AU2002238023B2 - Pyridine-substituted benzanilides as potassium ion channel openers - Google Patents

Pyridine-substituted benzanilides as potassium ion channel openers Download PDF

Info

Publication number
AU2002238023B2
AU2002238023B2 AU2002238023A AU2002238023A AU2002238023B2 AU 2002238023 B2 AU2002238023 B2 AU 2002238023B2 AU 2002238023 A AU2002238023 A AU 2002238023A AU 2002238023 A AU2002238023 A AU 2002238023A AU 2002238023 B2 AU2002238023 B2 AU 2002238023B2
Authority
AU
Australia
Prior art keywords
substituted
unsubstituted
compound according
compounds
group
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.)
Ceased
Application number
AU2002238023A
Other versions
AU2002238023A1 (en
Inventor
George Salvatore Amato
Paul Christopher Fritch
Grant Mcnaughton-Smith
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.)
Icagen Inc
Original Assignee
Icagen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2000/021308 external-priority patent/WO2001010380A2/en
Priority claimed from US09/776,791 external-priority patent/US6495550B2/en
Application filed by Icagen Inc filed Critical Icagen Inc
Publication of AU2002238023A1 publication Critical patent/AU2002238023A1/en
Application granted granted Critical
Publication of AU2002238023B2 publication Critical patent/AU2002238023B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

WO 02/062295 PCT/US02/03061 PYRIDINE-SUBSTITUTED BENZANILIDES AS POTASSIUM ION CHANNEL OPENERS CROSS-REFERENCES TO RELATED APPLICATIONS The present application claims priority to U.S. Provisional Patent Application Serial No. 60/147,221, filed on August 4, 1999, and is a continuation-in-part of U.S. Patent Application Serial No. 09/632,576, filed on August 4, 2000 the disclosure of each of which is incorporated herein by reference in its entirety for all purposes.
FIELD OF THE INVENTION This invention relates to the use of certain pyridine-substituted benzanilide derivatives as potassium channel openers and to the treatment of diseases modulated by potassium channels. Additionally, this invention relates to novel compounds that are useful as potassium channel openers.
BACKGROUND OF THE INVENTION Ion channels are cellular proteins that regulate the flow of ions, including calcium, potassium, sodium and chloride, into and out of cells. These channels are present in all human cells and affect such processes as nerve transmission, muscle contraction and cellular secretion. Among the ion channels, potassium channels are the most ubiquitous and diverse, being found in a variety of animal cells such as nervous, muscular, glandular, immune, reproductive, and epithelial tissue. These channels allow the flow of potassium in and/or out of the cell under certain conditions. For example, the outward flow of potassium ions upon opening of these channels makes the interior of the cell more negative, counteracting depolarizing voltages applied to the cell. These channels are regulated, by calcium sensitivity, voltage-gating, second messengers, extracellular ligands, and ATP-sensitivity.
Potassium channels have now been associated with a number of physiological processes, including regulation of heartbeat, dilation of arteries, release of insulin, excitability of nerve cells, and regulation of renal electrolyte transport. Potassium channels are made by alpha subunits that fall into at least 8 families, based on predicted structural and functional similarities (Wei et al., Neuropharmacology 35(7): 805-829 (1997)). Three of these families (Kv, eag-related, and KQT) share a common motif of six WO 02/062295 PCT/US02/03061 transmembrane domains and are primarily gated by voltage. Two other families, CNG and SK/IK, also contain this motif but are gated by cyclic nucleotides and calcium, respectively. The three other families of potassium channel alpha subunits have distinct patterns oftransmembrane domains. Slo family potassium channels, or BK channels have seven transmembrane domains (Meera et al., Proc. Natl. Acad. Sci. U.S.A. 94(25): 14066-71 (1997)) and are gated by both voltage and calcium or pH (Schreiber et al., J.
Biol. Chem. 273: 3509-16 (1998)). Another family, the inward rectifier potassium channels (Kir), belong to a structural family containing two transmembrane domains, and an eighth functionally diverse family (TP, or "two-pore") contains two tandem repeats of this inward rectifier motif.
Potassium channels are typically formed by four alpha subunits, and can be homomeric (made of identical alpha subunits) or heteromeric (made of two or more distinct types of alpha subunits). In addition, potassium channels made from Kv, KQT and Slo or BK subunits have often been found to contain additional, structurally distinct auxiliary, or beta, subunits. These subunits do not form potassium channels themselves, but instead they act as auxiliary subunits to modify the functional properties of channels formed by alpha subunits. For example, the Kv beta subunits are cytoplasmic and are known to increase the surface expression of Kv channels and/or modify inactivation kinetics of the channel (Heinemann et al., J Physiol. 493: 625-633 (1996); Shi et al., Neuron 16(4): 843-852 (1996)). In another example, the KQT family beta subunit, minK, primarily changes activation kinetics (Sanguinetti et al., Nature 384: 80-83 (1996)).
Slo or BK potassium channels are large conductance potassium channels found in a wide variety of tissues, both in the central nervous system and periphery. They play a key role in the regulation of processes such as neuronal integration, muscular contraction and hormone secretion. They may also be involved in processes such as lymphocyte differentiation and cell proliferation, spermatocyte differentiation and sperm motility. Three alpha subunits of the Slo family have been cloned, Slol, Slo2, and Slo3 (Butler et al., Science 261: 221-224 (1993); Schreiber et al., J Biol. Chem., 273: 3509-16 (1998); and Joiner et al., Nature Neurosci. 1: 462-469 (1998)). These Slo family members have been shown to be voltage and/or calcium gated, and/or regulated by intracellular pH.
Certain members of the Kv family of potassium channels were recently renamed (see Biervert, et al., Science 279: 403-406 (1998)). KvLQTl was re-named KCNQ1, and the KvLQT1-related channels (KvLR1 and KvLR2) were renamed KCNQ2 WO 02/062295 PCT/US02/03061 and KCNQ3, respectively. More recently, a fourth member of the KCNQ subfamily was identified (KCNQ4) as a channel expressed in sensory outer hair cells (Kubisch, et al., Cell 96(3): 437-446 (1999)).
KCNQ2 and KCNQ3 have been shown to be nervous system-specific potassium channels associated with benign familial neonatal convulsions a class of idiopathic generalized epilepsy (see, Leppert, et al., Nature 337: 647-648 (1989)).
These channels have been linked to M-current channels (see, Wang, et al., Science 282: 1890-1893 (1998)). The discovery and characterization of these channels and currents provides useful insights into how these voltage dependent (Kv) potassium channels function in different environments, and how they respond to various activation mechanisms. Such information has now led to the identification of modulators of KCNQ2 and KCNQ3 potassium channels or the M-current, and the use of such modulators as therapeutic agents. The modulators are the subject of the present invention.
KCNQ4 (Kubsich et al., Cell 96(3): 437 (1999)), KCNQ5 (Kananura et al., Neuroreport 11 (9):2063 (2000)), KCNQ 3/5 (Wickenden et al., Br. J. Pharma 132: 381 (2001)) and KCNQ6 have also recently been described.
SUMMARY OF THE INVENTION The present invention provides pyridine-substituted benzanilide compounds, and pharmaceutically acceptable salts thereof ("compounds of the invention"), which are useful in the treatment of diseases through the modulation of potassium ion flux through voltage-dependent potassium channels.
In one aspect, the present invention provides compounds having a structure according to Formula I: x
Y
Ar N H
(I)
in which the symbol Ar' represents a member selected from the group consisting of aryl, substituted aryl, heteroaryl and substituted heteroaryl. The letter X represents a member selected from the group consisting of O, S and in which R' is H, (C1-Cs)alkyl, substituted (CI-Cs)alkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C4)alkyl, substituted aryl(C1-C 4 )alkyl, CN, WO 02/062295 PCT/US02/03061
-C(O)R
2
-OR
3 -C(O)NR R 4 or -S(O)zNR 3
R
4 The symbol R 2 represents a member selected from the group consisting of (Ci-Cs)alkyl, substituted (Ci-Cs)alkyl, cycloalkyl, substituted cycloalkyl, heteroalkyl, substituted heteroalkyl, heterocyclyl, substituted heterocyclyl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl and substituted aryl(Ci-C 4 )alkyl. R 3 and R 4 are each members independently selected from the group consisting of hydrogen, (Ci-Cs)alkyl, substituted (Ci-Cs)alkyl, cycloalkyl, substituted cycloalkyl, heteroalkyl, substituted heteroalkyl, heterocyclyl, substituted heterocyclyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl and substituted aryl(Ci-C 4 )alkyl. Alternatively, R 3 and R 4 can be combined with the nitrogen to which each is attached to form a 6- or 7-membered ring, optionally having additional heteroatoms at the ring vertices. The letter Y represents a member selected from the group consisting of halogen, C1-C 4 alkyl, C 1
-C
4 substituted alkyl, -OCH 3 and
OCF
3 In another aspect, the present invention provides pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a compound having a structure according to Formula II:
X
Ar 1 N A H
(II)
in which the symbols Ar' and Ar 2 independently represent aryl, substituted aryl, heteroaryl and substituted heteroaryl. The letter represents O, S or N-R 1 in which R 1 is a H, (CI-Cs)alkyl, substituted (C 1 -Cs)alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl, substituted aryl(C 1
-C
4 )alkyl, CN, -C(O)R 2
-OR
3
C(O)NR
3
R
4 or -S(O) 2
NR
3
R
4 The symbol R 2 represents (Ci-Cs)alkyl, substituted (C1- Cs)alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl or substituted aryl(Ci-C 4 )alkyl. R 3 and R 4 are each members independently selected from the group consisting of hydrogen, (Ci-Cs)alkyl, substituted (C1-Cs)alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(CI-C 4 )alkyl and substituted aryl(CI-C 4 )alkyl.
Alternatively, R 3 and R 4 can be combined with the nitrogen to which each is attached to form a 6- or 7-membered ring optionally having additional heteroatoms at the ring vertices.
In a further aspect, the present invention provides compounds having a structure according to Formula III: WO 02/062295 PCT/US02/03061 Iu-'I H I R Q H H V (III).
In Formula III, the symbols Y' and Y 2 independently represent H, methyl, methoxy, trifluoromethoxy, -CF 3 or halo, with the proviso that both Y' and Y 2 are not H. The symbols V and X independently represent H, halo, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, NO 2 CN, CF 3
C(O)NR'R
12 and
C(O)R
1 3 The symbols R 1 1
R
1 2 and R 1 3 independently represent substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, substituted or unsubstituted carbocycle, substituted or unsubstituted heterocycle, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. R' 1 and R 1 2 can be joined into a ring. Q and W independently represent -(CR 2
R
3 )t-(CH 2
-(CH
2 )n-(CR2R 3 )t, or wherein R 2 and R 3 are independently F, substituted or unsubstituted lower alkyl or substituted or unsubstituted lower heteroalkyl, wherein R and R 3 are optionally joined to form a cyclic structure which is a member selected from the group consisting of cycloalkyls and heterocycles, or R 2 and R 3 together with the carbon to which they are attached form carbonyl Q can optionally be a bond between the phenyl ring and Z, and W can optionally be a bond between Z and R The symbol Z represents -N(R 4
-N(R
4
-C(O)N(R
4
-N(R
4
-N(R
4 and -SO 2
N(R
4 wherein R 4 and R 5 are members independently selected from the group consisting of H, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. R' is optionally joined together with either X or
R
4 to form a substituted or unsubstituted heterocycle. The symbol m represents an integer from 0 to 2, inclusive; n represents and integer from 0 and 3, inclusive; and t represents an integer from 0 to 2, inclusive.
In yet another aspect, the present invention provides a method for modulating ion flux through voltage dependent potassium channels, comprising contacting a cell containing the target ion channels with a compound according to Formulae I-IV.
WO 02/062295 PCT/US02/03061 In still another aspect, the present invention provides a method for the treatment of diseases through modulation of ion flux through voltage dependent potassium channels, the method comprising treating the host with an effective amount of a potassium channel opening compound of Formula I-IV.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 displays structures of representative compounds of the invention.
DETAILED DESCRIPTION OF THE INVENTION AND THE PREFERRED EMBODIMENTS Abbreviations and Definitions: The abbreviations used herein have their conventional meaning within the chemical and biological arts. For example: CHO, Chinese hamster ovary; EBSS, Earl's Balanced Salt Solution; KCNQ, potassium channel Q; KCNQ2, potassium channel Q2, hSK, Ca 2 activated small conductance potassium channels; SDS, sodium dodecyl sulfate; Et 3 N, triethylamine; MeOH, methanol; and DMSO, dimethylsulfoxide.
Where substituent groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents which would result from writing the structure from right to left, -CH 2 0is intended to also recite -OCH 2 The term "alkyl," by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e.
C
1 -Cio means one to ten carbons). Examples of saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3propynyl, 3-butynyl, and the higher homologs and isomers. The term "alkyl," unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail WO 02/062295 PCT/US02/03061 below, such as "heteroalkyl." Alkyl groups which are limited to hydrocarbon groups are termed "homoalkyl".
The term "alkylene" by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified, but not limited, by
CH
2
CH
2 CH2CH 2 and further includes those groups described below as "heteroalkylene." Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. A "lower alkyl" or "lower alkylene" is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
The terms "alkoxy," "alkylamino" and "alkylthio" (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.
The term "heteroalkyl," by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and at least one heteroatom selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) 0, N and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to,
CH
2
-CH
2
-O-CH
3
-CH
2
-CH
2
-NH-CH
3
-CH
2
-CH
2
-N(CH
3
)-CH
3
-CH
2
-S-CH
2
-CH
3
-CH
2
CH
2
,-S(O)-CH
3
-CH
2
-CH
2 -S(0) 2
-CH
3
-CH=CH-O-CH
3 -Si(CH 3 3
-CH
2
-CH=N-OCH
3 and -CH=CH-N(CH 3
)-CH
3 Up to two heteroatoms may be consecutive, such as, for example, -CH 2
-NH-OCH
3 and -CH 2 -O-Si(CH 3 3 Similarly, the term "heteroalkylene" by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH 2
-CH
2
-S-CH
2
-CH
2 and -CH 2
-S-CH
2
CH
2
-NH-CH
2 For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula -C(0) 2 represents both -C(0) 2 and -R'C(0) 2 The terms "cycloalkyl" and "heterocycloalkyl", by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of "alkyl" and "heteroalkyl", respectively. Additionally, for heterocycloalkyl, a heteroatom WO 02/062295 PCT/US02/03061 can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include, but are not limited to, 1 -(1,2,5,6-tetrahydropyridyl), 1piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran- 2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1 -piperazinyl, 2piperazinyl, and the like.
The terms "halo" or "halogen," by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
Additionally, terms such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl. For example, the term "halo(Ci-C 4 )alkyl" is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
The term "aryl" means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent which can be a single ring or multiple rings (preferably from 1 to 3 rings) which are fused together or linked covalently. The term "heteroaryl" refers to aryl groups (or rings) that contain from one to four heteroatoms selected from N, 0, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule through a heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, 1 -naphthyl, 2-naphthyl, 4-biphenyl, 1 -pyrrolyl, 2-pyrrolyl, 3pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl.
Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.
For brevity, the term "aryl" when used in combination with other terms aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the term "arylalkyl" is meant to include those radicals in which an aryl group is attached to an alkyl group benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom a methylene group) has been WO 02/062295 PCT/US02/03061 replaced by, for example, an oxygen atom phenoxymethyl, 2-pyridyloxymethyl, 3- (1-naphthyloxy)propyl, and the like).
Each of the above terms "alkyl," "heteroalkyl," "aryl" and "heteroaryl") are meant to include both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below.
Substituents for the alkyl and heteroalkyl radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of groups selected from, but not limited to: halogen, -CO 2 -CONR'R", -OC(0)NR'R", NR"C(0)R', -NR"C(0) 2 -S(0) 2
-S(O)
2 NR'R", -NRSO 2 -CN and -NO 2 in a number ranging from zero to where m' is the total number of carbon atoms in such radical. and each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, aryl substituted with 1-3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups. When a compound of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R", and groups when more than one of these groups is present. When R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a or 7-membered ring. For example, -NR'R" is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term "alkyl" is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl
-CF
3 and -CH 2
CF
3 and acyl -C(O)CH 3 -C(0)CF 3 -C(O)CH20CH 3 and the like).
Similar to the substituents described for the alkyl radical, substituents for the aryl and heteroaryl groups are varied and are selected from, for example: halogen, -halogen,
-CO
2 -CONR'R", -OC(O)NR'R", -NR"C(0) 2
R',
-S(0) 2 -S(0) 2
NR'R",
-NRSO
2 -CN and -NO 2
-N
3 -CH(Ph) 2 fluoro(Ci-C 4 )alkoxy, and fluoro(Ci-
C
4 )alkyl, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where and are preferably independently WO 02/062295 PCT/US02/03061 selected from hydrogen, (C1-Cs)alkyl and heteroalkyl, unsubstituted aryl and heteroaryl, (unsubstituted aryl)-(Ci-C 4 )alkyl, and (unsubstituted aryl)oxy-(Ci-C 4 )alkyl. When a compound of the invention includes more than one R group, for example, each of the R groups is independently selected as are each and groups when more than one of these groups is present.
Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula wherein T and U are independently -CRR'- or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula
A-(CH
2 wherein A and B are independently -S(O) 2 -S(0) 2 NR'- or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula where s and d are independently integers of from 0 to 3, and X is -S(O) 2 or S(0) 2 The substituents R, R" and are preferably independently selected from hydrogen or substituted or unsubstituted (Ci-C 6 )alkyl.
As used herein, the term "heteroatom" is meant to include oxygen nitrogen sulfur and silicon (Si).
The term "pharmaceutically acceptable salts" is meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, WO 02/062295 PCT/US02/03061 hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
The neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
In addition to salt forms, the present invention provides compounds, which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention. Additionally, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, geometric isomers and individual isomers are encompassed within the scope of the present invention.
WO 02/062295 PCT/US02/03061 The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium 3 iodine-125 (1251) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
Introduction The present invention provides compounds which, inter alia, are useful in the treatment of diseases through the modulation of potassium ion flux through voltagedependent potassium channels. More particularly, the invention provides compounds, compositions and methods that are useful in the treatment of central or peripheral nervous system disorders migraine, ataxia, Parkinson's disease, bipolar disorders, trigeminal neuralgia, spasticity, mood disorders, brain tumors, psychotic disorders, myokymia, seizures, epilepsy, hearing and vision loss, Alzheimer's disease, age-related memory loss, learning deficiencies, anxiety and motor neuron diseases), and as neuroprotective agents to prevent stroke and the like). Compounds of the invention have use as agents for treating convulsive states, for example that following grand mal, petit mal, psychomotor epilepsy or focal seizure. The compounds of the invention are also useful in treating disease states such as gastroesophogeal reflux disorder and gastrointestinal hypomotility disorders.
Moreover, compounds of the invention are useful in the treatment of pain, for example, neuropathic pain, inflammatory pain, cancer pain, migraine pain, and musculoskeletal pain. The compounds are also useful to treat conditions, which may themselves be the origin of pain, for example, inflammatory conditions, including arthritic conditions rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis and gouty arthritis) and non-articular inflammatory conditions herniated, ruptured and prolapsed disc syndrome, bursitis, tendonitis, tenosynovitis, fibromyalgia syndrome, and other conditions associated with ligamentous sprain and regional musculoskeletal strain).
Particularly preferred compounds of the invention are less ulcerogenic than other antiinflammatory agents ibuprofen, naproxen and aspirin). Furthermore, the compounds of the invention are useful in treating conditions and pain associated with abnormally raised skeletal muscle tone.
WO 02/062295 PCT/US02/03061 The compounds of the invention are also of use in treating anxiety (e.g.
anxiety disorders). Anxiety disorders are defined in the Diagnostic and Statistical Manual of Mental Disorders (Third Edition-revised 1987, published by the American Psychiatric Association, Washington, see, pages 235 to 253), as psychiatric conditions having symptoms of anxiety and avoidance behavior as characteristic features. Included amongst such disorders are generalized anxiety disorder, simple phobia and panic disorder.
Anxiety also occurs as a symptom associated with other psychiatric disorders, for example, obsessive compulsive disorder, post-traumatic stress disorder, schizophrenia, mood disorders and major depressive disorders, and with organic clinical conditions including, but not limited to, Parkinson's disease, multiple sclerosis, and other physically incapacitating disorders.
The development of therapeutic agents, which act on potassium ion channels has received considerable recent attention. One group has described a family of N-alkyl benzamides that act by blocking potassium channels (see, PCT/US98/02364, published as WO 98/37068). In contrast, the benzanilides provided herein act by opening potassium channels.
In view of the above-noted discovery, the present invention provides compounds, compositions, and methods for increasing ion flux in voltage-dependent potassium channels, particularly those channels responsible for the M-current. As used herein, the term "M-current," "channels responsible for the M-current" and the like, refers to a slowly activating, non-inactivating, slowly deactivating voltage-gated K+ channel.
M-current is active at voltages close to the threshold for action potential generation in a wide variety of neuronal cells, and thus, is an important regulator of neuronal excitability.
Recently, members of the voltage-dependent potassium channel family were shown to be directly involved in diseases of the central or peripheral nervous system. The benzanilides provided herein are now shown to act as potassium channel openers, particularly for KCNQ2 and KCNQ3, KCNQ4, KCNQ5 and KCNQ6 as well as the heteromultimer channels such as KCNQ2/3, KCNQ3/5 or the M-current.
Description of the Embodiments I. MODULATORS OF VOLTAGE-DEPENDENT POTASSIUM CHANNELS In view of the above surprising discovery, the present invention provides in one aspect, compounds according to Formula I: WO 02/062295 PCT/US02/03061 x Y Ar N H
(I)
in which the symbol Ar' represents a member selected from the group consisting of aryl, substituted aryl, heteroaryl and substituted heteroaryl. The letter X represents a member selected from the group consisting of O, S and in which R 1 is H, (Ci-Cs)alkyl, substituted (Ci-Cs)alkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(C 1
-C
4 )alkyl, substituted aryl(Ci-C 4 )alkyl, CN,
-C(O)R
2
-OR
3
-C(O)NR
3
R
4 or -S(O) 2
NR
3
R
4 The symbol R 2 represents a member selected from the group consisting of (C1-Cs)alkyl, substituted (Ci-Cs)alkyl, cycloalkyl, substituted cycloalkyl, heteroalkyl, substituted heteroalkyl, heterocyclyl, substituted heterocyclyl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl and substituted aryl(Ci-C 4 )alkyl. R 3 and R 4 are each members independently selected from the group consisting of hydrogen, (Ci-Cs)alkyl, substituted (C 1 -Cs)alkyl, cycloalkyl, substituted cycloalkyl, heteroalkyl, substituted heteroalkyl, heterocyclyl, substituted heterocyclyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C 4 )alkyl and substituted aryl(C1-C 4 )alkyl. Alternatively, R 3 and R 4 can be combined with the nitrogen to which each is attached to form a 6- or 7-membered ring, optionally having additional heteroatoms at the ring vertices. The letter Y represents a member selected from the group consisting of halogen, C1-C 4 alkyl, C 1
-C
4 substituted alkyl, -OCH 3 and
OCF
3 In one group of preferred embodiments, Ar 1 is selected from phenyl, substituted phenyl, indolyl, substituted indolyl, benzofuranyl, substituted benzofuranyl, furanyl, substituted furanyl, thienyl, substituted thienyl, isothiazolyl, substituted isothiazolyl, pyrazolyl or substituted pyrazolyl. Still further preferred are those embodiments in which Ar' is substituted phenyl, substituted or unsubstituted 2-indolyl and substituted or unsubstituted 2-thienyl. In yet another group of preferred embodiments, X is O.
In those preferred embodiments, in which Ar' is substituted, the Ar' substituents are preferably halogen, alkyl, halo(Ci-C 4 )alkyl, (C1-C 4 )alkoxy, halo(Ci-
C
4 )alkoxy, nitro, cyano, -N R 7 C(O) R 8 -N R 7
R
8 phenyl and/or substituted phenyl. The symbols R 7 and R 8 independently represent hydrogen, (C1-Cs)alkyl, substituted (Ci-Cs)alkyl, cycloalkyl, substituted cycloalkyl, heteroalkyl, substituted heteroalkyl, WO 02/062295 PCT/US02/03061 heterocyclyl, substituted heterocyclyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryl(Ci-C4)alkyl and substituted aryl(CI-C 4 )alkyl. Alternatively, R 7 and R 8 are combined with the nitrogen to which it is attached to form a 6- or 7-membered ring, optionally having additional heteroatoms at the ring vertices.
In a further aspect, the present invention provides compounds having a structure according to Formula III: Y1 H OY H 0 H 1 Y2 R,W ZN H H
QH
V (III).
In Formula III, the symbols Y' and Y 2 independently represent H, methyl, methoxy, trifluoromethoxy, -CF 3 or halo, with the proviso that both Y' and Y 2 are not H. The symbols V and X independently represent H, halo, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, NO 2 CN, CF 3 C(O)NR' R 12 and
C(O)R
13 The symbols R 1
R
12 and R 13 independently represent substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, substituted or unsubstituted carbocycle, substituted or unsubstituted heterocycle, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. and R 12 can be joined into a ring. Q and W independently represent -(CR 2
R
3 )t-(CH 2
-(CH
2 )n-(CR 2
R
3 )t, -C(R4)=C(R 5 or wherein R 2 and R 3 are independently H, F, substituted or unsubstituted lower alkyl or substituted or unsubstituted lower heteroalkyl, wherein R 2 and R 3 are optionally joined to form a cyclic structure which is a member selected from the group consisting of cycloalkyls and heterocycles, or R 2 and R 3 together with the carbon to which they are attached form carbonyl Q can optionally be a bond between the phenyl ring and Z, and W can optionally be a bond between Z and R 1 The symbol Z represents -N(R 4
-N(R
4
-C(O)N(R
4
-N(R
4
-N(R
4
-(CR
2
R
3 and -SO 2
N(R
4 wherein R 4 and R 5 are members independently selected from the group consisting of H, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. R 1 is optionally joined together with either X or R 4 to form a substituted or unsubstituted heterocycle. The WO 02/062295 PCT/US02/03061 symbol m represents an integer from 0 to 2, inclusive; n represents and integer from 0 and 3, inclusive; and t represents an integer from 0 to 2, inclusive.
In yet a further group of preferred embodiments, the compounds of the invention have a structure according to Formula IV: I o H
R
6
(IV).
In Formula IV, R 1 is R 6 and R 6 is selected from H, halogen, substituted or unsubstituted alkyl, halo(C 1
-C
4 )alkyl, nitro, cyano, substituted or unsubstituted phenyl,
R
9
R
9 RYNH-; R 9 NH-; R 9 NHS(0) 2
R
9 S(0) 2 with the proviso that both X and R 6 are not H. The symbol R 9 represents a member selected from aryl, and alkylaryl; when there is more than one R 9 group per molecule, each R 9 group is independently selected.
The symbol Y represents a member selected from halogen, C1-C 4 alkyl, -OCH3, and
-OCF
3 In a preferred embodiment, the compounds according to Formula IV, include an arylalkyl group at R 9 in which the aryl component of the arylalkyl group is unsubstituted aryl, or an aryl group that is substituted by from 1 to 3 halogen moieties.
More preferably, the alkyl component of the arylalkyl group is a C 1
-C
4 alkyl group.
In another preferred embodiment, the arylalkyl group according to R 9 is a substituted or unsubstituted heteroarylalkyl group. Preferred heteroarylalkyl groups include (Ci-C 4 )alkylpyridyl groups.
Certain combinations of the above preferred embodiments form a group of particularly preferred compounds. Accordingly, representative preferred compounds of the present invention are set forth in FIG. 1, appended hereto.
Also within the scope of the present invention are compounds of the invention that function as poly- or multi-valent species, including, for example, species such as dimers, trimers, tetramers and higher homologs of the compounds of the invention or reactive analogues thereof. The poly- and multi-valent species can be assembled from a single species or more than one species of the invention. For example, a dimeric construct can be "homo-dimeric" or "heterodimeric." Moreover, poly- and multi-valent constructs in which a compound of the invention or reactive analogues thereof are attached to an oligomeric or polymeric framework polylysine, dextran, WO 02/062295 PCT/US02/03061 hydroxyethyl starch and the like) are within the scope of the present invention. The framework is preferably polyfunctional having an array of reactive sites for attaching compounds of the invention). Moreover, the framework can be derivatized with a single species of the invention or more than one species of the invention.
Moreover, the present invention includes compounds within the motif set forth in Formulae I-IV, which are functionalized to afford compounds having a watersolubility that is enhanced relative to analogous compounds that are not similarly functionalized. Methods of enhancing the water-solubility of organic compounds is known in the art. Such methods include, but are not limited to, functionalizing an organic nucleus with a permanently charged moiety, quaternary ammonium, or a group that is charged at a physiologically relevant pH, e.g. carboxylic acid, amine. Other methods include, appending to the organic nucleus hydroxyl- or amine-containing groups, e.g.
alcohols, polyols, polyethers, and the like. Representative examples include, but are not limited to, polylysine, polyethyleneimine, poly(ethyleneglycol) and poly(propyleneglycol). Suitable functionalization chemistries and strategies for these compounds are known in the art. See, for example, Dunn, et al., Eds.
POLYMERIC DRUGS AND DRUG DELIVERY SYSTEMS, ACS Symposium Series Vol. 469, American Chemical Society, Washington, D.C. 1991.
Preparation of Potassium Channel Openers Compounds of the present invention can be prepared using readily available starting materials or known intermediates. Briefly, the synthesis ofN-aryl benzamides involves formation of a single amide bond from a "carbonyl component" a carboxylic acid, carboxylic acid chloride, ester or an activated form of a carboxylic acid, for example, a symmetrical or mixed anhydride) and an "amine component" an aniline, aniline derivative, amino heterocycle, and the like). General and specific procedures for the preparation of the present compounds are provided in the examples below.
Other compounds of the present invention can be prepared using standard procedures as outlined in Scheme 1 below. In this scheme, an N-phenyl benzamide (i, wherein D 1 and D 2 represent substituents, including multiple substituents on the aryl groups) can be treated with reagents such as Lawessons's reagent to provide the thioamides, ii. Alkylation of ii, with, for example, methyl iodide produces iii which can WO 02/062295 PCT/US02/03061 be converted to target structures iv, v and vi. Thus, treatment of iii with sodium hydride (or another suitable base) and sulfamide provides the sulfamoylimino derivative, iv.
Similarly, treatment of iii with sodium hydride or another base, followed by cyanamide provides v. Conversion ofv to vi can be accomplished with HC1.
One of skill in the art will recognize that other compounds of the present invention can be prepared from intermediates such as iii. For example, treatment of iii with a primary or secondary amine will provide amidine derivatives that are useful as described or they can be further derivatized.
SCHEME 1 0 N cID2 N0 D H Lawesson's reagent D1-- H ii Methyl iodide
II
NaH, H 2
NSO
2
NH
2 NaH, H 2
NCN
S NC H2N N v HCI iv v H 2 N NkN D2 D
H
vi Other methods of preparing the benzanilides of the invention will be apparent to, and are readily accessible by those of skill in the art.
WO 02/062295 PCT/US02/03061 Methods for preparing dimers, trimers and higher homologs of small organic molecules, such as those of the present invention, as well as methods of functionalizing a polyfunctional framework molecule are well known to those of skill in the art. For example, an aromatic amine of the invention is converted to the corresponding isothiocyanate by the action ofthiophosgene. The resulting isothiocyanate is coupled to an amine of the invention, thereby forming either a homo- or hetero-dimeric species. Alternatively, the isothiocyanate is coupled with an amine-containing backbone, such as polylysine, thereby forming a conjugate between a polyvalent framework and a compound of the invention. If it is desired to prepare a hetereofuntionalized polyvalent species, the polylysine is underlabeled with the first isothiocyanate and subsequently labeled with one or more different isothiocyanates. Alternatively, a mixture of isothiocyanates is added to the backbone. Purification proceeds by, for example, size exclusion chromatography, dialysis, nanofiltration and the like.
II. ASSAYS FOR MODULATORS OF KCNQ CHANNELS Assays for determining the ability of a compound of the invention to open a potassium ion channel are generally known in the art. One of skill in the art is able to determine an appropriate assay for investigating the activity of a selected compound of the invention towards a particular ion channel. For simplicity, portions of the following discussion focuses on KCNQ2 as a representative example, however, the discussion is equally applicable to other potassium ion channels.
KCNQ monomers as well as KCNQ alleles and polymorphic variants are subunits of potassium channels. The activity of a potassium channel comprising KCNQ subunits can be assessed using a variety of in vitro and in vivo assays, measuring current, measuring membrane potential, measuring ion flux, potassium or rubidium, measuring potassium concentration, measuring second messengers and transcription levels, using potassium-dependent yeast growth assays, and using voltage-sensitive dyes, radioactive tracers, and patch-clamp electrophysiology.
Furthermore, such assays can be used to test for inhibitors and activators of channels comprising KCNQ. Such modulators of a potassium channel are useful for treating various disorders involving potassium channels discussed herein and known to those of skill in the art. Exemplary disorders include, but are not limited to, central and peripheral nervous system disorders migraine, ataxia, Parkinson's disease, bipolar disorders, spasticity, mood disorders, brain tumors, psychotic disorders, myokymia, WO 02/062295 PCT/US02/03061 seizures, epilepsy, hearing and vision loss, Alzheimer's disease, age-related memory loss, learning deficiencies, and motor neuron diseases, and can also be used as neuroprotective agents to prevent stroke and the like). Such modulators are also useful for investigation of the channel diversity provided by KCNQ and the regulation/modulation of potassium channel activity provided by KCNQ.
Modulators of the potassium channels are tested using biologically active KCNQ, either recombinant or naturally occurring, or by using native cells, like cells from the nervous system expressing the M-current. KCNQ can be isolated, co-expressed or expressed in a cell, or expressed in a membrane derived from a cell. In such assays, KCNQ2 is expressed alone to form a homomeric potassium channel or is co-expressed with a second subunit another KCNQ family member, preferably KCNQ3) so as to form a heteromeric potassium channel. Modulation is tested using one of the in vitro or in vivo assays described above. Samples or assays that are treated with a potential potassium channel inhibitor or activator are compared to control samples without the test compound, to examine the extent of modulation. Control samples (untreated with activators or inhibitors) are assigned a relative potassium channel activity value of 100.
Activation of channels comprising KCNQ2 is achieved when the potassium channel activity value relative to the control is 110%, more preferably 130%, more preferably 170% higher. Compounds that increase the flux of ions will cause a detectable increase in the ion current density by increasing the probability of a channel comprising KCNQ2 being open, by decreasing the probability of it being closed, by increasing conductance through the channel, and increasing the number or expression of channels.
Changes in ion flux may be assessed by determining changes in polarization electrical potential) of the cell or membrane expressing the potassium channel comprising, for example, KCNQ2, KCNQ2/3 or the M-current. A preferred means to determine changes in cellular polarization is by measuring changes in current or voltage with the voltage-clamp and patch-clamp techniques, using the "cell-attached" mode, the "inside-out" mode, the "outside-out" mode, the "perforated cell" mode, the "one or two electrode" mode, or the "whole cell" mode (see, Ackerman et al., New Engl. J. Med. 336:1575-1595 (1997)). Whole cell currents are conveniently determined using the standard methodology (see, Hamil et al., Pflugers. Archiv. 391:85 (1981).
Other known assays include: radiolabeled rubidium flux assays and fluorescence assays using voltage-sensitive dyes (see, Vestergarrd-Bogind et al,, J Membrane Biol.
88:67-75 (1988); Daniel et al., J. Pharmacol. Meth. 25:185-193 (1991); Holevinsky et al., WO 02/062295 PCT/US02/03061 J. Membrane Biology 137:59-70 (1994)). Assays for compounds capable of inhibiting or increasing potassium flux through the channel proteins comprising KCNQ2 or heteromultimers of KCNQ subunits can be performed by application of the compounds to a bath solution in contact with and comprising cells having a channel of the present invention (see, Blatz et al., Nature 323:718-720 (1986); Park, J. Physiol. 481:555- 570 (1994)). Generally, the compounds to be tested are present in the range from 1 pM to 100 mM.
The effects of the test compounds upon the function of the channels can be measured by changes in the electrical currents or ionic flux or by the consequences of changes in currents and flux. Changes in electrical current or ionic flux are measured by either increases or decreases in flux of ions such as potassium or rubidium ions. The cations can be measured in a variety of standard ways. They can be measured directly by concentration changes of the ions or indirectly by membrane potential or by radiolabeling of the ions. Consequences of the test compound on ion flux can be quite varied.
Accordingly, any suitable physiological change can be used to assess the influence of a test compound on the channels of this invention. The effects of a test compound can be measured by a toxin binding assay. When the functional consequences are determined using intact cells or animals, one can also measure a variety of effects such as transmitter release dopamine), hormone release insulin), transcriptional changes to both known and uncharacterized genetic markers northern blots), cell volume changes in red blood cells), immunoresponses T cell activation), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as Ca2+, or cyclic nucleotides.
Preferably, the KCNQ2, or other KCNQ channel constituents that is a part of the potassium channel used in the assay will have the sequence provided in PCT/US98/13276 or a conservatively modified variant thereof. Alternatively, the KCNQ2 of the assay will be derived from a eukaryote.
KCNQ2 orthologs will generally confer substantially similar properties on a channel comprising such KCNQ2, as described above. In a preferred embodiment, the cell placed in contact with a compound that is suspected to be a KCNQ2 homolog is assayed for increasing or decreasing ion flux in a eukaryotic cell, an oocyte of Xenopus Xenopus laevis) or a mammalian cell such as a CHO or HeLa cell.
Channels that are affected by compounds in ways similar to KCNQ2 are considered homologs or orthologs of KCNQ2.
WO 02/062295 PCT/US02/03061 III. PHARMACEUTICAL COMPOSITIONS OF POTASSIUM CHANNEL
OPENERS
In another aspect, the present invention provides pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a compound according to Formula II, above.
Formulation of the Compounds (Compositions) The compounds of the present invention can be prepared and administered in a wide variety of oral, parenteral and topical dosage forms. Thus, the compounds of the present invention can be administered by injection, that is, intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally, or intraperitoneally.
Also, the compounds described herein can be administered by inhalation, for example, intranasally. Additionally, the compounds of the present invention can be administered transdermally. Accordingly, the present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier or excipient and either a compound of Formulae I-IV or a pharmaceutically acceptable salt of a compound of Formulae I-IV.
For preparing pharmaceutical compositions from the compounds of the present invention, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substance, which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
In powders, the carrier is a finely divided solid, which is in a mixture with the finely divided active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
The powders and tablets preferably contain from 5% or 10% to 70% of the active compound. Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. The term "preparation" is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component WO 02/062295 PCT/US02/03061 with or without other carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.
For preparing suppositories, a low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.
Also included are solid form preparations, which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
The pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
The quantity of active component in a unit dose preparation may be varied or adjusted from 0.1 mg to 10000 mg, more typically 1.0 mg to 1000 mg, most typically 10 mg to 500 mg, according to the particular application and the potency of the active component. The composition can, if desired, also contain other compatible therapeutic agents.
WO 02/062295 PCT/US02/03061 IV. METHODS FOR INCREASING ION FLOW IN VOLTAGE-DEPENDENT POTASSIUM CHANNELS In yet another aspect, the present invention provides methods for increasing ion flow through voltage dependent potassium channels in a cell, comprising contacting a cell containing the target ion channels with a compound of Formulae I-IV, above.
The methods provided in this aspect of the invention are useful for the diagnosis of conditions that can be treated by modulating ion flux through voltagedependent potassium channels, or for determining if a patient will be responsive to therapeutic agents which act by opening potassium channels. In particular, a patient's cell sample can be obtained and contacted with a compound of Formula I-IV and the ion flux can be measured relative to a cell's ion flux in the absence of a compound of Formula I-IV. An increase in ion flux will typically indicate that the patient will be responsive to a therapeutic regimen of ion channel openers.
V. METHODS FOR TREATING CONDITIONS MEDIATED BY VOLTAGE- DEPENDENT POTASSIUM CHANNELS In still another aspect, the present invention provides a method for the treatment of diseases or conditions mediated, at least in part, by voltage-dependent potassium channels. In this method, a subject suffering from such a condition or disease is administered an effective amount of a compound of Formulae I-IV.
The compounds provided herein are useful as potassium channel openers and find therapeutic utility via modulation of voltage-dependent potassium channels in the treatment of diseases or conditions. The potassium channels that are typically opened are described herein as voltage-dependent potassium channels such as the KCNQ potassium channels. As noted above, these channels may include homomultimers and heteromultimers ofKCNQ2, KCNQ3, KCNQ4, KCNQ5 and KCNQ6. A heteromultimer of two proteins, KCNQ2 and KCNQ3 is referred to as, for example, KCNQ2/3. The conditions that can be treated with the compounds and compositions of the present invention may include, but are not limited to, central or peripheral nervous system disorders migraine, ataxia, Parkinson's disease, bipolar disorders, spasticity, mood disorders, brain tumors, psychotic disorders, myokymia, seizures, epilepsy, hearing and vision loss, Alzheimer's disease, age-related memory loss, learning deficiencies, and WO 02/062295 PCT/US02/03061 motor neuron diseases, and as neuroprotective agents to prevent stroke and the like)).
In therapeutic use for the treatment of epilepsy or other neurological conditions, the compounds utilized in the pharmaceutical method of the invention are administered at the initial dosage of about 0.001 mg/kg to about 1000 mg/kg daily. A daily dose range of about 0.1 mg/kg to about 100 mg/kg is more typical. The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being employed. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages, which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired.
The materials, methods and devices of the present invention are further illustrated by the examples, which follow. These examples are offered to illustrate, but not to limit the claimed invention.
EXAMPLES
Example 1 sets forth representative methods of preparing 2-substituted aminopyridines of use in preparing the compounds of the invention. The representative methods include the reduction of nitropyridines, rearrangement of nicotinic acids, and the displacement reactions of 2-halopyridines.
Examples 2 and 3 set forth a representative methods of preparing the benzanilides of the invention. Example 2 provides a method of preparing a benzanilide form an acid chloride. Example 3 provides a method of preparing a benzanilide from a carboxylic acid by generating the acid chloride in situ.
Example 4, 5, 6, and 7 set forth methods of elaborating the benzanilide nucleus. Example 4 provides a method of preparing 4-amino substituted benzanilides via a nucleophilic displacement. Example 5 provides a method for reducing an aromatic nitro group to the corresponding amine. Example 6 provides a method for preparing hydroxyl amine compounds. Example 7 provides a method for preparing sulfonamides. Example 8 sets forth the characterization of the compounds prepared by the methods of Examples 1-7.
WO 02/062295 PCT/US02/03061 Examples 8 and 9 set forth the preparation of a number of representative compounds of the invention in which the benzanilide nucleus is elaborated by selected groups. Example 10 sets forth the results of the physical characterization of the compounds prepared by Examples 8 and 9. Example 11 sets forth an assay useful in the evaluation of the activity towards potassium ion channels, KCNQ2, of selected compounds of the invention.
General In the examples below, unless otherwise stated, temperatures are given in degrees Celsius operations were carried out at room or ambient temperature, or (typically a range of from about 18-25 0 C; evaporation of solvent was carried out using a rotary evaporator under reduced pressure (typically, 4.5-30 mmHg) with a bath temperature of up to 60 0 C; the course of reactions was typically followed by TLC and reaction times are provided for illustration only; melting points are uncorrected; products exhibited satisfactory 1 H-NMR and/or microanalytical data; yields are provided for illustration only; and the following conventional abbreviations are also used: mp (melting point), L (liter(s)), mL (milliliters), mmol (millimoles), g (grams), mg (milligrams), min (minutes), and h (hours).
General Experimental.
Unless otherwise specified, all solvents (HPLC grade) and reagents were purchased from suppliers and used without further purification. Reactions were conducted under a blanket of argon unless otherwise stated. Analytical thin layer chromatography (tlc) was performed on Whatman Inc. 60 silica gel plates (0.25 mm thickness).
Compounds were visualized under UV lamp (254 nM) or by developing with KMnO 4 /KOH, ninhydrin or Hanessian's solution. Flash chromatography was done using silica gel from Selcetro Scientific (particle size 32-63). 'H NMR, 9F NMR and "C NMR spectra were recorded on a Varian 300 machine at 300 MHz, 282 MHz and 75.7 MHz, respectively. Melting points were recorded on a Electrothermal IA9100 apparatus and were uncorrected.
WO 02/062295 PCT/US02/03061 EXAMPLE 1 Preparation of 1.1 Reduction of nitropyridines Referring to Scheme 2, the desired aminopyridines (II) are prepared by reducing the corresponding nitropyridines One skilled in the art will recognize that there are several methods to accomplish step 1. Tin chloride in DMF, hydrogenation using catalytic palladium and sodium borohydride in the presence of catalytic nickel chloride are known methods.
Scheme 2
XNO
2 Step 1
XNH
2 X N X N I II 1. la Synthesis of 5-amino-2-bromovpridine Tin (II) chloride hydrate (0.78g, 3.5 mmol) was added to a stirring solution of 5-nitro-2-bromopyridine (0.24 g, 1.2 mmol) in DMF (5 mL) at RT. After 2h, 6N NaOH (2 mL) was added and the suspension was stirred vigorously for 10 min. The organics were extracted with diethyl ether (2 x 10 mL), washed with brine (2 x 10 mL) and dried (Na 2
SO
4 The filtered solution was then concentrated under reduced pressure to afford the desired product as a yellow oil (0.178g, which was used without further purification.
1.2 Rearrangement of nicotinic acids Rearrangement of the corresponding nicotinic acids (III) (Scheme 3) using a modified Schmidt reaction, followed by deprotection of the aniline group generated the desired aminopyridines (IV) as the corresponding TFA salts.
Scheme 3 COOH Step 1 NH 2
.TFA
X HIN X N ffl IV WO 02/062295 PCT/US02/03061 1.2 a Synthesis of 5-amino-2-methylpvridine (TFA salt) A solution of diphenylphosphorylazide (430 pL, 2 mmol), triethylamine (278 kiL, 2 mmol) and 6-methyl-nicotinic acid (274 mg, 2 mmol) in t-butanol (30 mL) was heated at reflux for 4h. The solution was cooled to RT and poured into water mL). The organics were extracted with ether (3 x 20 mL), washed with brine (2 x 10 mL) and dried (Na 2 S04). Column chromatography (1:1 hexane/ethyl acetate) of the bocprotected aminopyridine gave the intermediate as a white solid (156 mg, 38%).
The desired 5-amino-2-methylpyridine-TFA salt was generated in situ by stirring in a 20% TFA/DCM solution (2 mL) for 4h. The solution was concentrated under reduced pressure to afford a semi-solid, which was used without further purification.
1.2 b Synthesis of5-amino-2-trifluoromethvl)ovridine (TFA salt) A solution of diphenylphosphorylazide (644 tL, 3 mmol), triethylamine (417 uL, 3 mmol) and 6-(trifluoromethyl)-nicotinic acid (573 mg, 3 mmol) in t-butanol (50 mL) was heated at reflux for 4h, then cooled to RT and poured into water (50 mL).
The organics were extracted with ether (3 x 20 mL), washed with brine (2 x 10 mL) and dried (Na 2
SO
4 Column chromatography (1:1 hexane/ethyl acetate) of the boc-protected aniline gave the intermediate as a white solid (389 mg, The desired 5-amino-2-methylpyridine-TFA salt was generated in situ by stirring in a 20% TFA/DCM solution (2 mL) for 4h. The solution was concentrated under reduced pressure to afford a semi-solid, which was used without further purification.
1.3: Displacement of 2-halopvridines.
Several aminopyridines, which are not readily accessible via the methods outlined in schemes 1 or 2, may be synthesized via nucleophilic displacement of 2chloropyridines as depicted in Scheme 4.
Scheme 4 N 0 2 1) NucH, solvent 2 N 2) Pd/C, H 2 Nuc N V
VI
1.3a Synthesis of 5-amino-2-fluoropvridine A mixture of 5-nitro-2-chloropyridine (2.0 g, 12.6 mmol) and anhydrous potassium fluoride (2.2 g, 38 mmol) in a combination of sulfalone (6 mL) and benzene (4 WO 02/062295 PCT/US02/03061 mL) was stirred at RT for 20 min. The benzene was then removed by distillation. The resulting mixture was heated at 150 °C for 12h. The mixture was cooled to RT whereupon water (60 mL) was added. The desired product was separated from the solution via steam distillation. Extraction of the distillate with diethyl ether (2 x 10 mL) followed by drying (Na 2
SO
4 and concentration gave 5-nitro-2-fluoropyridine as a water white oil (1.3 g, 73%).
Palladium on charcoal (20 mg, cat) was added to a stirring solution of 5-nitro-2-fluoropyridine (100 mg, 0.7 mmol) in dichloromethane (3 mL) at RT. 1 atmosphere of hydrogen gas was then applied to the solution and the mixture was stirred at RT for lh. The mixture was passed through a short plug of celite and the resulting solution, containing the desired 5-amino-2-fluoropyridine, was used without further purification.
EXAMPLE 2 2.1 Preparation of Benzanilides from Acid Chlorides Benzanilides VIII) were prepared by reacting acid chlorides VII) with aminopyridines II, IV and VI) as shown in Scheme 5. The reaction was typically conducted in the presence of a tertiary amine base such as triethylamine in an organic solvent such as dichloromethane or tetrahydrofuran, and at room temperature.
Scheme C Step 1 A A H Y VY
VIII
A aryl or heteroaryl 2.2 General experimental for Scheme A solution of acid chloride (VII) (1 mmol) in a dry solvent acetonitrile, THF, DCM) (3 mL) was added dropwise to a stirring solution of aminopyridine (II or VI) (1 mmol) and N, N-diisopropylethylamine (1.2 mmol) in a dry solvent acetonitrile, THF, DCM) (5 mL) at RT. The resulting solution was stirred for an additional lh. If TLC analysis indicated presence of starting aniline the solution was heated at 55 °C for another lh. After cooling to room temperature ethyl acetate WO 02/062295 PCT/US02/03061 mL) was added and the solution was washed with water (2 x 10 mL) and dried (Na 2 S04).
The solvent was removed under reduced pressure and the crude material was purified by column chromatography (hexanes/ethyl acetate) or by crystallization (hexane/dichloromethane). The products were typically white solids (50-98%).
Compounds prepared via this procedure include, 1-15, 25, 27, 29 and 42.
2.2a Preparation ofN-[2-chloro-5-pvridvl]-3-(trifluoromethvl)benzamide (3) To a stirring solution of 5-amino-2-chloropyridine (129 mg, 1 mmol) and N,N-diisopropylethylamine (209 ptL, 1.2 mmol) in dry acetonitrile (5 mL) was added 3-(trifluoromethyl)benzoyl chloride (151 pIL, 1 mmol). The resulting solution was heated at 55 0 C for 3h. After cooling to room temperature ethyl acetate (10 mL) was added and the solution was washed with water (2 x 10 mL) and dried (Na 2 SO4). The solvent was removed under reduced pressure and the crude material was purified by column chromatography hexane/ethyl acetate) to afford the desired product as a white solid (284 mg, 94%).
EXAMPLE 3 Preparation ofBenzanilides from Acids Benzanilides (VIII) may be also be prepared from acids (IX) by initially converting them to their acid chlorides (VII). Acids (IX) were treated with oxalyl chloride in the presence of catalytic N, N-dimethylformamide in an organic solvent such as dichloromethane or tetrahydrofuran preferably at 0 The acid chloride, generated in situ was then reacted with aminopyridines (II, IV or VI) in the presence of a tertiary amine base such as triethylamine in an organic solvent such as dichloromethane or tetrahydrofuran. The reactions were typically performed at RT.
Scheme 6 W 0 W 0
"X
OH
X
A OH A H IX VIII 3.1 General experimental for Scheme 6 Oxalyl chloride (1.05 mmol) was added dropwise to a stirring suspension of acid (IX) (1 mmol) and DMF (0.1 mmol) in dry DCM (5 mL) at 0 OC. Once addition WO 02/062295 PCT/US02/03061 was complete the reaction was allowed to warm to RT and stirred for a further 45 min whereupon the reaction was a clear solution. This solution was added dropwise to a stirring solution of aminopyridine (II or VI) (0.95 mmol) and N, N-diisopropylethylamine (2.2 mmol) in DCM (5 mL) at RT. After 30 min the organics were washed with aqueous IN NaOH (10 mL), brine (10 mL) and dried (Na 2
SO
4 The filtered solution was concentrated under reduced pressure and the crude product was purified by column chromatography (hexanes/ethyl acetate) or by crystallization (hexane/dichloromethane).
Compounds prepared via this procedure include, compounds 16-24, 26, 28 and 37.
EXAMPLE 4 Preparation of 4-Amino Substituted Benzamides via Nucleophilic Displacement Aryl fluorides (Scheme 7) possessing strongly electron withdrawing groups in either the ortho or para positions were displaced with primary or secondary amines under elevated temperatures in a polar organic solvent such as DMSO or NMP to yield compounds of the formula (XI).
Scheme 7 0 5 X 0 A N N N H H F X R 1
R
2 N x A NO 2 CN, F A= NO 2 CN, F 4.1 General experimental for Scheme 7 A solution of amine (1.1 mmol) and (1 mmol) in either dry NMP or DMSO (3 mL) was heated at 120 °C for 12h. After cooling to RT, water (10 mL) and ethyl acetate (10 mL) were added. After vortexing for several minutes the organic layer was removed and dried (Na 2 S04). The solvent was removed under reduced pressure and the residue purified by column chromatography (hexanes/ethyl acetate or acetone/chloroform). The products (XI) were obtained as white solids (30-50%).
Compounds prepared via this procedure include, compounds 30-34.
WO 02/062295 PCT/US02/03061 EXAMPLE Reduction of Aromatic Nitro to Amine Scheme 8 outlines a general synthetic route to compounds of formulae (XIII) and (XIV).
Scheme 8 0 IT X 0 Y, 2 N step 1
N
X
XIII
0 0
X
N step 2 0 N N
H
2 N R N
H
XIII H XIV 5.1 General experimental for Scheme 8 Compounds of structure (XII) were prepared using either general procedure B or C. Tin (II) chloride hydrate (22 mmol) was added to a stirring solution of (XII) in DMF (20 mL) at RT. After 14h, 6N NaOH (6 mL) was added and the suspension was stirred vigorously for 10 min. The organics were extracted with ethyl acetate (2 x mL), washed with brine (2 x 10 mL) and dried (Na 2
SO
4 The filtered solutions were concentrated under reduced pressure and the crude products were purified by column chromatography (hexanes/ethyl acetate; 1:2) to afford the desired intermediates (XIII) as a white solids (60-90%).
The intermediates (XIII) (0.2 mmol) were coupled with either acid chlorides (VI) or acids (0.2 mmol) using the methods described in general procedures A and B. The desired products (XIV) were obtained as tan solids Compounds prepared via this procedure include, compound EXAMPLE 6 Preparation of 4-Hydroxyamino Compounds (XV).
Hydroxyamines (XV) were prepared according to the synthetic route outlined in Scheme 9.
WO 02/062295 PCT/US02/03061 Scheme 9 0 X HO X H
H
Xi H XV 6.1 General Experimental for Scheme 9 Tin (II) chloride hydrate (3 mmol) was added to a stirring solution of nitrobenzamide (XII) (1 mmol) in DMF (5 mL) at RT. After 3h, 6N NaOH (6 mL) was added and the suspension was stirred vigorously for 10 min. The organics were extracted with ethyl acetate (2 x 20 mL), washed with brine (2 x 10 mL) and dried (NazSO 4 The filtered solution was then concentrated under reduced pressure to afford the crude product. Purification by column chromatography (ethyl acetate) gave the desired products (XV) as beige solids (50-70%).
EXAMPLE 7 Preparation of Sulfonamides Sulfonamides XVIII) were prepared using the chemistry outlined in Scheme 10. Intermediate (XVII) were generated by coupling an aminopyridine (II or IV) with an activated form of (XVI). Subsequent coupling of the sulfonyl group with an amine generated the desired sulfonamides (XVIII) Scheme 0 0 Y X C102S OH N IN CCI0S XI C XVII H
H
CIOs0 R 2 R, NO 2
S
XVII XVIII 7.1 General Experimental for Scheme Oxalyl chloride (175 gtL, 2 mmol) was added dropwise to a stirring solution of (XVI) (440 mg, 2 mmol) and DMF (20 pL, cat) in THF (8 mL) at 0 OC. After WO 02/062295 PCT/US02/03061 addition was complete the reaction was allowed to warm to RT. After 30 min the reaction was cooled back to 0 oC whereupon a solution of (II or IV) (1.9 mmol) and N, Ndiisopropylethylamine (700 pL, 4 mmol) in THF (2.3 mL) was added. After stirring for min at RT, this solution of (XVII) (0.18 M) was used directly without further manipulation.
A solution of (XVII) (5.5 mL, 1 mmol) in THF was added to a stirring solution of amine (1 mmol) and N, N-diisopropylethylamine (3 mmol) in THF (2 mL) at RT. After lh, water (10 mL) and ethyl acetate (10 mL) were added. The organic layer was separated, washed with water (5 mL), aqueous IN NaOH (5 mL), aqueous IN HCI mL) and then dried (Na 2
SO
4 The solvent was removed under reduced pressure and the residue purified by column chromatography (hexanes/ethyl acetate; The products were obtained as white solids Compounds prepared via this procedure include, compounds 36 and EXAMPLE 8 Example 8 sets forth the results of the characterization of representative compounds prepared by the methods of Examples 1 through 7. The compounds were characterized using a combination of melting point, 'H NMR and mass spectrometry.
The results of the characterization are presented below. The structures for the compounds set forth below are provided in FIG. 1.
3.4-Dichloro-N-pvridin-3-vl-benzamide mp 165-166 'H NMR (300 MHz, DMSO-d 6 6 10.58 (1H, brs), 8.87 (1H, d, J 2.3 Hz), 8.30 (1H, dd, J= 4.7, 1.4 Hz), 8.19 (1H, d, J= 1.9Hz), 8.16-8.12 (IH, 7.91 (1H, dd, J= 8.4, 2.1Hz), 7.80 (1H, d, J= and 7.38 (1H, dd, J= 8.4, 4.7Hz); MS (ESI) m/z: 266.9 3.4-Dichloro-N-(6-chloro-pyridin-3-vl-benzamide mp 188-189 OC;tH NMR (300 MHz, CDCl 3 8 7.36 (lH,d, J 8.7 Hz), 7.58 (1H, d, J 9.3 Hz), 7.70 (1H, dd, J= 9.4, Hz), 7.90 (1H, brs), 7.96 (1H, d, J= 1.9 Hz), 8.24 (1H, dd, J= 8.7, 2.8 Hz) and 8.48 (1H, d, J=2.8 Hz); 1 C NMR (75 MHz, DMSO-d 6 6 124.7, 128.6, 130.2, 131.4, 131.9, 134.8, 135.4, 142.0, 144.9 and 164.0; MS (ESI) m/z: 301.1 N-(6-Chloro-pvridin-3-vl)-3-trifluoromethyl-benzamide mp 139-140 'H NMR (300 MHz, CDC1 3 8 7.35 (1H, d, J 8.7 Hz), 7.63 (1H, t, J 7.8 Hz), 7.82 (1H, d, J= 7.8 Hz), 8.06 (1H, d, J= 7.8 Hz), 8.11 (1H, brs), 8.25 (1H, dd, J 8.7, 2.9 Hz), 8.36 (1H, s) and 8.50 (1H, d, J= 2.6 Hz); "F NMR (282 MHz, CDC1 3 6 -63.6; 'C NMR (75 MHz, DMSO-d 6 8 124.2, 124.2, 124.6, 129.0, 129.7, 130.6, 130.9, 133.7, 134.6, 141.2 and 164.8; MS (ESI) m/z: 301.2 WO 02/062295 WO 02/62295PCT/US02/03061 N-(6-Chloro-pyridin-3-yl)-3,4-difluoro-benzamide mp; 164 'H NNiR (300 MHz, DMSJ-d 6 8 7.51 (1H, d, J =8.7 Hz), 7.59-7.68 (1H, in), 7.84-7.88 (1H, 8.03 (1H, ddd, J= 11.3, 7.9, 2.1 Hz), 8.19-8.23 (1H, mn), 8.25 (1H, d, J= 2.0 Hz) and 10.64 (1H, s); '9 M (282 MHz, DMSO-d 6 85-112.9 -115.2 13 C NMR (75 MHz, DMS0-
Q
6 6 117.9 (dd, J= 18.3, 49.8 Hz), 124.7, 125.7 (dd, J= 3.4, 6.9 Hz), 131.5 131.9, 135.3, 142.1,145.0, 149.3 (dd, J= 14.7, 201.0 Hz), 152.8 (dd, J= 12.6, 205.0 Hz), 164.6; MS (ESI) m/z: 269.1 3-Chloro-N-(6-chloro-pvridin-3-yl)-benzainide mp 153-154 'H INMR (300 MHz, DMSO-d 6 5 10.67 (1H, brs), 8.74 (1H, d, J 2.6 Hz), 8.22 (1H, dd, J= 8.7, 2.8 Hz), 7.99 (1H, d, J= 1.7 Hz), 7.90 (1H, d, J= 7.8 Hz), 7.69 (1H, d, 1= 7.1 Hz), 7.58 (1H, t, J= 7.8 Hz) and 7.51 (1H, d, J= 8.7 Hz); MS (ESI) 267.0 BiphenvLI-4-carboxylic acid (6-chloro-pyiidin-3-yl)-amide mp 227-229 00; 'H N"MR (300 MHz, DMSO-d 6 8 10.62 (IH, bis), 8.81 (IH, d, J 2.3 Hz), 8.26 (1H, dd, 1= 8.7, 2.4 Hz), 8.06 (2H, d, J= 8.2 Hz), 7.85 (2H, d, 1= 8.2 Hz), 7.75 (2H, d, J= 7.5 Hz), 7.53- 7.47 (3H, m) and 7.42 (1H, q, 1= 7.1 Hz); MS (ESI) m/z: 309.2 6-Chloro-N-(6-chloro-pyridin-3-yl)-nicotinamide mp 228 lH NMR (300 MVIIz, DMSO-d6 5 8 10.81 (1H, brs), 8.92 (1H, d, J 2.3 Hz), 8.73 (1H, d, J= 2.6Hz), 8.32 (1H, dd, 3= 8.4, 2.4 Hz), 8.19 (1H, dd, J= 8.7, 2.8 Hz), 7.71 (1H, d, J= 8.4 Hz) and 7.52 (1H1, d, J= 8.7 Hz); MS (ESI) m/z: 268.1 3,4-Difluoro-N-(6-methyl-vridin-3-yl)-benzainide 'H NN/R (300 MiEz, DMSOd 6 8 10.42 (LH, brs), 8.73 (1H, d, J1 2.3 Hz), 8.04-7.97 (2H, mn), 7.86-7.82 (1H, mn), 7.90 (1H, dt, J= 10.4, 8.4 Hz), 7.23 (1H, d, J= 8.5 Hz) and 2.42 (3h, 9 F NMR (282 MHz, DMSO-1 6 5 -133.1 to -133.3 (1H, mn) and -137.1 (1H, q, J= 10.7Hz); MS (ESI) rn/z: 249.0 [M+HII+.
N-(6-Chloro-pvridin-3-yl)-3-fluoro-benzainide mp 160 'HNMR (300 MHz, DMSO-d 6 8 10.63 (1H, brs), 8.75 (1H, d, J1 2.8 Hz), 8.20 (1H, dd, J= 8.7, 2.8 Hz), 7.79 (1H1, d, J= 7.8 Hz), 7.75 (1H, d, J= 11.1 Hz), 7.62-7.55 (IH, m) and 7.53-7.43 (2H, in); "F NMIR (282 MHz, DMSO-d,) 85-112.0 8.5Hz); MS (ESI) m/z: 25 1.0 N-(6-Ghloro-pvridin-3-yl)-3-(trifluorornethyl)-benzamide mp 169-170 'H NMR (300 MHz, DMSO-d 6 8 10.81 (1H, brs), 8.75 (1H, di, J 2.8 Hz), 8.22 (1H, dd, J= 8.7, 2.8 Hz), 8.13 (1IH, d, J= 8.2 Hz), 7.91 (211, d, J= 8.4 Hz) and 7.52 (2H, d, 1= 8.7 Hz); 19F NMIR (282 MHz, DMSO-d 6 8 -61.4 MS (ESI) m/z: 301.2 N-(6-Chloro:pyridin-3-yl)-4-fluoro-3-trifluoromethvl-benzamide mp 149-150 00; 'H1 NMR (300 MHz, DMSO-d 6 8 10.63 (1H1, brs), 8.76 (1H, di, J 2.6 Hz), 8.22 (111, dd, J= 8.7, 2.8 Hz), 7.98 (2H1, d, 1= 8.7 Hz), 7.63 (1H, d, J= 8.7 Hz) and 7.52 (111, d, J= 8.7 Hz); MS (ESI) m/z: 319.1 N-(6-Chiloro-nvridin-3-vl)-3-fluoro-4-trifluoromehvl-benzainide mp 182'00; 'H NMR (300 MHz, DMSO-d 6 6 10.78 (1H1, brs), 8.75 (1H1, d, J1 2.3 Hz), 8.37-8.32 (2H, mi), 8.22 (111, dd, J= 8.7, 2.6 Hz), 7.73 (111, mn) and 7.54 (111, d, J= 8.7 Hz); 9 F NNJR (282 MVIHz, DMSO-d 6 8 -60.1 (3F, in), 110.7 MS (ESI) m/z: 319.1 WO 02/062295 WO 02/62295PCT/US02/03061 N-(6-Chloro-pyridin-3-vl)-4-fluoro-benzamide mp 163-164 I H NNvR (3 00 MHz, DMSO-d 6 8 10.58 (1H, brs), 8.76 (1H, d, J 2.6 Hz), 8.22 (1H, dd, J= 8.7, 2.6 Hz), 8.04 (IH, d, J= 8.7 Hz), 8.02 (1H, d, J= 8.7 Hz), 7.51 (1H, d, J= 8.7 Hz) and 7.39 (2H, t, J= 8.8 Hz); 9 F NN4R (282 MHz, DMSO-d 6 86-107.7 MS (ESI) m/z: 319.0 N-(6-Chloro-pvridin-3-yl)-4-chloro-benzamide mp 197-199 'H NMIR (300 MHz, DMSO-d 6 8 10.63 (1IH, brs), 8.76 (111, d, J 2.6 Hz), 8.22 (111, dd, J= 8.7, 2.8 Hz), 7.98 (2H, d, J= 8.7 Hz), 7.63 (1H, d, J= 8.7 Hz) and 7.52 (111, d, J= 8.7 Hz); MS (ESI) m/z: 267.0 5,6-Difluoro-N-(6-fluoro-pyridin-3-yl)-nicotinamnide mp 13 5-137 NMR (300 MHz, DMSO-d 6 8 10.58 (1H, brs), 8.52 (1H, 8.29-8.23 (1H, in), 8.03-7.97 (1H, in), 7.87-7.82 (1H, mn), 7.65-7.56 (1H, mn) and 7.19 (1H,dd, J= 8.9, 3.1 Hz); 9 F NMR (282 MHz, DMSO-1 6 8 -73.6 (1F, d, J= 6.5Hz), -132.9 (1H, q, J= 10.7Hz) and -137.1 (1H, q, J= 10.711z); MS (ESI) m/z: 253.0 N-(6-Chloro-pvridin-3-yl)-3-methyl-4-nritro-benzainide mp 192-193 0 C; 'H1 NIVMR (300 MHz, DMSO-d 6 8 10.81 (1H, brs), 8.74 (1H, d, J 2.6 Hz), 8.21 (1H, dd, J= 8.7, 2.8 Hz), 8.09 (1H, d, J= 8.4 Hz), 8.02 (1H, 7.94 (1H, dd, J= 8.5, 1.6 Hz), 7.52 (111, d, J= 8.7H-z) and 2.58 (3H, MS (EST) 292.2 LM+HV'.
5-Fluoro-1H-indole-2-carboxylic acid (6-chloro-pidin-3-yl)-amide mp 290 'H NMIR (300 MHz, DMS0-l 6 8 11.92 brs), 10.58 (1H,brs), 8.79 (1H, 8.25 (1H1, dd, 1= 8.7 and 2.4 Hz), 7.53-7.41 (4H, m) and 7.10 (1H1, t, J= 7.1 Hz); 9 F NMR (282 MHz, DMS0-cl 6 8 -123.0 MS (ESI) m/z: 290.0 5-Chloro-lH-indole-2-carboxvlic acid (6-chloro-p3ridin-3-vl)-amide mp 296' 0 C; I NNVR (300 MHz, DMSO-d 6 8 12.03 (1H, brs), 10.62 (1H, brs), 8.80 (1H, d, J= 2.6 Hz), 8.24 (111, dd, J= 8.7 and 2.6 Hz), 7.79 (111, s) 7.52 (1H, d, J= 8.7 Hz), 7.42 (1H, d, J= 8.9 Hz), 7.41 (1H, s) and 7.23 (1H, dcl, J= 8.7, 1.9 Hz); MS (ESI) ink: 306.0 5-Chloro-benzofuran-2-carboxylic acid (6-chloro-pyridin-3-fl-aiie (19) m22-223 0 C; I H NMR (3 00 MvHz, DMSO-d 6 8 11. 00 (1 H, brs), 8.7 8 (1 H, d, J 2.6 Hz), 8.24 (1 H, dcl, J= 8.7, 2.8 Hz), 7.90 (lIH, d, J= 2.1 Hz), 7.76 (1H, 7.73 (1H, d, J= 8.9 Hz) and 7.51 (2H, d, J= 8.9 Hz); MS (ESI) m/z: 306.9 5-Chloro-thioliene-2-carboxylic acid (6-chloro-pvridin-3-vl)-amide mp 215-216 c; 'H NN'R (300 MHz, DMSO-1 6 8 10.61 (111, brs), 8.68 (1H, d, J 2.8 Hz), 8.14 (1H, dcl, J= 8.7, 2.6 Hz), 7.88 (1H, d, J= 4.2 Hz), 7.50 (1H, d, J= 8.5 Hz) and 7.27 (1H, d, J= 4.0 Hz); 1 3 C NMvR (75 MIHz, DMSO-d 6 8 159.7, 144.8, 142.0, 138.5, 135.3, 135.2, 131.5, 130.4, 129.0 and 124.7.0; MS (ESI) tn/z: 273.0 5-Chloro-furan-2-carboxylic acid (6-chloro-pvridin-3-yl)-ainide mp 143-144' 0 C; 'H NMVR (300 MHz, DMSO-d 6 8 10.59 (1H, brs), 8.72 (1H, d, J 2.4 Hz), 8.18 (1H, dd, J= 8.7, 2.6 Hz), 7.50 (11H, d, J= 8.7 Hz), 7.43 (1H, d, J= 3.5 Hz) and 6.76 (111, d, J= 3.7 Hz); MS (EST) rn/z: 257.1 [M+H]V.
WO 02/062295 WO 02/62295PCT/US02/03061 4,5-Dichloro-isoffiiazole-3-carboxvlic acid (6-chloro-pvridin-3-yl)-arnide mp 199- 201 OC; 'H NMvR (300 MHz, DMSO-d 6 5 11.16 (1H, brs), 8.75 (1H, d, J =2.6 Hz), 8.20 (1H, dd, J= 8.7, 2.8 Hz) and 7.51 (1H, d, J= 8.7 Hz); MS (ESI) rn/z: 3O7.9[M+H]+.
3-Methyl-IH-indole-2-carboxylic acid (6-chloro-pyridin-3-yl)-amnide np 184-185 0 C; 'H NMR (300 M]Hz, DMSO-d 6 6 10.64 (11H, brs), 8.75 (1H, d, J =2.6 Hz), 8.21 (1H, dd, J= 8.7, 2.8 Hz), 7.68 (1H1, d, J= 8.0 Hz), 7.54 (1H, d, J= 8.5 Hz), 7.50 (2H, d, J= 8.9 Hz), 7.34 (1H1, 7.31 (111, t, J= 7.1 Hz), 7.13 (1H, t, J= 7.1 Hz) and 3.98 (3H1, MS (ESI) m/z: 286 .1 IIM+H]+.
-Ethyl-1IH-indole-2-carbo2vLic acid (6-chloro-ipvridin-3-vl)-amnide mp 270 0 C; 111 NMR (300 MHz, DMSO-d 6 6 11.67 (1H, brs), 10.49 (1H, brs), 8.80 (111, 8.25 (11, dd, J= 8.7 and 2.8 Hz), 7.51 (111, d, J= 8.7 Hz), 7.46 (1H, 7.36 (1H, d, J= 8.7 Hz), 7.35 (1 H, 7. 10 (1 H, d, J= 8.5 Hz), 2.66 (2H1, q, J= 7.7 Hz) and 1.21 (11H, t, 7.7 Hz); 1 3
C
NMR (75 Mffz, DMSO0-d 6 6 160.6, 144.3, 141.7, 13 6.2, 13 6.0, 131.1, 127.6, 125.6, 124.7, 120.4, 112.8, 104.8, 28.8 and 16.8; MS (ESI) rn/z: 300.2 IiM+H]+.
N-(6-Chloro-pyridin-3-vl)-benzamide mp 163-164 0 C; 'H NIVR (3 00 MHz, DMSOd 6 8 10.59 (1H1, brs), 8.74 (111, d, J =2.6 Hz), 8.21 (1H1, dd, J= 8.7, 2.8 Hz), 7.94 (1H1, s), 7.92 (1H, d, f= 1.6 Hz), 7.59 (111, d, J= 7.1 Hz) and 7.55-7.49 (3H, in); MS (ESI) m/z: 233.0 1H-Indole-2-carboxvlic acid (6-chloro-pvridin-3-vl)-amide mp 260-263 'H NMR (300 NMz, DMSO-d 6 6 11.85 (111, brs), 10.62 (1H, brs), 8.78 (1H, d, J 2.6 Hz), 8.23 (1H1, dd, J= 8.7, 2.8 Hz), 7.66 (1H, d, J= 8.0 Hz), 7.51 (111, d, J= 8.5 Hz), 7.45 (1H, d, J= 8.4 Hz), 7.41-7.40 (1H, in), 7.23 (1H, t, J= 7.1 Hz) and 7.06 (1H, d, J= 7.3 Hz); MS (ESI) m/z: 272.0 BenzoUblthiophene-2-carboxylic acid (6-chloro-pyridin-3-yl)-ainide (7:mp 226-227 'C; 'H NMR (300 MHz, DMSO-d 6 6 10.86 (111, brs), 8.75 (111, d, J= 2.6 Hz), 8.34 (iH, s), 8.21 (1H, dd, J= 8.7, 2.8 Hz), 8.06-7.99 (211, m) and 7.54-7.48 (311, in); MS (ESI) m/z: 289.1 5-Fluoro-1H-indole-2-carboxylic acid (6-mgthyl-pvridin-3-yl)-amide mp 303-304 0 C; 'H NMR (300 Mi~z, DMSO-d 6 6 11.88 (1H1, brs), 10.36 (1H, brs), 9.05 (1H1, d, J 1.9 Hz), 8.06 (111, dt, J= 8.4, 2.1 Hz), 7.45 (211, in), 7.39 (1H, 7.24 (111, d, J= 8.3 Hz), 7.08 (1H1, dt, J= 9.2, 2.4 Hz) and 2.43 (311, 1 9 F NrvR (282 MIHz, DMSO-d 6 6 -123.2; MS (ESI) 270.2 3,4-Difluoro-N-(6-trifluoromethvl-pyridin-3-vl)-benzamnide mp 1 75-176 111 NM (300 MHz, DMSO-d 6 6 10.84 (1H1, brs), 9.05 (1H, d, J 1.8 Hz), 8.45 (1H, dd, J= 1.9 Hz), 8.07 (11, ddd, J= 9.9, 7.6, 2.1 Hz), 7.93(111, d, J= 8.5 Hz) and 7.89-7.86 in); "F NMLR (282 MIHz, DMS0-l 6 6 -65.7 (3F, 132.5 (1iF, in) and -137.0 (IF, in); MS (ESI) m/z: 303.1 N-(6-Chloro-pyridin-3-yl)-3-fluoro-4-pvrrolidin-1 -yl-benzamide (391.: mp 215-216'CG; 111 NAMI (300 MHz, DMSO-1 6 6 10.23 (1H, brs), 8.71 (1H, d, J =2.6 Hz), 8.18 (1H1, dcl, J= 8.7, 2.4 Hz), 7.70 (111, 7.65 (111, d, J= 3.1 Hz), 7.46 (1H1, d, J= 8.9 Hz), 6.73 (111, t, 1= WO 02/062295 PCT/US02/03061 Hz), 3.42-3.40 (4H, m) and 1.91-1.88 (4H, mi);' 9 F NMR (282 MHz, DMSO-d 6 6 129.0 MS (ESI) m/z: 320.2 N-(6-Chloro-pyridin-3-vl)-3-fluoro-4-morpholin-4-vl-benzamide mp 228-229 OC; 'H NMR (300 MHz, DMSO-d 6 6 10.41 (1H, 8.75 (1H, d, J= 2.6 Hz), 8.20 (1H, dd, J= 8.7 and 2.6 Hz), 7.79 (1H, 7.74 (11, d, J= 7.7 Hz), 7.50 (1H, d, J= 8.7 Hz), 7.13 (1H, t, J= 9.2 Hz), 3.74 (41H1, m) and 3.12 (4H, 3 C NMR (75 MHz, DMSO-d 6 164.8, 155.7, 152.5, 144.4, 143.3 J= 7 Hz), 142.0, 135.9, 131.4, 127.3 J= 7Hz), 125.5, 124.6, 118.7 J= 3 Hz), 66.5 and 50.4 J= 5 Hz); MS (ESI),m/z: 336.2 N-(6-Chloro-pyvridin-3-vyl)-3-fluoro-4-imiidazol-1-vl-benzamide mp 215-218 oC; 'H NMR (300 MHz, DMSO-d 6 6 10.71 8.78 (1H, 8.24 (1H, dt, J= 8.7, 1.6 Hz), 8.17 (1H, 8.07 (1H, dd, J= 12.0, 1.6 Hz), 7.96 (1H, dd, J= 8.4, 1.7 Hz), 7.88 (1H, t, J= 8.2 Hz), 7.69 (1H, 7.53 (1H, d, J= 8.7 Hz) and 7.17 (1H, 9 F NMR (282 MHz, DMSO-d 6 6 -123.1 J= 8.8 Hz); MS (ESI) m/z: 317.1 N-(6-Chlorop ridin-3-v-3-fluoro-4-(pidin-2-lmh-ino-benzamide mp 210-211 'H NMR (300 MHz, DMSO-d 6 6 10.20 (1H, brs), 8.69 (1H, d, J 2.6 Hz), 8.50 (1H, d, J= 4.7 Hz), 8.15 (1H, dd, J= 8.7, 2.8 Hz), 7.75-7.64 (2H, 7.45 (1H, d, J= 8.7 Hz), 7.31 (1H, d, J= 8.0 Hz), 7.25 (IH, dd, J= 6.4, 5.1Hz), 6.63 (1H, t, J= 8.7 Hz) and 4.49 (2H, d, J= 5.9 Hz); 9 F NMR (282 MHz, DMSO-d 6 6 -134.3 MS (ESI) m/z: 357.0 N-(6-Chloro-Dridin-3-yl)-4-dimetLhylamino-3-fluoro-benzamide mp 170-171 C; 'H NMR (300 MHz, DMSO-d 6 6 10.32 (1H, brs), 8.76 (1H, 8.21 (1H, d, J= 8.7 Hz), 7.75 (1H, 7.71 (1H, d, J= 5.8 Hz), 7.59 (1H, d, J= 8.7 Hz), 7.02 (1H, t, J= 9.2 Hz) and 2.92 (61H1, 9 F NMR (282 MHz, DMSO-d 6 6 -122.6 J= 10.7 Hz); MS (ESI) m/z: 294.2 Pridine-2-carboxylic acid [4-(6-chloro-pridin-3-vlcarbamovl-phenv11 -amide mp 258-260 oC; 'H NMR (300 MHz, DMSO-d 6 6 10.92 (11, 10.50 (1H, 8.78 (1H, d, J= 2.6 Hz), 8.75 (1H, d, J= 4.7 Hz), 8.24 (1H, dd, J= 8.7 and 2.6 Hz), 8.17 (1H, d, J= 7.8 Hz), 8.09 (2H, d, J= 7.8 Hz), 8.08 (1H, 7.98 (2H, d, J= 8.7 Hz), 7.70 (1H, dd, J= 6.4, 5.0 Hz) and 7.50 (1H, d, J= 8.7 Hz); 3 C NMR (75 MHz, DMSO-d 6 6 165.8, 163.5, 150.0, 149.0, 144.4, 142.3, 141.9, 138.8, 136.1, 131.4, 129.4, 129.2, 127.7, 124.6, 123.1 and 120.1; MS (ESI) m/z: 353.2 N-(6-Chloro-pvridin-3-vl)-4-(morpholine-4-sulfonv1)-benzamide 'H NMR (300 MHz, DMSO-d 6 6 10.88 (1H, 8.78 (1H, d, J= 2.6 Hz), 8.24 (1H, dd, J= 8.7 and 2.6 Hz), 8.19 (21H1, d, J= 8.5 Hz), 7.90 (2H, d, J= 8.5 Hz), 7.50 (1H, d, J= 8.7 Hz), 3.62 (4H, m) and 2.90 (4H, 3 C NMR (75 MHz, DMSO-ds) 6 165.3, 144.9, 142.1, 138.8, 137.9, 135.7, 131.5, 129.4, 128.3, 124.7, 65.8 and 46.8; MS (ESI) m/z: 382.1 N-(6-Chloro-pyridin-3-yl)-3-fluoro-4-nitro-benzainide mp 205 OC; 'H NMR (300 MHz, DMSO-d 6 6 10.87 (1H, brs), 8.69 (1H, d, J 2.4 Hz), 8.32 (1H, t, J= 7.8 Hz), 8.22 (1H, dd, J= 8.7, 2.6 Hz), 8.10(11H1, dd, J= 11.8, 1.6 Hz), 7.96 (1H, d, J= 8.5 Hz) and 7.53 (1H, d, J= 8.7 Hz); MS (ESI) m/z: 294.0 WO 02/062295 PCT/US02/03061 N-(6-Chloro-pyridin-3-vl)-4-hvdroxvamino-benzamide dec 200 0 C; 'H NMR (300 MHz, DMSO-d 6 8 10.24 (1H, brs), 8.86 (1H, 8.75 (1H, d, J 2.4 Hz), 8.61 (1H, s), 8.22 (1H, dd, 1= 8.7, 2.8 Hz), 7.83 (2H, d, J= 8.5 Hz) and 7.47 (1H, d, J= 8.7 Hz); MS (ESI) m/z: 261.9 N-(6-Chloro-pvridin-3-l)-3-fluoro-4-_hydroxamino-benzamnide 1 H NMR (300 MHz, DMSO-d 6 8 10.16 (1H, brs), 8.74 (1H, d, J= 2.6 Hz), 8.19 (1H, dt, J 8.7, 2.8 Hz), 7.66 (1H, dd, J= 12.7, 1.7 Hz), 7.60 (1H, d, J= 8.4 Hz), 7.47 (1H, d, J= 8.7 Hz), 6.80 (1H, t, J= 8.9 Hz) and 5.93 (211, brs); MS (ESI) m/z: 280.0 N-(6-Chloro-pvridin-3-yl)-4-methvlsulfamovl-benzamide mp 186-189 0 C; 1 H NMR (300 MHz, DMSO-d 6 8 10.78 (1H, brs), 8.77 (In, d, 2= 2.3 Hz), 8.23 (1H, dt, 2= 8.7, 2.8 Hz), 8.13 (2H, d, 2= 8.4 Hz), 7.99 (IH, d, 2= 8.2 Hz), 7.63 (1H, q, 2= 5.2 Hz), 7.53 (1H, d, J= 8.7 Hz) and 2.43 (3H, MS (ESI) nz/z: 326.2 4-Amino-N-(6-chloro-vridin-3 -vl)-3 -fluoro-benzamide mp 193 0 C; 'H NMR (3 00 MHz, DMSO-d 6 8 10.16 (1H, brs), 8.73 (1 H, d, J 2.6 Hz), 8.20 (11H, dd, J= 8.7, 2.8 Hz), 7.66 (in, dd, J= 12.7, 1.7 Hz), 7.59 (1H, dd, 2= 8.4, 1.9 Hz), 7.46 (1H, d, J= 8.7 Hz), 6.80 (1H, t, J= 8.7 Hz) and 5.94 (2H, MS (ESI) n/z: 266.0 N-(6-Chloropyridin-3-vl)-4-nitro-benzamide mp 193 0 C; 'H NMR (300 MHz, DMSO-d 6 5 10.88 (1H, brs), 8.77 (1H, d, 2= 2.3 Hz), 8.39 (11, 8.36 (1H, 8.23 (IH, dt, 2= 8.7, 2.8 Hz), 8.20 (11H, 8.17 (11H, 7.63 (1H, q, 2= 5.2 Hz) and 7.54 (11H, d, J= 8.7 Hz): MS (ESI) m/z: 275.9 N-(6-Chloro-pvridin-3-vl-3-chloro-4-fluoro-benzamide mp 173 'H NMR (300 MHz, DMSO-d 6 6 10.65 (1H, brs), 8.74 (11, 8.23-8.18 (2H, 8.01-7.97 (1H, i), 7.61 (1H, t, 2= 9.1 Hz) and 7.52 (1H, d, 2= 8.7 Hz): MS (ESI) mn/z: 285.0 WO 02/062295 PCT/US02/03061 EXAMPLE 9 Example 9 sets forth representative methods for elaborating the ring structures of the substituted benzanilides of the invention 9.1 Synthesis of Amino-Modified Pvridvl-Benzanilides Scheme 11 for X"=C1 0 x" pyridine, THF N S, N for X"OH H SHN (CICO) 2 DMF, THF F V then pyridine XIX XX V XXI X O I RR'NH, DMSO, x N
H
H 110-120 F 65-85% v XXI v XXII 9. la General Method for XXT When X =Cl An aniline XX (1.55 mmol) was dissolved in dry THF (5 mL) and treated with pyridine (0.075 mL, 0.6 equivalents) followed by a 4-fluorobenzoyl chloride XIX (1.1 equivalents; slow addition; a precipitate forms immediately). After 2 h, saturated NaHCO 3 solution (15 mL) was added. After 30 min of vigorous stirring, the mixture was diluted with EtOAc (15 mL). The organic layer was washed with water (1x5 mL), dried (Na 2
SO
4 and filtered. Removal of the solvent provided the products XXI as solids, in high yields and with high purity (typically >95% by LC/MS).
9.1b General Method for XXI When X OH A 4-fluorobenzoic acid XIX (1.7 mmol, 1.1 equivalents) was dwassolved in dry THF (5 mL) and treated with dry DMF (3 drops) followed by oxalyl chloride (1.3 equivalents; slow addition; gas evolution; mild exotherm). After 1 h, an aniline XX (1.55 mmol) was added (a precipitate forms immediately) and 2 h later, saturated NaHCO 3 solution (15 mL) was added. After 30 min of vigorous stirring, the mixture was diluted with EtOAc (15 mL). The organic layer was washed with water (1 x 5 mL) and silica gel (2 g) was added. Removal of the solvent provided a solid, which was applied to WO 02/062295 PCT/US02/03061 a column of silica gel x and eluted with EtOAc/hexanes. The products XXI were thus obtained as solids in good yields and with high purity by LC/MS).
9. 1c General Method for Preparing Compounds ofMotif XXI Compound XXI (0.10 mmol) and RRNH (3 equivalents) were heated at 110-120 °C in dry DMSO (0.3 mL) for 20 h. The mixture was brought to room temperature and diluted with water (0.3 mL), saturated NaHCO 3 solution (0.3 mL), EtOAc (4 mL) and brine (2 mL). The organic layer was washed with brine (2x1 mL) and silica gel (400 mg) was added. Removal of the solvent provided a solid, which was applied to a column of silica gel x and eluted with EtOAc/hexanes. The products XXI were obtained as solids in good yields and with high purity by LC/MS).
9.2 Synthesis ofAryl-Modified Benzanilides Scheme 12 x .o ArOH, K 2
CO
3 X N H DMSO, 110-120 oC, F 20 h, 70-95% ArO V XXI V XXIII 9.2a General Method for Preparing XXIII Compound XXI (0.15 mmol), a phenol (1.2 equivalents) and K 2 CO3 were heated at 110-120 C in dry DMSO (0.5 mL) for 20 h. The mixture was brought to room temperature and diluted with water (0.7 mL), EtOAc (3 mL) and brine (1 mL). The organic layer was washed with brine (2x0.5 mL) silica gel (400 mg) was added.
Removal of the solvent provided a solid, which was applied to a column of silica gel x and eluted with EtOAc/hexanes. The products XXIII were obtained as solids in good yields and with high purity by LC/MS).
WO 02/062295 PCT/US02/03061 9.3 Preparation of Thioether-Modified Benzanilides Scheme 13 X O N RSH, Cs 2
CO
3 RN I X 'N.N HDMSO, rt, 4 h 2 d, H F 20-90% RS v
XXIV
9.3a General Method for Preparing Compounds ofMotifXXIV A 0.2-0.4 M suspension of XXI in DMSO, Cs 2
CO
3 (1-2 eq.) and alkyl or arylsulfide (1.2-3 eq.) was agitated at rt or up to 75 °C for 4 h to 2 days until the reaction was judged to have gone to completion (TLC or LCMS). The reaction mixture was partitioned between ethyl acetate and water. The aqueous layer was washed with water, 1 N NaOH solution water, and brine, dried (Na 2 S04) and concentrated.
Chromatography on SiO 2 trituration or recrystallization afforded products XXIV as solids in good yields and with high purities by LC/MS).
9.4 Preparation of Oxidized Thioether-Modified Benzanilides Scheme 14
X
N' N MCPBA, CH 2 C1 2
X
H rt, 12 h, 20-90% 1 H RS RSO2 V XXIV v Xx 9.4a General Method forPreparing Compounds ofMotifXXV A 0.1-0.25 M solution of sulfide XXIV in 2:1 CH 2 CL/THF was treated with mCPBA (2 eq.) and agitated at rt overnight. The reaction mixture was concentrated in vacuo and the residue was partitioned between ethyl acetate and water. The organic layer was washed with 1 N NaOH solution water, and brine, dried (Na 2
SO
4 and concentrated. Chromatography on SiO 2 trituration or recrystallization afforded pure product XXV as solids in good yields and with high purity by LC/MS).
WO 02/062295 WO 02/62295PCT/US02/03061 EXAMPLE Example 10 sets forth the results of the characterization of representative compounds prepared by the methods of Examples 8 and 9. The compounds were characterized using a combination of malting point, 1H INMR and mass spectrometry.
The results of the characterization are presented below. The structures for the compounds set forth below are provided in FIG. 1.
N-(6-Chloropyridin-3-yl)-3-fluoro-4-(2-phenethylamino)-benzamide (156): 'H NM'R (CDCl 3
CD
3 OD) 8 8.40 J 2.4 Hz, 1H), 8.32 (dd, J 2.6, 8.7 Hz, 1H), 7.54-7.61 (in, 2H1), 7.19-7.34 (in, 6H), 6.70 J 8.0 Hz, 1H), 3.47 J 7.0 Hz, 2H1), 2.94 J Hz, 211).
N-(6-Chloropvridin-3-vl)-3-fluoro-4-(2-(2-pvridyl)ethvlainino)-benzainide (122): 1 NMR (CDCl 3
CD
3 OD) 8 8.51 (dd, J 1.6, 5.7 Hz, 1H1), 8.41 J 2.4 Hz, 1H1), 8.31 (dd, J 2.8, 8.7 Hz, 1H), 7.53-7.66 (in, 3H), 7.28 J 9.0 Hz, 1H), 7.15-7.19 (mn, 2H), 6.72 IJ 8.5 Hz, 1 3.60 J 7. 0 Hz, 2H), 3. 10 J 6.9 Hz, 2H).
N-(6-Chloropvridin-3-vl)-3-fluoro-4-(2-(3-pyridvl)eth lamino)-benzainide (123): 1H1 NN4R (CDCl 3
CD
3 OD) 8 8.39-8.41 (in, 3H1), 8.30 (dd, J 2.6, 8.7 Hz, I 7.5.3-7.63 (in, 311), 7.23-7.28 (mn, 2H), 6.66 J 8.2 Hz, 1H1), 3.48 J1 7.0 Hz, 2H1), 2.93 J Hz, 2H).
N-(6-Chloropvridin-3-vl)-3-fluoro-4-(2-(4-pyridyl)ethylainino)-benzainide (124): 'H NMR (CDCl,, CD 3 OD) 6 8.43 J 2.1 Hz, 2H), 8.39 J 2.5 Hz, IH), 8.32 (dd, I 2.8, 8.7 Hz, 111), 7.56-7.63 (in, 2H), 7.27 J 9 Hz, 1H), 7.15 J 6.0 Hz, 111), 6.66 J =8.4 Hz, 111), 3.50 J 7.2 Hz, 211), 2.93 J 7.0 Hz, 2H).
N-(6-Chlorovrdin-3-yl)-4-(3.4-difluorophenoxv)-3-fluorobenzamide (210) 'H NMR (CDCl 3
CD
3 OD) 6 8.42 J 2.6 Hz, 1H1), 8.33 (dd, J 2.6, 4.5 Hz, 111), 7.80 (dd, J 2.1, 11.2 Hz, 1H), 7.70 (dd, J 1.0, 8.5 Hz, 1H), 7.30 J 8.7 Hz, 1H1), 7.00-7.17 (in, 211), 6.72-6.88 (in, 2H); MS(ESI): 379 (Mu).
WO 02/062295 PCT/US02/03061 N-(6-Chloropyridin-3-fl-3-fluoro-4-(fqran-2-yLmethlsulfanl)-benzamide (134): 'H NMR (DMSO-d 6 6 10.56 11), 8.76 1H), 8.22 J 8.7 Hz, 1H), 7.82-7.74 (m, 2H), 7.67 (dd, J 7.8, 8.0, 1H), 7.57 1H), 7.51 J 8.5 Hz, 1H), 6.38-6.32 2H), 4.42 2H).
N-(6-Chloro-pyridin-3-vl)-3-fluoro-4-phenethylsulfan-benzanide (135): 'H NMR (DMSO-d 6 6 10.55 1H), 8.77 1H), 8.22 J 8.5 Hz, 1H), 7.85-7.75 2H), 7.61 (dd, J 7.9, 7.9 Hz, 11), 7.51 J 8.7 Hz, 1H), 7.36-7.18 5 3.39-3.22 (m, 2H), 2.92 J 7.7 Hz, 211).
N-(6-Chloro-pvridin-3-yl)-3-fluoro-4-(2-phenvl-ethanesulfonvl)-benzamide (141): 'H NMR (DMSO-d 6 8 10.82 11), 8.76 11), 8.22 J 8.7 Hz, 1H), 8.05 J 10.7 Hz, 1H), 7.93 J 8.2 Hz, 1H), 7.81 (dd, J 7.1, 7.1 Hz, 1H), 7.62-7.52 3H), 7.19 J 8.2 Hz, 1H), 4.92 2H) N-(6-Chloro-tpidin-3-vl)-3-fluoro-4tphelmethanesulfoyl-benzamide (142): '1 NNR (DMSO-d 6 6 10.56 lH), 8.75 1H), 8.19 J 8.5, 1H), 7.76 J 9.4, 2H), 7.66- 7.34 4 7.18-7.06 2H), 4.36 211) N-(6-Chloro-vridin-3-vl)-3-fluoro-4-(thiphen-2-ylmethanesulfonyl)-benzamide (146): 'H NMR (DMSO-d 6 6 10.57 1H), 8.76 1H), 8.21 J 8.5 Hz, 1H), 7.84-7.58 (m, 4 7.52 J 8.3 Hz, 1H), 7.40 J 5.4 Hz, 1H), 7.07 111), 6.93 J 4.9 Hz, 1H), 4.64 2H).
N-(6-Chloro-in-(yl)-3-fluoro-4-(2 y147): 'H NMR (DMSO-d 6 6 10.56 IH), 8.76 J 2.3 Hz, 11), 8.51 J 4.5 Hz, 1H), 8.22 (dd, J 2.4, 8.7 Hz, 1H), 7.85-7.68 31), 7.63 (dd, J 7.6, 8.2 Hz, 1H), 7.51 J 8.7 Hz, 1H), 7.33 J 7.8 Hz, 1H), 7.25 (dd, J 5.6, 6.6, 1H), 3.47 J 7.3 Hz, 1H), 3.09 J 7.5 Hz, 2H).
N-(6-Chloro-pvridin-3yv1)-3-fluoro-4-2-:pridin-4-v-ethanesufoflY)-benzamide (155): 'H NMR (DMSO-d 6 6 10.55 1H), 8.76 IH), 8.47 J 5.2, 21), 8.22 J 8.7 WO 02/062295 PCT/US02/03061 Hz, 1H), 7.87-7.74 2H), 7.62 (dd, J 7.8, 8.2 Hz, 1H), 7.49 J 8.7, 1H), 7.30 J 5.3 Hz, 21), 3.40 J 7.3 Hz, 2H), 2.94 J 7.4 Hz, 21).
N-(6-Chloro-pyridin-3-vl)-3-fluoro-4-(thiohen-2-ylmethanesulfonyl)-benzamide (150): 'H NMR (DMSO-d 6 8 10.84 1H), 8.76 1H), 8.22 J 6.1 Hz, 1H), 8.04 J 10.6 Hz, 1H), 7.91 I 8.0 Hz, 1H), 7.83 J 7.7 Hz, 11), 7.58-7.46 2H), 6.95 21), 5.12 21).
N-(6-Chloro-pyridin-3-vl)-3-fluoro-4-(4-fluoro-phenvlmethanesulfonvl)-benzamide (151). 1H NMR (DMSO-d 6 6 10.82 11), 8.76 1H), 8.22 J 8.7 Hz, 1H), 7.90 J 8.2 Hz, 1H), 7.76 (dd, J 7.1, 7.9 Hz, 11), 7.55 J 8.7 Hz, 1H), 7.30-7.23 (m, 2H), 7.15 (dd, J 8.7, 8.9 Hz, 21), 4.85 2H) N-(6-Chloro-pvyrdin-3-yl)-3-fluoro-4-(pyidin-2-ylsulfa )-benzamide (157) 11 NMR (DMSO-d 6 8 10.71 111), 8.78 J 2.3 Hz, 1H), 8.37 J 4.0, 1H), 8.23 (dd, J 8.6 Hz, 1H), 7.95-7.67 4 7.53 J 8.7, 11), 7.28-7.16 2H).
4-(2-Chlorophenlsulfanvl)-N-(6-chloro- idin-3-v1)-3-fluoro-benzamide (158): 'H NMR (DMSO-d 6 6 10.65 11), 8.76 J 2.6 Hz, IH), 8.21 (dd, J 2.6, 8.7 Hz, 1H), 7.91 J 10.4 Hz, 11), 7.80 J 8.1 Hz, 1H), 7.62 J 7.4 Hz, 11), 7.52 J 8.7 Hz, 1H), 7.44-7.24 4 H).
N-(6-Chloro-pyridin-3-yl)-3-fluoro-4-(2-fluoro-phenvlsulfanl)-benzamide (159): 11 NMR (DMSO-d 6 6 10.61 11), 8.74 J 2.4 Hz, 1H), 8.20 (dd, J 2.6, 8.7, 1H), 7.87 J= 10.6, 1H), 7.72 J= 10.2 Hz, 1H), 7.60-7.45 3H), 7.40 (dd, I 8.7, Hz, 11), 7.30 (dd, J 7.6, 7.7 Hz, 1H), 7.18 (dd, J 7.8, 7.9 Hz, 1H) 4-tert-Butvlsulfanvl-N-(6-chloro-pyridin-3-l)-3-fluoro-benzarnide (160) 'H NMR (DMSO-d 6 6 10.69 1H), 8.78 1H), 8.23 J 6.1 Hz, 1H), 7.92-7.70 3H), 5.53 J 8.4 Hz, 11), 1.29 9 H).
4-Butvlsulfanyl-N-(6-chloro-vridin-3-yl)-3-fluoro-benzaiide (161): 11 NMR (DMSOd 6 6 10.56 1H), 8.76 1H), 8.22 J 6.3 Hz, IR), 7.88-7.70 21), 7.60-7.46 WO 02/062295 PCT/US02/03061 2H), 3.07 J 7.2 Hz, 2H), 1.63-1.50 (ni, 21), 1.48-1.28 2H), 0.89 J Hz, 3H).
N-(6-Chloro-tpyrdin-3-yl)-3-fluoro-4-isobtvlsulfanl-benzamide (163): 11 NMR (DMSO-d 6 8 10.55 11), 8.77 8.22 J 6.9 Hz, 1H), 7.83-7.72 2H), 7.60-7.44 2H), 2.96 J 6.5 Hz, 211), 1.88-1.70 1H), 1.01 J 6.4 Hz, 6 H).
N-(6-Chloro-pyridin-3-yl)-3-fluoro-4-(pvridin-4-ylsulfavl)-benzamide (164): 1 H NMR (DMSO-d 6 6 10.73 1H), 8.78 1H), 8.42 J 4.2 Hz, 2H), 8.23 J 8.7 Hz, 1H), 7.98 J 9.9 Hz, 1H), 7.91 J 8.2 Hz), 7.82 (dd, J 7.3, 7.8 Hz, 11), 7.53 (d, 1 8.7 Hz, 111), 7.12 J 5.7 Hz, 2H) N-(6-Chloro-pyridin-3-vl)-3-fluoro-4-(4-fluoro-phenlsulfanvfl-benzamide (165): 11 NMR [(CD 3 2 CO)] 6 9.98 11), 8.75 J 2.4 Hz, 1H), 8.37 (dd, J 1.2, 2.8 Hz, 1H), 7.84-7.74 2H), 7.66-7.57 2H), 7.50 J 8.7 Hz, IR), 7.34-7.25 2H), 7.14 (dd, J 7.6, 8.0 Hz, 11).
4-(2-Chloro-benzenesulfonyl)-N-(6-chloro-pvridin-3-yl)-3-fluoro-benzamide lH NNMR [(CD 3 2 CO)] 610.09 1H), 8.76 J 2.7 Hz, 1H), 8.43-8.29 (ni, 3H), 8.11 (dd, J 1.6, 8.2 Hz, 1H), 7.87 (dd, J 1.5, 10.7 Hz, 1H), 7.84-7.71 21), 7.64 (dd, J 1.4, 7.6 Hz, 1H), 7.47 J 8.7 Hz, 1H).
N-(6-Chloro-Vyridin-3-vl)-3-fluoro-4-(2-fluoro-benzenesulfonvl)-benzamide (167): lH NMR [(CD 3 2 CO)1 6 10.09 1H), 8.76 J 2.7 Hz, 11), 8.35-8.26 21), 8.19 (dd, J 7.3, 7.7 flz, 1H), 8.11 J 8.0 Hz, 11), 7.91-7.82 21), 7.58 (dd, J 7.6, 7.7 Hz, 1H), 7.48 J 8.7 Hz, 1H), 7.37 (dd, J 8.5, 10.3 Hz, 1H).
N-(6-Chloro-pyridin-3-l)-3-fluoro-4-(2-methyl-propane-2-sulfonvl)-benzamide (168): 11 NMR (DMSO-d 6 6 8.77 11), 8.23 J 8.7 Hz, 11), 8.08-7.85 311), 7.55 (d, J 8.9 Hz, 11), 1.30 9 H).
4-(Butane- l-sulfonvl)-N-(6-chloro-pvrdin-3-vl)-3-fluoro-benzamide (169): 'H NMR (DMSO-d 6 8 10.85 11), 8.77 111), 8.21 1H), 8.08-7.86 3H), 7.55 J 9.1 WO 02/062295 WO 02/62295PCT/US02/03061 Hz, 1H1), 3.43 J 7.5 Hz, 211), 1.60-1.44 (in, 2H1), 1.41-1.23 (in, 2H1), 0.84 J Hz, 3H1).
N-(6-Chloro-pvridin-3-yl)-3-fluoro-4-(p2ropane-2-sulfonvll)-benzamnide (170): 1H1 N4MR (DMSO-d 6 6 10.85 8.76 11H), 8.22 J 8.5 Hz, 11H), 8.07-7.92 (in, 3H), 7.55 J =8.7 Hz, 1H1), 3.58-3.48 (in, 1H1), 1.22 J 6.8 Hz, 6H).
N-(6-Chloro-pvrLidin-3-yl)-3-fluoro-4-(2-methvil-propane-l1-sulfonvl)-benzainide (171): 'H NMIR (DMSO-d 6 8 8.77 1H), 8.23 J 8.7 Hz, 1H1), 8.06-7.98 (in, 3H1), 7.55 (d, J 8.9 Hz, 1H), 3.38 J 6.2 Hz, 2H), 2.12-2.00 (in, 1H), 0.99 J 6.8 Hz, 6H1).
N-(6-Chloro-pvridin-3-yl)-3-fluoro-4-(pvrazin-2-ylmeth Isulfan l-benzamide (175): 111 NMIR (DMSO-d 6 6 10.55 1H1), 8.75(d, J 2.4 Hz, 1HI), 8.72 1H), 8.55 111), 1 J 2.5 Hz, 111), 8.20 (dd, J 2.5, 8.6 Hz), 7.82-7.72 (in, 211), 7.68 (dd, J =7.7, 7.9 Hz, 111), 7.50 J 8.7, 1H), 4.53 211).
N-(6-Chloro-pyridin-3-yl)-3-fluoro-4-(2-pyrazin-2-yl-ethvlsulfan I -benzaniide (176): 1H1 NNvI (DMSO-d 6 8 10.56 111), 8.77 1H1), 8.59 111), 8.57 1H1), 8.49 111), 8.22 J 8.9 Hz, 111), 7.88-7.74 (in, 2H), 7.63 (dd, J 8.0, 8.0 Hz, 111), 7.51 J 8.7 Hz, 111), 3.50 J 7.3, 211), 3.14 J 7.3 Hz, 211).
EXAMPLE 11 This examnple illustrates a KCNQ2 screening protocol for evaluating compounds of the present invention.
Cells expressing voltage-gated K- channels, such as KCNQ2-like channels were loaded with 86 Rb+ by culture in media containing 86 RbCI. Following loading, culture media was removed and the cells were washed in EBSS to remove residual traces of 86 Rb+. Cells were preincubated with drug (0.01 30 [tM in EBSS) and then 8 6 Rb+ efflux was stimulated by exposing cells to EBSS solution supplemented with a sub-maximal concentration of KCl (generally 7-20 mm) in the continued presence of drug. After a suitable efflux period, the EBSS/ KCI solution was removed fromt the cells and the 8 6 Rb± content determined by Cherenkov counting (Wallac Trilux). Cells were t then lysed with a SDS solution and the 86 Rb content of the lysate determined. Percent 86 Rb+ efflux was calculated according to: ;Z (86Rb+ content in EBSS/ 86 Rb content in EBSS 86 Rb content of the lysate)) 100 Efflux was normalized to the maximal 86 Rb+ efflux that induced by a high Cc concentration of KCI, generally 30-135 mM).
The compounds of the invention (FIG.1) were prepared according to the general 00 Cc methods set forth in the examples and they were assayed using the above-described method. The activity of the assayed compounds ranged from about 30% to greater than about 70% efflux.
It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgement or any form of suggestion that the prior art forms part of the common general knowledge in Australia.

Claims (19)

1. A compound having the formula: R ZW' wherein Y is a member selected from H, methyl, methoxy, trifluoromethoxy, -CF 3 or halo; V and X are members independently selected from H, halo, substituted or unsubstituted lower homoalkyl and CF 3 R 1 is selected from unsubstituted lower alkyl, unsubstituted lower heteroalkyl, substituted or unsubstituted carbocycle; S; CN 3 N N Cl Cl' CN. ONII ;N N N O N N H 3 H 3 C H3C CH 30 1-2 1-2 H 3 C H 3 C ;and H 3 C substituted or unsubstituted phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl,
4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2- Sthiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3- pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2- O benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 3-quinolyl, and 6-quinolyl; and Q and W are members independently selected from -(CR 2 R 3 )t-(CH 2 -(CH 2 )n-(CR 2 R 3 )t 00 -C(R 4 )=C(R 5 and wherein 2 3 NC( R and R are members independently selected from H, F, substituted or Sunsubstituted lower alkyl or substituted or unsubstituted lower heteroalkyl, CI in which R 2 and R 3 are optionally joined to form a cyclic structure which is a member selected from the group consisting of cycloalkyl and heterocycle groups, Z represents -N(R 4 -N(R 4 and -SO 2 N(R 4 wherein R 4 and R 5 are members independently selected from the group consisting of H, substituted or unsubstituted lower alkyl, substituted or unsubstituted lower heteroalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, in which R' is not joined together with either X or R 4 to form a substituted or unsubstituted heterocycle; m is an integer from 0 to 2, inclusive; n is an integer from 0 to 3, inclusive; and t is an integer from 0 to 2, inclusive. 2. The compound according to claim 1, wherein Y is a member selected from chloro and methyl. 3. The compound according to claim 1, wherein V and X are members independently selected from the group consisting of halo and -CF 3 4. The compound according to claim 1, wherein Z is 1 0 O N 0"' 00q tm (N (N 0~ The compound according to claim 1, wherein Z is a member selected from the group consisting of-S- and SO 2
6. The compound according to claim 1, wherein R 4 is H.
7. The compound according to claim 1, wherein R' from the group consisting of: FCN cI Cl S H3 N N N S/ *0 N V^-s.N- m is a member selected CH 3 1-2 1-2 H 3 C H 3 C ;and H 3 C H3C
8. The compound according to claim 3, wherein n is an integer from 0 to 2, inclusive; and t is an integer from 0 to 1, inclusive.
9. The compound according to claim 1, having a structure which is a member selected from the group consisting of the compounds set forth in FIG. 1. The compound according to claim 1, wherein Z is -NH-.
11. The compound according to claim 1, wherein R' is substituted or unsubstituted phenyl.
12. The compound according to claim 1, wherein t is the integer 0.
13. The compound according to claim 1, wherein n is the integer 0. O
14. The compound according to claim 1, wherein n is 0 and t is the integer 0. (N, The compound according to claim 1, wherein R' is substituted or 00 C unsubstituted is I-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, N pyrazinyl.
16. The compound according to claim 1, wherein R' is substituted or unsubstituted 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4- isoxazolyl.
17. The compound according to claim 1, wherein R' is substituted or unsubstituted 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3- thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2- benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3- quinolyl, and 6-quinolyl.
18. The compound according to claim 1, wherein R' is substituted or unsubstituted l-naphthyl, 2-naphthyl, or 4-biphenyl.
19. A method of increasing ion flow through voltage-dependent potassium channels in a cell, said method comprising contacting said cell with a voltage-dependent potassium channel-opening amount of a compound of any one of claims 1 to 18. The method according to claim 19, wherein said voltage-dependent potassium channel is responsible for the M-current.
21. The method according to claim 19, wherein said voltage-dependent potassium channel comprises KCNQ subunits. S" 22. A method of treating a central or peripheral nervous system disorder or tb3 condition through modulation of a voltage-dependent potassium channel, said method comprising administering to a subject in need of such treatment, an effective amount of a O compound of any one of claims 1 to 18.
23. The method according to claim 22, wherein said disorder or condition is 00 selected from the group consisting of migraine, ataxia, Parkinson's disease, bipolar disorders, C spasticity, mood disorders, brain tumors, psychotic disorders, myokymia, seizures, epilepsy, hearing loss, vision loss, Alzheimer's disease, age-related memory loss, learning deficiencies, motor neuron diseases, and stroke.
24. The method according to claim 23, wherein said disorder or condition is hearing loss. The method according to claim 23, wherein said disorder or condition is epilepsy or seizures.
26. A method of treating pain, anxiety or bipolar disorder through modulation of a voltage-dependent potassium channel, said method comprising administering to a subject in need of such treatment, an effective amount of a compound of any one of claims 1 to 18.
27. A composition comprising a pharmaceutically acceptable excipient and a compound of any one of claims 1 to 18.
AU2002238023A 1999-08-04 2002-02-01 Pyridine-substituted benzanilides as potassium ion channel openers Ceased AU2002238023B2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US14722199P 1999-08-04 1999-08-04
US60/147221 1999-08-04
PCT/US2000/021308 WO2001010380A2 (en) 1999-08-04 2000-08-04 Benzanilides as potassium channel openers
US09/776,791 US6495550B2 (en) 1999-08-04 2001-02-02 Pyridine-substituted benzanilides as potassium ion channel openers
US09/776,791 2001-02-02
PCT/US2002/003061 WO2002062295A2 (en) 2001-02-02 2002-02-01 Pyridine-substituted benzanilides as potassium ion channel openers

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU67585/00A Division AU779034B2 (en) 1999-08-04 2000-08-04 Benzanilides as potassium channel openers

Publications (2)

Publication Number Publication Date
AU2002238023A1 AU2002238023A1 (en) 2003-02-13
AU2002238023B2 true AU2002238023B2 (en) 2008-03-13

Family

ID=39248184

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002238023A Ceased AU2002238023B2 (en) 1999-08-04 2002-02-01 Pyridine-substituted benzanilides as potassium ion channel openers

Country Status (1)

Country Link
AU (1) AU2002238023B2 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5869812A (en) * 1981-10-22 1983-04-26 Chugai Pharmaceut Co Ltd Blood sugar level depressing agent
JPS59181257A (en) * 1983-03-31 1984-10-15 Chugai Pharmaceut Co Ltd Ureidobenzamide derivative
JPH10259176A (en) * 1997-03-17 1998-09-29 Japan Tobacco Inc New amide derivative having vascularization inhibiting action and its use
WO1998042672A1 (en) * 1997-03-24 1998-10-01 Kyowa Hakko Kogyo Co., Ltd. Benzene derivatives
WO1999051131A1 (en) * 1998-04-08 1999-10-14 Andre Labruyere Slotted cup holder
AU730100B2 (en) * 1997-02-27 2001-02-22 Cellomics, Inc. A system for cell-based screening
AU779034B2 (en) * 1999-08-04 2005-01-06 Icagen, Inc. Benzanilides as potassium channel openers

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5869812A (en) * 1981-10-22 1983-04-26 Chugai Pharmaceut Co Ltd Blood sugar level depressing agent
JPS59181257A (en) * 1983-03-31 1984-10-15 Chugai Pharmaceut Co Ltd Ureidobenzamide derivative
AU730100B2 (en) * 1997-02-27 2001-02-22 Cellomics, Inc. A system for cell-based screening
JPH10259176A (en) * 1997-03-17 1998-09-29 Japan Tobacco Inc New amide derivative having vascularization inhibiting action and its use
WO1998042672A1 (en) * 1997-03-24 1998-10-01 Kyowa Hakko Kogyo Co., Ltd. Benzene derivatives
WO1999051131A1 (en) * 1998-04-08 1999-10-14 Andre Labruyere Slotted cup holder
AU779034B2 (en) * 1999-08-04 2005-01-06 Icagen, Inc. Benzanilides as potassium channel openers

Similar Documents

Publication Publication Date Title
US6737422B2 (en) Benzanilides as potassium channel openers
US6495550B2 (en) Pyridine-substituted benzanilides as potassium ion channel openers
US6593349B2 (en) Bisarylamines as potassium channel openers
US8329713B2 (en) Fused ring heterocycles as potassium channel modulators
CA2505195C (en) Quinazolinones as potassium channel modulators
AU2002359636B2 (en) Pyrimidines as novel openers of potassium ion channels
AU2002238023B2 (en) Pyridine-substituted benzanilides as potassium ion channel openers
AU2002238023A1 (en) Pyridine-substituted benzanilides as potassium ion channel openers

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: ADD PRIORITY DETAILS 779034 4 AUG 2000

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired
NA Applications received for extensions of time, section 223

Free format text: AN APPLICATION TO EXTEND THE TIME FROM 04 AUG 2008 TO 04 JUN 2009 IN WHICH TO PAY A RENEWAL FEE HASBEEN FILED .

NB Applications allowed - extensions of time section 223(2)

Free format text: THE TIME IN WHICH TO PAY A RENEWAL FEE HAS BEEN EXTENDED TO 04 JUN 2009.

MK14 Patent ceased section 143(a) (annual fees not paid) or expired