US20130109672A1 - Activators of human pyruvate kinase - Google Patents

Activators of human pyruvate kinase Download PDF

Info

Publication number
US20130109672A1
US20130109672A1 US13/643,594 US201113643594A US2013109672A1 US 20130109672 A1 US20130109672 A1 US 20130109672A1 US 201113643594 A US201113643594 A US 201113643594A US 2013109672 A1 US2013109672 A1 US 2013109672A1
Authority
US
United States
Prior art keywords
oxo
sulfonamide
alkyl
tetrahydroquinoline
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/643,594
Inventor
Matthew B. Boxer
Min Shen
Douglas S. Auld
Craig J. Thomas
Martin J. Walsh
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.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US13/643,594 priority Critical patent/US20130109672A1/en
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOXER, MATTHEW B., THOMAS, CRAIG J., SHEN, MIN, WALSH, MARTIN J., AULD, DOUGLAS S.
Publication of US20130109672A1 publication Critical patent/US20130109672A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/227Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/14Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D223/16Benzazepines; Hydrogenated benzazepines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/341,4-Oxazines; Hydrogenated 1,4-oxazines condensed with carbocyclic rings
    • C07D265/361,4-Oxazines; Hydrogenated 1,4-oxazines condensed with carbocyclic rings condensed with one six-membered ring

Definitions

  • Pyruvate kinase is a critical metabolic enzyme operating at the ultimate step in glycolysis where it catalyzes the transfer of a phosphate group from phosphoenolpyruvate to adenosine diphosphate (ADP), yielding one molecule of pyruvate and one molecule of adenosine triphosphate (ATP).
  • ADP adenosine diphosphate
  • ATP adenosine triphosphate
  • L gene produces two different mRNAs that differ only in the first exon to produce the L (liver specific) and R (red blood cell) specific isozymes.
  • M1 isozyme that is found in most adult tissues and the M2 isozyme that is present in fetal tissues and is found to be re-expressed in tumors. Therefore, after embryonic development, adult tissues switch to either express PK-M1 or the tissue specific L or R isozymes. However, in all tumors or cell lines of cancer lineage (including those typically expressing either the L or R isozymes), PK gene expression reverts entirely to the M2 isoform.
  • PK is a tetrameric enzyme composed of four identical monomers that form a dimer of dimers in the final tetrameric structure.
  • the M2, L, and R isozymes are activated by fructose-1,6-bis phosphate (FBP) that binds to a flexible loop region at the interface of the two dimers.
  • FBP fructose-1,6-bis phosphate
  • Activation of PK shifts the enzyme to a state showing high affinity for phosphoenolpyruvate (PEP).
  • PEP phosphoenolpyruvate
  • the M1 isoform is not regulated by FBP and displays only high affinity PEP binding similar to the activated state of PK.
  • PK-M2 Various phosphotyrosine peptides can bind to PK-M2 near the activation loop that results in the removal of FBP from the enzyme which effectively down-regulates PK-M2 activity. These peptides are present in exacerbated levels in cancer cells.
  • PK-M2 When PK-M2 is activated, glucose is converted to pyruvate.
  • PK-M2 When PK-M2 is inactivated, a build-up of glycolytic intermediates occurs which intermediates can be diverted towards nucleotide and lipid biosynthesis required for cell growth and proliferation.
  • the present invention provides compounds that are activators of the M2 isoform of human pyruvate kinase.
  • the present invention provides compositions comprising these compounds and methods of using these compound as therapeutic agents in the treatment or prevention of cancer.
  • the invention provides a compound of formula (I):
  • a 1 and A 2 are each individually R′ or R′′;
  • R is H or C 1 -C 4 alkyl
  • L is SO 2 or CO
  • R′ is a fused bicyclic ring, wherein one ring of the bicyclic ring is phenyl which is linked to the NR-L moiety at the nitrogen atom or the sulfur atom when L is SO 2 or the carbon atom when L is CO and the other ring of the bicyclic ring is an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, wherein R′ is optionally substituted on one or both rings with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C)—C 4 )alkyl-(C 1 -C 4 )alkyl, (C 1 -C 4 )alkyl-CO—, and heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents
  • R′′ is phenyl, benzyl, or heteroaryl, which is optionally substituted with one or more substituents selected from the group consisting of halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxyl, cyano, alkylenedioxy, aryl, heteroaryl, benzyl, B(OH) 2 , and C 1 -C 4 alkyl substituted with one or more halogens, or is phenyl optionally fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of halogen, C 1 -C 4 alkyl, C 1 -C 1 alkoxyl, alkylenedioxy, aryl, heteroaryl, benzyl, and C 1 -C 4 alkyl substituted with one or more halogens;
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound or salt of the invention and a pharmaceutically acceptable carrier.
  • the invention further provides a method for treating cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of the invention to a mammal afflicted therewith.
  • the invention provides a compound of formula (I):
  • a 1 and A 2 are each individually R′ or R′′;
  • R is H or C 1 -C 4 alkyl
  • L is SO 2 or CO
  • R′ is a fused bicyclic ring, wherein one ring of the bicyclic ring is phenyl which is linked to the NR-L moiety at the nitrogen atom or the sulfur atom when L is SO 2 or the carbon atom when L is CO and the other ring of the bicyclic ring is an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, wherein R′ is optionally substituted on one or both rings with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, and heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH,
  • R′′ is phenyl, benzyl, or heteroaryl, which is optionally substituted with one or more substituents selected from the group consisting of halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxyl, cyano, alkylenedioxy, aryl, heteroaryl, benzyl, B(OH) 2 , and C 1 -C 4 alkyl substituted with one or more halogens, or is phenyl optionally fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxyl, alkylenedioxy, aryl, heteroaryl, benzyl, and C 1 -C 4 alkyl substituted with one or more halogens;
  • a 1 is R′′ and A 2 is R′.
  • the phenyl ring of the bicyclic ring of R′ is fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, (C 1 -C 4 )alkyl-CO—, and heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl.
  • the cyclyl or heterocyclyl of R′ or R′′ is a five-membered, six-membered, or seven-membered ring.
  • the heterocyclyl contains one or two heteroatoms.
  • R is methyl or H.
  • R′ is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • X 1 and X 2 are each individually O, NR 6 , or CR 7 R 8 ;
  • any NH—CH 2 moiety within the ring containing X 1 and X 2 is optionally replaced with a N ⁇ CH moiety;
  • any methylene of the ring containing X 1 and X 2 is optionally replaced by a carbonyl
  • n and m are each individually 0, 1, or 2, and wherein n m is 0 to 2;
  • each R′ is individually H, halogen, alkyl, alkoxyl, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl;
  • R 6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl; and
  • R 7 and R 8 are each individually II, halogen, alkyl, alkoxyl, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl.
  • one R′ is at the ortho position relative to the carbon attached to the NR-L moiety.
  • one R 1 is H, F, Cl, Br, methyl, N(Me) 2 , NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
  • R′ is optionally substituted with one or more substituents selected from the group consisting of methyl and acetyl.
  • R′′ is 3,4-dihydroquinolin-2(1H)-onyl, indolin-2-onyl, 4,5-dihydro-1H-benzo[b]azepin-2(3H)-onyl, 2H-benzo[b][1,4]oxazin-3(4H)-onyl, 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-onyl, 1-(indolin-1-yl)ethanonyl, 1-methyl-1H-indolyl, 1-acetyl-2-methylindolinyl, 6-chloro-2-oxoindolinyl, 3,3-dichloro-2-oxoindolinyl, 7-((2-(dimethyl)
  • R′′ is
  • X 3 is N or CH wherein when X 3 is N, the NR-L moiety is linked to a C of the ring containing X 3 ;
  • R 2 and R 3 are each individually halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxyl, cyano, B(OH) 2 , phenyl, C 1 -C 4 alkyl substituted with one or more halogens, or taken together form alkylenedioxyl.
  • R 2 and R 3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH) 2 , or taken together form alkylenedioxyl.
  • R′′ is 3,4-dimethylphenyl, 3-chlorophenyl, meta-tolyl, 3-methoxyphenyl, 3-fluorophenyl, 3-trifluoromethylphenyl, biphenyl-3-yl, pyridine-3-yl, 4-chlorophenyl, para-tolyl, 4-methoxyphenyl, 4-fluorophenyl, ortho-tolyl, 2-methoxyphenyl, 2-fluorophenyl, naphthalen-2-yl, naphthalen-1-yl, 2,3-dihydrobenzo[b][1,4]dioxin-6-yl, benzyl, 3-chloro-4-methylphenyl, 3,4-dichlorophenyl, 5-chloro-2-methylphenyl, 3-cyanophenyl, 3-chloro-2-methylphenyl, 3-phenylboronic acid, 4-fluoro-3-methylphenyl, 3-fluoromethylphenyl, 3-fluor
  • the compound of formula (I) is a compound of formula (II):
  • X 1 and X 2 are each individually O, NR 6 , or CR 7 R 8 ;
  • X 3 is N or CH wherein when X 3 is N, the NR-L moiety is linked to a C of the ring containing X 3 ;
  • any NH—CH 2 moiety within the ring containing X 1 and X 2 is optionally replaced with a N ⁇ CH moiety;
  • any methylene of the ring containing X 1 and X 2 is optionally replaced by a carbonyl
  • n and m are each individually 0, 1, or 2, and wherein n+m is 0 to 2;
  • k is 0 or 1
  • R 1 is H, halogen, alkyl, alkoxyl, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl;
  • R 2 and R 3 are each individually H, halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxyl, cyano, B(OH) 2 , phenyl, C 1 -C 4 alkyl substituted with one or more halogens, or taken together form alkylenedioxy or phenyl fused to a CH:CH moiety of the ring containing X 3 ;
  • R 6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl; and
  • R 7 and R 8 are each individually H, halogen, alkyl, alkoxyl, NH 2 , NH—(C 1 -C 4 )alkyl, N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH 2 is further optionally substituted with one or more substituents selected from the group consisting of NH 2 , OH, NH—(C 1 -C 4 )alkyl and N—(C 1 -C 4 )alkyl-(C 1 -C 4 )alkyl.
  • R 1 is H, F, Cl, Br, methyl, N(Me) 2 , NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
  • R 2 and R 3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH) 2 , or taken together form alkylenedioxyl or phenyl fused to a CH:CH moiety of the ring containing X 3 .
  • the compounds exclude a compound of formula (II) wherein R 2 is a methyl in the para position relative to the NR-L moiety, R 3 is a methyl in a meta position relative to the NR-L moiety, X 3 is CH, k is zero, R is H, L is SO 2 , R 1 is H, X 1 is NR 6 , R 6 is H, X 2 is CR 7 R 8 , R 7 and R 8 are each H, n is 1, m is zero, and the methylene adjacent to X 1 is replaced with a carbonyl.
  • FBP Fructose-1,6-bis phosphate
  • alkyl means a straight-chain or branched alkyl substituent containing from, for example, 1 to about 6 carbon atoms, preferably from 1 to about 4 carbon atoms, more preferably from 1 to 2 carbon atoms.
  • substituents include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, and the like.
  • alkoxyl means any alkyl substituent attached as a substituent via an oxygen atom.
  • alkenyl means a linear alkenyl substituent containing at least one carbon-carbon double bond and from, for example, about 2 to about 6 carbon atoms (branched alkenyls are about 3 to about 6 carbons atoms), preferably from about 2 to about 5 carbon atoms (branched alkenyls are preferably from about 3 to about 5 carbon atoms), more preferably from about 3 to about 4 carbon atoms.
  • substituents include propenyl, isopropenyl, n-butenyl, sec-butenyl, isobutenyl, tert-butenyl, pentenyl, isopentenyl, hexenyl, and the like.
  • Alkylenedioxy means a —O—(CH 2 ) q —O— group, where q is from 1 to about 6, preferably from 1 to about 4, more preferably from 1 to 2.
  • alkynyl means a linear alkynyl substituent containing at least one carbon-carbon triple bond and from, for example, 2 to about 6 carbon atoms (branched alkynyls are about 3 to about 6 carbons atoms), preferably from 2 to about 5 carbon atoms (branched alkynyls are preferably from about 3 to about 5 carbon atoms), more preferably from about 3 to about 4 carbon atoms.
  • substituents examples include propynyl, isopropynyl, n-butynyl, sec-butynyl, isobutynyl, tert-butynyl, pentynyl, isopentynyl, hexynyl, and the like.
  • cyclyl as used herein encompasses cycloalkyl and cycloalkenyl.
  • Cycloalkyl as used herein, means a cyclic alkyl substituent containing from, for example, about 3 to about 8 carbon atoms, preferably from about 4 to about 7 carbon atoms, and more preferably from about 4 to about 6 carbon atoms. Examples of such substituents include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like.
  • cycloalkenyl as used herein, means the same as the term “cycloalkyl,” however one or more double bonds are present.
  • Examples include cyclopentenyl and cyclohexenyl.
  • the cyclic alkyl groups may be unsubstituted or further substituted. Examples of substitutions include halogens, alkyoxy groups, and alkyl groups such as methyl groups, ethyl groups, and the like.
  • Cyclyl also encompasses cycloalkyl and cycloalkenyl in which a heteroatom is exocyclic.
  • the heteroatom for example, may be N, O, or S.
  • a methylene group of the cyclyl can be replaced with a carbonyl.
  • a cyclyl group may be fused to another ring, including another cyclyl, heterocyclyl, aryl, or heteroaryl.
  • a fused bicyclic ring is any ring of cyclyl, heterocyclyl, aryl, or heteroaryl fused with another cyclyl, heterocyclyl, aryl, or heteroaryl.
  • heteroaryl refers to a monocyclic or bicyclic 5- or 6-membered aromatic ring system containing one or more heteroatoms selected from the group consisting of O, N, S, and combinations thereof.
  • suitable monocyclic heteroarylgroups include but are not limited to furanyl, thiopheneyl, pyrrolyl, pyrazolyl, imidazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and triazinyl.
  • the heteroaryl group can be attached at any available position on the heteroaryl group.
  • a thiopheneyl group can be attached at the 2-position or the 3-position of the thiopheneyl group.
  • a pyridyl group can be attached at the 2-, 3-, or 4-position of the pyridyl group.
  • Suitable bicyclic heterocycloaryl groups include monocylic heterocycloaryl rings fused to a C 6 -C 10 aryl or heteroaryl ring.
  • Non-limiting examples of bicyclic heterocycloaryl groups include benzofuran, benzothiophene, quinoline, and isoquinoline.
  • the heteroaryl group is optionally substituted with 1, 2, 3, 4, or 5 substituents as recited herein, wherein the optional substituent can be present at any open position on the heteroaryl group.
  • heteroaryl oxide refers to an oxidized heteroaryl group as that term is defined herein, wherein one or more of the heteroatoms comprising the heteroaryl group is oxidized.
  • heteroaryl oxide groups include pyridine N-oxide, pyrimidine N-oxide, and pyrazine N-oxide.
  • heterocyclyl refers to a cyclic group, which may be aromatic or non-aromatic, or saturated or unsaturated, having one or more heteroatoms such as O, N, or S.
  • heterocyclyl groups include pyridyl, piperidinyl, piperazinyl, pyrazinyl, pyrolyl, pyranyl, tetrahydropyranyl, tetrahydrothiopyranyl, pyrrolidinyl, furanyl, tetrahydrofuranyl, thiophenyl, tetrahydrothiophenyl, purinyl, pyrimidinyl, thiazolyl, thiazolidinyl, thiazolinyl, oxazolyl, triazolyl, tetrazolyl, tetrazinyl, benzoxazolyl, morpholinyl, thiophorpholinyl, quinolinyl, and is
  • halo or “halogen,” as used herein, means a substituent selected from Group VIIA, such as, for example, fluorine, bromine, chlorine, and iodine.
  • aryl refers to an unsubstituted or substituted aromatic carbocyclic substituent, as commonly understood in the art, and the term “C 6 -C 10 aryl” includes phenyl and naphthenyl. It is understood that the term aryl applies to cyclic substituents that are planar and comprise 4n+2 ⁇ electrons, according to Hückel's Rule. An aryl group may be fused to another ring, including a cyclyl, heteroaryl, heterocyclyl, or another aryl.
  • a CH 2 —CH 2 moiety is any ethylene moiety wherein there is a carbon-carbon single bond.
  • a CH ⁇ CH moiety is any vinyl moiety that contains a carbon-carbon double bond.
  • a NH—CH 2 moiety contains a nitrogen-carbon single bond, and a N ⁇ CH contains a nitrogen-carbon double bond.
  • a CH:CH moiety contains a carbon-carbon bond intermediate between a single and a double bond, such as in an aromatic system, for example in the carbon-carbon bonds in benzene or the nitrogen-carbon bond in pyridine.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound or salt of any of the embodiments described above and a pharmaceutically acceptable carrier.
  • phrases “pharmaceutically acceptable salt” is intended to include nontoxic salts synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, Pa., 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977).
  • Suitable bases include inorganic bases such as alkali and alkaline earth metal bases, e.g., those containing metallic cations such as sodium, potassium, magnesium, calcium and the like.
  • suitable bases include sodium hydroxide, potassium hydroxide, sodium carbonate, and potassium carbonate.
  • Suitable acids include inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic, methanesulfonic acid, benzenesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, maleic acid, tartaric acid, fatty acids, long chain fatty acids, and the like.
  • Preferred pharmaceutically acceptable salts of inventive compounds having an acidic moiety include sodium and potassium salts.
  • Preferred pharmaceutically acceptable salts of inventive compounds having a basic moiety include hydrochloride and hydrobromide salts.
  • the compounds of the present invention containing an acidic or basic moiety are useful in the form of the free base or acid or in the form of a pharmaceutically acceptable salt thereof.
  • any salt of this invention is usually not of a critical nature, so long as the salt as a whole is pharmacologically acceptable and as long as the counterion does not contribute undesired qualities to the salt as a whole.
  • solvates refers to a molecular complex wherein the solvent molecule, such as the crystallizing solvent, is incorporated into the crystal lattice.
  • the solvent incorporated in the solvate is water, the molecular complex is called a hydrate.
  • Pharmaceutically acceptable solvates include hydrates, alcoholates such as methanolates and ethanolates, acetonitrilates and the like. These compounds can also exist in polymorphic forms.
  • the invention contemplates embodiments in which a compound having a chiral center is a substantially pure enantiomer thereof, a racemic mixture thereof, or a mixture containing any proportion of the two enantiomers thereof.
  • the invention also contemplates all stereoisomers and diastereoisomers of the compounds described herein.
  • the present invention is further directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one compound or salt described herein.
  • the pharmaceutically acceptable carrier be one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
  • compositions for parenteral administration that comprise a solution of the inventive compound or salt dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous isotonic sterile injection solutions.
  • Such solutions can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the compound or salt of the present invention may be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl-1,3-dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethyl
  • Oils useful in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations can contain preservatives and buffers.
  • such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17.
  • HLB hydrophile-lipophile balance
  • the quantity of surfactant in such formulations will typically range from about 5 to about 15% by weight.
  • Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • Topical formulations including those that are useful for transdermal drug release, are well-known to those of skill in the art and are suitable in the context of the invention for application to skin.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as a therapeutically effective amount of the inventive compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules, (c) powders, (d) suspensions in an appropriate liquid, and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • the compound or salt of the present invention can be made into aerosol formulations to be administered via inhalation.
  • the compounds are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of active compound are 0.01%-20% by weight, preferably 1%-10%.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • Such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
  • Mixed esters such as mixed or natural glycerides may be employed.
  • the surfactant may constitute 0.1%-20% by weight of the composition, preferably 0.25%-5%. The balance of the composition is ordinarily propellant.
  • a carrier can also be included as desired, e.g., lecithin for intranasal delivery.
  • aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations may be used to spray mucosa.
  • pressurized propellants such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • non-pressured preparations such as in a nebulizer or an atomizer.
  • Such spray formulations may be used to spray mucosa.
  • the compound or salt of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • the compound or salt of the present invention may be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.
  • inclusion complexes such as cyclodextrin inclusion complexes, or liposomes.
  • Liposomes serve to target the compounds to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of the inventive compound.
  • Liposomes useful in the present invention include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like.
  • the active agent to be delivered is incorporated as part of a liposome, alone or in conjunction with a suitable chemotherapeutic agent.
  • liposomes filled with a desired inventive compound or salt thereof can be directed to the site of a specific tissue type, hepatic cells, for example, where the liposomes then deliver the selected compositions.
  • Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, for example, liposome size and stability of the liposomes in the blood stream.
  • a ligand to be incorporated into the liposome can include, for example, antibodies or fragments thereof specific for cell surface determinants of the targeted tissue type.
  • a liposome suspension containing a compound or salt of the present invention may be administered intravenously, locally, topically, etc. in a dose that varies according to the mode of administration, the agent being delivered, and the stage of disease being treated.
  • Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached.
  • the present inventive method typically will involve the administration of about 0.001 to about 300 mg of one or more of the compounds described above per kg body weight of the individual. The administration can involve about 0.001 mg, about 0.01 mg, about 0.1 mg, about 1 mg, about 5 mg, about 10 mg, about 20 mg, about 50 mg, about 100 mg, about 200 mg, or about 300 mg or more of one or more of the compounds described above per kg body weight of the individual.
  • the administration can involve about 300 mg, about 200 mg, about 100 mg, about 50 mg, about 20 mg, about 10 mg, about 5 mg, about 1 mg, about 0.1 mg, about 0.01 mg, or about 0.001 mg or less of one or more of the compounds described above per kg body weight of the individual.
  • the administration can be bounded by any two of the aforementioned endpoints.
  • the administration can be about 0.001 mg to about 200 mg, about 0.001 mg to about 1 mg, about 0.01 mg to about 50 mg, about 0.1 mg to about 20 mg, about 1 mg to about 10 mg, about 1 mg to about 20 mg, about 10 mg to about 50 mg, or any other combination of endpoints, of one or more of the compounds described above per kg body weight of the individual.
  • the present invention further provides a method of treating a disease responsive to activation of human PK-M2 comprising administering to a patient in need thereof a therapeutically effective amount of any of the compounds described herein or a pharmaceutically acceptable salt thereof.
  • the invention further provides any of the compounds described herein or a pharmaceutically acceptable salt thereof for use in treating a disease responsive to activation of human PK-M2.
  • the invention further provides the use of a compound or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a disease responsive to activation of human PK-M2 of a patient, wherein the compound is any of the compounds described herein or a pharmaceutically acceptable salt thereof
  • the disease responsive to activation of PK-M2 can be caused by or associated with, e.g., the function PKM2.
  • diseases may include, e.g., cancer, obesity, diabetes, atherosclerosis, restenosis, autoimmune diseases, and proliferation-dependent diseases.
  • Cancers include, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic (myeloid) leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic (myeloid) leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (e.g., Hodgkin's disease or non-Hodgkin's disease), Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angio
  • the cancers may include bone cancer, brain cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the oral cavity, cancer of the vulva, esophageal cancer, gastrointestinal carcinoid tumor, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, nasopharynx cancer, non-small cell lung cancer, peritoneum, omentum, and mesentery cancer, pharynx cancer, rectal cancer, renal cancer, small intestine cancer, soft tissue cancer, stomach cancer, thyroid cancer, and ureter cancer.
  • Adipose tissue expresses PKM2. Additionally, activators of PKM2, described herein, may be useful in the treatment of type II diabetes, as the activation of PKM2 may allow for decreased lipid production and increased oxidative phosphorylation in adipose tissue. This effect should decrease adiposity, which is known to contribute to type 2 diabetes.
  • diseases include autoimmune diseases and proliferative diseases.
  • Activators of PKM2, described herein may be used to treat, e.g., autoimmune diseases or proliferative diseases.
  • Autoimmune disorders include, e.g., type I diabetes, Crohn's disease, multiple sclerosis, arthritis, rheumatoid arthritis, systemic lupus erythematosus, autoimmune (Hashimoto's) thyroiditis, autoimmune liver diseases (e.g., hepatitis and primary biliary cirrhosis), hyperthyroidism (e.g., Graves' disease and thyrotoxicosis), insulin-resistant diabetes, autoimmune adrenal insufficiency (e.g., Addison's disease), autoimmune oophoritis, autoimmune orchitis, autoimmune hemolytic anemia, paroxysmal cold hemoglobinuria, Behcet's disease, autoimmune thrombocytopenia, autoimmune neutropenia, pernic
  • Proliferative diseases include, e.g., cancer (e.g., benign and malignant), benign prostatic hyperplasia, psoriasis, abnormal keratinization, lymphoproliferative disorders (e.g., a disorder in which there is abnormal proliferation of cells of the lymphatic system), chronic rheumatoid arthritis, arteriosclerosis, restenosis, and diabetic retinopathy.
  • cancer e.g., benign and malignant
  • benign prostatic hyperplasia e.g., psoriasis
  • abnormal keratinization e.g., lymphoproliferative disorders
  • lymphoproliferative disorders e.g., a disorder in which there is abnormal proliferation of cells of the lymphatic system
  • chronic rheumatoid arthritis e.g., arteriosclerosis, restenosis, and diabetic retinopathy.
  • Proliferative diseases are described in U.S. Pat. Nos. 5,639,600 and 7,
  • inventive methods can provide any amount of any level of treatment or prevention in a mammal.
  • the treatment or prevention provided by the inventive method can include treatment or prevention of one or more conditions or symptoms of the diseases described herein being treated or prevented.
  • prevention can encompass delaying the onset of the disease, or a symptom or condition thereof.
  • the invention further provides a use of a compound or salt of the invention in the manufacture of a medicament for treating disease responsive to activation of PK-M2.
  • the medicament typically is a pharmaceutical composition as described herein.
  • the dose administered to a human in accordance with the present invention should be sufficient to effect the desired response.
  • Such responses include reversal or prevention of the bad effects of the disease responsive to activation of PK-M2 for which treatment is desired or to elicit the desired benefit.
  • dosage will depend upon a variety of factors, including the age, condition, and body weight of the human, as well as the source, particular type of the cancer, and extent of cancer in the human.
  • the size of the dose will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular compound and the desired physiological effect. It will be appreciated by one of skill in the art that various conditions or disease states may require prolonged treatment involving multiple administrations.
  • the compounds of the invention can be prepared as follows.
  • the synthetic elaboration of substituted 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides can begin with a standard coupling between commercially available sulfonyl chlorides and substituted anilines, according to Scheme I.
  • This example illustrates methods used in preparing exemplary compounds of the invention.
  • the mobile phase consisted of acetonitrile and water (each containing 0.1% trifluoroacetic acid). A gradient of 10% to 50% acetonitrile over 8 minutes was used during the purification. Fraction collection was triggered by UV detection (220 nM). Analytical analysis was performed on an Agilent LC/MS (Agilent Technologies, Santa Clara, Calif., USA).
  • Method 1 A 7 minute gradient of 4% to 100% Acetonitrile (containing 0.025% trifluoroacetic acid) in water (containing 0.05% trifluoroacetic acid) was used with an 8 minute run time at a flow rate of 1 mL/min.
  • Method 2 A 3 minute gradient of 4% to 100% Acetonitrile (containing 0.025% trifluoroacetic acid) in water (containing 0.05% trifluoroacetic acid) was used with a 4.5 minute run time at a flow rate of 1 mL/min.
  • N-(3,4-dimethylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide (0.01 g, 0.029 mmol) was dissolved in acetonitrile (0.5 ml) and dimethylamine (2.0 M THF) (0.029 ml, 0.057 mmol) was added followed by triethylamine (6.00 ⁇ l, 0.043 mmol).
  • the solution was heated in a microwave at 180° C. for 1 h, cooled to RT, diluted with DMSO (0.5 mL) and purified by directly injecting to a Waters® reverse phase purification system.
  • AC 50 values were determined utilizing the luminescent pyruvate kinase-luciferase coupled assay (Inglese, J. et al, Quantitative high-throughput screening: a titration-based approach that efficiently identifies biological activities in large chemical libraries. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 11473-11478).
  • ATP phosphatidylcholine
  • PEP phosphatidylcholine
  • LDH phosphatidylcholine
  • NADH phosphatidylcholine
  • Reagents and solvents were purchased from Sigma, Alfa Aesar (Ward Hill, Mass., USA), Acros (Morris Plains, N.J., USA), Enamine (Monmouth Jct., NJ, USA), Oakwood Products (West Columbia, S.C., USA), Matrix Scientific (Columbia, S.C., USA) or Chem-Impex International (Wood Dale, Ill., USA).
  • nL of compound in DMSO were delivered with a 1,536-pin array tool, and 1 ⁇ L of enzyme mix in assay buffer (final concentration, 0.1 nM pyruvate kinase, 50 mM imidazole pH 7.2, 0.05% BSA, 4° C.) was added.
  • Microtiter plates were incubated at r.t. for 1 hour and 2 uL of luciferase detection mix (Kinase-Glo from Promega, Madison, Wis., USA) at 4° C. protected from light, was added and luminescence was read with a ViewLux (Perkin Elmer, Waltham, Mass., USA) using a 2 second exposure/plate (with 2 ⁇ binning).
  • the final concentration of DMSO was 0.5% and found not to affect the assay signal.
  • qHTS uses an inter-plate dilution method where the first plate contains the highest concentration of a set of compounds in DMSO, while subsequent plates contain the same compounds in the same well locations, but at successive lower concentrations.
  • the rate was calculated as a plate throughput of 18 plates/hr or approximately 7 samples/sec on the Kalypsys robotic system which means that a 7 point CRC was obtained every second on the robotic system.
  • the second examination involved the modification of the core 3,4-dihydroquinolin-2(1H)-one heterocycle.
  • Numerous, related heterocyclic sulfonyl chlorides were examined after coupling to 3,4-dimethylaniline to maintain uniformity with the lead from the primary screen.
  • Results detailed in Table 1 demonstrate that the 3,4-dihydroquinolin-2(1H)-one heterocycle retains the best combination of potency and maximum response.
  • Other heterocycles included the related 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one and the modestly divergent 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-one and 1-(indolin-1-yl)ethanone heterocycles.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Indole Compounds (AREA)
  • Quinoline Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Disclosed are pyruvate kinase M2 activators which are compounds of Formula (I), including those of Formula (II), wherein A1, A2, L, R, R1 to R3, X1 to X3, k, n, and m are as defined herein, that are useful in treating a number of diseases that are treatable by the activation of PKM2, for example, cancer. A1-NR-L-A2 (I).
Figure US20130109672A1-20130502-C00001

Description

    CROSS-REFERENCE TO A RELATED APPLICATION
  • The present application claims the benefit of U.S. provisional patent application No. 61/329,158, filed Apr. 29, 2010, the disclosure of which is incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • Pyruvate kinase (PK) is a critical metabolic enzyme operating at the ultimate step in glycolysis where it catalyzes the transfer of a phosphate group from phosphoenolpyruvate to adenosine diphosphate (ADP), yielding one molecule of pyruvate and one molecule of adenosine triphosphate (ATP). In humans there are two pyruvate kinase genes and each produces two distinct gene products by alternative splicing. The L gene produces two different mRNAs that differ only in the first exon to produce the L (liver specific) and R (red blood cell) specific isozymes. Splicing of a single exon within the M gene produces the M1 isozyme that is found in most adult tissues and the M2 isozyme that is present in fetal tissues and is found to be re-expressed in tumors. Therefore, after embryonic development, adult tissues switch to either express PK-M1 or the tissue specific L or R isozymes. However, in all tumors or cell lines of cancer lineage (including those typically expressing either the L or R isozymes), PK gene expression reverts entirely to the M2 isoform.
  • PK is a tetrameric enzyme composed of four identical monomers that form a dimer of dimers in the final tetrameric structure. In humans, the M2, L, and R isozymes are activated by fructose-1,6-bis phosphate (FBP) that binds to a flexible loop region at the interface of the two dimers. Activation of PK shifts the enzyme to a state showing high affinity for phosphoenolpyruvate (PEP). In contrast, the M1 isoform is not regulated by FBP and displays only high affinity PEP binding similar to the activated state of PK.
  • Tumor cells undergo a metabolic transformation that is required to supply the biochemical precursors necessary for rapid cell growth and proliferation. Knock-down of PKM2 and re-expression of PKM1 has been shown to significantly diminish the proliferation of cancer cells in vivo such that even when tumors do grow, they have delayed formation and re-expression of PKM2.
  • Various phosphotyrosine peptides can bind to PK-M2 near the activation loop that results in the removal of FBP from the enzyme which effectively down-regulates PK-M2 activity. These peptides are present in exacerbated levels in cancer cells. When PK-M2 is activated, glucose is converted to pyruvate. However, when PK-M2 is inactivated, a build-up of glycolytic intermediates occurs which intermediates can be diverted towards nucleotide and lipid biosynthesis required for cell growth and proliferation.
  • Methods for detecting activators of PK-M2 are known. However, there is a desire for the identification of new activators of PK-M2.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides compounds that are activators of the M2 isoform of human pyruvate kinase. In addition, the present invention provides compositions comprising these compounds and methods of using these compound as therapeutic agents in the treatment or prevention of cancer.
  • The invention provides a compound of formula (I):

  • A1-NR-L-A2  (I)
  • wherein A1 and A2 are each individually R′ or R″;
  • wherein R is H or C1-C4 alkyl;
  • wherein L is SO2 or CO;
  • wherein R′ is a fused bicyclic ring, wherein one ring of the bicyclic ring is phenyl which is linked to the NR-L moiety at the nitrogen atom or the sulfur atom when L is SO2 or the carbon atom when L is CO and the other ring of the bicyclic ring is an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, wherein R′ is optionally substituted on one or both rings with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH2, NH—(C1-C4)alkyl, N—(C)—C4)alkyl-(C1-C4)alkyl, (C1-C4)alkyl-CO—, and heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
  • wherein R″ is phenyl, benzyl, or heteroaryl, which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, alkylenedioxy, aryl, heteroaryl, benzyl, B(OH)2, and C1-C4 alkyl substituted with one or more halogens, or is phenyl optionally fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C1 alkoxyl, alkylenedioxy, aryl, heteroaryl, benzyl, and C1-C4 alkyl substituted with one or more halogens;
  • or a pharmaceutically acceptable salt thereof.
  • The invention also provides a pharmaceutical composition comprising a compound or salt of the invention and a pharmaceutically acceptable carrier.
  • The invention further provides a method for treating cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of the invention to a mammal afflicted therewith.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In accordance with an embodiment, the invention provides a compound of formula (I):

  • A1-NR-L-A2  (I)
  • wherein A1 and A2 are each individually R′ or R″;
  • wherein R is H or C1-C4 alkyl;
  • wherein L is SO2 or CO;
  • wherein R′ is a fused bicyclic ring, wherein one ring of the bicyclic ring is phenyl which is linked to the NR-L moiety at the nitrogen atom or the sulfur atom when L is SO2 or the carbon atom when L is CO and the other ring of the bicyclic ring is an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, wherein R′ is optionally substituted on one or both rings with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, and heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
  • wherein R″ is phenyl, benzyl, or heteroaryl, which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, alkylenedioxy, aryl, heteroaryl, benzyl, B(OH)2, and C1-C4 alkyl substituted with one or more halogens, or is phenyl optionally fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxyl, alkylenedioxy, aryl, heteroaryl, benzyl, and C1-C4 alkyl substituted with one or more halogens;
  • or a pharmaceutically acceptable salt thereof.
  • In an embodiment, A1 is R″ and A2 is R′.
  • In another embodiment, the phenyl ring of the bicyclic ring of R′ is fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, (C1-C4)alkyl-CO—, and heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
  • In certain embodiments, the cyclyl or heterocyclyl of R′ or R″ is a five-membered, six-membered, or seven-membered ring.
  • In particular embodiments, the heterocyclyl contains one or two heteroatoms.
  • In preferred embodiments, R is methyl or H.
  • In any embodiments of the above, R′ is
  • Figure US20130109672A1-20130502-C00002
  • wherein X1 and X2 are each individually O, NR6, or CR7R8;
  • wherein any CH2—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a CH═CH moiety;
  • wherein any NH—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a N═CH moiety;
  • wherein any methylene of the ring containing X1 and X2 is optionally replaced by a carbonyl;
  • wherein n and m are each individually 0, 1, or 2, and wherein n m is 0 to 2;
  • wherein each R′ is individually H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl;
  • wherein R6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
  • wherein R7 and R8 are each individually II, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
  • In another embodiment, one R′ is at the ortho position relative to the carbon attached to the NR-L moiety.
  • In another embodiment, one R1 is H, F, Cl, Br, methyl, N(Me)2, NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
  • Additional embodiments include where R′ is optionally substituted with one or more substituents selected from the group consisting of methyl and acetyl.
  • In keeping with the embodiments above, R″ is 3,4-dihydroquinolin-2(1H)-onyl, indolin-2-onyl, 4,5-dihydro-1H-benzo[b]azepin-2(3H)-onyl, 2H-benzo[b][1,4]oxazin-3(4H)-onyl, 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-onyl, 1-(indolin-1-yl)ethanonyl, 1-methyl-1H-indolyl, 1-acetyl-2-methylindolinyl, 6-chloro-2-oxoindolinyl, 3,3-dichloro-2-oxoindolinyl, 7-((2-(dimethylamino)ethyl)(methyl)amino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinolinyl, 7-(3-(dimethylamino)pyrrolidin-1-yl)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(diethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, (S)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, or (R)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl.
  • In any of the embodiments above, R″ is
  • Figure US20130109672A1-20130502-C00003
  • wherein X3 is N or CH wherein when X3 is N, the NR-L moiety is linked to a C of the ring containing X3;
  • wherein R2 and R3 are each individually halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, B(OH)2, phenyl, C1-C4 alkyl substituted with one or more halogens, or taken together form alkylenedioxyl.
  • Another embodiment is where R2 and R3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH)2, or taken together form alkylenedioxyl.
  • In a particular embodiment of the compounds described above, R″ is 3,4-dimethylphenyl, 3-chlorophenyl, meta-tolyl, 3-methoxyphenyl, 3-fluorophenyl, 3-trifluoromethylphenyl, biphenyl-3-yl, pyridine-3-yl, 4-chlorophenyl, para-tolyl, 4-methoxyphenyl, 4-fluorophenyl, ortho-tolyl, 2-methoxyphenyl, 2-fluorophenyl, naphthalen-2-yl, naphthalen-1-yl, 2,3-dihydrobenzo[b][1,4]dioxin-6-yl, benzyl, 3-chloro-4-methylphenyl, 3,4-dichlorophenyl, 5-chloro-2-methylphenyl, 3-cyanophenyl, 3-chloro-2-methylphenyl, 3-phenylboronic acid, 4-fluoro-3-methylphenyl, 3-fluoro-4-methylphenyl, 4-chloro-3-methylphenyl, or 3-chloro-4-fluorophenyl.
  • In a particular embodiment, the compound of formula (I) is a compound of formula (II):
  • Figure US20130109672A1-20130502-C00004
  • wherein X1 and X2 are each individually O, NR6, or CR7R8;
  • wherein X3 is N or CH wherein when X3 is N, the NR-L moiety is linked to a C of the ring containing X3;
  • wherein any CH2—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a CH═CH moiety;
  • wherein any NH—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a N═CH moiety;
  • wherein any methylene of the ring containing X1 and X2 is optionally replaced by a carbonyl;
  • wherein n and m are each individually 0, 1, or 2, and wherein n+m is 0 to 2;
  • wherein k is 0 or 1;
  • wherein R1 is H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl;
  • wherein R2 and R3 are each individually H, halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, B(OH)2, phenyl, C1-C4 alkyl substituted with one or more halogens, or taken together form alkylenedioxy or phenyl fused to a CH:CH moiety of the ring containing X3;
  • wherein R6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
  • wherein R7 and R8 are each individually H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
  • In a particular embodiment of formula II, R1 is H, F, Cl, Br, methyl, N(Me)2, NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
  • In an embodiment of formula II, the moiety
  • Figure US20130109672A1-20130502-C00005
  • is optionally substituted with one or more substituents selected from the group consisting of methyl and acetyl.
  • In yet another embodiment of formula II, the moiety
  • Figure US20130109672A1-20130502-C00006
  • is 3,4-dihydroquinolin-2(1H)-onyl, indolin-2-onyl, 4,5-dihydro-1H-benzo[b]azepin-2(3H)-onyl, 2H-benzo[b][1,4]oxazin-3(4H)-onyl, 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-onyl, 1-(indolin-1-yl)ethanonyl, 1-methyl-1,1-indolyl, 1-acetyl-2-methylindolinyl, 6-chloro-2-oxoindoline, 3,3-dichloro-2-oxoindolinyl, 7-((2-(dimethylamino)ethyl)(methyl)amino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinolinyl, 7-(3-(dimethylamino)pyrrolidin-1-yl)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(diethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, (S)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, or (R)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl.
  • In another embodiment of formula II, R2 and R3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH)2, or taken together form alkylenedioxyl or phenyl fused to a CH:CH moiety of the ring containing X3.
  • In accordance with an embodiment of formula II, the moiety
  • Figure US20130109672A1-20130502-C00007
  • is 3,4-dimethylphenyl, 3-chlorophenyl, meta-tolyl, 3-methoxyphenyl, 3-fluorophenyl, 3-trifluoromethylphenyl, biphenyl-3-yl, pyridin-3-yl, 4-chlorophenyl, para-tolyl, 4-methoxyphenyl, 4-fluorophenyl, ortho-tolyl, 2-methoxyphenyl, 2-fluorophenyl, naphthalen-2-yl, naphthalen-1-yl, 2,3-dihydrobenzo[b][1,4]dioxin-6-yl, benzyl, 3-chloro-4-methylphenyl, 3,4-dichlorophenyl, 5-chloro-2-methylphenyl, 3-cyanophenyl, 3-chloro-2-methylphenyl, 3-phenylboronic acid, 4-fluoro-3-methylphenyl, 3-fluoro-4-methylphenyl, 4-chloro-3-methylphenyl, or 3-chloro-4-fluorophenyl.
  • Specific examples of the compound described above include
    • N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxo-2,3,4,5-tetrahydro-1H-benzo[b]azepine-7-sulfonamide,
    • N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-4-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxo-2,3-dihydro-1H-benzo[d]imidazole-5-sulfonamide,
    • N-(3,4-dimethylphenyl)-1,3-dimethyl-2-oxo-2,3-dihydro-1H-benzo[d]imidazole-5-sulfonamide,
    • 1-acetyl-N-(3,4-dimethylphenyl)indoline-5-sulfonamide,
    • N-(3,4-dimethylphenyl)-1-methyl-1H-indole-5-sulfonamide,
    • N-(3-chlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 2-oxo-N-m-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide.
    • N-(3-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 2-oxo-N-(3-(trifluoromethyl)phenyl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(biphenyl-3-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 2-oxo-N-(pyridin-3-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(4-chlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 2-oxo-N-p-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(4-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(4-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 2-oxo-N-o-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(2-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(2-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(naphthalen-2-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(naphthalen-1-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-benzyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 7-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-(methylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxo-7-(piperidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 6-chloro-N-(3,4-dimethyl phenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
    • 6-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
    • 6-chloro-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
    • 6-bromo-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
    • N-(3,4-dimethylphenyl)-6-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
    • N-(3,4-dimethylphenyl)-3-oxo-6-phenyl-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
    • N-(3,4-dimethylphenyl)-N-methyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-carboxamide,
    • N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dichlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 6-chloro-N-(5-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
    • N-(3-cyanophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 1-acetyl-N-(3,4-dimethylphenyl)-2-methyl indoline-5-sulfonamide,
    • N-(5-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 6-chloro-N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
    • N-(3-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3-chloro-4-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 3-(2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamido)phenylboronic acid,
    • N-(4-fluoro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dichlorophenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3-fluoro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(4-chloro-3-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 6-chloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
    • N-(4-chloro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 6-chloro-N-(3-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
    • 3,3-dichloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
    • 7-((2-(dimethylamino)ethyl)(methyl)amino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 7-(3-(dimethylamino)pyrrolidin-1-yl)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • 7-(diethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • (S)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • (R)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • N-(3,4-dimethylphenyl)-7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • (S)—N-(3-chloro-4-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
    • (S)—N-(4-fluoro-3-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide, and
    • (S)—N-(3-chloro-4-fluorophenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide.
  • In one embodiment of the invention, the compounds exclude a compound of formula (II) wherein R2 is a methyl in the para position relative to the NR-L moiety, R3 is a methyl in a meta position relative to the NR-L moiety, X3 is CH, k is zero, R is H, L is SO2, R1 is H, X1 is NR6, R6 is H, X2 is CR7R8, R7 and R8 are each H, n is 1, m is zero, and the methylene adjacent to X1 is replaced with a carbonyl.
  • Certain molecule modulators of human PK activity are known. Fructose-1,6-bis phosphate (FBP) (compound 1) is required to allosterically activate human PKM2, PKL, and PKR. NCGC00185916 (compound 2), NCGC00186527 (compound 3), and others listed in International Patent Application No. PCT/US09/60237 (incorporated herein by reference) also activate human PKM2.
  • Figure US20130109672A1-20130502-C00008
  • Referring now to terminology used generically herein, the term “alkyl” means a straight-chain or branched alkyl substituent containing from, for example, 1 to about 6 carbon atoms, preferably from 1 to about 4 carbon atoms, more preferably from 1 to 2 carbon atoms. Examples of such substituents include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, and the like. The term “alkoxyl” means any alkyl substituent attached as a substituent via an oxygen atom.
  • The term “alkenyl,” as used herein, means a linear alkenyl substituent containing at least one carbon-carbon double bond and from, for example, about 2 to about 6 carbon atoms (branched alkenyls are about 3 to about 6 carbons atoms), preferably from about 2 to about 5 carbon atoms (branched alkenyls are preferably from about 3 to about 5 carbon atoms), more preferably from about 3 to about 4 carbon atoms. Examples of such substituents include propenyl, isopropenyl, n-butenyl, sec-butenyl, isobutenyl, tert-butenyl, pentenyl, isopentenyl, hexenyl, and the like. “Alkylenedioxy” means a —O—(CH2)q—O— group, where q is from 1 to about 6, preferably from 1 to about 4, more preferably from 1 to 2.
  • The term “alkynyl,” as used herein, means a linear alkynyl substituent containing at least one carbon-carbon triple bond and from, for example, 2 to about 6 carbon atoms (branched alkynyls are about 3 to about 6 carbons atoms), preferably from 2 to about 5 carbon atoms (branched alkynyls are preferably from about 3 to about 5 carbon atoms), more preferably from about 3 to about 4 carbon atoms. Examples of such substituents include propynyl, isopropynyl, n-butynyl, sec-butynyl, isobutynyl, tert-butynyl, pentynyl, isopentynyl, hexynyl, and the like.
  • The term “cyclyl” as used herein encompasses cycloalkyl and cycloalkenyl. “Cycloalkyl” as used herein, means a cyclic alkyl substituent containing from, for example, about 3 to about 8 carbon atoms, preferably from about 4 to about 7 carbon atoms, and more preferably from about 4 to about 6 carbon atoms. Examples of such substituents include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like. The term “cycloalkenyl,” as used herein, means the same as the term “cycloalkyl,” however one or more double bonds are present. Examples include cyclopentenyl and cyclohexenyl. The cyclic alkyl groups may be unsubstituted or further substituted. Examples of substitutions include halogens, alkyoxy groups, and alkyl groups such as methyl groups, ethyl groups, and the like. “Cyclyl” also encompasses cycloalkyl and cycloalkenyl in which a heteroatom is exocyclic. The heteroatom, for example, may be N, O, or S. For example, a methylene group of the cyclyl can be replaced with a carbonyl. A cyclyl group may be fused to another ring, including another cyclyl, heterocyclyl, aryl, or heteroaryl. A fused bicyclic ring is any ring of cyclyl, heterocyclyl, aryl, or heteroaryl fused with another cyclyl, heterocyclyl, aryl, or heteroaryl.
  • The term “heteroaryl,” as used herein, refers to a monocyclic or bicyclic 5- or 6-membered aromatic ring system containing one or more heteroatoms selected from the group consisting of O, N, S, and combinations thereof. Examples of suitable monocyclic heteroarylgroups include but are not limited to furanyl, thiopheneyl, pyrrolyl, pyrazolyl, imidazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and triazinyl. The heteroaryl group can be attached at any available position on the heteroaryl group. For example, a thiopheneyl group can be attached at the 2-position or the 3-position of the thiopheneyl group. A pyridyl group can be attached at the 2-, 3-, or 4-position of the pyridyl group. Suitable bicyclic heterocycloaryl groups include monocylic heterocycloaryl rings fused to a C6-C10 aryl or heteroaryl ring. Non-limiting examples of bicyclic heterocycloaryl groups include benzofuran, benzothiophene, quinoline, and isoquinoline. The heteroaryl group is optionally substituted with 1, 2, 3, 4, or 5 substituents as recited herein, wherein the optional substituent can be present at any open position on the heteroaryl group.
  • The term “heteroaryl oxide,” as used herein, refers to an oxidized heteroaryl group as that term is defined herein, wherein one or more of the heteroatoms comprising the heteroaryl group is oxidized. Non-limiting examples of heteroaryl oxide groups include pyridine N-oxide, pyrimidine N-oxide, and pyrazine N-oxide.
  • The term “heterocyclyl” refers to a cyclic group, which may be aromatic or non-aromatic, or saturated or unsaturated, having one or more heteroatoms such as O, N, or S. Examples of heterocyclyl groups include pyridyl, piperidinyl, piperazinyl, pyrazinyl, pyrolyl, pyranyl, tetrahydropyranyl, tetrahydrothiopyranyl, pyrrolidinyl, furanyl, tetrahydrofuranyl, thiophenyl, tetrahydrothiophenyl, purinyl, pyrimidinyl, thiazolyl, thiazolidinyl, thiazolinyl, oxazolyl, triazolyl, tetrazolyl, tetrazinyl, benzoxazolyl, morpholinyl, thiophorpholinyl, quinolinyl, and isoquinolinyl. A heterocyclyl group may be fused to another ring, including a cyclyl, aryl, heteroaryl, or another heterocyclyl.
  • The term “halo” or “halogen,” as used herein, means a substituent selected from Group VIIA, such as, for example, fluorine, bromine, chlorine, and iodine.
  • The term “aryl” refers to an unsubstituted or substituted aromatic carbocyclic substituent, as commonly understood in the art, and the term “C6-C10 aryl” includes phenyl and naphthenyl. It is understood that the term aryl applies to cyclic substituents that are planar and comprise 4n+2π electrons, according to Hückel's Rule. An aryl group may be fused to another ring, including a cyclyl, heteroaryl, heterocyclyl, or another aryl.
  • A CH2—CH2 moiety is any ethylene moiety wherein there is a carbon-carbon single bond. A CH═CH moiety is any vinyl moiety that contains a carbon-carbon double bond. A NH—CH2 moiety contains a nitrogen-carbon single bond, and a N═CH contains a nitrogen-carbon double bond. A CH:CH moiety contains a carbon-carbon bond intermediate between a single and a double bond, such as in an aromatic system, for example in the carbon-carbon bonds in benzene or the nitrogen-carbon bond in pyridine.
  • The present invention also provides a pharmaceutical composition comprising a compound or salt of any of the embodiments described above and a pharmaceutically acceptable carrier.
  • The phrase “pharmaceutically acceptable salt” is intended to include nontoxic salts synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, Pa., 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977).
  • Suitable bases include inorganic bases such as alkali and alkaline earth metal bases, e.g., those containing metallic cations such as sodium, potassium, magnesium, calcium and the like. Non-limiting examples of suitable bases include sodium hydroxide, potassium hydroxide, sodium carbonate, and potassium carbonate. Suitable acids include inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic, methanesulfonic acid, benzenesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, maleic acid, tartaric acid, fatty acids, long chain fatty acids, and the like. Preferred pharmaceutically acceptable salts of inventive compounds having an acidic moiety include sodium and potassium salts. Preferred pharmaceutically acceptable salts of inventive compounds having a basic moiety (e.g., a pyridyl group) include hydrochloride and hydrobromide salts. The compounds of the present invention containing an acidic or basic moiety are useful in the form of the free base or acid or in the form of a pharmaceutically acceptable salt thereof.
  • It should be recognized that the particular counterion forming a part of any salt of this invention is usually not of a critical nature, so long as the salt as a whole is pharmacologically acceptable and as long as the counterion does not contribute undesired qualities to the salt as a whole.
  • It is further understood that the above compounds and salts may form solvates, or exist in a substantially uncomplexed form, such as the anhydrous form. As used herein, the term “solvate” refers to a molecular complex wherein the solvent molecule, such as the crystallizing solvent, is incorporated into the crystal lattice. When the solvent incorporated in the solvate is water, the molecular complex is called a hydrate. Pharmaceutically acceptable solvates include hydrates, alcoholates such as methanolates and ethanolates, acetonitrilates and the like. These compounds can also exist in polymorphic forms.
  • The invention contemplates embodiments in which a compound having a chiral center is a substantially pure enantiomer thereof, a racemic mixture thereof, or a mixture containing any proportion of the two enantiomers thereof. The invention also contemplates all stereoisomers and diastereoisomers of the compounds described herein.
  • The present invention is further directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one compound or salt described herein.
  • It is preferred that the pharmaceutically acceptable carrier be one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
  • The choice of carrier will be determined in part by the particular compound of the present invention chosen, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following formulations for oral, aerosol, parenteral, subcutaneous, intra-arterial, intravenous, intramuscular, intraperitoneal, intrathecal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
  • The pharmaceutical composition can be administered parenterally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly. Thus, the invention provides compositions for parenteral administration that comprise a solution of the inventive compound or salt dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous isotonic sterile injection solutions.
  • Overall, the requirements for effective pharmaceutical carriers for parenteral compositions are well known to those of ordinary skill in the art. See, e.g., Banker and Chalmers, eds., Pharmaceutics and Pharmacy Practice, J. B. Lippincott Company, Philadelphia, pp. 238-250 (1982), and Toissel, ASHP Handbook on Injectable Drugs, 4th ed., pp. 622-630 (1986). Such solutions can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound or salt of the present invention may be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl-1,3-dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants.
  • Oils useful in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • The parenteral formulations can contain preservatives and buffers. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • Topical formulations, including those that are useful for transdermal drug release, are well-known to those of skill in the art and are suitable in the context of the invention for application to skin.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as a therapeutically effective amount of the inventive compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules, (c) powders, (d) suspensions in an appropriate liquid, and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • The compound or salt of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. The compounds are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of active compound are 0.01%-20% by weight, preferably 1%-10%. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. The surfactant may constitute 0.1%-20% by weight of the composition, preferably 0.25%-5%. The balance of the composition is ordinarily propellant. A carrier can also be included as desired, e.g., lecithin for intranasal delivery. These aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations may be used to spray mucosa.
  • Additionally, the compound or salt of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • It will be appreciated by one of ordinary skill in the art that, in addition to the aforedescribed pharmaceutical compositions, the compound or salt of the present invention may be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes. Liposomes serve to target the compounds to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of the inventive compound. Liposomes useful in the present invention include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations, the active agent to be delivered is incorporated as part of a liposome, alone or in conjunction with a suitable chemotherapeutic agent. Thus, liposomes filled with a desired inventive compound or salt thereof, can be directed to the site of a specific tissue type, hepatic cells, for example, where the liposomes then deliver the selected compositions. Liposomes for use in the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, for example, liposome size and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, for example, Szoka et al., Ann. Rev. Biophys. Bioeng., 9, 467 (1980), and U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369. For targeting to the cells of a particular tissue type, a ligand to be incorporated into the liposome can include, for example, antibodies or fragments thereof specific for cell surface determinants of the targeted tissue type. A liposome suspension containing a compound or salt of the present invention may be administered intravenously, locally, topically, etc. in a dose that varies according to the mode of administration, the agent being delivered, and the stage of disease being treated.
  • Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. The present inventive method typically will involve the administration of about 0.001 to about 300 mg of one or more of the compounds described above per kg body weight of the individual. The administration can involve about 0.001 mg, about 0.01 mg, about 0.1 mg, about 1 mg, about 5 mg, about 10 mg, about 20 mg, about 50 mg, about 100 mg, about 200 mg, or about 300 mg or more of one or more of the compounds described above per kg body weight of the individual. Alternatively, or in addition, the administration can involve about 300 mg, about 200 mg, about 100 mg, about 50 mg, about 20 mg, about 10 mg, about 5 mg, about 1 mg, about 0.1 mg, about 0.01 mg, or about 0.001 mg or less of one or more of the compounds described above per kg body weight of the individual. Thus, the administration can be bounded by any two of the aforementioned endpoints. For example, the administration can be about 0.001 mg to about 200 mg, about 0.001 mg to about 1 mg, about 0.01 mg to about 50 mg, about 0.1 mg to about 20 mg, about 1 mg to about 10 mg, about 1 mg to about 20 mg, about 10 mg to about 50 mg, or any other combination of endpoints, of one or more of the compounds described above per kg body weight of the individual.
  • The present invention further provides a method of treating a disease responsive to activation of human PK-M2 comprising administering to a patient in need thereof a therapeutically effective amount of any of the compounds described herein or a pharmaceutically acceptable salt thereof.
  • The invention further provides any of the compounds described herein or a pharmaceutically acceptable salt thereof for use in treating a disease responsive to activation of human PK-M2.
  • The invention further provides the use of a compound or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating a disease responsive to activation of human PK-M2 of a patient, wherein the compound is any of the compounds described herein or a pharmaceutically acceptable salt thereof
  • The disease responsive to activation of PK-M2 can be caused by or associated with, e.g., the function PKM2. These diseases may include, e.g., cancer, obesity, diabetes, atherosclerosis, restenosis, autoimmune diseases, and proliferation-dependent diseases.
  • Cancers include, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic (myeloid) leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic (myeloid) leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (e.g., Hodgkin's disease or non-Hodgkin's disease), Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, (malignant) mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma (including alveolar), colon carcinoma (colon cancer), pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct/intrahepatic bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma (lung cancer), small cell lung carcinoma, bladder carcinoma (urinary bladder cancer), epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). The cancers may include bone cancer, brain cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the oral cavity, cancer of the vulva, esophageal cancer, gastrointestinal carcinoid tumor, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, nasopharynx cancer, non-small cell lung cancer, peritoneum, omentum, and mesentery cancer, pharynx cancer, rectal cancer, renal cancer, small intestine cancer, soft tissue cancer, stomach cancer, thyroid cancer, and ureter cancer.
  • Other diseases include diabetes and obesity. Adipose tissue expresses PKM2. Additionally, activators of PKM2, described herein, may be useful in the treatment of type II diabetes, as the activation of PKM2 may allow for decreased lipid production and increased oxidative phosphorylation in adipose tissue. This effect should decrease adiposity, which is known to contribute to type 2 diabetes.
  • Additionally diseases include autoimmune diseases and proliferative diseases. Activators of PKM2, described herein, may be used to treat, e.g., autoimmune diseases or proliferative diseases. Autoimmune disorders include, e.g., type I diabetes, Crohn's disease, multiple sclerosis, arthritis, rheumatoid arthritis, systemic lupus erythematosus, autoimmune (Hashimoto's) thyroiditis, autoimmune liver diseases (e.g., hepatitis and primary biliary cirrhosis), hyperthyroidism (e.g., Graves' disease and thyrotoxicosis), insulin-resistant diabetes, autoimmune adrenal insufficiency (e.g., Addison's disease), autoimmune oophoritis, autoimmune orchitis, autoimmune hemolytic anemia, paroxysmal cold hemoglobinuria, Behcet's disease, autoimmune thrombocytopenia, autoimmune neutropenia, pernicious anemia, pure red cell anemia, autoimmune coagulopathies, myasthenia gravis, experimental allergic encephalomyelitis, autoimmune polyneuritis, pemphigus and other bullous diseases, rheumatic carditis, Goodpasture's syndrome, postcardiotomy syndrome, Sjogren's syndrome, polymyositis, dermatomyositis, and scleroderma. Autoimmune disorders are described in U.S. Pat. Nos. 5,891,435 and 6,773,705, hereby incorporated by reference.
  • Proliferative diseases include, e.g., cancer (e.g., benign and malignant), benign prostatic hyperplasia, psoriasis, abnormal keratinization, lymphoproliferative disorders (e.g., a disorder in which there is abnormal proliferation of cells of the lymphatic system), chronic rheumatoid arthritis, arteriosclerosis, restenosis, and diabetic retinopathy. Proliferative diseases are described in U.S. Pat. Nos. 5,639,600 and 7,087,648, hereby incorporated by reference.
  • The terms “treat” and “prevent,” as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete treatment or prevention. Rather, there are varying degrees of treatment or prevention of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the inventive methods can provide any amount of any level of treatment or prevention in a mammal. Furthermore, the treatment or prevention provided by the inventive method can include treatment or prevention of one or more conditions or symptoms of the diseases described herein being treated or prevented. Also, for purposes herein, “prevention” can encompass delaying the onset of the disease, or a symptom or condition thereof.
  • The invention further provides a use of a compound or salt of the invention in the manufacture of a medicament for treating disease responsive to activation of PK-M2. The medicament typically is a pharmaceutical composition as described herein.
  • One skilled in the art will appreciate that suitable methods of utilizing a compound and administering it to a human for the treatment of disease states, in particular, diseases responsive to activation of PK-M2, which would be useful in the method of the present invention, are available. Although more than one route can be used to administer a particular compound, a particular route can provide a more immediate and more effective reaction than another route. Accordingly, the described methods are merely exemplary and are in no way limiting.
  • The dose administered to a human in accordance with the present invention should be sufficient to effect the desired response. Such responses include reversal or prevention of the bad effects of the disease responsive to activation of PK-M2 for which treatment is desired or to elicit the desired benefit. One skilled in the art will recognize that dosage will depend upon a variety of factors, including the age, condition, and body weight of the human, as well as the source, particular type of the cancer, and extent of cancer in the human. The size of the dose will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular compound and the desired physiological effect. It will be appreciated by one of skill in the art that various conditions or disease states may require prolonged treatment involving multiple administrations.
  • The compounds of the invention can be prepared as follows. For example, the synthetic elaboration of substituted 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides can begin with a standard coupling between commercially available sulfonyl chlorides and substituted anilines, according to Scheme I.
  • Figure US20130109672A1-20130502-C00009
  • Conditions and reagents: (i) Hunig's base, DMF, rt, 1 h; (ii) Ar1—B(OH)2, tetrakis (3 mol %), Na2CO3, 1,2,-DME/H2O, μW, 120° C., 20 min; (iii) R′″N, MeCN, μW, 180° C., 1 h.
  • The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
  • Example 1
  • This example illustrates methods used in preparing exemplary compounds of the invention.
  • General Methods Used for all Exemplary Compounds.
  • All air or moisture sensitive reactions were performed under positive pressure of nitrogen with oven-dried glassware. Anhydrous solvents such as tetrahydrofuran (THF), toluene, dichloromethane, N,N-dimethylforamide (DMF), acetonitrile, methanol and triethylamine were obtained from Sigma-Aldrich (St. Louis, Mo., USA). Preparative purification was performed on a Waters® (Milford, Mass., USA) semi-preparative HPLC. The column used was a Phenomenex (Torrance, Calif., USA) Luna C18 (5 micron, 30×75 mm) at a flow rate of 45 mL/min. The mobile phase consisted of acetonitrile and water (each containing 0.1% trifluoroacetic acid). A gradient of 10% to 50% acetonitrile over 8 minutes was used during the purification. Fraction collection was triggered by UV detection (220 nM). Analytical analysis was performed on an Agilent LC/MS (Agilent Technologies, Santa Clara, Calif., USA).
  • Method 1: A 7 minute gradient of 4% to 100% Acetonitrile (containing 0.025% trifluoroacetic acid) in water (containing 0.05% trifluoroacetic acid) was used with an 8 minute run time at a flow rate of 1 mL/min. A Phenomenex Luna C18 column (3 micron, 3×75 mm) was used at a temperature of 50° C.
  • Method 2: A 3 minute gradient of 4% to 100% Acetonitrile (containing 0.025% trifluoroacetic acid) in water (containing 0.05% trifluoroacetic acid) was used with a 4.5 minute run time at a flow rate of 1 mL/min. A Phenomenex Gemini Phenyl column (3 micron, 3×100 mm) was used at a temperature of 50° C.
  • Purity determination was performed using an Agilent Diode Array Detector. Mass determination was performed using an Agilent 6130 mass spectrometer with electrospray ionization in the positive mode. 1H NMR spectra were recorded on Varian (Palo Alto, Calif., USA) 400 MHz spectrometers. Chemical Shifts are reported in ppm with tetramethylsilane (TMS) as internal standard (0 ppm) for CDCl3 solutions or undeuterated solvent (DMSO-H6 at 2.49 ppm) for DMSO-d6 solutions. All of the analogues for assay have purity greater than 95% based on both analytical methods. High resolution mass spectrometry was recorded on Agilent 6210 Time-of-Flight LC/MS system. Confirmation of molecular formula was accomplished using electrospray ionization in the positive mode with the Agilent Masshunter software (version B.02).
  • General Procedure for Synthesis of Compounds 4-31, 35, 37-39, 41, and 43-60
  • The methods of synthesizing compound 4 were generally followed for all compounds 4-31, 35, 37-39, 41, and 43-60.
  • Compound 4. N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00010
  • 2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonyl chloride (0.2 g, 0.814 mmol) was dissolved in DMF (2 ml) and 3,4-dimethylaniline (0.118 g, 0.977 mmol) was added followed by the dropwise addition of DIPEA (0.213 ml, 1.221 mmol). The reaction was stirred at RT for 1 h then purified by directly injecting to a Waters® reverse phase purification system.
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.40 (s, 1H), 9.91 (br. s., 1H), 7.56 (s, 1H), 7.51 (dd, J=8.41, 1.96 Hz, 1H), 6.95 (d, J=8.22 Hz, 1H), 6.90 (d, J=8.22 Hz, 1H), 6.86 (s, 1H), 6.81 (dd, J=8.02, 1.96 Hz, 1H), 2.90 (t, J=7.53 Hz, 2H), 2.46 (t, J=7.63 Hz, 2H), 2.10 (s, 3H), 2.08 (s, 3H). LC/MS: Method 1, retention time: 5.744 min; HRMS: m/z (M+)=454.0872 (Calculated for C19H22N2O7S2=454.0868).
  • Compound 5. N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide
  • Figure US20130109672A1-20130502-C00011
  • 2-oxoindoline-5-sulfonyl chloride (0.189 g, 0.814 mmol) was dissolved in DMF (2 ml) and 3,4-dimethylaniline (0.118 g, 0.977 mmol) was added followed by the dropwise addition of DIPEA (0.213 ml, 1.221 mmol). The reaction was stirred at RT for 1 h then purified by directly injecting to a Waters® reverse phase purification system.
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.70 (s, 1H), 9.87 (s, 1H), 7.44-7.62 (m, 2H), 6.67-6.98 (m, 4H), 3.50 (s, 2H), 2.05 (m, 6H). LC/MS: Method 1, retention time: 4.833 min; HRMS: m/z (M+)=316.0878 (Calculated for C16H16N2O3S=316.0882).
  • Compound 6. N-(3,4-dimethylphenyl)-2-oxo-2,3,4,5-tetrahydro-1H-benzo[b]azepine-7-sulfonamide
  • Figure US20130109672A1-20130502-C00012
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.89 (br. s., 1H), 9.77 (br. s., 1H), 7.38-7.67 (m, 2H), 7.00 (d, J=8.2 Hz, 1H), 6.93 (d, J=8.0 Hz, 1H), 6.61-6.86 (m, 2H), 2.57-2.72 (m, 2H), 1.93-2.22 (m, 10H). LC/MS: Method 1, retention time: 5.081 min; HRMS: m/z (M+)=344.1195 (Calculated for C18H20N2O3S=344.1195).
  • Compound 7. N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide
  • Figure US20130109672A1-20130502-C00013
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.90 (br. s., 1H), 9.97 s., 1H), 7.17-7.32 (m, 2H), 7.01 (d, J=8.8 Hz, 1H), 6.94 (d, J=8.2 Hz, 1H), 6.83 (d, J=1.8 Hz, 1H), 6.76 (dd, J=8.1, 2.1 Hz, 1H), 4.62 (s, 2H), 2.07 (m, 6H). LC/MS: Method 1, retention time: 5.133 min; HRMS: m/z (M+)=332.0823 (Calculated for C16H16N2O4S=332.0831).
  • Compound 8. N-(3,4-dimethylphenyl)-4-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide
  • Figure US20130109672A1-20130502-C00014
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.93 (s, 1H), 7.25-7.43 (m, 2H), 7.06 (d, J=8.4 Hz, 1H), 6.94 (d, J=8.0 Hz, 1H), 6.86 (s, 1H), 6.79 (dd, J=8.0, 2.2 Hz, 1H), 4.70 (s, 2H), 3.19 (s, 3H), 2.07 (m, 6H). LC/MS: Method 1, retention time: 5.519 min; HRMS: m/z (M+)=346.0989 (Calculated for C17H18N2O4S=346.0987).
  • Compound 9. N-(3,4-dimethylphenyl)-2-oxo-2,3-dihydro-1H-benzo[d]imidazole-5-sulfonamide
  • Figure US20130109672A1-20130502-C00015
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 11.02 (br. s., 1H), 10.89 (br. s., 1H), 9.83 (br. s., 1H), 7.31 (dd, J=8.2, 1.6 Hz, 1H), 7.21 (d, J=1.6 Hz, 1H), 6.96 (d, J=8.2 Hz, 1H), 6.90 (d, J=8.2 Hz, 1H), 6.81 (s, 1H), 6.75 (dd, J=8.0, 2.0 Hz, 1H), 2.05 (m, 6H). LC/MS: Method 1, retention time: 4.535 min; HRMS: m/z (M+)=317.0837 (Calculated for C15H15N3O3S=317.0834).
  • Compound 10. N-(3,4-dimethylphenyl)-1,3-dimethyl-2-oxo-2,3-dihydro-1,1-benzo[d]imidazole-5-sulfonamide
  • Figure US20130109672A1-20130502-C00016
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.87 (s, 1H), 7.33-7.52 (m, 2H), 7.21 (d, J=8.0 Hz, 1H), 6.90 (d, J=8.0 Hz, 1H), 6.85 (d, J=1.8 Hz, 1H), 6.77 (dd, J=8.1, 2.1 Hz, 1H), 3.23-3.37 (s, 6H), 2.05 (m, 6H). LC/MS: Method 1, retention time: 5.179 min; HRMS: m/z (M+)=345.1152 (Calculated for C17H19N3OS=345.1147).
  • Compound 11. 1-acetyl-N-(3,4-dimethylphenyl)indoline-5-sulfonamide
  • Figure US20130109672A1-20130502-C00017
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.86 (s, 1H), 8.02 (d, J=8.8 Hz, 1H), 7.41-7.56 (m, 2H), 6.91 (d, J=8.2 Hz, 1H), 6.82 (s, 1H), 6.75 (dd, J=7.9, 1.9 Hz, 1H), 4.08 (t, J=8.5 Hz, 2H), 3.11 (t, J=8.4 Hz, 2H), 2.12 (s, 3H), 2.05 (m, 6H). LC/MS: Method 1, retention time: 5.338 min; HRMS: m/z (M+)=344.1196 (Calculated for C18H20N2O3S=344.1195).
  • Compound 12. N-(3,4-dimethylphenyl)-1-methyl-1H-indole-5-sulfonamide
  • Figure US20130109672A1-20130502-C00018
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 9.82 (s, 1H), 7.96 (d, J=1.2 Hz, 1H), 7.40-7.55 (m, 3H), 6.80-6.91 (m, 2H), 6.72-6.80 (m, 1H), 6.55 (d, J=2.9 Hz, 1H), 3.76 (s, 3H), 2.01 (m, 6H). Method 1, retention time: 5.895 min; HRMS: m/z (M+)=314.1095 (Calculated for C17H18N2O2S=314.1089).
  • Compound 13. N-(3-chlorophenyl)-2-oxo-1,2,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00019
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.41 (s, 2H), 7.46-7.63 (m, 2H), 7.22 (t, J=8.1 Hz, 1H), 6.98-7.13 (m, 3H), 6.90 (d, J=8.4 Hz, 1H), 2.89 (t, J=7.6 Hz, 2H), 2.35-2.48 (m, 2H). LC/MS: Method 1, retention time: 4.933 min; HRMS: m/z (M+)=336.0333 (Calculated for C15H13ClN2O3S=336.0335).
  • Compound 14. 2-oxo-N-m-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00020
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (s, 1H), 10.03 (s, 1H), 7.41-7.61 (m, 2H), 7.06 (t, J=7.8 Hz, 1H), 6.82-6.93 (m, 3H), 6.78 (d, J=7.2 Hz, 1H), 2.87 (t, J=7.6 Hz, 2H) 2.39-2.45 (t, J=7.8 Hz, 2H), 2.16 (s, 3H). LC/MS: Method 1, retention time: 4.741 min; HRMS: m/z (M+)=316.0886 (Calculated for C16H16N2O3S=316.0882).
  • Compound 15. N-(3-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00021
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (s, 1H), 10.12 (s, 1H), 7.45-7.60 (m, 2H), 7.08 (t, J=8.3 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 6.60-6.69 (m, 2H), 6.44-6.60 (m, 1H), 3.62 (s, 3H), 2.88 (t, J=7.6 Hz, 2H), 2.39-2.45 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 4.513 min; HRMS: m/z (M+)=332.0830 (Calculated for C16H16N2O4S=332.0831).
  • Compound 16. N-(3-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00022
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.28-10.52 (m, 2H), 7.43-7.65 (m, 2H), 7.13-7.31 (m, 1H), 6.85-6.99 (m, 3H), 6.71-6.85 (m, 1H), 2.89 (t, J=7.6 Hz, 2H), 2.36-2.46 (t, J=7.1 Hz, 2H). LC/MS: Method 1, retention time: 4.650 min; m/z (M+)=320.0628 (Calculated for C15H13FN2O3S=320.0631).
  • Compound 17. 2-oxo-N-(3-(trifluoromethyl)phenyl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00023
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.56 (s, 1H), 10.41 (s, 1H), 7.51-7.64 (m, 2H), 7.45 (t, J=8.1 Hz, 1H), 7.24-7.39 (m, 3H), 6.90 (d, J=8.4 Hz, 1H), 2.88 (t, J=7.6 Hz, 2H), 2.37-2.45 (t, J=7.9 Hz, 2H). LC/MS: Method 1, retention time: 5.151 min; HRMS: m/z (M+)=370.0596 (Calculated for C16N13F3N2O3S=370.0599).
  • Compound 18. N-(biphenyl-3-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00024
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (s, 1H), 10.23 (br. s., 1H), 7.51-7.65 (m, 2H), 7.44 (dt, J=15.0, 7.6 Hz, 4H), 7.16-7.38 (m, 4H), 7.06 (dd, J=6.8, 1.6 Hz, 1H), 6.90 (d, 18.4 Hz, 1H), 2.88 (t, J=7.5 Hz, 2H), 2.42 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 5.406 min; HRMS: m/z (M+)=378.1039 (Calculated for C21H18N2O3S=378.1038).
  • Compound 19. 2-oxo-N-(pyridin-3-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00025
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.33-10.53 (m, 2H), 8.15-8.33 (m, 2H), 7.57 (s, 1H), 7.46-7.54 (m, 2H), 7.29 (dd, J=8.2, 4.7 Hz, 1H), 6.90 (d, 1=8.4 Hz, 1H), 2.89 (t, J=7.6 Hz, 2H), 2.39-2.44 (t, J=7.4 Hz, 2H). LC/MS: Method 1, retention time: 2.984 min; HRMS: m/z (M+)=303.0683 (Calculated for C14H13N3O3S=303.0678).
  • Compound 20. N-(4-chlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00026
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.40 (s, 1H), 10.27 (s, 1H), 7.45-7.60 (m, 2H), 7.20-7.33 (m, 2H), 7.06 (d, J=8.8 Hz, 2H), 6.89 (d, J=8.4 Hz, 1H), 2.88 (t, J=7.6 Hz, 2H), 2.37-2.47 (t. J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 4.938 min; HRMS: m/z (M+)=336.0328 (Calculated for C15H13ClN2O3S=336.0335).
  • Compound 21. 2-oxo-N-p-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00027
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.37 (s, 1H), 9.93 (s, 1H), 7.52 (s, 1H), 7.46 (dd, 0.1=8.4, 2.0 Hz, 1H), 6.91-7.05 (m, 4H), 6.86 (d, J=8.4 Hz, 1H), 2.87 (t, d=7.6 Hz, 2H), 2.38-2.44 (t, J=7.5 Hz, 2H), 2.15 (s, 3H). LC/MS: Method 1, retention time: 4.747 min; HRMS: m/z (M+)=316.0879 (Calculated for C16H16N2O3S=316.0882).
  • Compound 22. N-(4-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00028
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.37 (s, 1H), 9.72 (s, 1H), 7.47 (s, 1H), 7.40 (dd, J=8.3, 2.1 Hz, 1H), 6.90-6.97 (m, 2H), 6.86 (d, J=8.2 Hz, 1H), 6.68-6.83 (m, 2H), 3.63 (s, 3H), 2.86 (t, J=7.5 Hz, 2H), 2.36-2.47 (t, J=7.6 Hz, 2H). LC/MS: Method 1, retention time: 4.422 min; HRMS: m/z (M+)=332.0830 (Calculated for C16H16N2O4S=332.0831).
  • Compound 23. N-(4-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00029
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.39 (s, 1H), 10.04 (s, 1H), 7.50 (s, 1H), 7.45 (dd, J=8.3, 2.1 Hz, 1H), 7.04 (d, J=6.7 Hz, 4H), 6.87 (d, J=8.4 Hz, 1H), 2.87 (t, J=7.6 Hz, 2H), 2.38 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 4.580 min; HRMS: m/z (M+)=320.0633 (Calculated for C15H13FN2O3S=320.0631).
  • Compound 24. 2-oxo-N-o-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00030
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.40 (s, 1H), 9.33 (br. s., 1H), 7.32-7.50 (m, 2H), 6.96-7.14 (m, 3H), 6.83-6.96 (m, 2H), 2.86 (t, J=7.5 Hz, 2H), 2.40 (t, J=7.4 Hz, 2H), 2.01 (s, 3H). LC/MS: Method 1, retention time: 4.656 min; HRMS: m/z (M+)=316.0870 (Calculated for C16H16N2O3S=316.0882).
  • Compound 25. N-(2-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00031
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.36 (s, 1H), 9.21 (s, 1H), 7.50 (s, 1H), 7.43 (dd, J=8.4, 2.0 Hz, 1H), 7.16 (dd, J=7.8, 1.4 Hz, 1H), 7.06 (td, J=7.8, 1.6 Hz, 1H), 6.70-6.92 (m, 3H), 3.50 (s, 3H), 2.86 (t, J=7.5 Hz, 2H), 2.41 (t, J=7.2 Hz, 2H). LC/MS: Method 1, retention time: 4.580 min; HRMS: m/z (M+) 332.0833 (Calculated for C16H16N2O4S=332.0831).
  • Compound 26. N-(2-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00032
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.40 (s, 1H), 9.95 (s, 1H), 7.42-7.54 (m, 2H), 7.20 (t, J=7.9 Hz, 1H), 7.02-7.16 (m, 3H), 6.89 (d, J=8.4 Hz, 1H), 2.87 (t, J=7.5 Hz, 2H), 2.38 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 4.455 min; m/z (M+)=320.0629 (Calculated for C15H13FN2O3S=320.0631).
  • Compound 27. N-(naphthalen-2-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00033
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.35 (br. s., 2H), 7.66-7.82 (m, 3H), 7.48-7.66 (m, 3H), 7.32-7.48 (m, 2H), 7.26 (dd, J=8.8, 2.0 Hz, 1H), 6.86 (d, J=8.2 Hz, 1H), 2.85 (t, J=7.5 Hz, 2H), 2.40 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 5.077 min; HRMS: m/z (M+)=352.0883 (Calculated for C19H16N2O3S=352.0882).
  • Compound 28. N-(naphthalen-1-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00034
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.36 (s, 1H), 10.01 (s, 1H), 8.01 (d, J=8.2 Hz, 1H), 7.84 (d, J=7.8 Hz, 1H), 7.73 (d, J=8.2 Hz, 1H), 7.28-7.52 (m, 5H), 7.12 (d, J=7.2 Hz, 1H), 6.83 (d, J=8.2 Hz, 1H), 2.81 (t, J=7.5 Hz, 2H), 2.40 (t, J=8.1 Hz, 2H). LC/MS: Method 1, retention time: 4.938 mm; HRMS: m/z (M+)=352.0883 (Calculated for C19H16N2O3S=352.0882).
  • Compound 29. N-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00035
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (s, 1H), 9.78 (s, 1H), 7.39-7.54 (m, 2H), 6.88 (d, J=8.4 Hz, 1H), 6.67 (d, J=8.6 Hz, 1H), 6.42-6.58 (m, 2H), 4.12 (m, 4H), 2.88 (t, J=7.6 Hz, 2H), 2.39 (t, J=7.8 Hz, 2H). LC/MS: Method 1, retention time: 4.363 min; HRMS: m/z (M+)=360.0781 (Calculated for C17H16N2O5S=360.0780).
  • Compound 30. N-benzyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00036
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (s, 1H), 7.93 (t, J=6.4 Hz, 1H), 7.47-7.58 (m, 2H), 7.11-7.34 (m, 5H), 6.91 (d, J=8.0 Hz, 1H), 3.92 (d, J=6.3 Hz, 2H), 2.88 (t, J=7.5 Hz, 2H), 2.39 (t, J=7.5 Hz, 2H). LC/MS: Method 1, retention time: 4.528 min; HRMS: m/z (M+)=316.0882 (Calculated for C16H16N2O3S=316.0882).
  • Compound 31. N-(3,4-dimethylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00037
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.42 (s, 1H), 10.14 (s, 1H), 7.56 (d, J=7.6 Hz, 1H), 6.88-7.02 (m, 1H), 6.75-6.88 (m, 2H), 6.67 (d. J=11.3 Hz, 1H), 2.85 (t, J=7.5 Hz, 2H), 2.37 (t, J=7.2 Hz, 2H), 2.06 (m, 6H). LC/MS: Method 1, retention time: 5.105 min; HRMS: m/z (M+)=348.0949 (Calculated for C17H17FN2O3S=348.0944).
  • Compound 35. 6-chloro-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide
  • Figure US20130109672A1-20130502-C00038
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.38 (br. s., 1H), 10.12 (br. s., 1H), 7.76 (s, 1H), 6.60-7.13 (m, 4H), 2.81 (t, J=7.5 Hz, 2H), 2.39 (t, J=7.7 Hz, 2H), 2.03 (m, 6H)LC/MS: Method 1, retention time: 5.188 min; HRMS: m/z (M+)=364.0652 (Calculated for C17H17ClN2O3S=364.0648).
  • Compound 37. 6-chloro-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide
  • Figure US20130109672A1-20130502-C00039
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 11.05 (s, 1H), 10.25 (s, 1H), 7.39 (s, 1H), 6.69-7.13 (m, 4H), 4.62 (s, 2H), 2.05 (m, 6H). LC/MS: Method 1, retention time: 5.308 min; HRMS: m/z (M+)=366.0446 (Calculated for C16H15ClN2O4S=366.0441).
  • Compound 38. 6-bromo-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide
  • Figure US20130109672A1-20130502-C00040
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.89 (s, 1H), 10.21 (s, 1H), 7.57 (s, 1H), 7.32 (s, 1H), 6.94 (d, J=8.2 Hz, 1H), 6.85 (s, 1H), 6.77 (dd, J=8.1, 2.1 Hz, 1H), 4.65 (s, 2H), 2.06 (d, 6H). LC/MS: Method 1, retention time: 5.394 min; HRMS: m/z (M+)=409.9939 (Calculated for C16H15BrN2O4S=409.9936).
  • Compound 39. N-(3,4-dimethylphenyl)-6-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide
  • Figure US20130109672A1-20130502-C00041
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.93 (s, 1H), 10.04 (br. s., 1H), 7.29 (s, 1H), 6.92 (d, J=8.2 Hz, 1H), 6.80 (s, 1H), 6.66-6.79 (m, 2H), 4.56 (s, 2H), 2.41 (s, 3H), 2.11 (m, 6H). LC/MS: Method 1, retention time: 5.202 min. HRMS: m/z (M+)=346.0993 (Calculated for C17H18N2O4S=346.0987).
  • Compound 41. N-(3,4-dimethylphenyl)-N-methyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00042
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.44 (br. s., 1H), 7.32 (s, 1H), 7.17-7.28 (m, 1H), 6.98-7.16 (m, 1H), 6.83-6.98 (m, 2H), 6.72 (d, J=7.8 Hz, 1H), 3.02 (s, 3H), 2.88 (t, J=7.52 Hz, 2H), 2.46 (t, J=7.61 Hz, 2H), 2.15 (s, 3H), 2.12 (s, 3H). LC/MS: Method 1, retention time: 5.457 min; Method 2, retention time: 3.889 min. HRMS: m/z (M+)=344.1199 (Calculated for C18H20N2O3S2=344.1195).
  • Compound 43. N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00043
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.41 (s, 1H), 10.24 (br. s., 1H), 7.55 (s, 1H), 7.51 (dd, J=8.4, 2.0 Hz, 1H), 7.17 (d, J=8.2 Hz, 1H), 7.08 (d, J=2.2 Hz, 1H), 6.95 (dd, J=8.2, 2.2 Hz, 1H), 6.90 (d, J=8.4 Hz, 1H), 2.89 (t, J=7.6 Hz, 2H), 2.41 (t, J=7.4 Hz, 2H), 2.17 (s, 3H). Method 1, retention time: 5.150 min. HRMS: m/z (M+)=350.0489 (Calculated for C16H15ClN2O3S=350.0492).
  • Compound 44. N-(3,4-dichlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00044
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.54 (s, 1H), 10.42 (s, 1H), 7.51-7.61 (m, 2H), 7.46 (d, J=8.8 Hz, 1H), 7.24 (d, J=2.5 Hz, 1H), 7.05 (dd, J=8.7, 2.4 Hz, 1H), 6.92 (d, J=8.4 Hz, 1H), 2.90 (t, J=7.6 Hz, 3H), 2.49 (t, J=7.6 Hz, 3H). Method 1, retention time: 5.279 min. HRMS: m/z (M+)=369.9942 (Calculated for C15H12Cl2N2O3S=369.9942).
  • Compound 45. 6-chloro-N-(5-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide
  • Figure US20130109672A1-20130502-C00045
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.47 (s, 1H), 9.89 (hr. s., 1H), 7.66 (s, 1H), 7.09-7.22 (m, 2H), 7.06 (d, J=2.0 Hz, 1H), 7.00 (s, 1H), 2.87 (t, J=7.6 Hz, 2H), 2.39-2.46 (t, J=7.3 Hz, 2H); Method 1, retention time: 5.328 min. HRMS: m/z (M+)=384.0106 (Calculated for C16H14C12N2O3S=384.0102).
  • Compound 46. N-(3-cyanophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00046
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.60 (s, 1H), 10.41 (s, 1H), 7.51-7.63 (m, 2H), 7.30-7.51 (m, 4H), 6.90 (d, J=8.2 Hz, 1H), 2.89 (t, J=7.5 Hz, 2H), 2.40-2.44 (t, J=7.5 Hz, 2H); Method 1, retention time: 4.389 min. HRMS: m/z (M+)=327.0672 (Calculated for C26H13N3O3S=327.0678).
  • Compound 47. 1-acetyl-N-(3,4-dimethylphenyl)-2-methylindoline-5-sulfonamide
  • Figure US20130109672A1-20130502-C00047
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 9.88 (br. s., 1H), 7.99 (br. s., 1H), 7.48-7.71 (m, 2H), 6.93 (d, J=6.1 Hz, 1H), 6.71-6.88 (m, 2H), 4.62 (br. s., 1H), 2.21 (s, 3H), 2.07 (m, 6H), 1.16 (d, J=3.5 Hz, 3H); Method 1, retention time: 5.574 min. HRMS: m/z (M+) 358.1353 (Calculated for C19H22N2O3S=358.1351).
  • Compound 48. N-(5-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00048
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.47 (s, 1H), 9.94 (s, 1H), 7.46 (d, J=7.8 Hz, 1H), 7.10-7.24 (m, 2H), 7.07 (d, J=1.8 Hz, 1H), 6.72 (d, J=11.3 Hz, 1H), 2.84 (t. J=7.6 Hz, 2H), 2.39-2.45 (t, J=7.6 Hz, 2H), 2.05 (s, 3H); Method 1, retention time: 5.197 min. HRMS: m/z (M+)=368.0389 (Calculated for C16H14ClFN2O3S=368.0398).
  • Compound 49. 6-chloro-N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide
  • Figure US20130109672A1-20130502-C00049
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.55 (br. s., 1H), 10.44 (s, 1H), 7.84 (s, 1H), 7.16 (d, J=8.2 Hz, 1H), 7.08 (d, J=2.2 Hz, 1H), 6.85-6.99 (m, 2H), 2.91 (t, J=7.6 Hz, 2H), 2.41 (t, J=7.5 Hz, 2H), 2.16 (s, 3H); Method 1, retention time: 5.405 min. HRMS: m/z (M+) 384.0094 (Calculated for C16N14Cl2N2O3S=384.0102).
  • Compound 50. N-(3-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00050
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.49 (s, 1H), 9.96 (s, 1H), 7.42 (d, J=7.6 Hz, 1H), 7.29 (d, J=7.8 Hz, 1H), 7.11 (t, J=8.0 Hz, 1H), 6.95 (d, J=7.6 Hz, 1H), 6.74 (d, J=11.3 Hz, 1H), 2.84 (t, J=7.6 Hz, 2H), 2.39 (t, J=7.6 Hz, 2H), 2.17 (s, 3H); Method 1, retention time: 5.192 min. HRMS: m/z (M+)=368.0394 (Calculated for C16H14ClFN2O3S=368.0398).
  • Compound 51. N-(3-chloro-4-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00051
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.52 (s, 1H), 10.45 (s, 1H), 7.61 (d. J=7.8 Hz, 1H), 7.18 (d, J=8.4 Hz, 1H), 7.10 (d, J=2.2 Hz, 1H), 6.96 (dd, J=8.2, 2.2 Hz, 1H), 6.69 (d, J=11.3 Hz, 1H), 2.88 (t, J=7.6 Hz, 2H), 2.38-2.46 (t, J=7.4 Hz, 2H), 2.17 (s, 3H); Method 1, retention time: 5.289 min. HRMS: m/z (M+)=368.0387 (Calculated for C16H14ClFN2O3S 368.0398).
  • Compound 52. 3-(2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamido)phenylboronic acid
  • Figure US20130109672A1-20130502-C00052
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.36 (s, 1H), 9.97 (s, 1H), 7.96 (s, 1H), 7.34-7.58 (m, 3H), 7.01-7.21 (m, 2H), 6.86 (d, J=8.2 Hz, 1H), 3.13 (m, 2H), 2.86 (t, J=7.5 Hz, 2H), 2.42 (t, J=7.6 Hz, 2H); Method 1, retention time: 3.701 min. HRMS: m/z (M+)=345.0821 (Calculated for C15N15BN2O5S=345.0831).
  • Compound 53. N-(4-fluoro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00053
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.38 (s, 1H), 9.95 (br. s., 1H), 7.40-7.55 (m, 2H), 6.91-7.01 (m, 2H), 6.74-6.91 (m, 2H), 2.87 (t, J=7.5 Hz, 2H), 2.38-2.45 (t, J=7.5 Hz, 2H), 2.10 (s, 3H); Method 1, retention time: 4.909 min. HRMS: m/z (M+)=334.0781 (Calculated for C16H15FN2O3S=334.0787).
  • Compound 54. N-(3,4-dichlorophenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00054
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.84 (s, 1H), 10.48 (s, 1H), 7.66 (d, J=7.6 Hz, 1H), 7.48 (d, J=8.8 Hz, 1H), 7.26 (d, J=2.5 Hz, 1H), 7.06 (dd, J=8.8, 2.5 Hz, 1H), 6.71 (d, J=11.3 Hz, 1H), 2.89 (t, J=7.5 Hz, 2H), 2.39-2.47 (t, J=7.6 Hz, 2H); Method 1, retention time: 5.442 min. HRMS: m/z (M+)=387.9850 (Calculated for C15N11Cl2FN2O3S=387.9851).
  • Compound 55. N-(3-fluoro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00055
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.39 (s, 1H), 10.21 (br. s., 1H), 7.48-7.57 (m, 2H), 7.08 (t, J=8.5 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 6.72-6.84 (m, 2H), 2.88 (t, J-7.6 Hz, 2H), 2.41 (t, J=7.6 Hz, 2H), 2.06 (s, 3H); Method 1, retention time: 4.916 min. HRMS: m/z (M+)=334.0775 (Calculated for C16H15FN2O3S=334.0787).
  • Compound 56. N-(4-chloro-3-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00056
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.53 (s, 1H), 10.46 (s, 1H), 7.61 (d, J=7.8 Hz, 1H), 7.18 (d, J=8.4 Hz, 1H), 7.10 (d, J=2.2 Hz, 1H), 6.96 (dd, J=8.2, 2.2 Hz, 1H), 6.69 (d, J=11.3 Hz, 1H), 2.88 (t, J=7.5 Hz, 2H), 2.40 (t, J=7.5 Hz, 2H), 2.17 (s, 3H); Method 1, retention time: 5.294 min. HRMS: m/z (M+)=368.0388 (Calculated for C16N14ClFN2O1S 368.0398).
  • Compound 57. 6-chloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide
  • Figure US20130109672A1-20130502-C00057
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.78 (s, 1H), 10.13 (s, 1H), 7.77 (s, 1H), 6.86-6.95 (m, 2H), 6.84 (s, 1H), 6.71-6.82 (m, 1H), 3.50 (s, 2H), 2.06 (s, 3H), 2.04 (s, 3H); Method 1, retention time: 5.069 min. HRMS: m/z (M+)=350.0481 (Calculated for C16N15ClN2O3S=350.0492).
  • Compound 58. N-(4-chloro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00058
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.40 (br. s., 2H), 7.55 (m, 2H), 7.14 (m, 3H), 6.90 (d, J=8.4 Hz, 1H), 2.88 (t, J=7.4 Hz, 2H), 2.40 (t, J=7.5 Hz, 2H), 2.20 (s, 3H); Method 1, retention time: 5.160 min. HRMS: m/z (M+)=350.0487 (Calculated for C16H15ClN2O3S=350.0492).
  • Compound 59. 6-chloro-N-(3-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide
  • Figure US20130109672A1-20130502-C00059
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.48 (s, 1H), 9.89 (s, 1H), 7.61 (s, 1H), 7.28 (d, J=7.8 Hz, 1H), 7.09 (t, J=8.0 Hz, 1H), 7.02 (s, 1H), 6.92 (d, J=7.8 Hz, 1H), 2.86 (t, J=7.6 Hz, 2H), 2.39 (t, J=7.6 Hz, 2H), 2.20 (s, 3H); Method 1, retention time: 5.352 min. HRMS: m/z (M+)=384.0102 (Calculated for C16H14Cl2N2O1S=384.0102).
  • Compound 60. 3,3-dichloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide
  • Figure US20130109672A1-20130502-C00060
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 11.74 (s, 1H), 9.97 (s, 1H), 7.85 (s, 1H), 7.70 (d, J=8.2 Hz, 1H), 7.05 (d, J=8.2 Hz, 1H), 6.95 (d, J=8.0 Hz, 1H), 6.68-6.88 (m, 2H), 2.06 (s, 6H); Method 1, retention time: 5.779 min. HRMS: m/z (M+)=384.0088 (Calculated for C16H14Cl2N2O3S=384.0102).
  • General Procedure for the Synthesis of Compounds 32-34, 36, and 61-70
  • The methods of synthesizing compound 32 were generally followed for all compounds 32-34, 36, and 61-73.
  • Compound 32. 7-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00061
  • In a microwave vial, N-(3,4-dimethylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide (0.01 g, 0.029 mmol) was dissolved in acetonitrile (0.5 ml) and dimethylamine (2.0 M THF) (0.029 ml, 0.057 mmol) was added followed by triethylamine (6.00 μl, 0.043 mmol). The solution was heated in a microwave at 180° C. for 1 h, cooled to RT, diluted with DMSO (0.5 mL) and purified by directly injecting to a Waters® reverse phase purification system.
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.19 (s, 1H), 9.41 (s, 1H), 7.50-7.63 (s, 1H), 6.83-6.91 (m, 2H), 6.75-6.83 (m, 2H), 2.80 (t, J=7.5 Hz, 2H), 2.58 (s, 6H), 2.39 (m, 2H), 2.04 (m, 6H). LC/MS: Method 1, retention time: 5.170 min; HRMS: m/z (M+)=373.1461 (Calculated for C19H23N3O3S=373.1460).
  • Compound 33. N-(3,4-dimethylphenyl)-7-(methylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00062
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.10 (s, 1H), 9.83 (s, 1H), 7.33 (s, 1H), 6.92 (m, 2H), 6.65-6.8 (m, 2H), 6.12 (s, 1H), 2.85 (t, J=7.5 Hz, 2H), 2.69 (d, J=4.6 Hz, 3H), 2.35 (t, J=7.4 Hz, 2H), 2.06 (m, 6H). LC/MS: Method 1, retention time: 5.170 min; HRMS: m/z (M+)=359.1302 (Calculated for C18H21N3O3S=359.1304).
  • Compound 34. N-(3,4-dimethylphenyl)-2-oxo-7-(piperidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00063
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.21 (s, 1H), 8.93 (s, 1H), 7.61 (s, 1H), 6.88 (d, J=8.2 Hz, 1H), 6.78 (m, 2H), 6.71 (dd, J=8.1, 2.1 Hz, 1H), 2.81 (t, J=7.5 Hz, 2H), 2.70 (m, 4H), 2.40 (t, J=7.6 Hz, 2H), 2.03 (d, J=4.7 Hz, 6H), 1.73 (m, 4H), 1.48 (m, 2H). LC/MS: Method 1, retention time: 5.718 min; HRMS: m/z (M+)=413.1786 (Calculated for C22H27N3O3S=413.1773).
  • Compound 36. 6-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide
  • Figure US20130109672A1-20130502-C00064
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.19 (s, 1H), 9.41 (s, 1H), 7.58 (s, 1H), 6.84-6.93 (m, 2H), 6.67-6.84 (m, 2H), 2.80 (t, J=7.5 Hz, 2H), 2.58 (s, 6H), 2.39 (t, J=7.5 Hz, 2H), 1.99-2.07 (m, 6H). LC/MS: Method 1, retention time: 5.196 min; HRMS: m/z (M+)=373.1468 (Calculated for C19H23N3O3S=373.1460).
  • Compound 61. 7-((2-(dimethylamino)ethyl)(methyl)amino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00065
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.29 (br. s., 1H), 9.62 (br. s., 1H), 8.80 (s, 1H), 7.66 (s, 1H), 6.69-6.98 (m, 3H), 3.03-3.18 (m, 2H), 2.73-2.96 (m, 7H), 2.52 (s, 6H), 2.41 (t, J=7.6 Hz, 2H), 2.07 (s, 3H), 2.04 (s, 3H); Method 1, retention time: 4.090 min. HRMS: m/z (M+)=430.2044 (Calculated for C22H30N4O3S=430.2039).
  • Compound 62. N-(3,4-dimethylphenyl)-2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00066
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.12 (s, 1H), 9.41 (s, 1H), 7.56 (s, 1H), 6.89 (d, J=8.0 Hz, 1H), 6.80 (s, 1H), 6.73 (dd, J=8.0, 2.0 Hz, 1H), 6.63 (s, 1H), 3.54 (br. s., 24H), 3.03-3.19 (m, 4H), 2.75 (t, J=7.5 Hz, 3H), 2.37 (t, J=7.5 Hz, 3H), 2.05 (s, 3H), 2.04 (s, 3H), 1.85 (m, 4H); Method 1, retention time: 5.262 min. HRMS: m/z (M+)=399.1620 (Calculated for C21H25N3O3S=399.1617).
  • Compound 63. 7-(3-(dimethylamino)pyrrolidin-1-yl)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00067
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.27 (s, 1H), 9.35 (s, 1H), 7.60 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.79 (s, 2H), 6.72 (dd, J=8.1, 1.9 Hz, 1H), 3.23 (m, 1H), 3.09-3.18 (m, 1H), 3.03 (m, 1H), 2.70-2.88 (m, 6H), 2.46 (s, 6H), 2.39 (m, 2H), 2.06 (m, 9H); Method 1, retention time: 4.092 min. HRMS: m/z (M+)=442.2040 (Calculated for C23H30N4O3S=442.2039).
  • Compound 64. N-(3,4-dimethylphenyl)-7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00068
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.03 (s, 1H), 9.84 (s, 1H), 7.33 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.79 (d, J=1.8 Hz, 1H), 6.72 (dd, J==8.0, 2.2 Hz, 1H), 6.05-6.23 (m, 2H), 2.87 (d. J=4.5 Hz, 2H), 2.68 (t, J=7.4 Hz, 2H), 2.50 (s, 1H), 2.36 (t, J=7.4 Hz, 2H), 2.05 (m, 6H), 1.12 (s, 6H); Method 1, retention time: 5.052 min. HRMS: m/z (M+)=417.1723 (Calculated for C21H27N3O4S=417.1722).
  • Compound 65. N-(3,4-dimethylphenyl)-7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00069
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.03 (s, 1H), 9.92 (s, 1H), 7.34 (s, 1H), 6.92 (d, J=8.0 Hz, 1H), 6.77 (s, 1H), 6.71 (dd, J=8.1, 2.1 Hz, 1H), 6.17 (s, 1H), 5.52 (d, J=7.2 Hz, 1H), 3.37-3.50 (m, 1H), 2.68 (t, J=7.4 Hz, 3H), 2.36 (t, J=7.5 Hz, 3H), 2.06 (s, 3H), 2.05 (s, 3H), 1.07 (d, J=6.3 Hz, 6H); Method 1, retention time: 5.596 min. HRMS: m/z (M+)=387.1614 (Calculated for C20H25N3O3S=387.1617).
  • Compound 66. 7-(diethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00070
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.15 (s, 1H), 9.42 (br. s., 1H), 7.71 (s, 1H), 6.87 (d, J=8.0 Hz, 1H), 6.79 (d, J=2.9 Hz, 2H), 6.71 (dd, J=8.1, 2.1 Hz, 1H), 2.78-2.97 (m, 6H), 2.40 (t, J=7.6 Hz, 2H), 2.04 (s, 3H), 2.03 (s, 3H), 0.88 (t, J=7.1 Hz, 6H); Method 1, retention time: 4.328 min. HRMS: m/z (M+)=401.1775 (Calculated for C21H27N3O3S—401.1773).
  • Compound 67. N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00071
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.04 (s, 1H), 9.84 (s, 1H), 7.32 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.77 (s, 1H), 6.72 (d, J=8.0 Hz, 1H), 6.21 (s, 1H), 5.77 (d, J=6.3 Hz, 1H), 4.82 (br. s., 1H), 3.40-3.34 (m, 3H), 2.67 (t, J=7.4 Hz, 2H), 2.35 (t, J=7.3 Hz, 2H), 2.06 (s, 3H), 2.05 (s, 3H), 1.03 (d, J=5.1 Hz, 3H); Method 1, retention time: 4.943 min. HRMS: m/z (M+)=403.1564 (Calculated for C20H25N3O3S=403.1566).
  • Compound 68. (S)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00072
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.04 (s, 1H), 9.84 (s, 1H), 7.32 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.77 (s, 1H), 6.72 (d, J=8.0 Hz, 1H), 6.21 (s, 1H), 5.77 (d. J=6.3 Hz, 1H), 4.82 (br. s., 1H), 3.40-3.34 (m, 3H), 2.67 (t, J=7.4 Hz, 2H), 2.35 (t J=7.3 Hz, 2H), 2.06 (s, 3H), 2.05 (s, 3H), 1.03 (d, J=5.1 Hz, 3H); [α]D=−53 (c=1.0, MeOH). Method 1, retention time: 4.943 min. HRMS: m/z (M+)=403.1562 (Calculated for C20H25N3O3S=403.1566).
  • Compound 69. (R)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00073
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.04 (s, 1H), 9.84 (s, 1H), 7.32 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.77 (s, 1H), 6.72 (d, J=8.0 Hz, 1H), 6.21 (s, 1H), 5.77 (d, J=6.3 Hz, 1H), 4.82 (br. s., 1H), 3.40-3.34 (m, 3H), 2.67 (t, J=7.4 Hz, 2H), 2.35 (t, J=7.3 Hz, 2H), 2.06 (s, 3H), 2.05 (s, 3H), 1.03 (d, J=5.1 Hz, 3H); [α]D=53 (c=1.0, MeOH). Method 1, retention time: 4.943 min. HRMS: m/z (M+)=403.1565 (Calculated for C20H25N3O3S=403.1566).
  • Compound 70. N-(3,4-dimethylphenyl)-7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00074
  • 1H NMR (400 Hz, DMSO-d6) δ ppm 10.06 (br. s., 1H), 9.85 (br. s., 1H), 7.32 (s, 1H), 6.91 (d, J=8.0 Hz, 1H), 6.67-6.82 (m, 2H), 6.18 (s, 1H), 5.95 (br. s., 1H), 4.78 (br. s., 1H), 3.55 (t, J=5.4 Hz, 2H), 3.06 (t, J=5.3 Hz, 2H), 2.67 (t, J=7.4 Hz, 2H), 2.35 (t, J=7.3 Hz, 2H), 2.06 (s, 3H), 2.05 (s, 3H); Method 1, retention time: 4.752 min; HRMS: m/z (M+)=389.1404 (Calculated for C19H23N3O4S=389.1409).
  • Compound 71. (S)—N-(3-chloro-4-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00075
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.22 (s, 1H) 10.08 (s, 1H) 7.36 (s, 1H) 7.17 (d, J=8.22 Hz, 1H) 7.02 (d, J=1.56 Hz, 1H) 6.89 (dd, J=8.12, 1.66 Hz, 1H) 6.25 (s, 1H) 5.79 (d, J=5.87 Hz, 1H) 4.85 (br. s, 1H) 3.36 (m, 3H) 2.70 (t, J=7.34 Hz, 2H) 2.38 (t, J=7.43 Hz, 2H) 2.18 (s, 3H) 1.05 (d, J=5.48 Hz, 3H). Method 1, retention time: 4.996 min. HRMS: m/z (M+)=423.1018 (Calculated for C19H22ClN3O4S=423.1020).
  • Compound 72. (S)—N-(4-fluoro-3-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00076
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.06 (br. s., 1H) 9.94 (s, 1H) 7.33 (d, J=9.00 Hz, 1H) 6.73-7.04 (m, 3H) 6.24 (d. J=9.19 Hz, 1H) 5.79 (br. s., 1H) 4.85 (br. s., 1H) 3.33 (m, 3H) 2.69 (t, J=7.41 Hz, 2H) 2.38 (t, J=7.43 Hz, 2H) 2.11 (s, 3H) 1.03 (d, J=5.51 Hz, 3H). Method 1, retention time: 4.709 min. HRMS: m/z (M+)=407.1319 (Calculated for C19H22FN3O4S=407.1315).
  • Compound 73. (S)—N-(3-chloro-4-fluorophenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide
  • Figure US20130109672A1-20130502-C00077
  • 1H NMR (400 MHz, DMSO-d6) δ ppm 10.28 (br. s., 1H) 10.10 (s, 1H) 7.35-7.28 (m, 3H) 7.12-7.00 (m, 2H) 6.25 (m, 1H) 5.90 (br. s., 1H) 3.20 (m, 3H) 2.70 (t, J=7.41 Hz, 2H) 2.38 (t, J=7.43 Hz, 2H) 1.04 (d, J=5.51 Hz, 3H). Method 1, retention time: 4.881 min. HRMS: m/z (M+)=427.0773 (Calculated for C18H19ClFN3O4S=427.0769).
  • General Procedure for the Synthesis of Compound 40 Compound 40. N-(3,4-dimethylphenyl)-3-oxo-6-phenyl-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide
  • Figure US20130109672A1-20130502-C00078
  • To a microwave vial, 6-bromo-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide (0.03 g, 0.073 mmol), phenylboronic acid (0.018 g, 0.146 mmol), Tetrakis (2.53 mg, 2.188 μmol), sodium carbonate (2.0M aqueous solution) (0.109 ml, 0.219 mmol) and 1,2-DME (0.5 ml) were added. The vessel was sealed and heated under microwave irradiation at 120° C. for 20 minutes. The reaction was cooled to RT, filtered through a thiol-SPE column (Stratospheres) and the column rinsed with methanol (˜2 mL). The resultant solution was purified. 1H NMR (400 MHz, DMSO-d6) δ ppm: 11.01 (br. s., 1H), 9.57 (br. s., 1H), 7.46-7.58 (m, 1H), 7.32 (d, J=5.3 Hz, 3H), 7.09-7.25 (m, 2H), 6.91 (d, J=8.0 Hz, 1H), 6.73 (s, 1H), 6.48-6.68 (m, 2H), 4.66 (s, 2H), 1.94-2.14 (m, 6H). LC/MS: Method 1, retention time: 5.946 min; HRMS: m/z (M+)=408.1141 (Calculated for C22H20N2O4S=408.1144).
  • General Procedure for the Synthesis of Compound 42 Compound 42. N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-carboxamide
  • Figure US20130109672A1-20130502-C00079
  • 2-oxo-1,2,3,4-tetrahydroquinoline-6-carboxylic acid (0.075 g, 0.392 mmol) and 3,4-dimethylaniline (0.052 g, 0.432 mmol) were dissolved in DMF (1 ml) and EDC (0.083 g, 0.432 mmol) was added. The reaction was stirred at RT for 4 h, then directly purified by directly injecting to a Waters® reverse phase purification system.
  • 1H NMR (400 MHz, DMSO-d6) δ ppm: 10.29 (s, 1H), 9.86 (s, 1H), 7.67-7.80 (m, 2H), 7.38-7.53 (m, 2H), 7.04 (d, J=8.2 Hz, 1H), 6.89 (d, J=8.2 Hz, 1H), 3.13 (s, 3H), 2.92 (t, J=7.6 Hz, 2H), 2.45 (t, J=7.65 Hz, 2H), 2.16 (m, 6H). Method 1, retention time: 5.009 min; HRMS: m/z (M+)=294.1361 (Calculated for C18H18N2O2=294.1368).
  • Example 2
  • This example illustrates some of the properties of exemplary compounds of the invention.
  • Structure active relationship (SAR) explorations were done with 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides, modifying the core 3,4-dihydroquinolin-2(1H)-one heterocycle with the sulfonamide attachment at the 6 and 7 positions of the ring (see Scheme 1 above, which shows the link at C6). The related 2H-benzo[b][1,4]oxazin-3(4H)-one, 1H-benzo[d]imidazol-2(3H)-one, indolin-2-one, and several 3,4-dihydroquinolin-2(1H)-one analogues with F, Cl, and Br substitutions were also explored. Additional SAR explorations were also performed at the 7 position of the 3,4-dihydroquinolin-2(1H)-one heterocycle. To explore aryl analogues at this position, Suzuki-Miyaura couplings between the 7-bromo-3,4-dihydroquinolin-2(1H)-one moiety and selected aryl-boronic acids (see Scheme 1 above) were pursued. A second method for exploring SAR at the 7-position involved displacement of the aryl fluoride of the 7-fluoro-3,4-dihydroquinolin-2(1H)-one moiety with various amines (see Scheme 1 above).
  • AC50 values were determined utilizing the luminescent pyruvate kinase-luciferase coupled assay (Inglese, J. et al, Quantitative high-throughput screening: a titration-based approach that efficiently identifies biological activities in large chemical libraries. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 11473-11478).
  • Reagents.
  • Kinase-Glo was obtained from Promega (Madison, Wis., USA). ATP, PEP, LDH and NADH were from Sigma (St. Louis, Mo., USA). Reagents and solvents were purchased from Sigma, Alfa Aesar (Ward Hill, Mass., USA), Acros (Morris Plains, N.J., USA), Enamine (Monmouth Jct., NJ, USA), Oakwood Products (West Columbia, S.C., USA), Matrix Scientific (Columbia, S.C., USA) or Chem-Impex International (Wood Dale, Ill., USA).
  • Luminescent Pyruvate Kinase-Luciferase Coupled Assay.
  • Production of a luminescent signal based on the generation of ATP by pyruvate kinase was determined by using the ATP-dependent enzyme firefly luciferase. Three μL of substrate mix (at r.t.) in assay buffer (50 mM imidazole pH 7.2, 50 mM KCl, 7 mM MgCl2, 0.01% tween 20, 0.05% BSA) was dispensed into Kalypsys (San Diego, Calif., USA) white solid bottom 1,536 well microtiter plates using a bottle-valve solenoid-based dispenser (Kalypsys). The final concentrations of substrates in the assay were 0.1 mM ADP and 0.5 mM PEP. Twenty-three nL of compound in DMSO were delivered with a 1,536-pin array tool, and 1 μL of enzyme mix in assay buffer (final concentration, 0.1 nM pyruvate kinase, 50 mM imidazole pH 7.2, 0.05% BSA, 4° C.) was added. Microtiter plates were incubated at r.t. for 1 hour and 2 uL of luciferase detection mix (Kinase-Glo from Promega, Madison, Wis., USA) at 4° C. protected from light, was added and luminescence was read with a ViewLux (Perkin Elmer, Waltham, Mass., USA) using a 2 second exposure/plate (with 2× binning). The final concentration of DMSO was 0.5% and found not to affect the assay signal.
  • Data was normalized for AC50 values to control columns containing uninhibited enzyme (n), and AC100 inhibition (i) according the following equation: Activation (%)=[(c−n)/(n−i)]*100 where c=compound, n=DMSO neutral, i=no enzyme control. A % activity of 100% is approximately a 2-fold increase over basal assay signal (% Activation by FBP was variable but averaged 100%). Monitoring of activation was accomplished using enzyme at 3× the final concentration.
  • All compounds were screened using a qHTS approach, where compounds are assayed using at least seven concentrations to generate concentration-response curves for every compounds. Briefly, qHTS uses an inter-plate dilution method where the first plate contains the highest concentration of a set of compounds in DMSO, while subsequent plates contain the same compounds in the same well locations, but at successive lower concentrations. Using the protocol outlined above, the rate was calculated as a plate throughput of 18 plates/hr or approximately 7 samples/sec on the Kalypsys robotic system which means that a 7 point CRC was obtained every second on the robotic system.
  • Three primary structural aspects of the 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamide molecule were pursued, i.e. the two moieties of the 3,4-dimethylaniline and 3,4-dihydroquinolin-2(1H)-one and the sulfonamide linkage. The first SAR examinations surrounded the linkage between the two aromatic moieties (compounds 41 and 42). An N-methyl sulfonamide (compound 41) version of compound 4 had an AC50 of 23.09 μM and Max. Res. of 36.00%. An amide (compound 42) version of compound 4 had an AC50 of 40 μM and a Max. Res. of 5%.
  • The second examination involved the modification of the core 3,4-dihydroquinolin-2(1H)-one heterocycle. Numerous, related heterocyclic sulfonyl chlorides were examined after coupling to 3,4-dimethylaniline to maintain uniformity with the lead from the primary screen. Results detailed in Table 1 demonstrate that the 3,4-dihydroquinolin-2(1H)-one heterocycle retains the best combination of potency and maximum response. Other heterocycles included the related 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one and the modestly divergent 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-one and 1-(indolin-1-yl)ethanone heterocycles.
  • TABLE 1
    SAR of selected N-(3,4-dimethylphenyl)arylsulfonamides
    hPK, M2 hPK, M2
    # Z1 AC50 (μM) Max. Res.a
    Figure US20130109672A1-20130502-C00080
      4-12
     4  5  6  7  8  9 10 11 12 3,4-dihydroquinolin-2(1H)-one-6-sulfonamide indolin-2-one-6-sulfonamide 4,5-dihydro-1H-benzo[b]azepin-2(3H)-one-7-sulfonamide 2H-benzo[b][1,4]oxazin-3(4H)-one-6-sulfonamide 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-one-6-sulfonamide 1H-benzo[d]imidazol-2(3H)-one-5-sulfonamide 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-one-5-sulfonamide 1-(indolin-1-yl)ethanone-5-sulfonamide 1-methyl-1H-indole-5-sulfonamide  0.65 14.5  18.0  20.0   0.92 21.0   1.8   1.8  20.0  104% 130%  66%  93% 120%  59% 100%  65%  40%
    aMax. Res. value represents the % activation at 57 μM of compound. Each value is the mean with standard deviation from three replicate experiments.
  • The next examination involved alterations to the 3,4-dimethylaniline moiety and are detailed in Table 2. While the 3,4-dimethylaniline moiety was among the most potent analogues, the 3-chlorophenyl derivative (compound 13) possessed an equal degree of potency and maximum response. Selected SAR trends were noticed in this series including the positive effect of substitutions at the meta position relative to the ortho and para positions (for instance, see the values for fluoro substitution within compounds 16, 23 and 26).
  • TABLE 2
    SAR of selected 2-oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamide
    hPK, M2 hPK, M2
    # R″ AC50 (μM) Max. Res.a
    Figure US20130109672A1-20130502-C00081
      4,13-30
     4 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 3,4-dimethylphenyl 3-chlorophenyl meta-tolyl 3-methoxyphenyl 3-fluorophenyl 3-trifluoromethylphenyl biphenyl-3-yl pyridin-3-yl 4-chlorophenyl para-tolyl 4-methoxyphenyl 4-fluorophenyl ortho-tolyl 2-methoxyphenyl 2-fluorophenyl naphthalen-2-yl naphthalen-1-yl 2,3-dihydrobenzo[b][1,4]dioxin-6-yl benzyl  0.65  0.65  1.2   3.2   1.8  13   14   23    3.2   4.1  36   10    3.9  21    7.3   2.9  10   16   14   104% 100%  99%  91%  93%  96%  13%  36%  94    110     47%  99%  96%  60%  85%  87% 101%  86%  25%
    aMax. Res. value represents the % activation at 57 μM of compound. Each value is the mean with standard deviation from three replicate experiments.
  • The 6-position (by IUPAC nomenclature rules) in the 2H-benzo[b][1,4]oxazin-3(4H)-one heterocycle was examined (compounds 37-40). In general, this related position resulted in analogues with good potency and maximum response values (see Table 3). Increased size, however, was less effective as demonstrated by the piperidine analogue 34 and the aryl substituted analogue 40. Amine substitutions provided several analogues with good potency including the NHMe-containing derivative compound 33. The N(Me)-2-containing compounds 32 and 36 were both potent and fully activated the enzyme.
  • TABLE 3
    SAR of selected N-(3,4-dimethylphenyl)arylsulfonamides
    hPK, M2 hPK, M2
    # R1 AC50 (μM) Max. Res.a
    Figure US20130109672A1-20130502-C00082
    Figure US20130109672A1-20130502-C00083
    Figure US20130109672A1-20130502-C00084
     4 31 32 33 34 35 36 37 38 39 40 H F N(Me)2 NHMe 1-piperidine Cl N(Me)2 Cl Br Me phenyl  0.65  0.92  0.52  0.16 15    0.26  0.46  0.58 18    1.2  20.6  104% 115% 106%  53%  57% 104% 110%  95% 103% 106%  36%
    aMax. Res. value represents the % activation at 57 μM of compound. Each value is the mean with standard deviation from three replicate experiments.
  • Compounds 41-73 were also tested. The results are in Table 4.
  • TABLE 4
    Compound Structure AC50 (μM) Max. Res.
    41
    Figure US20130109672A1-20130502-C00085
    23.09 36.00
    42
    Figure US20130109672A1-20130502-C00086
    40 5
    43
    Figure US20130109672A1-20130502-C00087
    0.26 122.38
    44
    Figure US20130109672A1-20130502-C00088
    2.59 126.17
    45
    Figure US20130109672A1-20130502-C00089
    2.91 108.88
    46
    Figure US20130109672A1-20130502-C00090
    9.19 97.00
    47
    Figure US20130109672A1-20130502-C00091
    18.34 111.76
    48
    Figure US20130109672A1-20130502-C00092
    4.61 94.62
    49
    Figure US20130109672A1-20130502-C00093
    0.65 111.72
    50
    Figure US20130109672A1-20130502-C00094
    2.59 117.55
    51
    Figure US20130109672A1-20130502-C00095
    0.12 102.49
    52
    Figure US20130109672A1-20130502-C00096
    0.82 99.32
    53
    Figure US20130109672A1-20130502-C00097
    0.37 121.19
    54
    Figure US20130109672A1-20130502-C00098
    3.66 108.94
    55
    Figure US20130109672A1-20130502-C00099
    0.46 131.37
    56
    Figure US20130109672A1-20130502-C00100
    0.82 94.23
    57
    Figure US20130109672A1-20130502-C00101
    0.92 107.73
    58
    Figure US20130109672A1-20130502-C00102
    0.41 96.45
    59
    Figure US20130109672A1-20130502-C00103
    1.16 109.46
    60
    Figure US20130109672A1-20130502-C00104
    10.31 117.30
    61
    Figure US20130109672A1-20130502-C00105
    5.84 70.48
    62
    Figure US20130109672A1-20130502-C00106
    0.46 124.25
    63
    Figure US20130109672A1-20130502-C00107
    8.19 102.12
    64
    Figure US20130109672A1-20130502-C00108
    1.83 100.38
    65
    Figure US20130109672A1-20130502-C00109
    0.32 105.86
    66
    Figure US20130109672A1-20130502-C00110
    7.24 110.76
    67
    Figure US20130109672A1-20130502-C00111
    0.11 110.43
    68
    Figure US20130109672A1-20130502-C00112
    0.09 131.05
    69
    Figure US20130109672A1-20130502-C00113
    0.37 87.70
    70
    Figure US20130109672A1-20130502-C00114
    0.09 110.67
    71
    Figure US20130109672A1-20130502-C00115
    0.08 124
    72
    Figure US20130109672A1-20130502-C00116
    0.27 99
    73
    Figure US20130109672A1-20130502-C00117
    0.14 95
  • The same assay system was utilized to examine the activity of compound 4 and all related analogs versus PKL, PKM 1 and PKR pyruvate kinase isoforms. No activity for any of the compounds were found against PKL, PKM1, or PKR.
  • All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
  • The use of the terms “a” and “an” and “the” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to.”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
  • Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims (28)

1. A compound of the formula for:

R″—NR-L-R′;
wherein R is H or C1-C4 alkyl;
wherein L is SO2 or CO;
wherein R′ is a fused bicyclic ring, wherein one ring of the bicyclic ring is phenyl which is linked to the NR-L moiety at the nitrogen atom or the sulfur atom when L is SO2 or the carbon atom when L is CO and the other ring of the bicyclic ring is an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, wherein R′ is optionally substituted on one or both rings with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, (C1-C4)alkyl-CO—, and heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
wherein R″ is phenyl, benzyl, or heteroaryl, which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, alkylenedioxy, aryl, heteroaryl, benzyl, B(OH)2, and C1-C4 alkyl substituted with one or more halogens, or is phenyl optionally fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxyl, alkylenedioxy, aryl, heteroaryl, benzyl, and C1-C4 alkyl substituted with one or more halogens; and
with the proviso that the compound is not
Figure US20130109672A1-20130502-C00118
or a pharmaceutically acceptable salt thereof.
2. (canceled)
3. The compound or salt of claim 1, wherein the phenyl ring of the bicyclic ring of R′ is fused with an aryl, a heteroaryl, a cyclyl, or a heterocyclyl, each of which is optionally substituted with one or more substituents selected from the group consisting of aryl, heteroaryl, cyclyl, alkyl, alkoxyl, halogen, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, (C1-C4)alkyl-OC—, and heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
4. The compound or salt of claim 1, wherein the cyclyl or heterocyclyl of R′ or R″ is a five-membered, six-membered, or seven-membered ring.
5. The compound or salt of claim 1, wherein the heterocyclyl contains one or two heteroatoms.
6. The compound or salt of claim 1, wherein R is methyl or H.
7. The compound or salt of claim 1, wherein R′ is
Figure US20130109672A1-20130502-C00119
wherein X1 and X2 are each individually O, N-acetyl, NR6, or CR7R8;
wherein any CH2—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a CH═CH moiety;
wherein any NH—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a N═CH moiety;
wherein any methylene of the ring containing X1 and X2 is optionally replaced by a carbonyl;
wherein n and m are each individually 0, 1, or 2, and wherein n+m is 0 to 2;
wherein each R1 is individually H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl;
wherein R6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
wherein R7 and R8 are each individually H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
8. The compound or salt of claim 7, wherein one R1 is at the ortho position relative to the carbon attached to the NR-L moiety.
9. The compound or salt of claim 7, wherein one R1 is H, F, Cl, Br, methyl, N(Me)2, NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
10. The compound or salt of claim 1, wherein R′ is substituted with one or more substituents selected from the group consisting of methyl and acetyl.
11. The compound or salt of claim 1, wherein R′ is 3,4-dihydroquinolin-2(1H)-onyl, indolin-2-onyl, 4,5-dihydro-1H-benzo[b]azepin-2(3H)-onyl, 2H-benzo[b][1,4]oxazin-3(4H)-onyl, 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-onyl, 1-(indolin-1-yl)ethanonyl, 1-methyl-1H-indolyl, 1-acetyl-2-methylindolinyl, 6-chloro-2-oxoindolinyl, 3,3-dichloro-2-oxoindolinyl, 7-((2-(dimethylamino)ethyl)(methyl)amino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinolinyl, 7-(3-(dimethylamino)pyrrolidin-1-yl)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(diethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, (S)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, or (R)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl.
12. The compound or salt of claim 1, wherein R″ is
Figure US20130109672A1-20130502-C00120
wherein X3 is N or CH wherein when X3 is N, the NR-L moiety is linked to a C of the ring containing X3;
wherein R2 and R3 are each individually H, halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, B(OH)2, phenyl, C1-C4 alkyl substituted with one or more halogens, or taken together form alkylenedioxyl.
13. The compound or salt of claim 12, wherein R2 and R3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH)2, or taken together form alkylenedioxyl.
14. The compound or salt of claim 1, wherein R″ is 3,4-dimethylphenyl, 3-chlorophenyl, meta-tolyl, 3-methoxyphenyl, 3-fluorophenyl, 3-trifluoromethylphenyl, biphenyl-3-yl, pyridine-3-yl, 4-chlorophenyl, para-tolyl, 4-methoxyphenyl, 4-fluorophenyl, ortho-tolyl, 2-methoxyphenyl, 2-fluorophenyl, naphthalen-2-yl, naphthalen-1-yl, 2,3-dihydrobenzo[b][1,4]dioxin-6-yl, benzyl, 3-chloro-4-methylphenyl, 3,4-dichlorophenyl, 5-chloro-2-methylphenyl, 3-cyanophenyl, 3-chloro-2-methylphenyl, 3-phenylboronic acid, 4-fluoro-3-methylphenyl, 3-fluoro-4-methylphenyl, 4-chloro-3-methylphenyl, or 3-chloro-4-fluorophenyl.
15. The compound or salt of claim 1, wherein the compound of formula (I) is a compound of formula (II):
Figure US20130109672A1-20130502-C00121
wherein X1 and X2 are each individually O, N-acetyl, NR6, or CR7R8;
wherein X3 is N or CH wherein when X3 is N, the NR-L moiety is linked to a C of the ring containing X3;
wherein any CH2—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a CH═CH moiety;
wherein any NH—CH2 moiety within the ring containing X1 and X2 is optionally replaced with a N═CH moiety;
wherein any methylene of the ring containing X1 and X2 is optionally replaced by a carbonyl;
wherein n and m are each individually 0, 1, or 2, and wherein n+m is 0 to 2;
wherein k is 0 or 1;
wherein R1 is H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl;
wherein R2 and R3 are each individually H, halogen, C1-C4 alkyl, C1-C4 alkoxyl, cyano, B(OH)2, phenyl, C1-C4 alkyl substituted with one or more halogens, or taken together form alkylenedioxy or phenyl fused to a CH:CH moiety of the ring containing X3;
wherein R6 is H, alkyl, alkylcarboxy, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl; and
wherein R7 and R8 are each individually H, halogen, alkyl, alkoxyl, NH2, NH—(C1-C4)alkyl, N—(C1-C4)alkyl-(C1-C4)alkyl, aryl, heteroaryl, cyclyl, or heterocyclyl, each of which other than halogen and NH2 is further optionally substituted with one or more substituents selected from the group consisting of NH2, OH, NH—(C1-C4)alkyl and N—(C1-C4)alkyl-(C1-C4)alkyl.
16. The compound or salt of claim 15, wherein R1 is H, F, Cl, Br, methyl, N(Me)2, NHMe, 1-piperidinyl, 2-(dimethylamino)ethyl)(methyl)amino, pyrrolidin-1-yl, 3-(dimethylamino)pyrrolidin-1-yl, 2-hydroxy-2-methylpropylamino, isopropylamino, diethylamino, 1-hydroxypropan-2-ylamino, 2-hydroxyethylamino, or phenyl.
17. The compound or salt of claim 15, wherein
Figure US20130109672A1-20130502-C00122
is substituted with one or more substituents selected from the group consisting of methyl and acetyl.
18. The compound or salt of claim 15, wherein
Figure US20130109672A1-20130502-C00123
is 3,4-dihydroquinolin-2(1H)-onyl, indolin-2-onyl, 4,5-dihydro-1H-benzo[b]azepin-2(3H)-onyl, 2H-benzo[b][1,4]oxazin-3(4H)-onyl, 4-methyl-2H-benzo[b][1,4]oxazin-3(4H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, 1,3-dimethyl-1H-benzo[d]imidazol-2(3H)-onyl, 1-(indolin-1-yl)ethanonyl, 1-methyl-1H-indolyl, 1-acetyl-2-methylindolinyl, 6-chloro-2-oxoindoline, 3,3-dichloro-2-oxoindolinyl, 7-((2-(dimethylamino)ethyl)(methyl)amino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinolinyl, 7-(3-(dimethylamino)pyrrolidin-1-yl)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(diethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, 7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, (S)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl, or (R)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinolinyl.
19. The compound or salt of claim 15, wherein R2 and R3 are each individually H, F, Cl, Br, methyl, methoxy, cyano, trifluoromethyl, phenyl, B(OH)2, or taken together form alkylenedioxyl or phenyl fused to a CH:CH moiety of the ring containing X3.
20. The compound or salt of claim 15, wherein
Figure US20130109672A1-20130502-C00124
is 3,4-dimethylphenyl, 3-chlorophenyl, meta-tolyl, 3-methoxyphenyl, 3-fluorophenyl, 3-trifluoromethylphenyl, biphenyl-3-yl, pyridin-3-yl, 4-chlorophenyl, para-tolyl, 4-methoxyphenyl, 4-fluorophenyl, ortho-tolyl, 2-methoxyphenyl, 2-fluorophenyl, naphthalen-2-yl, naphthalen-1-yl, 2,3-dihydrobenzo[b][1,4]dioxin-6-yl, benzyl, 3-chloro-4-methylphenyl, 3,4-dichlorophenyl, 5-chloro-2-methylphenyl, 3-cyanophenyl, 3-chloro-2-methylphenyl, 3-phenylboronic acid, 4-fluoro-3-methylphenyl, 3-fluoro-4-methylphenyl, 4-chloro-3-methylphenyl, or 3-chloro-4-fluorophenyl.
21. The compound or salt of claim 1, wherein the compound is
N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
N-(3,4-dimethylphenyl)-2-oxo-2,3,4,5-tetrahydro-1H-benzo[b]azepine-7-sulfonamide,
N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide,
N-(3,4-dimethylphenyl)-4-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-6-sulfonamide,
N-(3,4-dimethylphenyl)-2-oxo-2,3-dihydro-1H-benzo[d]imidazole-5-sulfonamide,
N-(3,4-di methylphenyl)-1,3-dimethyl-2-oxo-2,3-dihydro-1H-benzo[d]imidazo le-5-sulfonamide,
1-acetyl-N-(3,4-dimethylphenyl)indoline-5-sulfonamide,
N-(3,4-dimethylphenyl)-1-methyl-1H-indole-5-sulfonamide,
N-(3-chlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
2-oxo-N-m-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
2-oxo-N-(3-(trifluoromethyl)phenyl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(biphenyl-3-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
2-oxo-N-(pyridin-3-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(4-chlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
2-oxo-N-p-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(4-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(4-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
2-oxo-N-o-tolyl-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(2-methoxyphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(2-fluorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(naphthalen-2-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(naphthalen-1-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-benzyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
7-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-(methylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-2-oxo-7-(piperidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
6-chloro-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
6-(dimethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
6-chloro-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
6-bromo-N-(3,4-dimethylphenyl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
N-(3,4-dimethylphenyl)-6-methyl-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
N-(3,4-dimethylphenyl)-3-oxo-6-phenyl-3,4-dihydro-2H-benzo[b][1,4]oxazine-7-sulfonamide,
N-(3,4-dimethylphenyl)-N-methyl-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-carboxamide,
N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dichlorophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
6-chloro-N-(5-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
N-(3-cyanophenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
1-acetyl-N-(3,4-dimethylphenyl)-2-methylindoline-5-sulfonamide,
N-(5-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
6-chloro-N-(3-chloro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
N-(3-chloro-2-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3-chloro-4-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
3-(2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamido)phenylboronic acid,
N-(4-fluoro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dichlorophenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3-fluoro-4-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(4-chloro-3-methylphenyl)-7-fluoro-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
6-chloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
N-(4-chloro-3-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
6-chloro-N-(3-chloro-2-methylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-7-sulfonamide,
3,3-dichloro-N-(3,4-dimethylphenyl)-2-oxoindoline-5-sulfonamide,
7-((2-(dimethylamino)ethyl)(methyl)amino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-2-oxo-7-(pyrrolidin-1-yl)-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
7-(3-(dimethylamino)pyrrolidin-1-yl)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-(2-hydroxy-2-methylpropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-(isopropylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
7-(diethylamino)-N-(3,4-dimethylphenyl)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
(S)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
(R)—N-(3,4-dimethylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
N-(3,4-dimethylphenyl)-7-(2-hydroxyethylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
(S)—N-(3-chloro-4-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide,
(S)—N-(4-fluoro-3-methylphenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide, or
(S)—N-(3-chloro-4-fluorophenyl)-7-(1-hydroxypropan-2-ylamino)-2-oxo-1,2,3,4-tetrahydroquinoline-6-sulfonamide.
22. A pharmaceutical composition comprising a compound or salt according to claim 1, and a pharmaceutically acceptable carrier.
23.-27. (canceled)
28. A method of treating a disease responsive to activation of human PK-M2 comprising administering to a patient in need thereof a therapeutically effective amount of a compound or salt according to claim 1.
29. The method according to claim 28, wherein the disease responsive to activation of human PK-M2 is cancer.
30. The method according to claim 29, wherein the cancer is leukemia, polycythemia vera, lymphoma, Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, sarcoma, or carcinoma.
31. The method according to claim 29, wherein the cancer is acute leukemia, acute lymphocytic leukemia, acute myeloid leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, Hodgkin's disease, non-Hodgkin's disease, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, malignant mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, alveolar rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, intrahepatic bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small cell lung carcinoma, urinary bladder cancer, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma, bone cancer, brain cancer, cancer of the anus, cancer of the anal canal, cancer of the anorectum, cancer of the eye, cancer of the joints, cancer of the neck, cancer of the gallbladder, cancer of the pleura, cancer of the nose, cancer of the nasal cavity, cancer of the middle ear, cancer of the oral cavity, cancer of the vulva, esophageal cancer, gastrointestinal carcinoid tumor, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, nasopharynx cancer, non-small cell lung cancer, peritoneum cancer, omentum cancer, mesentery cancer, pharynx cancer, rectal cancer, renal cancer, small intestine cancer, soft tissue cancer, stomach cancer, thyroid cancer, or ureter cancer.
32. The method according to claim 31, wherein the cancer is liver cancer.
US13/643,594 2010-04-29 2011-04-26 Activators of human pyruvate kinase Abandoned US20130109672A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/643,594 US20130109672A1 (en) 2010-04-29 2011-04-26 Activators of human pyruvate kinase

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US32915810P 2010-04-29 2010-04-29
US13/643,594 US20130109672A1 (en) 2010-04-29 2011-04-26 Activators of human pyruvate kinase
PCT/US2011/033852 WO2011137089A1 (en) 2010-04-29 2011-04-26 Activators of human pyruvate kinase

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/033852 A-371-Of-International WO2011137089A1 (en) 2010-04-29 2011-04-26 Activators of human pyruvate kinase

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/643,107 Continuation US9708267B2 (en) 2010-04-29 2015-03-10 Activators of human pyruvate kinase

Publications (1)

Publication Number Publication Date
US20130109672A1 true US20130109672A1 (en) 2013-05-02

Family

ID=44280778

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/643,594 Abandoned US20130109672A1 (en) 2010-04-29 2011-04-26 Activators of human pyruvate kinase
US14/643,107 Active US9708267B2 (en) 2010-04-29 2015-03-10 Activators of human pyruvate kinase

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/643,107 Active US9708267B2 (en) 2010-04-29 2015-03-10 Activators of human pyruvate kinase

Country Status (6)

Country Link
US (2) US20130109672A1 (en)
EP (1) EP2563761A1 (en)
JP (1) JP2013525438A (en)
AU (1) AU2011245441B2 (en)
CA (1) CA2797694A1 (en)
WO (1) WO2011137089A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017011408A1 (en) * 2015-07-10 2017-01-19 Loyola University Of Chicago Indoline and tetrahydroquinoline sulfonyl inhibitors of dimetalloenzymes and use of the same
WO2019169267A1 (en) * 2018-03-01 2019-09-06 Reaction Biology Corp. Quinoline and isoquinoline based hdac inhibitors and methods of use thereof
WO2022109292A1 (en) * 2020-11-20 2022-05-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Methods and materials for inhibiting cb1 activity
US20230192643A1 (en) * 2017-09-04 2023-06-22 C4 Therapeutics, Inc. Dihydroquinolinones for medical treatment

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010118063A2 (en) 2009-04-06 2010-10-14 Agios Pharmaceuticals, Inc. Therapeutic compositions and related methods of use
DK2448581T3 (en) 2009-06-29 2017-03-13 Agios Pharmaceuticals Inc Therapeutic compositions and methods for their applications
EP3241554B1 (en) 2009-06-29 2020-01-29 Agios Pharmaceuticals, Inc. Quinoline-8-sulfonamide derivatives having an anticancer activity
US9221792B2 (en) 2010-12-17 2015-12-29 Agios Pharmaceuticals, Inc N-(4-(azetidine-1-carbonyl) phenyl)-(hetero-) arylsulfonamide derivatives as pyruvate kinase M2 (PMK2) modulators
EP2655350B1 (en) * 2010-12-21 2016-03-09 Agios Pharmaceuticals, Inc. Bicyclic pkm2 activators
TWI549947B (en) 2010-12-29 2016-09-21 阿吉歐斯製藥公司 Therapeutic compounds and compositions
US9181231B2 (en) 2011-05-03 2015-11-10 Agios Pharmaceuticals, Inc Pyruvate kinase activators for use for increasing lifetime of the red blood cells and treating anemia
WO2012151451A1 (en) 2011-05-03 2012-11-08 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
WO2013027168A1 (en) * 2011-08-22 2013-02-28 Pfizer Inc. Novel heterocyclic compounds as bromodomain inhibitors
WO2013122897A1 (en) 2012-02-13 2013-08-22 Amgen Inc. Dihydrobenzoxazine and tetrahydroquinoxaline sodium channel inhibitors
WO2013134518A1 (en) 2012-03-09 2013-09-12 Amgen Inc. Sulfamide sodium channel inhibitors
US9458132B2 (en) * 2012-11-08 2016-10-04 Agios Pharmaceuticals, Inc Therapeutic compounds and compositions and their use as PKM2 modulators
WO2014139144A1 (en) 2013-03-15 2014-09-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
CN105272904B (en) * 2014-07-18 2019-04-09 中国科学院广州生物医药与健康研究院 N- phenylamide compound and its application
BR112017002053A2 (en) 2014-08-01 2018-01-30 Nuevolution A/S compound according to formula (i), pharmaceutical composition, and uses of a compound
FR3030518B1 (en) 2014-12-19 2018-03-23 Galderma Research & Development SULFONAMIDE DERIVATIVES AS REVERSE AGONISTS OF GAMMA RECTINTATIVE GAMMA ORPHELIN ASSOCIATED WITH RIN GAMMA RETINOIDS (T)
FR3030516B1 (en) 2014-12-19 2019-12-27 Galderma Research & Development BICYCLE SULFONAMIDE DERIVATIVES AS INVERTED AGONISTS OF THE ORPHAN GAMMA RECEPTOR ASSOCIATED WITH ROR GAMMA (T) RETINOIDS
MA44392B1 (en) 2015-06-11 2023-10-31 Agios Pharmaceuticals Inc METHODS OF USING PYRUVATE KINASE ACTIVATORS
TW202244048A (en) 2017-03-20 2022-11-16 美商佛瑪治療公司 Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators
AU2018316587B2 (en) 2017-08-15 2023-05-11 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
GB201810581D0 (en) 2018-06-28 2018-08-15 Ctxt Pty Ltd Compounds
BR112021005188A2 (en) 2018-09-19 2021-06-08 Forma Therapeutics, Inc. treating sickle cell anemia with a pyruvate kinase r activating compound
US20230055923A1 (en) 2018-09-19 2023-02-23 Forma Therapeutics, Inc. Activating pyruvate kinase r
HRP20231501T1 (en) 2019-06-18 2024-03-01 Pfizer Inc. Benzisoxazole sulfonamide derivatives
DE102019129527A1 (en) 2019-10-31 2021-05-06 Forschungszentrum Jülich GmbH Bromodomain Inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19755268A1 (en) * 1997-12-12 1999-06-17 Merck Patent Gmbh Benzamidine derivatives
US7259179B2 (en) * 2002-05-23 2007-08-21 Cytopia Research Pty Ltd Kinase inhibitors
WO2009084501A1 (en) * 2007-12-27 2009-07-09 Asahi Kasei Pharma Corporation Sultam derivative

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE907739C (en) * 1949-07-23 1954-02-18 Kalle & Co Ag Process for the production of copies, especially printing forms, with the aid of diazo compounds and light-sensitive material which can be used therefor
US3998828A (en) 1975-01-31 1976-12-21 Pfizer Inc. 4-[2-(1,3-Dialkyl-1,2,3,4-tetra-hydropyrimidine-2,4-dione-5-carboxamido)ethyl]-1-piperidine sulfonamide
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
JPS569749A (en) * 1979-07-04 1981-01-31 Mitsubishi Paper Mills Ltd Preparation of lithographic plate
DE2948434A1 (en) 1979-12-01 1981-06-11 Hoechst Ag, 6000 Frankfurt 1-PIPERIDINSULFONYL UREA AND METHOD FOR THE PRODUCTION THEREOF
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4593102A (en) 1984-04-10 1986-06-03 A. H. Robins Company, Inc. N-[(amino)alkyl]-1-pyrrolidine, 1-piperidine and 1-homopiperidinecarboxamides (and thiocarboxamides) with sulfur linked substitution in the 2, 3 or 4-position
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
JPH0667926B2 (en) 1985-11-12 1994-08-31 旭化成工業株式会社 Cyclic isoquinoline sulfonamide derivative
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5962490A (en) 1987-09-25 1999-10-05 Texas Biotechnology Corporation Thienyl-, furyl- and pyrrolyl-sulfonamides and derivatives thereof that modulate the activity of endothelin
BR9200951A (en) * 1991-03-21 1992-11-17 Hoffmann La Roche COMPOUNDS, PROCESS FOR ITS PRODUCTION, PHARMACEUTICAL PREPARATIONS AND USE
IT1252567B (en) 1991-12-20 1995-06-19 Italfarmaco Spa DERIVATIVES OF 5-ISOKINOLINSULPHONAMIDS INHIBITORS OF PROTEIN-KINASES
US5891435A (en) 1993-04-16 1999-04-06 Research Corporation Technologies, Inc. Methods and compositions for delaying or preventing the onset of autoimmune disease
US5639600A (en) 1994-08-05 1997-06-17 The Regents Of The University Of California Diagnosis and treatment of cell proliferative disease having clonal macrophage involvement
EP1586558A3 (en) 1995-01-20 2005-10-26 G.D. Searle LLC. Bis-sulfonamide hydroxyethylamino retroviral protease inhinitors
JPH09286783A (en) * 1996-02-19 1997-11-04 Fuji Photo Film Co Ltd Thermal recording material and diazo compound
DE19629335A1 (en) 1996-07-20 1998-01-22 Golden Records Ass Internation Plastic card
US6020357A (en) 1996-12-23 2000-02-01 Dupont Pharmaceuticals Company Nitrogen containing heteroaromatics as factor Xa inhibitors
AR016551A1 (en) 1997-07-30 2001-07-25 Smithkline Beecham Corp DERIVATIVES OF 2-OXINDOL, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE THEM AND THE USE OF THE SAME FOR THE MANUFACTURE OF MEDICINES
AR016384A1 (en) 1997-07-30 2001-07-04 Smithkline Beecham Corp INHIBITORS OF CASPASAS, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE SUCH INHIBITORS OF CASPASAS AND USE OF CASPASE INHIBITORS TO PREPARE A USEFUL MEDICINAL PRODUCT FOR THE TREATMENT OF APOPTOSIS AND DISORDERS ASSOCIATED WITH EXCESSIVE ACTIVITY ILL-1 CONVERT.
US7087648B1 (en) 1997-10-27 2006-08-08 The Regents Of The University Of California Methods for modulating macrophage proliferation using polyamine analogs
US6617171B2 (en) 1998-02-27 2003-09-09 The General Hospital Corporation Methods for diagnosing and treating autoimmune disease
JP2002536446A (en) 1999-02-09 2002-10-29 3−ディメンショナル ファーマシューティカルズ, インコーポレイテッド Heteroarylamidines, methylamidines and guanidines as protease inhibitors
US6211182B1 (en) 1999-03-08 2001-04-03 Schering Corporation Imidazole compounds substituted with a six or seven membered heterocyclic ring containing two nitrogen atoms
CO5170498A1 (en) 1999-05-28 2002-06-27 Abbott Lab BIARIL SULFONAMIDS ARE USEFUL AS CELL PROLIFERATION INHIBITORS
AU7625500A (en) 1999-09-30 2001-04-30 Smithkline Beecham Corporation Caspases and apoptosis
CA2410509A1 (en) * 2000-06-02 2001-12-13 Sugen, Inc. Indolinone derivatives as protein kinase/phosphatase inhibitors
AR032885A1 (en) 2000-08-16 2003-12-03 Actelion Pharmaceuticals Ltd BIS-SULFONAMIDS, PHARMACEUTICAL COMPOSITIONS, PROCEDURE TO PREPARE THEM AND THE USE OF THE SAME FOR THE PRODUCTION OF PHARMACEUTICAL COMPOSITIONS
AU2002319180B2 (en) 2001-05-23 2007-07-05 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Pyruvate-kinase as a novel target molecule
CA2479932A1 (en) 2002-03-28 2003-10-09 Eli Lilly And Company Piperazine substituted aryl benzodiazepines and their use as dopamine receptor antagonists for the treatment of psychotic disorders
WO2003106381A2 (en) 2002-06-12 2003-12-24 Exelixis, Inc. Human adam-10 inhibitors
GB0215650D0 (en) 2002-07-05 2002-08-14 Cyclacel Ltd Bisarylsufonamide compounds
WO2004076640A2 (en) 2003-02-25 2004-09-10 Ambion, Inc. Small-molecule inhibitors of angiogenin and rnases and in vivo and in vitro methods of using same
JP2005008624A (en) * 2003-05-27 2005-01-13 Taiho Yakuhin Kogyo Kk Sulfonamide derivative having tetrahydroquinoline skeleton, and its salt
WO2005074946A1 (en) 2004-02-10 2005-08-18 Adenobio N.V. Stable and active complexes of adenosine and adenosine phosphates with aminoalcohols for the treatment of pulmonary artery hypertension, cardiac failure and other diseases
EP1720874A4 (en) 2004-02-24 2010-03-03 Bioaxone Therapeutique Inc 4-substituted piperidine derivatives
RU2261246C1 (en) 2004-04-15 2005-09-27 Общество с ограниченной ответственностью "Исследовательский Институт Химического Разнообразия" (ООО "Исследовательский Институт Химического Разнообразия") Substituted 7-sulfonyl-benzo[b][1,4]diazepines (variants), methods for their preparing (variants), focused library and pharmaceutical composition
DE102004028862A1 (en) 2004-06-15 2005-12-29 Merck Patent Gmbh 3-aminoindazoles
ATE529106T1 (en) 2004-06-24 2011-11-15 Univ Temple ALPHA-, BETA-UNSATURATED SULFONES, SULFONE OXIDES, SULFONIMIDES, SULFINIMIDES, ACYLSULFONAMIDES AND ACYLSULFINAMIDES AND THERAPEUTIC USES THEREOF
TW200606152A (en) 2004-07-02 2006-02-16 Tanabe Seiyaku Co Piperidine compound and process for preparing the same
FR2872704B1 (en) 2004-07-12 2007-11-02 Laurent Schwartz PLURITHERAPY AGAINST CANCER
WO2006055880A2 (en) 2004-11-16 2006-05-26 Genzyme Corporation Diagnostic pkm2 methods and compositions
JP4058106B2 (en) 2005-02-18 2008-03-05 アストラゼネカ アクチボラグ Antibacterial piperidine derivatives
KR20080021030A (en) 2005-05-16 2008-03-06 버텍스 파마슈티칼스 인코포레이티드 Bicyclic derivatives as modulators of ion channels
AR054363A1 (en) 2005-05-23 2007-06-20 Astrazeneca Ab COMPOUNDS THAT DISPLAY MODULATORY ACTIVITY IN THE 5-HYDROXY-TRIPTAMINE 6 RECEIVER
FR2887881B1 (en) 2005-07-01 2009-10-09 Pierre Fabre Medicament Sa PROTEIN INHIBITORS KINASES
WO2007019346A1 (en) 2005-08-04 2007-02-15 Sirtris Pharmaceuticals, Inc. Benzothiazoles and thiazolopyridines as sirtuin modulators
WO2007076320A2 (en) 2005-12-22 2007-07-05 Smithkline Beecham Corporation Compounds
JP2009521444A (en) 2005-12-22 2009-06-04 メルク エンド カムパニー インコーポレーテッド Substituted piperidines as calcium channel blockers
WO2007076055A2 (en) * 2005-12-22 2007-07-05 Entremed, Inc. Compositions and methods comprising proteinase activated receptor antagonists
BRPI0706654A2 (en) 2006-01-20 2011-04-05 Smithkline Beecham Corp pharmaceutical composition, method for treating conditions or disorders in a mammal, use of a compound, and, compound
US8987474B2 (en) * 2006-04-07 2015-03-24 University Of South Florida Inhibition of Shp2/PTPN11 protein tyrosine phosphatase by NSC-87877, NSC-117199 and their analogs
US20090325944A1 (en) * 2006-04-12 2009-12-31 Suzanne Walker Kahne Methods and Compositions for Modulating Glycosylation
ATE469129T1 (en) 2006-07-11 2010-06-15 Sanofi Aventis N-Ä(1,5-DIPHENYL-1H-PYRAZOL-3-YL)METHYL SULFONAMIDE DERIVATIVES ANTAGONISTS OF THE CB1 RECEPTORS OF THE CANNABINOIDS
TW200817319A (en) * 2006-08-10 2008-04-16 Astellas Pharma Inc Sulfonamide compound or salt thereof
PE20080932A1 (en) 2006-11-10 2008-07-13 Wyeth Corp N-SUBSTITUTED PIPERIDINYL 4-ARYLSULFONAMIDES AS MODULATORS OF FRIZZLED-RELATED SECRETED PROTEIN 1 (SFRP-1)
WO2008107661A1 (en) 2007-03-05 2008-09-12 Biolipox Ab New methylenebisphenyl compounds useful in the treatment of inflammation
WO2008124838A1 (en) * 2007-04-10 2008-10-16 University Of Maryland, Baltimore Compounds that inhibit human dna ligases and methods of treating cancer
US7897776B2 (en) 2007-04-23 2011-03-01 Salutria Pharmaceuticals Llc Sulfonamide containing compounds for treatment of inflammatory disorders
US8552050B2 (en) 2007-08-16 2013-10-08 Beth Israel Deaconess Medical Center Activators of pyruvate kinase M2 and methods of treating disease
EP2195656A4 (en) * 2007-08-21 2011-10-19 Senomyx Inc Human t2r bitterness receptors and uses thereof
US20110105477A1 (en) * 2007-09-14 2011-05-05 The Regents Of The University Of Michigan Compositions and methods relating to hiv protease inhibition
WO2009055418A1 (en) * 2007-10-22 2009-04-30 Smithkline Beecham Corporation Pyridosulfonamide derivatives as pi3 kinase inhibitors
EP2219646A4 (en) * 2007-12-21 2010-12-22 Univ Rochester Method for altering the lifespan of eukaryotic organisms
GB0815947D0 (en) * 2008-09-02 2008-10-08 Univ Dundee Compounds
EP2344453B1 (en) * 2008-10-09 2016-12-28 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Activators of human pyruvate kinase
PL2470173T3 (en) * 2009-08-25 2016-11-30 Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors
SG186885A1 (en) 2010-06-04 2013-02-28 Albany Molecular Res Inc Glycine transporter-1 inhibitors, methods of making them, and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19755268A1 (en) * 1997-12-12 1999-06-17 Merck Patent Gmbh Benzamidine derivatives
US7259179B2 (en) * 2002-05-23 2007-08-21 Cytopia Research Pty Ltd Kinase inhibitors
WO2009084501A1 (en) * 2007-12-27 2009-07-09 Asahi Kasei Pharma Corporation Sultam derivative

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Zubkov, ca 151:33377, abstract only of Zhurnal Organ ta Farmatsevtichnoi Khimii, vol 6(3), p 39-44, 2008. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017011408A1 (en) * 2015-07-10 2017-01-19 Loyola University Of Chicago Indoline and tetrahydroquinoline sulfonyl inhibitors of dimetalloenzymes and use of the same
US10626087B2 (en) 2015-07-10 2020-04-21 Loyola University Of Chicago Indoline and tetrahydroquinoline sulfonyl inhibitors of dimetalloenzymes and use of the same
US20230192643A1 (en) * 2017-09-04 2023-06-22 C4 Therapeutics, Inc. Dihydroquinolinones for medical treatment
WO2019169267A1 (en) * 2018-03-01 2019-09-06 Reaction Biology Corp. Quinoline and isoquinoline based hdac inhibitors and methods of use thereof
WO2022109292A1 (en) * 2020-11-20 2022-05-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Methods and materials for inhibiting cb1 activity

Also Published As

Publication number Publication date
US20150183744A1 (en) 2015-07-02
WO2011137089A1 (en) 2011-11-03
AU2011245441A1 (en) 2012-11-15
AU2011245441B2 (en) 2014-12-18
JP2013525438A (en) 2013-06-20
EP2563761A1 (en) 2013-03-06
CA2797694A1 (en) 2011-11-03
US9708267B2 (en) 2017-07-18

Similar Documents

Publication Publication Date Title
US9708267B2 (en) Activators of human pyruvate kinase
KR102668900B1 (en) Heterocyclic compounds
US11053207B2 (en) Indoleamine-2,3-dioxygenase inhibitor and preparation method therefor
EP2344453B1 (en) Activators of human pyruvate kinase
JP6219882B2 (en) IRE-1α inhibitor
TW201738226A (en) Heterocyclic compound
US8791106B2 (en) Fused ring pyridine compound
JP2002308882A (en) Thienopyrimidine derivative
EP1723126A2 (en) Novel mch receptor antagonists
WO2013192610A2 (en) Pro-drugs of riluzole and their method of use for the treatment of amyotrophic lateral sclerosis
US20190077777A1 (en) Nicotinic acid derivatives, their preparation and the use thereof
JP2018087173A (en) Anti-malignant brain tumor therapeutic agent
AU2015201398A1 (en) Activators of human pyruvate kinase
RU2780892C2 (en) Heterocyclic compound
CN112752760B (en) Heterocyclic compounds
US11970483B2 (en) Heterocyclic compound
JP2023518299A (en) Androgen receptor modulation by small molecule enantiomers
EP3858823A1 (en) Condensed-cyclic compound

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BOXER, MATTHEW B.;SHEN, MIN;AULD, DOUGLAS S.;AND OTHERS;SIGNING DATES FROM 20121101 TO 20121109;REEL/FRAME:029408/0588

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE