated groups
  • C ⁇ O is a C1 alkylene that is substituted by ⁇ O, for example.
  • alkaryl represents an aryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein
  • alkheteroaryl refers to a heteroaryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein.
  • the alkylene and the aryl or heteroaryl group are each optionally substituted as described herein.
  • Heteroalkylene, heteroalkenylene and heteroalkynylene are similarly defined as divalent groups having a specified size, typically C1-C3, C1-C4, C1-C6, or C1-C8 for the saturated groups and C2-C3, C2-C4, C2-C6, or C2-C8 for the unsaturated groups.
  • heteroalkylene, heteroalkenylene or heteroalkynylene group replaces one carbon atom of the alkylene, alkenylene or alkynylene group to which the heteroform corresponds.
  • these heteroforms do not contain more than three contiguous heteroatoms.
  • alkylsulfonyl represents a heteroalkyl group that is described as an optionally substituted alkyl group, as described herein, that includes an —S(O) 2 — group.
  • amino represents —N(R N1 ) 2 , wherein each R N1 is, independently, H, OH, NO 2 , N(R N2 ) 2 , SO 2 OR N2 , SO 2 R N2 , SOR N2 , SO 2 N(R N2 ) 2 , SON(R N2 ) 2 , an N-protecting group, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, heterocyclyl (e.g., heteroaryl), alkheterocyclyl (e.g., alkheteroaryl), or two R N1 combine to form a heterocyclyl or an N-protecting group, and wherein each R N2 is, independently, H, alkyl, or aryl.
  • amino is —NH 2 , or —NHR N1 , wherein R N1 is, independently, OH, NO 2 , NH 2 , NR N2 2 , SO 2 OR N2 , SO 2 R N2 , SOR N2 , SO 2 N(R N2 ) 2 , SON(R N2 ) 2 , alkyl, or aryl, and each R N2 can be H, alkyl, or aryl.
  • the term “aminoalkyl,” as used herein, represents a heteroalkyl group, as defined hrein, that is described as an alkyl group, as defined herein, substituted by an amino group, as defined herein.
  • the alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group.
  • the alkyl moiety may comprise an oxo ( ⁇ O) substituent.
  • “Aromatic” moiety or “aryl” moiety refers to any monocyclic or fused ring bicyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system and includes a monocyclic or fused bicyclic moiety such as phenyl or naphthyl; “heteroaromatic” or “heteroaryl” also refers to such monocyclic or fused bicyclic ring systems containing one or more heteroatoms selected from O, S and N. The inclusion of a heteroatom permits inclusion of 5-membered rings to be considered aromatic as well as 6-membered rings.
  • typical aromatic/heteroaromatic systems include pyridyl, pyrimidyl, indolyl, benzimidazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, thienyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, benzoxazolyl, benzoisoxazolyl, imidazolyl and the like. Because tautomers are theoretically possible, phthalimido is also considered aromatic.
  • the ring systems contain 5-12 ring member atoms or 6-10 ring member atoms.
  • the aromatic or heteroaromatic moiety is a 6-membered aromatic rings system optionally containing 1-2 nitrogen atoms. More particularly, the moiety is an optionally substituted phenyl, pyridyl, indolyl, pyrimidyl, pyridazinyl, benzothiazolyl or benzimidazolyl, pyrazolyl, imidazolyl, isoxazolyl, thiazolyl, benzothiazolyl, indolyl. Even more particularly, such moiety is phenyl, pyridyl, or pyrimidyl and even more particularly, it is phenyl.
  • O-aryl or “O-heteroaryl” refers to aromatic or heteroaromatic systems which are coupled to another residue through an oxygen atom.
  • a typical example of an O-aryl is phenoxy.
  • arylalkyl refers to aromatic and heteroaromatic systems which are coupled to another residue through a carbon chain, saturated or unsaturated, typically of C1-C8, C1-C6, or more particularly C1-C4 or C1-C3 when saturated or C2-C8, C2-C6, C2-C4, or C2-C3 when unsaturated, including the heteroforms thereof.
  • arylalkyl thus includes an aryl or heteroaryl group as defined above connected to an alkyl, heteroalkyl, alkenyl, heteroalkenyl, alkynyl or heteroalkynyl moiety also as defined above.
  • Typical arylalkyls would be an aryl(C6-C12)alkyl(C1-C8), aryl(C6-C12)alkenyl(C2-C8), or aryl(C6-C12)alkynyl(C2-C8), plus the heteroforms.
  • a typical example is phenylmethyl, commonly referred to as benzyl.
  • Halo may be any halogen atom, especially F, Cl, Br, or I, and more particularly it is fluoro or chloro.
  • haloalkyl represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, Cl, Br, or I).
  • a haloalkyl may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens.
  • Haloalkyl groups include perfluoroalkyls.
  • the haloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.
  • hydroxy represents an —OH group.
  • hydroxyalkyl represents an alkyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by hydroxymethyl, dihydroxypropyl, and the like.
  • N-protecting group represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference.
  • N-protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, ⁇ -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine, and the like; sulfonyl-containing groups such as benzenesulfonyl, p-toluenesulfonyl, and the like; carbamate forming groups such as benzyloxycarbon
  • N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).
  • a substituent group e.g., alkyl, alkenyl, alkynyl, or aryl (including all heteroforms defined above) may itself optionally be substituted by additional substituents.
  • additional substituents e.g., alkyl, alkenyl, alkynyl, or aryl (including all heteroforms defined above
  • alkyl may optionally be substituted by the remaining substituents listed as substituents where this makes chemical sense, and where this does not undermine the size limit of alkyl per se; e.g., alkyl substituted by alkyl or by alkenyl would simply extend the upper limit of carbon atoms for these embodiments, and is not included.
  • substituents include, but are not limited to: C1-C6 alkyl or heteroaryl, C2-C6 alkenyl or heteroalkenyl, C2-C6 alkynyl or heteroalkynyl, halogen; aryl, heteroaryl, azido(—N 3 ), nitro (—NO 2 ), cyano (—CN), acyloxy(—OC( ⁇ O)R′), acyl (—C( ⁇ O)R′), alkoxy (—OR′), amido (—NR′C( ⁇ O)R′′ or —C( ⁇ O)NRR′), amino (—NRR′), carboxylic acid (—CO 2 H), carboxylic ester (—CO 2 R′), carbamoyl (—OC( ⁇ O)NR′R′′ or —NRC( ⁇ O)OR′), hydroxy (—OH), isocyano (—NC),
  • Typical optional substituents on aromatic or heteroaromatic groups include independently halo, CN, NO 2 , CF 3 , OCF 3 , COOR′, CONR′ 2 , OR′, SR′, SOR′, SO 2 R′, NR′ 2 , NR′(CO)R′,NR′C(O)OR′, NR′C(O)NR′ 2 , NR′SO 2 NR′ 2 , or NR′SO 2 R′, wherein each R 1 is independently H or an optionally substituted group selected from alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, and aryl (all as defined above); or the substituent may be an optionally substituted group selected from alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, O-aryl, O-heteroaryl and aryl
  • non-aromatic groups e.g., alkyl, alkenyl, and alkynyl groups
  • a non-aromatic group may also include a substituent selected from ⁇ O and ⁇ NOR 1 where R 1 is H or an optionally substituted group selected from alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteralkynyl, heteroaryl, and aryl (all as defined above).
  • an “effective amount” of an agent is that amount sufficient to effect beneficial or desired results, such as clinical results, and, as such, an “effective amount” depends upon the context in which it is being applied.
  • an agent e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1
  • an effective amount depends upon the context in which it is being applied.
  • an agent that is a modulator of a voltage-gated ion channel e.g., a sodium channel such as Na v 1.7 or Na v 1.8
  • an effective amount of an agent is, for example, an amount sufficient to achieve a change in sodium channel activity as compared to the response obtained without administration of the agent.
  • composition represents a composition containing a compound described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) formulated with a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
  • compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein.
  • unit dosage form e.g., a tablet, capsule, caplet, gelcap, or syrup
  • topical administration e.g., as a cream, gel, lotion, or ointment
  • intravenous administration e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use
  • a “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non-inflammatory in a patient.
  • Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration.
  • excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, B
  • prodrugs as used herein, represents those prodrugs of the compounds of the present invention that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • pharmaceutically acceptable salt represents those salts of the compounds described here (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P. H. Stahl and C. G. Wermuth), Wiley-VCH, 2008.
  • the salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
  • the compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts.
  • These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases.
  • the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases.
  • Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulphuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pe
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine and the like.
  • solvate means a compound as described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) where molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered.
  • solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof.
  • Suitable solvents are ethanol, water (for example, mono-, di-, and tri-hydrates), N-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), N,N′-dimethylformamide (DMF), N,N′-dimethylacetamide (DMAC), 1,3-dimethyl-2-imidazolidinone (DMEU), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like.
  • NMP N-methylpyrrolidinone
  • DMSO dimethyl sulfoxide
  • DMF N,N′-dimethylformamide
  • DMAC N,N′-dimethylacetamide
  • DMEU 1,3-dimethyl-2-imidazolidinone
  • DMPU
  • prevent refers to prophylactic treatment or treatment that prevents one or more symptoms or conditions of a disease, disorder, or conditions described herein (for example, pain (e.g., chronic or acute pain), epilepsy, Alzheimer's disease, Parkinson's disease, cardiovascular disease, diabetes, cancer, sleep disorders, obesity, psychosis such as schizophrenia, overactive bladder, renal disease, neuroprotection, addiction, and male birth control).
  • Preventative treatment can be initiated, for example, prior to (“pre-exposure prophylaxis”) or following (“post-exposure prophylaxis”) an event that precedes the onset of the disease, disorder, or conditions.
  • Preventive treatment that includes administration of a compound described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1), or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof, can be acute, short-term, or chronic.
  • the doses administered may be varied during the course of preventative treatment.
  • prodrug represents compounds that are rapidly transformed in vivo to the parent compound of the above formula, for example, by hydrolysis in blood.
  • Prodrugs of the compounds described herein may be conventional esters. Some common esters that have been utilized as prodrugs are phenyl esters, aliphatic (C1-C8 or C8-C24) esters, cholesterol esters, acyloxymethyl esters, carbamates, and amino acid esters. For example, a compound that contains an OH group may be acylated at this position in its prodrug form.
  • prodrugs of the compounds of the present invention are suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use.
  • the compounds of the invention may be coupled through conjugation to substances designed to alter the pharmacokinetics, for targeting, or for other reasons.
  • the invention further includes conjugates of these compounds.
  • polyethylene glycol is often coupled to substances to enhance half-life; the compounds may be coupled to liposomes covalently or noncovalently or to other particulate carriers. They may also be coupled to targeting agents such as antibodies or peptidomimetics, often through linker moieties.
  • the invention is also directed to compounds (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) when modified so as to be included in a conjugate of this type.
  • a condition or “treatment” of the condition e.g., the conditions described herein such as pain (e.g., chronic or acute pain), epilepsy, Alzheimer's disease, Parkinson's disease, cardiovascular disease, diabetes, cancer, sleep disorders, obesity, psychosis such as schizophrenia, overactive bladder, renal disease, neuroprotection, addiction, and male birth control
  • pain e.g., chronic or acute pain
  • epilepsy e.g., epilepsy
  • Alzheimer's disease e.g., chronic or acute pain
  • Parkinson's disease e.g., cardiovascular disease
  • diabetes e.g., cancer
  • sleep disorders e.g., obesity
  • psychosis e.g., schizophrenia, overactive bladder, renal disease, neuroprotection, addiction, and male birth control
  • Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable.
  • “Palliating” a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.
  • unit dosage form refers to a physically discrete unit suitable as a unitary dosage for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with any suitable pharmaceutical excipient or excipients.
  • exemplary, non-limiting unit dosage forms include a tablet (e.g., a chewable tablet), caplet, capsule (e.g., a hard capsule or a soft capsule), lozenge, film, strip, gelcap, and syrup.
  • the compounds of the invention contain one or more chiral centers.
  • the invention includes each of the isolated stereoisomeric forms as well as mixtures of stereoisomers in varying degrees of chiral purity, including racemic mixtures. It also encompasses the various diastereomers and tautomers that can be formed.
  • Compounds useful in the invention may also be isotopically labeled compounds.
  • Useful isotopes include hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, (e.g., 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl).
  • Isotopically labeled compounds can be prepared by synthesizing a compound using a readily available isotopically labeled reagent in place of a non-isotopically labeled reagent.
  • the compound e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1
  • the compound has the natural abundance of each element present in the compound.
  • the compounds described herein are also useful for the manufacture of a medicament useful to treat conditions requiring modulation of voltage-gated ion channel activity (e.g., sodium channel activity), and, in particular, Na v 1.7 or Na v 1.8 channel activity.
  • voltage-gated ion channel activity e.g., sodium channel activity
  • FIGS. 1A-1B show the modulation of ion channel activity by the compounds described herein.
  • FIGS. 2A-2C and 3 A- 3 C show data obtained in the spinal nerve ligation (SNL) assay for select compounds of the invention.
  • the invention features compounds that can modulate the activity of voltage-gated ion channels (e.g., voltage-gated sodium channels). These compounds can also be used to treat disorders such as pain, epilepsy, Parkinson's disease, mood disorders, psychosis (e.g., schizophrenia), tinnitus, amyotropic lateral sclerosis, glaucoma, ischaemia, spasticity disorders, obsessive compulsive disorder, restless leg syndrome, and Tourette syndrome.
  • Exemplary compounds described herein include compounds that have a structure according to the following formula,
  • each of R 1 , R 2 , R 3 , and R 4 is selected, independently, from H, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 haloalkyl, optionally substituted C6-C10 aryl, or optionally substituted 5 to 6-membered heteroaryl, where at least one of R 1 , R 2 , R 3 , and R 4 is halogen or optionally substituted C1-C6 haloalkyl;
  • R 5 is H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C10 heteroalkyl;
  • R 6 is —R 6A or —CH 2 R 6B ;
  • R 6A is NH 2 , optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, optionally substituted pyrimidine, optionally substituted C1-C10 amino alkyl, optionally substituted C1-C10 hydroxyalkyl, optionally substituted C1-C10 alkoxyalkyl, optionally substituted C1-C10 haloalkyl, or optionally substituted C1-C10 alkylsulfonyl; or R 6A has a structure according to
  • n is an integer between 0-4;
  • Z 1 is CH 2 , NH, NCH 3 , or O;
  • L 1 is —CH 2 , —CHR 4A , —CH 2 C( ⁇ O), —C( ⁇ O)CH 2 , —CH 2 C( ⁇ O)NH, —CH 2 C( ⁇ O)NHCH 2 , or —CH 2 NHC( ⁇ O)CH 2 ;
  • each R 2A and R 2C when present, is selected from OH, N(R 2B ) 2 , halogen, and unsubstituted C1-C3 alkyl, or two R 2A combine to form an oxo ( ⁇ O) group, and wherein no more than two R 2A combine to form an oxo group; and
  • each R 2B is, independently, H or unsubstituted C1-C6 alkyl
  • R 2D is H, OH, or NH 2 ;
  • R 6B is optionally substituted cyclopropyl, optionally substituted azetidine, optionally substituted cyclopentyl, optionally substituted pyrazole, optionally substituted pyrrole, optionally substituted pyrrolidine, optionally substituted thiazolidine, optionally substituted thiazolidine-1,1-dioxide, or optionally substituted pyrimidine.
  • R 5 is H.
  • R 5 is optionally substituted C1-C10 heteroalkyl.
  • R 2 and R 4 are both CF 3 , F, or Cl.
  • R 2 and R 3 are both CF 3 , F, or Cl.
  • R 6 is —CH 2 R 6B , and R 6B is optionally substituted azetidine.
  • R 6 is optionally substituted C1-C10 aminoalkyl.
  • the C1-C10 aminoalkyl includes an oxo ( ⁇ O) substituent, an alkoxy substituent, an N-sulfonyl group, or any combination thereof.
  • the compounds described herein are useful in the methods of the invention and, while not bound by theory, are believed to exert their desirable effects through their ability to modulate the activity of voltage-gated ion channels, e.g., the activity of sodium channels such as the Na v 1.7 and Na v 1.8 channels.
  • the compounds described herein can also be used for the treatment of certain conditions such as pain, epilepsy, migraine, Parkinson's disease, mood disorders, schizophrenia, psychosis, tinnitus, amyotropic lateral sclerosis, glaucoma, ischaemia, spasticity disorders, obsessive compulsive disorder, restless leg syndrome, and Tourette syndrome.
  • Na v 1 ⁇ -subunit isoforms Na v 1.1-1.9 (see, e.g., Yu et al., Genome Biolog, 4:207, 2003).
  • other conditions associated with voltage-dependent sodium channel activity include seizures (e.g., Na v 1.1), epilepsy (e.g., Na v 1.2), neurodegeneration (e.g., Na v 1.1, Na v 1.2), myotonia (e.g., Na v 1.4), arrhythmia (e.g., Na v 1.5), and movement disorders (e.g., Na v 1.6) as described in PCT Publication No. WO 2008/118758, herein incorporated by reference.
  • seizures e.g., Na v 1.1
  • epilepsy e.g., Na v 1.2
  • neurodegeneration e.g., Na v 1.1, Na v 1.2
  • myotonia e.g., Na v 1.4
  • arrhythmia
  • the expression of particular isoforms in particular tissues can influence the therapeutic effects of sodium channel modulators.
  • the Na v 1.4 and Na v 1.5 isoforms are largely found in skeletal and cardiac myocytes (see, e.g., Gold, Exp Neurol. 210(1): 1-6, 2008).
  • Voltage-dependent ion channels in pain-sensing neurons are currently of great interest in developing drugs to treat pain.
  • blocking voltage-dependent sodium channels in pain-sensing neurons can block pain signals by interrupting initiation and transmission of the action potential.
  • Na v 1.3 isoform has also been implicated in pain, e.g., pain associated with tissue injury (Gold, Exp Neurol. 210(1): 1-6, 2008).
  • the Na v 1.7 and Na v 1.8 channel subtypes act as major contributors to both inflammatory and neuropathic pain (vide infra).
  • mutations have been identified in the Na v 1.7 channel that lead either to a gain of channel function (Dib-Hajj et al., Brain 128:1847-1854, 2005) or more commonly to a loss of channel function (Chatelier et al., J. Neurophisiol. 99:2241-50, 2008). These mutations underlie human heritable disorders such as erythermalgia (Yang et al., J Med. Genet. 41(3) 171-4, 2004), paroxysmal extreme pain disorder (Fertleman et al., Neuron.
  • mice inflammatory and acute mechanosensory pain is reduced when Na v 1.7 is knocked out in Na v 1.8-positive neurons (Nassar et al., Proc Natl Acad Sci USA. 101(34):12706-11, 2004).
  • siRNA of Na v 1.7 attenuates inflammatory hyperalgesia (Yeomans et al., Hum Gene Ther. 16(2) 271-7, 2005).
  • the Na v 1.8 isoform is selectively expressed in sensory neurons and has been identified as a target for thre treatment of pain, e.g., chronic pain (e.g., Swanwick et al., Neurosci. Lett. 486:78-83, 2010).
  • the role of Na v 1.8 in inflammatory has also emerged using molecular techniques to knockdown Na v 1.8, which has been shown to reduce the maintenance of these different pain states.
  • Lacosamide is a functionalized amino acid that has shown effectiveness as an analgesic in several animal models of neuropathic pain and is currently in late stages of clinical development for epilepsy and diabetic neuropathic pain.
  • One mode of action that has been validated for lacosamide is inhibition of voltage-gated sodium channel activity by selective inhibition with the slow-inactivated conformation of the channel (Sheets et al., Journal of Pharmacology and Experimental Therapeutics, 326(1) 89-99 (2008)).
  • Modulators of sodium channels including clinically relevant compounds, can exhibit a pronounced state-dependent binding, where sodium channels that are rapidly and repeatedly activated and inactivated are more readily blocked.
  • voltage-gated sodium channels have four distinct states: open, closed, fast-inactivated and slow-inactivated.
  • Classic sodium channel modulators such as lidocaine, are believed to exhibit the highest affinity for the fast-inactivated state.
  • alteration of the slow inactivated state is also clinically relevant.
  • calcium channels directly affect membrane potential and contribute to electrical properties such as excitability, repetitive firing patterns and pacemaker activity. Calcium entry further affects neuronal functions by directly regulating calcium-dependent ion channels and modulating the activity of calcium-dependent enzymes such as protein kinase C and calmodulin-dependent protein kinase II.
  • An increase in calcium concentration at the presynaptic nerve terminal triggers the release of neurotransmitter, which also affects neurite outgrowth and growth cone migration in developing neurons.
  • Calcium channels mediate a variety of normal physiological functions, and are also implicated in a number of human disorders as described herein. For example, calcium channels also have been shown to mediate the development and maintenance of the neuronal sensitization and hyperexcitability processes associated with neuropathic pain, and provide attractive targets for the development of analgesic drugs (reviewed in Vanegas et al., Pain 85: 9-18 (2000)). Native calcium channels have been classified by their electrophysiological and pharmacological properties into T-, L-, N-, P/Q- and R-types (reviewed in Catterall, Annu Rev Cell Dev Biol 16: 521-555, 2000; Huguenard, Annu Rev Physiol 58: 329-348, 1996).
  • T-type channels activate at more positive potentials (high voltage-activated) and display diverse kinetics and voltage-dependent properties (Id.).
  • T-type channels can be distinguished by having a more negative range of activation and inactivation, rapid inactivation, slow deactivation, and smaller single-channel conductances.
  • ⁇ 1G , ⁇ 1H , and ⁇ 1I alternatively called Cav 3.1, Cav 3.2 and Cav 3.3 respectively.
  • T-type calcium channels are involved in various medical conditions. In mice lacking the gene expressing the 3.1 subunit, resistance to absence seizures was observed (Kim et al., Mol. Cell. Neurosci. 18(2): 235-245 (2001)). Other studies have also implicated the 3.2 subunit in the development of epilepsy (Su et al., J. Neurosci. 22: 3645-3655 (2002)). There is also evidence that some existing anticonvulsant drugs, such as ethosuximide, function through the blockade of T-type channels (Gomora et al., Mol. Pharmacol. 60: 1121-1132 (2001)).
  • T-type calcium channels are abnormally expressed in cancerous cells and that blockade of these channels may reduce cell proliferation in addition to inducing apoptosis.
  • Recent studies also show that the expression of T-type calcium channels in breast cancer cells is proliferation state dependent, i.e. the channels are expressed at higher levels during the fast-replication period, and once the cells are in a non-proliferation state, expression of this channel is minimal. Therefore, selectively blocking calcium channel entry into cancerous cells may be a valuable approach for preventing tumor growth (e.g., PCT Patent Application Nos.
  • T-type calcium channels may also be involved in still other conditions.
  • a recent study also has shown that T-type calcium channel antagonists inhibit high-fat diet-induced weight gain in mice.
  • administration of a selective T-type channel antagonist reduced body weight and fat mass while concurrently increasing lean muscle mass (e.g., Uebele et al., The Journal of Clinical Investigation, 119(6):1659-1667 (2009)).
  • T-type calcium channels may also be involved in pain (see for example: US Patent Publication No. 2003/0086980; PCT Publication Nos. WO 03/007953 and WO 04/000311).
  • epilepsy see also US Patent Application No.
  • T-type calcium channels have been implicated in diabetes (US Patent Publication No. 2003/0125269), sleep disorders (US Patent Publication No. 2006/0003985), Parkinson's disease and psychosis such as schizophrenia (US Patent Publication No. 2003/0087799); overactive bladder (Sui et al., British Journal of Urology International 99(2): 436-441 (2007); US Patent Publication No. 2004/0197825), renal disease (Hayashi et al., Journal of Pharmacological Sciences 99: 221-227 (2005)), anxiety and alcoholism (US Patent Publication No. 2009/0126031), neuroprotection, and male birth control.
  • modulation of ion channels by the compounds described herein can be measured according to methods known in the art (e.g., in the references provided herein).
  • Modulators of ion channels e.g., voltage gated sodium and calcium ion channels, and the medicinal chemistry or methods by which such compounds can be identified, are also described in, for example: Birch et al., Drug Discovery Today, 9(9):410-418 (2004); Audesirk, “Chapter 6-Electrophysiological Analysis of Ion Channel Function,” Neurotoxicology: Approaches and Methods, 137-156 (1995); Camerino et al., “Chapter 4: Therapeutic Approaches to Ion Channel Diseases,” Advances in Genetics, 64:81-145 (2008); Petkov, “Chapter 16-Ion Channels,” Pharmacology: Principles and Practice, 387-427 (2009); Standen et al., “Chapter 15-Patch Clamping Methods and Analysis of Ion Channels,” Principles of Medical Biology , Vol.
  • Exemplary conditions that can be treated using the compounds described herein include pain (e.g., chronic or acute pain), epilepsy, Alzheimer's disease, Parkinson's disease, diabetes; cancer; sleep disorders; obesity; mood disorders, psychosis such as schizophrenia; overactive bladder; renal disease, neuroprotection, and addiction.
  • the conidition can be pain (e.g., neuropathic pain or post-surgery pain), epilepsy, migraine, Parkinson's disease, mood disorders, schizophrenia, psychosis, tinnitus, amyotropic lateral sclerosis, glaucoma, ischaemia, spasticity disorders, obsessive compulsive disorder, restless leg syndrome, and Tourette syndrome.
  • Epilepsy as used herein includes but is not limited to partial seizures such as temporal lobe epilepsy, absence seizures, generalized seizures, and tonic/clonic seizures.
  • Cancer as used herein includes but is not limited to breast carcinoma, neuroblastoma, retinoblastoma, glioma, prostate carcinoma, esophageal carcinoma, fibrosarcoma, colorectal carcinoma, pheochromocytoma, adrenocarcinoma, insulinoma, lung carcinoma, melanoma, and ovarian cancer.
  • Acute pain as used herein includes but is not limited to nociceptive pain and post-operative pain.
  • Chronic pain includes but is not limited by: peripheral neuropathic pain (e.g., post-herpetic neuralgia, diabetic neuropathic pain, neuropathic cancer pain, HIV-associated neuropathy, erythromelalgia, failed back-surgery syndrome, trigeminal neuralgia, or phantom limb pain); central neuropathic pain (e.g., multiple sclerosis related pain, Parkinson disease related pain, post-stroke pain, post-traumatic spinal cord injury pain, lumbosacral radiculopathy, cervical radiculopathy, brachial radiculopathy, or pain in dementia); musculoskeletal pain such as osteoarthritic pain and fibromyalgia syndrome; inflammatory pain (e.g., inflammatory pain caused by rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis,
  • joint mobility can also improve as the underlying chronic pain is reduced.
  • use of compounds of the present invention to treat osteoarthritic pain inherently includes use of such compounds to improve joint mobility in patients suffering from osteoarthritis.
  • the compounds described herein can be tested for efficacy in any standard animal model of pain.
  • Various models test the sensitivity of normal animals to intense or noxious stimuli (physiological or nociceptive pain). These tests include responses to thermal, mechanical, or chemical stimuli.
  • Thermal stimuli usually involve the application of hot stimuli (typically varying between 42-55° C.) including, for example: radiant heat to the tail (the tail flick test), radiant heat to the plantar surface of the hindpaw (the Hargreaves test), the hotplate test, and immersion of the hindpaw or tail into hot water. Immersion in cold water, acetone evaporation, or cold plate tests may also be used to test cold pain responsiveness.
  • Tests involving mechanical stimuli typically measure the threshold for eliciting a withdrawal reflex of the hindpaw to graded strength monofilament von Frey hairs or to a sustained pressure stimulus to a paw (e.g., the Ugo Basile analgesiometer). The duration of a response to a standard pinprick may also be measured.
  • a chemical stimulus the response to the application or injection of a chemical irritant (e.g., capsaicin, mustard oil, bradykinin, ATP, formalin, acetic acid) to the skin, muscle joints or internal organs (e.g., bladder or peritoneum) is measured.
  • a chemical irritant e.g., capsaicin, mustard oil, bradykinin, ATP, formalin, acetic acid
  • peripheral sensitization i.e., changes in the threshold and responsiveness of high threshold nociceptors
  • sensitizing chemicals e.g., prostaglandins, bradykinin, histamine, serotonin, capsaicin, or mustard oil.
  • Central sensitization i.e., changes in the excitability of neurons in the central nervous system induced by activity in peripheral pain fibers
  • noxious stimuli e.g., heat
  • chemical stimuli e.g., injection or application of chemical irritants
  • electrical activation of sensory fibers e.g., electrical activation of sensory fibers.
  • SNL tests which involves the ligation of a spinal segmental nerve (Kim and Chung Pain (1992) 50: 355), the Seltzer model involving partial nerve injury (Seltzer, Pain (1990) 43: 205-18), the spared nerve injury (SNI) model (Decosterd and Woolf, Pain (2000) 87:149), chronic constriction injury (CCI) model (Bennett (1993) Muscle Nerve 16: 1040), tests involving toxic neuropathies such as diabetes (streptozocin model), pyridoxine neuropathy, taxol, vincristine, and other antineoplastic agent-induced neuropathies, tests involving ischaemia to a nerve, peripheral neuritis models (e.g., CFA applied peri-neurally), models of post-herpetic neuralgia using HSV infection, and compression models.
  • peripheral neuritis models e.g., CFA applied peri-neurally
  • models of post-herpetic neuralgia using HSV infection and compression
  • outcome measures may be assessed, for example, according to behavior, electrophysiology, neurochemistry, or imaging techniques to detect changes in neural activity.
  • hERG KCNH2 or K v 11.1 K + Channels: Screening for Cardiac Arrhythmia Risk,” Curr. Drug Metab. 9(9):965-70 (2008).
  • hERG K + channel is not inhibited or only minimally inhibited as compared to the inhibition of the primary channel targeted.
  • cytochrome p450 an enzyme that is required for drug detoxification.
  • Such compounds may be particularly useful in the methods described herein.
  • the compounds of the invention can be formulated as pharmaceutical or veterinary compositions.
  • a summary of such techniques is found in Remington: The Science and Practice of Pharmacy, 21 st Edition , Lippincott Williams & Wilkins, (2005); and Encyclopedia of Pharmaceutical Technology , eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, each of which is incorporated herein by reference.
  • the compounds described herein may be present in amounts totaling 1-95% by weight of the total weight of the composition.
  • the composition may be provided in a dosage form that is suitable for intraarticular, oral, parenteral (e.g., intravenous, intramuscular), rectal, cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intravesicular, intraurethral, intrathecal, epidural, aural, or ocular administration, or by injection, inhalation, or direct contact with the nasal, genitourinary, gastrointesitnal, reproductive or oral mucosa.
  • parenteral e.g., intravenous, intramuscular
  • rectal cutaneous, subcutaneous, topical, transdermal, sublingual, nasal, vaginal, intravesicular, intraurethral, intrathecal, epidural, aural, or ocular administration, or by injection, inhalation, or direct contact with the nasal, genitourinary, gastrointesitnal
  • the pharmaceutical composition may be in the form of, e.g., tablets, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols.
  • the compositions may be formulated according to conventional pharmaceutical practice.
  • the compounds described herein may be used alone, as mixtures of two or more compounds or in combination with other pharmaceuticals.
  • An example of other pharmaceuticals to combine with the compounds described herein e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) would include pharmaceuticals for the treatment of the same indication.
  • a compound in the treatment of pain, a compound may be combined with another pain relief treatment such as an NSAID, or a compound which selectively inhibits COX-2, or an opioid, or an adjuvant analgesic such as an antidepressant.
  • Another example of a potential pharmaceutical to combine with the compounds described herein e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1
  • the compounds will be formulated into suitable compositions to permit facile delivery.
  • Each compound of a combination therapy may be formulated in a variety of ways that are known in the art.
  • the first and second agents of the combination therapy may be formulated together or separately. Desirably, the first and second agents are formulated together for the simultaneous or near simultaneous administration of the agents.
  • the compounds of the invention may be prepared and used as pharmaceutical compositions comprising an effective amount of a compound described herein (e.g., a compound according to any of Formulas (I)-(XIII) or any of Compounds (1)-(236) of Table 1) and a pharmaceutically acceptable carrier or excipient, as is well known in the art.
  • a pharmaceutically acceptable carrier or excipient as is well known in the art.
  • the composition includes at least two different pharmaceutically acceptable excipients or carriers.
  • Formulations may be prepared in a manner suitable for systemic administration or topical or local administration.
  • Systemic formulations include those designed for injection (e.g., intramuscular, intravenous or subcutaneous injection) or may be prepared for transdermal, transmucosal, or oral administration.
  • the formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, preservatives and the like.
  • the compounds can be administered also in liposomal compositions or as microemulsions.
  • formulations can be prepared in conventional forms as liquid solutions or suspensions or as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions.
  • Suitable excipients include, for example, water, saline, dextrose, glycerol and the like.
  • Such compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, such as, for example, sodium acetate, sorbitan monolaurate, and so forth.
  • Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermal patches, transmucosal delivery and intranasal administration.
  • Oral administration is also suitable for compounds of the invention. Suitable forms include syrups, capsules, and tablets, as is understood in the art.
  • the dosage of the compounds of the invention may be, for example, 0.01-50 mg/kg (e.g., 0.01-15 mg/kg or 0.1-10 mg/kg).
  • the dosage can be 10-30 mg/kg.
  • Each compound of a combination therapy may be formulated in a variety of ways that are known in the art.
  • the first and second agents of the combination therapy may be formulated together or separately.
  • kits that contain, e.g., two pills, a pill and a powder, a suppository and a liquid in a vial, two topical creams, etc.
  • the kit can include optional components that aid in the administration of the unit dose to patients, such as vials for reconstituting powder forms, syringes for injection, customized IV delivery systems, inhalers, etc.
  • the unit dose kit can contain instructions for preparation and administration of the compositions.
  • the kit may be manufactured as a single use unit dose for one patient, multiple uses for a particular patient (at a constant dose or in which the individual compounds may vary in potency as therapy progresses); or the kit may contain multiple doses suitable for administration to multiple patients (“bulk packaging”).
  • the kit components may be assembled in cartons, blister packs, bottles, tubes, and the like.
  • Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients.
  • excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose
  • Two or more compounds may be mixed together in a tablet, capsule, or other vehicle, or may be partitioned.
  • the first compound is contained on the inside of the tablet, and the second compound is on the outside, such that a substantial portion of the second compound is released prior to the release of the first compound.
  • Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin, or olive oil.
  • Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.
  • Dissolution or diffusion controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound into an appropriate matrix.
  • a controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-hydroxymethacrylate, methacrylate hydrogels, 1,3 butylene glycol, ethylene glycol methacrylate, and/or polyethylene glycols.
  • the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.
  • liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • aqueous solutions suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • the oral dosage of any of the compounds of the combination of the invention will depend on the nature of the compound, and can readily be determined by one skilled in the art. Typically, such dosage is normally about 0.001 mg to 2000 mg per day, desirably about 1 mg to 1000 mg per day, and more desirably about 5 mg to 500 mg per day. Dosages up to 200 mg per day may be necessary.
  • Administration of each drug in a combination therapy can, independently, be one to four times daily for one day to one year, and may even be for the life of the patient. Chronic, long-term administration may be indicated.
  • reaction scheme and Examples are intended to illustrate the synthesis of a representative number of compounds. Accordingly, the Examples are intended to illustrate but not to limit the invention. Additional compounds not specifically exemplified may be synthesized using conventional methods in combination with the methods described herein.
  • tert-Butyl (2-((2-amino-3,5-bis(trifluoromethyl)phenyl)amino)-2-oxoethyl)carbamate (3) (1.4 g, mmol) was heated in THF/AcOH (95/5, 20 mL) using a microwave at 140° C. for 2.5 hours. The reaction was concentrated in vacuo, taken up in EtOAc, and washed with NaHCO 3 (saturated solution) to neutralize. The organic layer was separated, dried (Na 2 SO 4 ), and concentrated in vacuo.
  • tert-Butyl ((4,6-bis(trifluoromethyl)-1H-benzo[d]imidazol-2-yl)methyl)carbamate (4) (730 mg, 1.9 mmol) was taken up in EtOAc, and the solution was flushed with HCl (g) for 5 minutes. The resultant suspension was stirred at room temperature for 25 minutes then concentrated in vacuo.
  • the aqueous layer was subsequently separated, acidified with hydrochloric acid (2 M, 12 mL), extracted with ethyl acetate (3 ⁇ 30 mL), and dried over anhydrous sodium sulfate.
  • the resultant oil was taken up in an ethyl acetate:methanol mixture (1:1) and filtered. The filtrate was then concentrated in vacuo, and the product was purified by automated flash chromatography to afford the title compound (32) (confirmed by LCMS (positive ion mode)).
  • the mixture of isomers was taken up in THF/AcOH (95/5) and heated using a microwave at 140° C. for 2 hours. The reaction was concentrated in vacuo, taken up in EtOAc, and then washed with water (30 mL), saturated aqueous sodium bicarbonate (30 mL), and brine (30 mL). The organic layer was dried over anhydrous sodium sulfate then concentrated in vacuo.
  • the residue was purified by automated column chromatography (smooth gradient 20 ⁇ 70% ethyl acetate:petroleum ether), and this initial purification was followed by a second round of automated column chromatography (smooth gradient 0 ⁇ 50% ethyl acetate:dichloromethane) to afford the N-Boc-protected product (38) as clear colorless gum.
  • the product was taken up in HCl saturated ethyl acetate and stirred for two hours at room temperature. The clear mixture turned milky over time and was condensed in vacuo to give the HCl salt of the product (39) as a white solid (1.00 g, 51% over two steps).
  • tert-Butyl 3-oxopiperazine-1-carboxylate (4.0 g, 20 mmol) was stirred in dry DMF at room temperature under Ar. NaH (60% dispersion in mineral oil) (960 mg, 24 mmol) was added, and the reaction stirred for 30 minutes. Ethyl bromopropionate (56) (2.55 mL, 20 mmol) was added in one portion, and stirring continued for 14 hours. The reaction was partitioned between EtOAc and H 2 O.
  • tert-Butyl 4-(3-((2-amino-3,5-bis(trifluoromethyl)phenyl)amino)-3-oxopropyl)-3-oxopiperazine-1-carboxylate (59) (500 mg, 1.0 mmol) was heated in THF/AcOH (95:5, 2 mL) using a microwave at 140° C. for 2.5 hours. The residue was concentrated in vacuo to give crude tert-butyl 4-(2-(4,6-bis(trifluoromethyl)-1H-benzo[d]imidazol-2-yl)ethyl)-3-oxopiperazine-1-carboxylate (60).
  • tert-Butyl piperazine-1-carboxylate (62) (1 g, 5.4 mmol) and TEA (837 ⁇ L, 6.0 mmol) were stirred in DCM at room temperature.
  • Ethyl malonyl chloride (63) (810 ⁇ L, 5.4 mmol) was added in one portion, and the reaction stirred at room temperature for 1 hour.
  • the reaction was diluted with DCM and washed sequentially with NH 4 Cl (saturated solution) and NaHCO 3 (saturated solution). The organics were separated, dried (Na 2 SO 4 ), and concentrated in vacuo.
  • tert-Butyl 4-(3-((2-amino-3,5-bis(trifluoromethyl)phenyl)amino)-3-oxopropanoyl)piperazine-1-carboxylate (66) (650 mg, 1.31 mmol) was heated in THF/AcOH (95:5, 2 mL) using a microwave at 140° C. for 2.5 hours. The residue was concentrated in vacuo to give crude tert-butyl 4-(2-(4,6-bis(trifluoromethyl)-1H-benzo[d]imidazol-2-yl)acetyl)piperazine-1-carboxylate (67).
  • HEK 293F cell line stably expressing human Na v 1.7 was achieved by co-transfecting human SCN9A and human SCN1B cDNAs, subcloned into plasmid vectors and utilizing standard transfection techniques. Clones were selected using appropriate selection agents (0.3 mg/mL Zeocin and 0.8 mg/mL Geneticin) and maintained in Dulbecco's Modified Eagle medium, 10% fetal bovine serum, 1% non essential amino acids to ⁇ 80% confluence at 37° C. in a humidified incubator with 95% atmosphere and 5% CO 2 .
  • appropriate selection agents 0.3 mg/mL Zeocin and 0.8 mg/mL Geneticin
  • the internal recording solution contained (in mM): 129 CsF, 2 MgCl 2 , 11 EGTA, 10 HEPES, 6 NaCl, 3 Na 2 ATP adjusted to pH 7.2 with CsOH and 280 mOsm with sucrose.
  • the automated liquid handling facility of PatchXpress dispensed cells and added compound.
  • Modulation of Na v 1.7 channels by compounds was assessed by promoting the channels into the inactivated state using a conditioning voltage pulse of variable amplitude, followed by a brief hyperpolarizing pulse with a subsequent depolarized voltage step to measure the current amplitude in the presence and absence of compound. Exemplary data are provided in FIG. 1 .
  • the compounds described herein can also be assayed for modulation of other voltage gated channels (e.g., other Na + channel isoforms or Ca 2+ channels such as Ca v 3.2 T-type channels). Additional methods are known in the art. Exemplary data obtained according to these methods are also shown in FIG. 1 .
  • other voltage gated channels e.g., other Na + channel isoforms or Ca 2+ channels such as Ca v 3.2 T-type channels.
  • the compounds described herein can also be studied as modulators of voltage-gated Ca 2+ channels (e.g., Ca v 1.2, Ca v 2.2, Ca v 3.1, or Ca v 3.2 channels). Exemplary methods are described herein.
  • the culture media can be replaced with extracellular solution (ECS) containing (in mM): 142 CsCl, 10 D-glucose, 2 CaCl 2 , 1 MgCl 2 , 10 HEPES, pH adjusted to 7.4 with CsOH.
  • ECS extracellular solution
  • Borosilicate glass patch pipettes pulled on a P-97 micropipette puller (Sutter Instruments, Novato, Calif.) with typical resistances of 2-4 MW, can be backfilled with intracellular solution containing (in mM): 126.5 Cs-methanesulphonate, 2 MgCl 2 , 10 HEPES, 11 EGTA, 2 Na-ATP, pH adjusted to 7.3 CsOH. Voltages were recorded in the whole-cell configuration at room temperature ( ⁇ 21° C.) using an Axopatch 200B (Molecular Devices, Sunnyvale, Calif.) patch-clamp amplifier.
  • Axopatch 200B Molecular Devices, Sunnyvale, Calif.
  • Recordings can be low-pass filtered at 1 kHz ( ⁇ 3 dB 4-pole Bessel filter), digitized at 2 kHz with a Digidata 1322A interface (Molecular Devices), and acquired using pClamp 9.2 (Molecular Devices), with no leak subtraction being used.
  • Test compounds prepared as 30 mM stock solutions in DMSO and diluted in extracellular buffer, can be applied through a gravity driven multi-barrelled array of capillaries (24 gauge) connected to reservoirs controlled by solenoid valves. The effects of compounds on Ca v 3.2 slow and fast inactivation can then be evaluated using different voltage protocols. The voltage dependence of fast and slow channel inactivation can be examined using a two pulse protocol. Data were analyzed and fitted using OriginPro v.7.5 (OriginLab, Northampton, Mass.) software.
  • Cells were plated in 384-well, clear-bottom, black-walled, poly-D-lysine coated plates (Becton Dickinson, Franklin Lake, N.J.) 2 days prior to use in the FLIPR assay.
  • 100 ⁇ L of cells (1.4 ⁇ 10 6 cell/mL) containing doxycyline (Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression) were added to each well using a Multidrop (Thermo Scientific, Waltham, Mass.) and were maintained in 5% CO 2 incubator at 37° C. On the morning of the assay, cells were transferred to a 5% CO 2 incubator at 29° C.
  • Cells can then be washed with a wash buffer containing (in mM): 118 NaCl, 18.4 HEPES, 11.7 D-glucose, 2 CaCl 2 , 0.5 MgSO 4 , 4.7 KCl, 1.2 KH 2 PO 4 , pH adjusted to 7.2 with NaOH.
  • a wash buffer containing (in mM): 118 NaCl, 18.4 HEPES, 11.7 D-glucose, 2 CaCl 2 , 0.5 MgSO 4 , 4.7 KCl, 1.2 KH 2 PO 4 , pH adjusted to 7.2 with NaOH.
  • Cells were then rinsed with either a 2 mM KCl closed-state buffer (in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH) when performing the closed-state assay or 12.5 mM KCl inactivated-state buffer (in mM: 128 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 12.5 mM KCl, with the pH adjusted to 7.4 with NaOH) when performing the inactivated-state assay.
  • a 2 mM KCl closed-state buffer in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH
  • 12.5 mM KCl inactivated-state buffer in mM: 128 NaCl, 10 HEPES, 10 D-
  • Concentration-dependent response curves were generated from 5 mM stock solutions prepared in DMSO (Sigma-Aldrich) and diluted in either the 2 mM KCl buffer or 12.5 mM KCl buffer and incubated for 20 minutes at 29° C. in 5% CO 2 .
  • Calcium entry was evoked with an addition of 130 mM KCl stimulation buffer (in mM: 10.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 130 KCl, with the pH adjusted to 7.4 with NaOH) for both the closed-state or inactivated-state assay.
  • Fluo-4 fluorescence signal was assessed using FLIPR TETRA TM instrument (Molecular Devices, Sunnyvale, Calif.) for 3 minutes following the elevation of extracellular KCl using an illumination wavelength of 470-495 nm with emissions recorded at 515-575 nm.
  • Concentration-dependent response curves were obtained by comparing the fluorescence signal in the presence of compound and fitted with a logistic function (1) to obtain the concentration that inhibited 50% (IC 50 ) of the RLU signal using OriginPro v.7.5 software (OriginLab. Northampton, Mass.).
  • Cells were plated in 384-well, clear-bottom, black-walled, poly-D-lysine coated plates (Becton Dickinson, Franklin Lake, N.J.) 2 days prior to use in the FLIPR assay.
  • 100 ⁇ L of cells 2.0 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • 100 ⁇ L of cells 2.0 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • Concentration-dependent response curves were obtained by comparing the fluorescence signal in the presence of compound and fitted with a logistic function (1) to obtain the concentration that inhibited 50% (IC 50 ) of the RLU signal using OriginPro v.7.5 software (OriginLab, Northampton, Mass.).
  • Cells were plated in 384-well, clear-bottom, black-walled, poly-D-lysine coated plates (Becton Dickinson, Franklin Lake, N.J.) 2 days prior to use in the FLIPR assay.
  • 100 ⁇ L of cells 1.2 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • 100 ⁇ L of cells 1.2 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • Cells were then rinsed with either a 2 mM KCl closed-state buffer (in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH) when performing the closed-state assay or 7.6 mM KCl inactivated-state buffer (in mM: 130.9 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 7.6 mM KCl, with the pH adjusted to 7.4 with NaOH) when performing the inactivated-state assay.
  • a 2 mM KCl closed-state buffer in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH
  • 7.6 mM KCl inactivated-state buffer in mM: 130.9 NaCl, 10 HEPES, 10 D-
  • Concentration-dependent response curves were generated from 5 mM stock solutions prepared in DMSO (Sigma-Aldrich), diluted in either the 2 mM KCl buffer or 7.6 mM KCl buffer, and incubated for 20 minutes at 29° C. in 5% CO 2 .
  • Fluo-4 fluorescence signal was assessed using FLIPR instrument (Molecular Devices, Sunnyvale, Calif.) for 3 minutes following the elevation of extracellular KCl using an illumination wavelength of 470-495 nm with emissions recorded at 515-575 nm.
  • Concentration-dependent response curves were obtained by comparing the fluorescence signal in the presence of compound and fitted with a logistic function (1) to obtain the concentration that inhibited 50% (IC 50 ) of the RLU signal using OriginPro v.7.5 software (OriginLab, Northampton, Mass.).
  • Cells were plated in 384-well, clear-bottom, black-walled, poly-D-lysine coated plates (Becton Dickinson, Franklin Lake, N.J.) 2 days prior to use in the FLIPR assay.
  • 100 ⁇ L of cells 1.2 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • 100 ⁇ L of cells 1.2 ⁇ 10 6 cell/mL
  • doxycyline Sigma-Aldrich, 1.5 ⁇ g/mL; to induce channel expression
  • Cells were then rinsed with either a 2 mM KCl closed-state buffer (in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH) when performing the closed-state assay or 30 mM KCl inactivated-state buffer (in mM: 110.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 30 mM KCl, with the pH adjusted to 7.4 with NaOH) when performing the inactivated-state assay.
  • a 2 mM KCl closed-state buffer in mM: 138.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 2 KCl, with the pH adjusted to 7.4 with NaOH
  • 30 mM KCl inactivated-state buffer in mM: 110.5 NaCl, 10 HEPES, 10 D-glucos
  • Concentration-dependent response curves were generated from 5 mM stock solutions prepared in DMSO (Sigma-Aldrich), diluted in either the 2 mM KCl buffer or 30 mM KCl buffer, and incubated for 20 minutes at 29° C. in 5% CO 2 . Calcium entry was evoked with an addition of 130 mM KCl stimulation buffer (in mM: 10.5 NaCl, 10 HEPES, 10 D-glucose, 1 CaCl 2 , and 130 KCl, with the pH adjusted to 7.4 with NaOH).
  • Fluo-4 fluorescence signal was assessed using FLIPR TETRA TM instrument (Molecular Devices, Sunnyvale, Calif.) for 3 minutes following the elevation of extracellular KCl using an illumination wavelength of 470-495 nm with emissions recorded at 515-575 nm.
  • Concentration-dependent response curves were obtained by comparing the fluorescence signal in the presence of compound and fitted with a logistic function (1) to obtain the concentration that inhibited 50% (IC 50 ) of the RLU signal using OriginPro v.7.5 software (OriginLab, Northampton, Mass.).
  • the compound has very low activity with respect to the hERG K + channel, which is expressed in the heart: compounds that block this channel with high potency may cause reactions which are fatal. See, e.g., Bowlby et al., “hERG (KCNH2 or K v 11.1 K + Channels: Screening for Cardiac Arrhythmia Risk,” Curr. Drug Metab. 9(9):965-70 (2008)).
  • hERG KCNH2 or K v 11.1 K + Channels: Screening for Cardiac Arrhythmia Risk,” Curr. Drug Metab. 9(9):965-70 (2008).
  • the hERG K + channel is not inhibited or only minimally inhibited as compared to the inhibition of the primary channel targeted. Such compounds may be particularly useful in the methods described herein.
  • the Spinal Nerve Ligation is an animal model representing peripheral nerve injury generating a neuropathic pain syndrome. In this model experimental animals develop the clinical symptoms of tactile allodynia and hyperalgesia.
  • L5/L6 Spinal nerve ligation (SNL) injury was induced using the procedure of Kim and Chung (Kim et al., Pain 50:355-363 (1992)) in male Sprague-Dawley rats (Harlan; Indianapolis, Ind.) weighing 200 to 250 grams.
  • An exemplary protocol is provided below
  • the animals were anesthetized with isoflurane, the left L6 transverse process was removed, and the L5 and L6 spinal nerves were tightly ligated with 6-0 silk suture.
  • the wound was closed with internal sutures and external tissue adhesive. Rats that exhibit motor deficiency (such as paw-dragging) or failure to exhibit subsequent tactile allodynia can be excluded from further testing.
  • Sham control rats can undergo the same operation and handling as the experimental animals, but without SNL.
  • Baseline and post-treatment values for mechanical hyperalgesia were evaluated using a digital Randall-Selitto device (dRS; IITC Life Sciences, Woodland Hills, Calif.). Animals were allowed to acclimate to the testing room for a minimum of 30 minutes before testing. Animals were placed in a restraint sling that suspends the animal, leaving the hind limbs available for testing. Paw compression threshold was measured once at each time point for the ipsilateral and contralateral paws. The stimulus was applied to the plantar surface of the hind paw by a dome-shaped tip placed between the 3rd and 4th metatarsus, and pressure was applied gradually over approximately 10 seconds. Measurements are taken from the first observed nocifensive behavior of vocalization, struggle or withdrawal.
  • dRS digital Randall-Selitto device
  • a cut-off value of 300 g was used to prevent injury to the animal.
  • the mean and standard error of the mean (SEM) were determined for each paw for each treatment group.
  • Fourteen days after surgery, mechanical hyperalgesia was assessed and rats were assigned to treatment groups based on pre-treatment baseline values.
  • baseline behavioural testing data can be obtained prior to initiating drug delivery. At selected times after infusion of the Test or Control Article behavioural data can then be collected again.
  • FIGS. 2A-2C and 3 A- 3 C Exemplary data are shown in FIGS. 2A-2C and 3 A- 3 C for select compounds of the invention. Additional data are presented in Tables 2-4 below.
  • Table 2 shows Compound (I) and Compound (41) (30 mg/kg, p.o.) significantly decreased mechanical hyperalgesia at 2 and 4 hours after administration compared to vehicle treated animals.
  • Table 3 shows that administration of Compound (33) (30 mg/kg, p.o.) significantly decreased mechanical hyperalgesia at 1, 2 and 4 hours after administration compared to vehicle treated animals.
  • Exemplary data are also shown in FIG. 2C and FIG. 3C for Compound (110) and in Table 4 below.
  • Compound (110) is shown to significantly decrease mechanical hyperalgesia at two and four hours after administration compared to vehicle treated animals.
  • the assessment of tactile allodynia can consist of measuring the withdrawal threshold of the paw ipsilateral to the site of nerve injury in response to probing with a series of calibrated von Frey filaments (innocuous stimuli). Animals can be acclimated to the suspended wire-mesh cages for 30 min before testing. Each von Frey filament can be applied perpendicularly to the plantar surface of the ligated paw of rats for 5 sec. A positive response can be indicated by a sharp withdrawal of the paw. For rats, the first testing filament is 4.31. Measurements can be taken before and after administration of test articles. The paw withdrawal threshold can be determined by the non-parametric method of Dixon (Dixon, Ann. Rev. Pharmacol. Toxicol.
  • the protocol can be repeated until three changes in behaviour were determined (“up and down” method; Chaplan et al., J. Neurosci. Methods 53:55-63 (1994)).
  • the cut-off values for rats can be, for example, no less than 0.2 g and no higher than 15 g (5.18 filament); for mice no less than 0.03 g and no higher than 2.34 g (4.56 filament).
  • a significant drop of the paw withdrawal threshold compared to the pre-treatment baseline is considered tactile allodynia.
  • Rat SNL tactile allodynia can be tested for the compounds described herein at, e.g., 60 minutes comapred to baseline and post-SNL.
  • Hargreaves and colleagues can be employed to assess paw-withdrawal latency to a noxious thermal stimulus.
  • Rats may be allowed to acclimate within a Plexiglas enclosure on a clear glass plate for 30 minutes.
  • a radiant heat source e.g., halogen bulb coupled to an infrared filter
  • Paw-withdrawal latency can be determined by a photocell that halts both lamp and timer when the paw is withdrawn.
  • the latency to withdrawal of the paw from the radiant heat source can be determined prior to L5/L6 SNL, 7-14 days after L5/L6 SNL but before drug, as well as after drug administration.
  • a maximal cut-off of 33 seconds is typically employed to prevent tissue damage. Paw withdrawal latency can be thus determined to the nearest 0.1 second.
  • a significant drop of the paw withdrawal latency from the baseline indicates the status of thermal hyperalgesia.
  • Antinociception is indicated by a reversal of thermal hyperalgesia to the pre-treatment baseline or a significant (p ⁇ 0.05) increase in paw withdrawal latency above this baseline.
  • Data is converted to % anti hyperalgesia or % anti nociception by the formula: (100 ⁇ (test latency ⁇ baseline latency)/(cut-off ⁇ baseline latency) where cut-off is 21 seconds for determining anti hyperalgesia and 40 seconds for determining anti nociception.
  • Compounds can be evaluated for the protection against seizures induced by a 6 Hz, 0.2 ms rectangular pulse width of 3 s duration, at a stimulus intensity of 32 mA (CC97) applied to the cornea of male CF1 mice (20-30 g) according to procedures described by Barton et al, “Pharmacological Characterization of the 6 Hz Psychomotor Seizure Model of Partial Epilepsy,” Epilepsy Res. 47(3):217-27 (2001). Seizures are characterised by the expression of one or more of the following behaviours: stun, forelimb clonus, twitching of the vibrissae and Straub-tail immediately following electrical stimulation. Animals can be considered “protected” if, following pre-treatment with a compound, the 6 Hz stimulus failed to evoke a behavioural response as describe above.
  • the GAERS Genetic Absence Epilepsy Rats from France
  • the GAERS is noted for its long and frequently recurring absence seizure episodes.
  • Investigators have determined, using electrophysiological recordings from neurons within the thalamus, that the activity and expression of the low-voltage calcium channels is significantly increased in GAERS.
  • mice can be monitored for overt signs of impaired neurological or muscular function.
  • the rotarod procedure (Dunham et al., J. Am. Pharmacol. Assoc. 46:208-209 (1957)) is used to disclose minimal muscular or neurological impairment (MMI).
  • MMI minimal muscular or neurological impairment
  • animals may exhibit a circular or zigzag gait, abnormal body posture and spread of the legs, tremors, hyperactivity, lack of exploratory behavior, somnolence, stupor, catalepsy, loss of placing response and changes in muscle tone.
  • Male Wistar rats (P6 to P9 for voltage-clamp and P15 to P18 for current-clamp recordings) can be anaesthetized through intraperitoneal injection of Inactin (Sigma).
  • the spinal cord can then be rapidly dissected out and placed in an ice-cold solution protective sucrose solution containing (in mM): 50 sucrose, 92 NaCl, 15 D-Glucose, 26 NaHCO 3 , 5 KCl, 1.25 NaH 2 PO 4 , 0.5 CaCl 2 , 7 MgSO 4 ,1 kynurenic acid, and bubbled with 5% CO 2 /95% O 2 .
  • the meninges, dura, and dorsal and ventral roots can then removed from the lumbar region of the spinal cord under a dissecting microscope.
  • the “cleaned” lumbar region of the spinal cord may be glued to the vibratome stage and immediately immersed in ice cold, bubbled, sucrose solution.
  • 300 to 350 ⁇ m parasagittal slices can be cut to preserve the dendritic arbour of lamina I neurons, while 350 to 400 ⁇ m transverse slices can be prepared for voltage-clamped Na v channel recordings. Slices may be allowed to recover for 1 hour at 35° C.
  • Neurons may be visualized using IR-DIC optics (Zeiss Axioskop 2 FS plus, Gottingen, Germany), and neurons from lamina I and the outer layer of lamina II can be selected based on their location relative to the substantia gelatinosa layer. Neurons can be patch-clamped using borosilicate glass patch pipettes with resistances of 3 to 6 MW.
  • Preliminary exposure characteristics of the compounds can be evaluated using, e.g., an in vivo Rat Early Pharmacokinetic (EPK) study design to show bioavailability.
  • EPK in vivo Rat Early Pharmacokinetic
  • Male Sprague-Dawley rats can be dosed via oral (PO) gavage in a particular formulation.
  • Blood samples can then be collected from the animals at 6 timepoints out to 4 hours post-dose.
  • Pharmacokinetic analysis can then performed on the LC-MS/MS measured concentrations for each timepoint of each compound.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US14/002,943 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel Abandoned US20140187533A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/002,943 US20140187533A1 (en) 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161448923P 2011-03-03 2011-03-03
US201161448910P 2011-03-03 2011-03-03
US14/002,943 US20140187533A1 (en) 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel
PCT/CA2012/000193 WO2012116440A1 (en) 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2012/000193 A-371-Of-International WO2012116440A1 (en) 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/833,601 Division US9688615B2 (en) 2011-03-03 2015-08-24 Benzimidazole inhibitors of the sodium channel

Publications (1)

Publication Number Publication Date
US20140187533A1 true US20140187533A1 (en) 2014-07-03

Family

ID=46757322

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/002,943 Abandoned US20140187533A1 (en) 2011-03-03 2012-03-02 Benzimidazole inhibitors of the sodium channel
US14/833,601 Active US9688615B2 (en) 2011-03-03 2015-08-24 Benzimidazole inhibitors of the sodium channel
US15/602,089 Abandoned US20170260130A1 (en) 2011-03-03 2017-05-22 Benzimidazole Inhibitors of the Sodium Channel
US15/846,198 Active US10351514B2 (en) 2011-03-03 2017-12-18 Benzimidazole inhibitors of the sodium channel

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/833,601 Active US9688615B2 (en) 2011-03-03 2015-08-24 Benzimidazole inhibitors of the sodium channel
US15/602,089 Abandoned US20170260130A1 (en) 2011-03-03 2017-05-22 Benzimidazole Inhibitors of the Sodium Channel
US15/846,198 Active US10351514B2 (en) 2011-03-03 2017-12-18 Benzimidazole inhibitors of the sodium channel

Country Status (5)

Country Link
US (4) US20140187533A1 (de)
EP (2) EP2681200A4 (de)
CA (1) CA2828456C (de)
ES (1) ES2786298T3 (de)
WO (1) WO2012116440A1 (de)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9108953B2 (en) 2013-11-26 2015-08-18 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
US9255089B2 (en) 2013-03-28 2016-02-09 Gilead Sciences, Inc. Benzimidazolone derivatives as bromodomain inhibitors
US9458145B2 (en) 2013-05-09 2016-10-04 Gilead Sciences, Inc. Benzimidazole derivatives as bromodomain inhibitors
WO2018213426A1 (en) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels
WO2019014352A1 (en) 2017-07-11 2019-01-17 Vertex Pharmaceuticals Incorporated CARBOXAMIDES AS INHIBITORS OF SODIUM CHANNELS
US10208023B2 (en) 2013-03-01 2019-02-19 Mark G. DeGiacomo Heterocyclic inhibitors of the sodium channel
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
WO2021113627A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
WO2021231553A1 (en) * 2020-05-12 2021-11-18 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of cav3.2 activity and uses thereof
CN113692304A (zh) * 2019-02-06 2021-11-23 礼来公司 作为kcnq增强剂的1-((2-(2,2,2-三氟乙氧基)吡啶-4-基)甲基)脲衍生物
WO2022256708A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Solid dosage forms and dosing regimens comprising (2r,3s,4s,5r)-4-[[3-(3,4-difluoro-2-methoxy-phenyl)-4,5-dimethyl-5-(trifluoromethyl) tetrahydrofuran-2-carbonyl]amino]pyridine-2-carboxamide
WO2022256676A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran analogs as modulators of sodium channels
WO2022256842A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Hydroxy and (halo)alkoxy substituted tetrahydrofurans as modulators of sodium channels
WO2022256702A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran-2-carboxamides as modulators of sodium channels
WO2022256679A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamide analogs as modulators of sodium channels
WO2022256622A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
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
WO2023205463A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205778A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2024123815A1 (en) 2022-12-06 2024-06-13 Vertex Pharmaceuticals Incorporated Process for the synthesis of substituted tetrahydrofuran modulators of sodium channels

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2797896A1 (de) * 2011-12-28 2014-11-05 Allergan, Inc. Benzimidazolderivate als selektive blocker eines dauerhaften natriumstroms
EP2809655B1 (de) 2012-02-03 2015-08-12 Pfizer Inc Benziimidazol- und imidazopyridinderivate als natriumkanalmodulatoren
WO2014149207A2 (en) * 2013-03-15 2014-09-25 Dow Agrosciences Llc Benzimidazole-based insecticidal compositions and related methods
US9828348B2 (en) 2013-11-08 2017-11-28 Purdue Pharma L.P. Benzimidazole derivatives and use thereof
DK3080134T3 (en) 2013-12-13 2018-10-22 Vertex Pharma PRODRUGS OF PYRIDONAMIDS USED AS MODULATORS OF SODIUM CHANNELS
US9737545B2 (en) 2013-12-19 2017-08-22 Merck Sharp & Dohme Corp. HIV protease inhibitors
USRE49816E1 (en) 2014-01-10 2024-01-30 Cornell University Dipeptides as inhibitors of human immunoproteasomes
WO2015127549A1 (en) * 2014-02-27 2015-09-03 Hassan Pajouhesh Heterocyclic inhibitors of the sodium channel
JP6758282B2 (ja) 2014-08-18 2020-09-23 コーネル・ユニバーシティーCornell University ヒト免疫プロテアソーム阻害剤としてのジペプチド模倣薬
JP2018052817A (ja) * 2015-01-21 2018-04-05 大日本住友製薬株式会社 新規ベンズイミダゾール誘導体およびその医薬用途
ES2818529T3 (es) * 2015-09-11 2021-04-13 Sumitomo Dainippon Pharma Co Ltd Derivados de bencimidazol como inhibidores Nav 1.7 (subunidad alfa del canal de sodio, dependiente del voltaje, tipo IX (SCN9A)) para tratar el dolor, la disuria y la esclerosis múltiple
CN108473503A (zh) * 2015-09-30 2018-08-31 赛特温治疗公司 用于治疗疼痛的11,13-修饰的石房蛤毒素类化合物
CN108351169B (zh) 2015-10-15 2022-01-28 康奈尔大学 蛋白酶体抑制剂及其用途
GB201617630D0 (en) 2016-10-18 2016-11-30 Cellcentric Ltd Pharmaceutical compounds
CN110381941A (zh) * 2017-02-03 2019-10-25 代表亚利桑那大学的亚利桑那校董会 Crmp2的sumo相关修饰的小分子拮抗剂及其用途
KR20200012833A (ko) 2017-03-29 2020-02-05 사이트원 테라퓨틱스, 인코포레이티드 통증 치료용 11,13-개질된 색시톡신
CN110914276A (zh) 2017-03-29 2020-03-24 赛特温治疗公司 用于治疗疼痛的11,13-修饰的石房蛤毒素类化合物
US11203613B2 (en) 2017-10-11 2021-12-21 Cornell University Peptidomimetic proteasome inhibitors
JP7450534B2 (ja) 2017-10-19 2024-03-15 帝人ファーマ株式会社 ベンズイミダゾール誘導体及びその用途
CA3082375A1 (en) * 2017-11-13 2019-05-16 Ecolab Usa Inc. A novel one-pot homogeneous process for the large scale manufacture of 2-substituted benzimidazoles
US11529337B2 (en) 2018-02-12 2022-12-20 Vertex Pharmaceuticals Incorporated Method of treating pain
GB201806320D0 (en) 2018-04-18 2018-05-30 Cellcentric Ltd Process
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
WO2020115045A1 (en) * 2018-12-05 2020-06-11 Esteve Pharmaceuticals, S.A. Substituted 1h-benzo[d]imidazole derivatives having multimodal activity against pain and pain related conditions

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3655901A (en) * 1970-07-30 1972-04-11 Merck & Co Inc Method of inhibiting the formation of phenylethanalamine-n-methyl transferase with 2-aminobenzimidazoles
WO2007117399A2 (en) * 2006-03-31 2007-10-18 Janssen Pharmaceutica N.V. Benzoimidazol-2-yl pyrimidines and pyrazines as modulators of the histamine h4 receptor

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3271249A (en) * 1963-06-24 1966-09-06 S B Penick & Co Inc Dihalobenzimidazoles for poultry disease treatment
FR2104717A1 (en) * 1970-09-28 1972-04-21 Dynachim Sarl Organophosphorus pesticides stabilised with benzimidazole derivs - - for controlling insects and internal parasites of animals
DE3915756A1 (de) * 1989-05-13 1990-11-29 Bayer Ag Cyclopropanoylaminosaeureamid-derivate
EP0593557B1 (de) * 1991-07-05 1996-01-31 MERCK SHARP & DOHME LTD. Aromatische verbindungen, diese enthaltende pharmazeutische zusammensetzungen und ihre therapeutische anwendung
CA2110514A1 (en) * 1991-07-05 1993-01-21 Raymond Baker Aromatic compounds, pharmaceutical compositions containing them and their use in therapy
AU5447094A (en) * 1992-10-21 1994-05-09 Regents Of The University Of Michigan, The Polysubstituted benzimidazoles as antiviral agents
US5674889A (en) * 1993-02-22 1997-10-07 Merck, Sharp & Dohme, Ltd. Aromatic compounds, compositions containing them and their use in therapy
TW263498B (de) * 1993-11-10 1995-11-21 Takeda Pharm Industry Co Ltd
US5565568A (en) * 1995-04-06 1996-10-15 Eli Lilly And Company 2-acylaminopropanamides as tachykinin receptor antagonists
US5624677A (en) 1995-06-13 1997-04-29 Pentech Pharmaceuticals, Inc. Controlled release of drugs delivered by sublingual or buccal administration
US20030125269A1 (en) 1998-08-26 2003-07-03 Ming Li T-type calcium channel
PL366335A1 (en) * 2000-07-26 2005-01-24 Smithkline Beecham P.L.C. Aminopiperidine quinolines and their azaisosteric analogues with antibacterial activity
PA8535601A1 (es) * 2000-12-21 2002-11-28 Pfizer Derivados benzimidazol y piridilimidazol como ligandos para gabaa
WO2003007953A1 (en) 2001-07-18 2003-01-30 Washington University Methods and compositions for modulating t-type calcium channels
KR100534556B1 (ko) 2001-10-26 2005-12-08 주식회사 오리엔트바이오 알파1g 단백질의 기능을 억제하여 간질을 일으키지 않게하는 방법
KR20030037081A (ko) 2001-11-02 2003-05-12 한국과학기술연구원 T 타입 칼슘채널을 조절하여 복통을 억제하는 방법
GB0126781D0 (en) 2001-11-07 2002-01-02 Medical Res Council Modulation
GB0211833D0 (en) * 2002-05-22 2002-07-03 Univ London Sodium channel regulators and modulators
DE10227511A1 (de) 2002-06-19 2004-01-08 Max-Delbrück-Centrum für Molekulare Medizin Mittel zur medikamentösen Behandlung von akuten und chronischen Schmerzen
US20060003985A1 (en) 2002-10-17 2006-01-05 Renger John J Enhancement of sleep with t-type calcium channel antagonists
WO2004042351A2 (en) * 2002-10-30 2004-05-21 Merck & Co., Inc. Piperidinyl-alpha-aminoamide modulators of chemokine receptor activity
JP2007520996A (ja) 2003-01-15 2007-08-02 ミレニアム・ファーマシューティカルズ・インコーポレイテッド 44390、54181、211、5687、884、1405、636、4421、5410、30905、2045、16405、18560、2047、33751、52872、14063、20739、32544、43239、44373、51164、53010、16852、1587、2207、22245、2387、52908、69112、14990、18547、115、579、15985、15625、760、18603、2395、2554、8675、32720、4809、14303、16816、17827、32620、577、619、1423、2158、8263、15402、16209、16386、21165、30911、41897、1643、2543、9626、13231、32409、84260、2882、8203、32678または55053を用いて泌尿器科障害を処置するための方法および組成物
CN1902181A (zh) * 2003-10-28 2007-01-24 沃泰克斯药物股份有限公司 可用作离子通道调控剂的苯并咪唑
WO2005077082A2 (en) 2004-02-11 2005-08-25 Univertsity Of Virginia Patent Foundation INHIBITING CAV3 ISOFORMS AND THE δ25B SPLICE VARIENTS FOR THE DIAGNOSIS AND TREATMENT OF CANCER
US20060025438A1 (en) * 2004-02-25 2006-02-02 Salter-Cid Luisa M Amine-based and amide-based inhibitors of semicarbazide-sensitive amine oxidase (SSAO) enzyne activity and VAP-1 mediated adhesion useful for treatment of diseases
US20060003020A1 (en) 2004-03-11 2006-01-05 The Regents Of The University Of Michigan Anti-metastatic ability of mibefradil and gadolinium
US7723562B2 (en) 2004-05-04 2010-05-25 Korea Institute Of Science And Technology Mice lacking alpha 1G showing enhanced novelty-seeking and alcohol preference and therapeutic methods for mood disorders by modulating alpha 1G T-type calcium channels
WO2006040646A1 (en) * 2004-10-14 2006-04-20 Pfizer, Inc. Benzimidazole or indole amides as inhibitors of pin1
US20060089316A1 (en) * 2004-10-25 2006-04-27 Brown Truman R Method for reducing a susceptibility to tumor formation induced by 3-deoxyglucosone and precursors thereof
TW200730494A (en) 2005-10-10 2007-08-16 Glaxo Group Ltd Novel compounds
TW200728258A (en) 2005-10-10 2007-08-01 Glaxo Group Ltd Novel compounds
EP2545923A1 (de) * 2005-12-22 2013-01-16 Alchemia Limited Antibakterielle Wirkstoffe
AU2007233179A1 (en) * 2006-03-31 2007-10-11 Astrazeneca Ab Bicyclic benzimidazole compounds and their use as metabotropic glutamate receptor potentiators
TW200815353A (en) * 2006-04-13 2008-04-01 Euro Celtique Sa Benzenesulfonamide compounds and their use
EA016076B1 (ru) * 2006-07-06 2012-01-30 Глэксо Груп Лимитед Замещенные n-фенилметил-5-оксопролин-2-амиды в качестве антагонистов р2х7-рецептора и способы их применения
GB0613836D0 (en) * 2006-07-13 2006-08-23 Univ Greenwich New medical use of triazine derivatives
JP5539717B2 (ja) * 2006-07-14 2014-07-02 塩野義製薬株式会社 オキシム化合物およびその使用
TW200827340A (en) * 2006-11-10 2008-07-01 Astrazeneca Ab Chemical compounds
ES2551709T3 (es) * 2006-12-15 2015-11-23 Janssen Pharmaceutica, N.V. Inhibidores Bencimidazola TRPV1
WO2008118758A1 (en) 2007-03-23 2008-10-02 Icagen, Inc. Inhibitors of ion channels
BRPI0809930A2 (pt) 2007-04-03 2014-10-07 Glaxo Group Ltd Derivados de carboxamida imidazolidina como moduladores de p2x7
GB0706630D0 (en) 2007-04-04 2007-05-16 Glaxo Group Ltd Novel compounds
WO2010029299A1 (en) * 2008-09-12 2010-03-18 Biolipox Ab Pyrimidinone derivaties for use as medicaments
US8609696B2 (en) * 2008-12-18 2013-12-17 Boehringer Ingelheim International Gmbh Serotonin 5-HT2B receptor inhibitors
WO2010074193A1 (ja) * 2008-12-26 2010-07-01 大日本住友製薬株式会社 新規2環性複素環化合物
EP2448932B1 (de) * 2009-07-02 2014-03-05 Sanofi Neue 6-morpholin-4-yl-pyrimidin-4-(3h)-on-derivate und pharmazeutische zubereitung daraus als akt(pkb)-phosphorylierungshemmer
US20120220603A1 (en) * 2009-09-04 2012-08-30 Zalicus Pharmaceuticals Ltd. Substituted heterocyclic derivatives for the treatment of pain and epilepsy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3655901A (en) * 1970-07-30 1972-04-11 Merck & Co Inc Method of inhibiting the formation of phenylethanalamine-n-methyl transferase with 2-aminobenzimidazoles
WO2007117399A2 (en) * 2006-03-31 2007-10-18 Janssen Pharmaceutica N.V. Benzoimidazol-2-yl pyrimidines and pyrazines as modulators of the histamine h4 receptor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Chemical Abstracts Registry Number 825616-92-0, indexed in the Registry file on STN CAS ONLINE February 4, 2005. *
PubChem CID 21484168 - National Center for Biotechnology Information. PubChem Compound Database; CID=21484168, http://pubchem.ncbi.nlm.nih.gov/summary/summary.cgi?cid=21484168 (accessed April 18, 2015), create date December 5, 2007. *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10208023B2 (en) 2013-03-01 2019-02-19 Mark G. DeGiacomo Heterocyclic inhibitors of the sodium channel
US9255089B2 (en) 2013-03-28 2016-02-09 Gilead Sciences, Inc. Benzimidazolone derivatives as bromodomain inhibitors
US9458145B2 (en) 2013-05-09 2016-10-04 Gilead Sciences, Inc. Benzimidazole derivatives as bromodomain inhibitors
US10017501B2 (en) 2013-05-09 2018-07-10 Gilead Sciences, Inc. Benzimidazole derivatives as bromodomain inhibitors
US9108953B2 (en) 2013-11-26 2015-08-18 Gilead Sciences, Inc. Quinoline derivatives as bromodomain inhibitors
WO2018213426A1 (en) 2017-05-16 2018-11-22 Vertex Pharmaceuticals Incorporated Deuterated pyridone amides and prodrugs thereof as modulators of sodium channels
US11603351B2 (en) 2017-07-11 2023-03-14 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
WO2019014352A1 (en) 2017-07-11 2019-01-17 Vertex Pharmaceuticals Incorporated CARBOXAMIDES AS INHIBITORS OF SODIUM CHANNELS
US10647661B2 (en) 2017-07-11 2020-05-12 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
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
CN113692304A (zh) * 2019-02-06 2021-11-23 礼来公司 作为kcnq增强剂的1-((2-(2,2,2-三氟乙氧基)吡啶-4-基)甲基)脲衍生物
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
WO2021113627A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
US11919887B2 (en) 2019-12-06 2024-03-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
US11834441B2 (en) 2019-12-06 2023-12-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
WO2021231553A1 (en) * 2020-05-12 2021-11-18 Arizona Board Of Regents On Behalf Of The University Of Arizona Small molecule inhibitors of cav3.2 activity and uses thereof
WO2022256842A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Hydroxy and (halo)alkoxy substituted tetrahydrofurans as modulators of sodium channels
WO2022256679A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamide analogs as modulators of sodium channels
WO2022256622A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
WO2022256702A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran-2-carboxamides as modulators of sodium channels
US11827627B2 (en) 2021-06-04 2023-11-28 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
WO2022256676A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofuran analogs as modulators of sodium channels
WO2022256708A1 (en) 2021-06-04 2022-12-08 Vertex Pharmaceuticals Incorporated Solid dosage forms and dosing regimens comprising (2r,3s,4s,5r)-4-[[3-(3,4-difluoro-2-methoxy-phenyl)-4,5-dimethyl-5-(trifluoromethyl) tetrahydrofuran-2-carbonyl]amino]pyridine-2-carboxamide
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
WO2023205463A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2023205778A1 (en) 2022-04-22 2023-10-26 Vertex Pharmaceuticals Incorporated Heteroaryl compounds for the treatment of pain
WO2024123815A1 (en) 2022-12-06 2024-06-13 Vertex Pharmaceuticals Incorporated Process for the synthesis of substituted tetrahydrofuran modulators of sodium channels

Also Published As

Publication number Publication date
US9688615B2 (en) 2017-06-27
CA2828456C (en) 2021-05-04
EP3009427B1 (de) 2019-12-18
US20150361032A1 (en) 2015-12-17
WO2012116440A1 (en) 2012-09-07
EP2681200A4 (de) 2015-05-27
ES2786298T3 (es) 2020-10-09
CA2828456A1 (en) 2012-09-07
US10351514B2 (en) 2019-07-16
US20170260130A1 (en) 2017-09-14
US20180111894A1 (en) 2018-04-26
EP3009427A1 (de) 2016-04-20
EP2681200A1 (de) 2014-01-08

Similar Documents

Publication Publication Date Title
US10351514B2 (en) Benzimidazole inhibitors of the sodium channel
US8629149B2 (en) Oxopiperazine derivatives for the treatment of pain and epilepsy
US20140113898A1 (en) Bisarylsulfone and dialkylarylsulfone compounds as calcium channel blockers
US20120220564A1 (en) Selective calcium channel modulators
WO2013131018A1 (en) Biaryl inhibitors of the sodium channel
US20120220603A1 (en) Substituted heterocyclic derivatives for the treatment of pain and epilepsy
US10208023B2 (en) Heterocyclic inhibitors of the sodium channel
US20120245137A1 (en) Aryl sulphone derivatives as calcium channel blockers
US11236078B2 (en) Heterocyclic inhibitors of the sodium channel
JP2008523072A (ja) タンパク質キナーゼの阻害剤
KR20080027864A (ko) N-(헤테로아릴)-1-헤테로아릴알킬-1h-인돌-2-카르복스아미드 유도체, 그의 제법 및 용도
KR20210021519A (ko) 통합 스트레스 반응 경로의 저해제
US20140371212A1 (en) Bace inhibitors
US20070185098A1 (en) Inhibitors of protein kinases
US20090012010A1 (en) Amino acid derivatives as calcium channel blockers
US20230026696A1 (en) Trpv4 receptor ligands
US20170107203A1 (en) Heterocyclic inhibitors of the sodium channel

Legal Events

Date Code Title Description
AS Assignment

Owner name: ZALICUS PHARMACEUTICALS LTD., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAJOUHESH, HASSAN;HOLLAND, RICHARD;ZHANG, LINGYUN;AND OTHERS;SIGNING DATES FROM 20131017 TO 20131028;REEL/FRAME:031995/0183

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: EPIRUS BIOPHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZALICUS PHARMACEUTICALS LTD.;REEL/FRAME:038100/0291

Effective date: 20150929

AS Assignment

Owner name: DEGIACOMO, INTERIM TRUSTEE, MARK G., MASSACHUSETTS

Free format text: COURT APPOINTMENT;ASSIGNOR:EPIRUS BIOPHARMACEUTICALS, INC.;REEL/FRAME:041099/0031

Effective date: 20160726

AS Assignment

Owner name: DEGIACOMO, INTERIM TRUSTEE, MARK G., MASSACHUSETTS

Free format text: COURT APPOINTMENT;ASSIGNOR:EPIRUS BIOPHARMACEUTICALS, INC.;REEL/FRAME:041518/0503

Effective date: 20160726

AS Assignment

Owner name: ANABIOS CORPORATION, CALIFORNIA

Free format text: ASSET PURCHASE AGREEMENT;ASSIGNOR:DEGIACOMO, INTERIM TRUSTEE, MARK G.;REEL/FRAME:062078/0813

Effective date: 20180824