WO2012092442A1 - Therapeutic compounds and compositions - Google Patents

Therapeutic compounds and compositions Download PDF

Info

Publication number
WO2012092442A1
WO2012092442A1 PCT/US2011/067752 US2011067752W WO2012092442A1 WO 2012092442 A1 WO2012092442 A1 WO 2012092442A1 US 2011067752 W US2011067752 W US 2011067752W WO 2012092442 A1 WO2012092442 A1 WO 2012092442A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
phenyl
carbonyl
sulfonamide
nmr
Prior art date
Application number
PCT/US2011/067752
Other languages
French (fr)
Inventor
Francesco G. Salituro
Jeffrey O. Saunders
Shunqi Yan
Original Assignee
Agios Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agios Pharmaceuticals, Inc. filed Critical Agios Pharmaceuticals, Inc.
Priority to JP2013547663A priority Critical patent/JP5989664B2/en
Priority to MX2013007541A priority patent/MX342274B/en
Priority to AU2011352085A priority patent/AU2011352085B2/en
Priority to EP11811286.1A priority patent/EP2658543B1/en
Priority to BR112013016811A priority patent/BR112013016811A2/en
Priority to CN201180068645.3A priority patent/CN103491960B/en
Priority to CA2823401A priority patent/CA2823401C/en
Publication of WO2012092442A1 publication Critical patent/WO2012092442A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07D215/04Heterocyclic 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 only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to the ring carbon atoms
    • C07D215/06Heterocyclic 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 only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to the ring carbon atoms having only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/04Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems

Definitions

  • Cancer cells rely primarily on glycolysis to generate cellular energy and biochemical intermediates for biosynthesis of lipids and nucleotides, while the majority of "normal" cells in adult tissues utilize aerobic respiration. This fundamental difference in cellular metabolism between cancer cells and normal cells, termed the Warburg Effect, has been exploited for diagnostic purposes, but has not yet been exploited for therapeutic benefit.
  • PK Pyruvate kinase
  • L and R isoforms are expressed in liver and red blood cells
  • Ml isoform is expressed in most adult tissues
  • M2 isoform is a splice variant of Ml expressed during embryonic development. All tumor cells exclusively express the embryonic M2 isoform.
  • FBP fructose- 1,6-bisphosphate
  • PKM2 is also expressed in adipose tissue and activated T-cells.
  • the activation of PKM2 may be effective in the treatment of, e.g., obesity, diabetes, autoimmune conditions, and proliferation-dependent diseases, e.g., benign prostatic hyperplasia (BPH).
  • BPH benign prostatic hyperplasia
  • Current inhibitors of pyruvate kinase are not selective, making it difficult to treat disease related to pyruvate kinase function.
  • PKM2 phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form.
  • Compounds that bind to PKM2 and lock the enzyme in the active confirmation will lead to the loss of allosteric control of PKM2 needed for shunting biochemical intermediates from glycolysis into biosynthesis of nucleotides and lipids.
  • the activation of PKM2 i.e., activators of PKM2
  • PLM2 pyruvate kinase M2
  • W, X, Y and Z are each independently selected from CH or N;
  • Q and Q 1 are independently selected from a bond or NR b ;
  • A is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl
  • L is a bond, -C(O)-, -(CR c R c ) m -, -OC(O)-, -(CR c R c ) m -OC(0)-, -(CR c R c ) m -C(0)-, -NR b C(S)-, or -NR b C(0)- (wherein the point of the attachment to R 1 is on the left-hand side);
  • R 1 is selected from alkyl, carbocycle, aryl, heteroaryl, and heterocyclyl; each of which is substituted with 0-5 occurrences of R d ;
  • each R 3 is independently selected from halo, haloalkyl, alkyl, hydroxyl and -OR a , or two adjacent R taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
  • each R a is independently selected from alkyl, acyl, hydroxyalkyl and haloalkyl;
  • each R d is independently selected from halo, haloalkyl, haloalkoxy, alkyl, alkynyl, nitro, cyano, hydroxyl, -C(0)R a , -OC(0)R a , -C(0)OR a , -SR a , -NR a R b and -OR a , or two R d taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
  • n 0, 1, or 2;
  • n 1, 2 or 3;
  • h 0, 1, 2;
  • g 0, 1 or 2;
  • p 0, 1 or 2; and provided that the compound of formula (I) is not
  • compositions comprising a compound of Formula I or a pharmaceutically acceptable salt thereof and the use of such compositions in methods of treating diseases and conditions that are related to reduced pyruvate kinase function (e.g., PKM2 function), including, e.g., cancer, diabetes, obesity, autoimmune disorders, and benign prostatic hyperplasia (BPH).
  • diseases and conditions that are related to reduced pyruvate kinase function (e.g., PKM2 function)
  • PKM2 function e.g., cancer, diabetes, obesity, autoimmune disorders, and benign prostatic hyperplasia (BPH).
  • a method of modulating e.g., increasing or decreasing the level of PKM2 activity and/or glycolysis (e.g., modulating the endogenous ability of a cell in the patient to down regulate PKM2) in a patient in need thereof.
  • the method comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby modulating (e.g., increasing or decreasing) the level of PKM2 activity and/or glycolysis in the patient.
  • the method includes identifying or selecting a patient who would benefit from activation of PKM2. For example, the patient can be identified on the basis of the level of PKM2 activity in a cell of the patient for treatment of cancer associated with PKM2 function.
  • a method of inhibiting cell proliferation in a patient in need thereof comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby inhibiting cell proliferation in the patient.
  • the compound described herein is administered at a dosage and frequency sufficient to increase lactate production or oxidative phosphorylation.
  • halo or halogen refers to any radical of fluorine, chlorine, bromine or iodine.
  • alkyl refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms.
  • Q-C 12 alkyl indicates that the group may have from 1 to 12 (inclusive) carbon atoms in it.
  • the term “alkyl” refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 6 carbon atoms.
  • the term “alkyl” refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 4 carbon atoms.
  • haloalkyl refers to an alkyl in which one or more hydrogen atoms are replaced by halo, and includes alkyl moieties in which all hydrogens have been replaced by halo (e.g., perfluoroalkyl).
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-12 carbon atoms and having one or more double bonds.
  • alkenyl groups include, but are not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups.
  • One of the double bond carbons may optionally be the point of attachment of the alkenyl substituent.
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-6 carbon atoms and having one or more double bonds.
  • alkenyl refers to a monovalent straight or branched hydrocarbon chain containing 2-4 carbon atoms and having one or more double bonds.
  • alkynyl refers to a monovalent straight or branched hydrocarbon chain containing 2-12 carbon atoms and characterized in having one or more triple bonds.
  • alkynyl groups include, but are not limited to, ethynyl, propargyl, and 3-hexynyl.
  • One of the triple bond carbons may optionally be the point of attachment of the alkynyl substituent.
  • alkylamino and “dialkylamino” refer to -NH(alkyl) and -NH(alkyl) 2 radicals respectively.
  • aralkylamino refers to a -NH(aralkyl) radical.
  • alkylaminoalkyl refers to a (alkyl)NH-alkyl- radical.
  • dialkylaminoalkyl refers to a (alkyl) 2 N-alkyl- radical.
  • mercapto refers to an -SH radical.
  • thioalkoxy refers to an -S-alkyl radical.
  • thioaryloxy refers to an -S-aryl radical.
  • alkoxy refers to an -O-alkyl radical.
  • aryl refers to a monocyclic, bicyclic, or tricyclic aromatic hydrocarbon ring system.
  • aryl moieties include, but are not limited to, phenyl, naphthyl, and anthracenyl.
  • arylalkyl or “aralkyl” refer to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group.
  • Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of “arylalkyl” or “aralkyl” include benzyl, 2-phenylethyl, 3-phenylpropyl, 9-fluorenyl, benzhydryl, and trityl groups.
  • Carbocyclyl refers to a non-aromatic, monocyclic, bicyclic, or tricyclic hydrocarbon ring system.
  • Carbocyclyl groups include fully saturated ring systems (e.g., cycloalkyls), and partially saturated ring systems.
  • cycloalkyl as employed herein includes saturated cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 3 to 12 carbons. Any ring atom can be substituted (e.g., by one or more substituents). Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclohexyl, methylcyclohexyl, adamantyl, and norbornyl.
  • heteroaryl refers to a fully aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms selected independently from N, O, or S if monocyclic, bicyclic, or tricyclic, respectively).
  • heterocyclyl refers to a nonaromatic, 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively).
  • the heteroatom may optionally be the point of attachment of the heterocyclyl substituent.
  • heterocyclyl examples include, but are not limited to, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, pyrrolinyl, pyrimidinyl, and pyrrolidinyl.
  • Bicyclic and tricyclic ring systems containing one or more heteroatoms and both aromatic and non-aromatic rings are considered to be heterocyclyl groups according to the present definition.
  • Such bicyclic or tricyclic ring systems may be alternately characterized as being an aryl or a heteroaryl fused to a carbocyclyl or heterocyclyl, particularly in those instances where the ring bound to the rest of the molecule is required to be aromatic.
  • heteroarylalkyl and “heteroaralkyl”, as used herein, refers to an alkyl group substituted with a heteroaryl group.
  • heterocyclylalkyl refers to an alkyl group substituted with a heterocyclyl group.
  • acyl refers to an alkylcarbonyl, carbocyclecarbonyl, arylcarbonyl,
  • heterocyclylcarbonyl or heteroarylcarbonyl substituent, any of which may be further substituted (e.g., by one or more substituents).
  • All ring systems i.e, aryl, heteroaryl, carbocyclyl, cycloalkyl, heterocyclyl, etc.
  • ring system portions of groups e.g., the aryl portion of an aralkyl group
  • any alkyl substituent is optionally further substituted with one or more of -OH, -0-(Ci-C 4 alkyl), halo, -NH 2 , -NH(C C 4 alkyl), or -N(C C 4 alkyl) 2 ; and
  • any carbon atom on a phenyl, carbocycle (e.g., cycloalkyl), heteroaryl or heterocycle substituent is optionally further substituted with one or more of -(Ci-C 4 alkyl), -(C C 4 fluoroalkyl), -OH, -0-(C C 4 alkyl), -0-(C C 4 fluoroalkyl), halo, -NH 2 , -NH(CrC 4 alkyl), or -N(C C 4 alkyl) 2 .
  • All heterocyclyl ring systems (and any heterocyclyl substituents on any ring system) are optionally substituted on one or more any substitutable nitrogen atom with -Ci-C 4 alkyl, oxo, fluoro-substituted C C 4 alkyl, or acyl.
  • substituted refers to the replacement of a hydrogen atom by another group.
  • oxo refers to an oxygen atom, which forms a carbonyl when attached to carbon, an N-oxide when attached to nitrogen, and a sulfoxide or sulfone when attached to sulfur.
  • selective is meant at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, or 10-fold greater activation of PKM2 than PKM1.
  • activator means an agent that (measurably) increases the activity of PKM2 or causes PKM2 activity to increase to a level that is greater than PKM2's basal levels of activity.
  • the activator may mimic the effect caused by a natural ligand (e.g., FBP).
  • the activator effect caused by a compound provided herein may be to the same, or to a greater, or to a lesser extent than the activating effect caused by a natural ligand, but the same type of effect is caused.
  • a compound provided herein can be evaluated to determine if it is an activator by measuring either directly or indirectly the activity of the pyruvate kinase when subjected to said compound.
  • the activity of PKM2 can be measured, for example, by monitoring the concentration of a substrate such as ATP or NADH, for example, as described herein.
  • Compounds Described herein are compounds and compositions that activate PKM2.
  • Compounds that activate PKM2 can be used to treat disorders such as neoplastic disorders (e.g., cancer) or fat related disorders (e.g., obesity).
  • a compound of formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising a compound of formula (I) or a
  • W, X, Y and Z are each independently selected from CH
  • Q and Q 1 are independently selected from a bond or NR b ;
  • A is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl
  • L is a bond, -C(O)-, -(CR c R c ) m -, -OC(O)-, -(CR c R c ) m -OC(0)-, -(CR c R c ) m -C(0)-, - NR b C(S)-, or -NR b C(0)- (wherein the point of the attachment to R 1 is on the left-hand side);
  • R 1 is selected from alkyl, carbocycle, aryl, heteroaryl, and heterocyclyl; each of which is substituted with 0-5 occurrences of R d ;
  • each R 3 is independently selected from halo, haloalkyl, alkyl, hydroxyl and -OR a , or two adjacent R taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
  • each R a is independently selected from alkyl, acyl, hydroxyalkyl and haloalkyl;
  • each R b is independently selected from hydrogen and alkyl
  • each R c is independently selected from hydrogen, halo, alkyl, alkoxy and halo alkoxy or two R c taken together with the carbon atoms to which they are attached form an optionally substituted cycloalkyl;
  • each R d is independently selected from halo, haloalkyl, haloalkoxy, alkyl, alkynyl, nitro, cyano, hydroxyl, -C(0)R a , -OC(0)R a , -C(0)OR a , -SR a , -NR a R b and -OR a , or two R d taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
  • n 0, 1, or 2;
  • n 1, 2 or 3;
  • h 0, 1, 2;
  • g 0, 1 or 2;
  • p 0, 1 or 2; and provided that the compound of formula (I) is not
  • p is 1 or 2. In one aspect of this embodiment, p is 2 and the compound has the formula la:
  • p is 1 or 2; and each R 4 is independently selected from (5 , )-alkyl, (R)-alkyl, (S)- phenyl, and (R)-phenyl.
  • g is 1, h is 1 ; p is 1 or 2; and each R 4 is independently selected from ( ⁇ -methyl, (R)-methyl, (5 , )-ethyl, (R)-ethyl, (S)- isopropyl, (R)-isopropyl, ( ⁇ -phenyl, and (R)-phenyl.
  • p is 2 and the two R 4 taken together with the carbon atoms to which they are attached form a phenyl ring that is fused to the piperazine ring.
  • n 1 or 2.
  • A is an optionally substituted bicyclic heteroaryl.
  • W, X, Y, Z and the carbons to which they are attached form a phenyl ring.
  • W, X, Y, Z and the carbons to which they are attached form a pyridyl ring.
  • W, X and Y are CH and Z is N.
  • X, Y and Z are CH and W is N.
  • W, X, Y, Z and the carbon atoms to which they are attached form a pyrimidyl ring.
  • W, X, Y, Z and the carbon atoms to which they are attached form a pyridazinyl ring.
  • the ring comprising W, X, Y and Z is unsubstituted (i.e., n is 0). In some embodiments, the ring comprising W, X, Y and Z is mono substituted (i.e., n is 1).
  • R is selected from fluoro, chloro methyl, ethyl, CF 3 , methoxy, and OCF .
  • Q is NR b and Q 1 is a bond. In some aspects of these
  • R b is methyl. In other aspects of these embodiments, R b is hydrogen (H).
  • L is a bond
  • L is -(CR c R c ) m - and m is 1. In some aspects of these
  • each R c is hydrogen. In other aspects of these embodiments, one R c is methyl and the other R c is hydrogen. In some aspects of these embodiments, one R c is -CF 3 and one R c is hydrogen. In some aspects of these embodiments, both R c are methyl. In some aspects of these embodiments, two R c taken together with the carbon to which they are attached form a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • L is ethyl or n-propyl.
  • L is -C(O)-.
  • L is -O-C(O)-.
  • L is -(CR c R c ) m -C(0)- and m is 1. In some aspects of these embodiments, each R c is hydrogen. In some aspects of these embodiments, one R c is methyl and one R c is hydrogen. In some aspects of these embodiments, both R c are methyl. In some embodiments, L is -(CR c R c ) m -0-C(0)- and m is 1 or 2. In some aspects of these embodiments, each R c is hydrogen.
  • L is selected from bond, -C(O)-, -OC(O)-, -CH 2 -OC(0)-, - -OC(0)-, -C(CH 3 ) 2 -C(0)-, -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 3 -, -CH(CH 3 )-, -CH(CF 3 )-, -C(CH 3 ) 2 -,
  • R 1 is selected from methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 1,2,3- thiadiazol-5-yl, l,2,3-thiadiazol-4-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, lH-imidazol-4-yl, lH-imidazol-2-yl, lH-pyrazol-3-yl, lH-pyrazol-4-yl, lH-pyrazol-5-yl, pyrazin-2-yl, oxazol-4-yl, isoxazol-5-yl, tetrahydrofuran-2-yl, tetrahydrofuran-3-y
  • R 1 is substituted with one or more substituents independently selected from fluoro, chloro, methyl, CF , and methoxy.
  • the compound of Formula I is selected from any one of the compounds set forth in the Examples, Table 1 or Table 2.
  • the compounds described herein can be made using a variety of synthetic techniques as set forth in the Examples. As can be appreciated by the skilled artisan, methods of synthesizing additional compounds of the formulae herein will be evident to those of ordinary skill in the art by appropriate modifications of the exemplified schemes. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d.
  • the compounds provided herein may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included within the scope. Unless otherwise indicated when a compound is named or depicted by a structure without specifying the
  • the compounds provided herewith may also contain linkages (e.g., carbon-carbon bonds) or substituents that can restrict bond rotation, e.g. restriction resulting from the presence of a ring or double bond.
  • linkages e.g., carbon-carbon bonds
  • substituents that can restrict bond rotation, e.g. restriction resulting from the presence of a ring or double bond.
  • the compounds provided herein may also comprise one or more isotopic substitutions.
  • H may be in any isotopic form, including ] ⁇ , 2 H (D or deuterium), and 3 H (T or tritium);
  • C may be in any isotopic form, including 12 C, 13 C, and 14 C;
  • O may be in any isotopic form, including 16 0 and 18 0; and the like.
  • the compounds provided herein may also be represented in multiple tautomeric forms, in such instances, expressly includes all tautomeric forms of the compounds described herein, even though only a single tautomeric form may be represented (e.g., alkylation of a ring system may result in alkylation at multiple sites; all such reaction products are expressly included). All such isomeric forms of such compounds are expressly included.
  • a salt for example, can be formed between an anion and a positively charged substituent (e.g., amino) on a compound described herein. Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate. Likewise, a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein.
  • a positively charged substituent e.g., amino
  • Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate.
  • a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein.
  • Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion.
  • Examples of prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing active compounds.
  • the compounds provided herein may be modified by appending appropriate functionalities to enhance selected biological properties, e.g., targeting to a particular tissue.
  • modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • the compounds described herein can be evaluated for ability to activate PKM2 by methods known in the art. Exemplary methods include contacting the compound with a cell-based assay which allows assessment of the ability to modulate (e.g., activate) PKM2. E.g., the candidate compound can be contacted with a cell and measuring the consumption of oxygen or production of lactate.
  • a change in cellular phosphoenolpyruvate, a change in glycerol-phosphate, a change in ribose or deoxyribose, a change in lipid synthesis, or a change in glucose conversion to lipid or nucleic acids or amino acids or protein can also be used to evaluate a compound for its ability to modulate PKM2 (e.g., activate PKM2).
  • the evaluation could also include measuring a change in pyruvate or a determination of an alteration in mitochondrial membrane potential, e.g., as measured by fluorescent potentiometric dyes.
  • PKM1 and PKM2 for use in the screening/testing method may be produced by any method known in the art for expression of recombinant proteins.
  • nucleic acids that encode the desired polypeptide may be introduced into various cell types or cell-free systems for expression.
  • Eukaryotic (e.g., COS, HEK293T, CHO, and NIH cell lines) and prokaryotic (e.g., E. coli) expression systems may be generated in which a PKM sequence is introduced into a plasmid or other vector, which is then used to transform living cells.
  • Constructs in which the PKM cDNA contains the entire open reading frame, or biologically active fragment thereof, are inserted in the correct orientation into an expression plasmid and may be used for protein expression.
  • Prokaryotic and eukaryotic expression systems allow for the expression and recovery of fusion proteins in which the PKM protein is covalently linked to a tag molecule on either the amino terminal or carboxy terminal side, which facilitates identification and/or purification.
  • tags that can be used include hexahistidine, HA, FLAG, and c-myc epitope tags.
  • An enzymatic or chemical cleavage site can be engineered between the PKM protein and the tag molecule so that the tag can be removed following purification.
  • the activity of the PKM enzyme measured in the screening/testing assay may be measured by, e.g., monitoring the concentration of a substrate (e.g., ATP or NADH) present in the reaction mixture.
  • a substrate e.g., ATP or NADH
  • Pyruvate produced by the enzymatic activity of pyruvate kinase, is converted into lactate by lactate dehydrogenase, which requires the consumption of NADH (NADH ⁇ NAD+).
  • NADH ⁇ NAD+ lactate dehydrogenase
  • the activity of PKM2 can be indirectly measured by monitoring the consumption of NADH through, e.g., fluorescence assays.
  • the activity of the PKM2 enzyme can be directly monitored by measuring the production of ATP, as ATP is produced when phosphoenolpyruvate is converted to pyruvate.
  • Methods for monitoring the amount of substrate in a reaction mixture include, e.g., absorbance, fluorescence, Raman scatter
  • the screening procedure requires the presence of specific components in the reaction mixture.
  • Components utilized in the assay include, e.g., a nucleoside diphosphate (e.g., ADP), phosphoenolpyruvate, NADH, lactate dehydrogenase, FBP, a reducing agent (e.g., dithiothreitol), a detergent (e.g., Brij 35), glycerol, and a solvent (e.g., DMSO).
  • a nucleoside diphosphate e.g., ADP
  • phosphoenolpyruvate phosphoenolpyruvate
  • NADH e.g., lactate dehydrogenase
  • FBP e.g., FBP
  • a reducing agent e.g., dithiothreitol
  • a detergent e.g., Brij 35
  • glycerol e.g., glycerol
  • solvent
  • Compounds useful as PKM2 activators are those demonstrate specificity and activation of PKM2 enzyme in the absence of FBP to a level greater than that of 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99, or 100% in the presence of FBP. Furthermore, compounds can be evaluated in the presence or absence of a phosphotyrosine peptide. Phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form. Compounds that bind to PKM2 and lock the enzyme in the active confirmation even in the presence of a
  • phosphotyrosine peptide will lead to the loss of allosteric control of PKM2 needed for shunting the biochemical intermediates from glycolysis into biosynthesis of other intermediates. This, in turn, will lead to inhibition of growth of cancer cells, activated immune cells and fat cells.
  • Described herein is a method to measure the activity of PKM2 activators in living cells and tissue.
  • a method to measure the activity of PKM2 activators in living cells and tissue One of ordinary skill in the art would recognize and understand that this method can be adapted to high throughput format, and can accommodate a variety of cell lines and growth conditions.
  • cells are treated with a compound described herein (i.e., a PKM2 activator). This compound is capable of entering the cell and binding to PKM2, inducing an activated conformation. The excess unbound compound is washed away with PBS, and the cells are lysed by snap-freezing on dry ice, followed by addition of a detergent-containing lysis buffer.
  • the lysate, in which activated PKM2 remains intact, is removed and added to a chemical cocktail including the chemicals necessary to measure pyruvate kinase activity, in an assay that is coupled to the LDHa enzyme.
  • the amount of pyruvate kinase activity that is measured is normalized to the total protein content in the lysate, and related to the concentration of PKM2 activator that was added to the cell. This allows an AC5 0 (concentration at which PKM2 is activated 50%) value to be derived.
  • the total fold-increase in activity over mock- treated cells can also be calculated, and the "maximum level of activation" can be used to distinguish between compounds that fully activate PKM2 and compounds that can only partially activate PKM2.
  • tissue for example, in a cell tumor
  • animals harboring the tissue/tumor of interest are dosed with a compound.
  • the tissue/tumor is harvested from the animal, snap-frozen, and then lysed and homogenized. The amount of pyruvate kinase activity in this lysate can then be quantitated as described above.
  • This lysis buffer (without PMSF) is available from Cell Signaling Technology as a lOx stock (#9803) 1 mM PMSF is added fresh from a 100 mM stock solution made up in isopropanol.
  • the stock solution can be stored at 4 degrees indefinitely.
  • the cells On the second day (Day 2), the cells should be 70-90% confluent.
  • the cells are then treated with a compound described herein dissolved in media at final assay concentrations in a 96-well assay block (500 uL) (Costar, #3956).
  • the final DMSO concentration is 0.1% (0.5 into 500 uL).
  • Compound dilutions in DMSO are prepared so that the final DMSO concentration is constant at all compound concentrations.
  • the media for the assay is RPMI-1640 (no phenol red, with L- glutamine @300 mg/L).
  • the media is then aspirated carefully from the cells using a multi-channel aspirator. 100 ⁇ L ⁇ of media w/compounds is added onto cells with a multichannel pipette. Each compound concentration is then assayed in triplicate (a duplicate assay is also sufficient).
  • the cells are treated for 1-4 hrs (this time is determined empirically compared to DMSO reference treatment).
  • PBS containing calcium and magnesium
  • lysis buffer is cooled on ice.
  • the cells are lysed and the pyruvate kinase activity is assayed.
  • the remaining media is aspirated and the cells are washed 2x with 100 uL ice-cold PBS.
  • the PBS is removed, and the cell plate frozen on dry ice for 5 minutes.
  • the cells are lysed in 50 ⁇ L ⁇ cold lysis buffer. Cells are subsequently kept on ice for 5 minutes, and then agitated on a plate shaker for 5 minutes (repeat 3x). Remove 10 ⁇ L ⁇ for protein quantitation (or use OD 2 8o on entire plate).
  • pyruvate kinase assay master mix 170 uL of pyruvate kinase assay master mix was added to each well (see end for recipe). 10 uL of cell lysate was then transferred into each well. The assay was initiated upon addition of 20 uL of ADP solution. The rates were then calculated against the initial rates to determine pyruvate kinase specific activity.
  • the concentration and type of detergent in the lysis buffer can be varied to accommodate the specific physicochemical properties of the specific PKM2 activator. For instance, the interaction between some PKM2 activators and PKM2 can be disrupted by higher detergent concentrations, but preserved when cells are lysed with lower detergent concentrations.
  • a method for treating or preventing a disease, condition or disorder as described herein comprising administering a compound, a pharmaceutically acceptable salt of a compound or pharmaceutical composition comprising a compound described herein (e.g., a compound of formula (I), (I-a), (II), in Examples, Table 1 or Table 2).
  • the compounds and compositions described herein can be administered to cells in culture, e.g. in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below.
  • the term "treat” or “treatment” is defined as the application or administration of a compound, alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a disorder (e.g., a disorder as described herein), a symptom of a disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder, or one or more symptoms of the disorder.
  • a disorder e.g., a disorder as described herein
  • an amount of a compound effective to treat a disorder refers to an amount of the compound which is effective, upon single or multiple dose administration to a subject, in treating a cell, or in curing, alleviating, relieving or improving a subject with a disorder beyond that expected in the absence of such treatment.
  • a compound alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a predisposition toward a disorder, with the purpose to prevent the occurrence of at least one symptom of the disorder or to delay onset of at least one symptom of the disorder).
  • an amount of a compound effective to prevent a disorder refers to an amount effective, upon single- or multiple-dose administration to the subject, in preventing or delaying the occurrence of the onset or recurrence of a disorder or a symptom of the disorder.
  • the term "subject” is intended to include human and non-human animals.
  • exemplary human subjects include a human patient having a disorder, e.g., a disorder described herein or a normal subject.
  • non-human animals includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals, e.g., sheep, dog, cat, cow, pig, etc.
  • a compound or composition described herein can be used to treat a neoplastic disorder.
  • a "neoplastic disorder” is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth.
  • Exemplary neoplastic disorders include: carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • the disclosed methods are useful in the prevention and treatment of cancer, including for example, solid tumors, soft tissue tumors, and metastases thereof.
  • the disclosed methods are also useful in treating non-solid cancers.
  • Exemplary solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary.
  • Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
  • a compound described herein is administered with one or morechemotherapies.
  • Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. "Chemotherapy” usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g., with the duplication of DNA or the separation of newly formed chromosomes. Most forms of
  • chemotherapy target all rapidly dividing cells and are not specific for cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
  • chemotherapeutic agents used in cancer therapy include, for example, antimetabolites (e.g., folic acid, purine, and pyrimidine derivatives) and alkylating agents (e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, toposimerase inhibitors and others).
  • antimetabolites e.g., folic acid, purine, and pyrimidine derivatives
  • alkylating agents e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, toposimerase inhibitors and others.
  • agents include Aclarubicin, Actinomycin, Alitretinon, Altretamine, Aminopterin, Aminolevulinic acid, Amrubicin, Amsacrine, Anagrelide, Arsenic trioxide, Asparaginase, Atrasentan, Belotecan, Bexarotene, endamustine, Bleomycin, Bortezomib, Busulfan, Camptothecin, Capecitabine, Carboplatin, Carboquone, Carmofur, Carmustine, Celecoxib, Chlorambucil,
  • Fluorouracil (5FU), Fotemustine, Gemcitabine, Gliadel implants, Hydroxycarbamide, Hydroxyurea, Idarubicin, Ifosfamide, Irinotecan, Irofulven, Ixabepilone, Larotaxel, Leucovorin, Liposomal doxorubicin, Liposomal daunorubicin, Lonidamine,
  • the chemotherapy agents can be used in combination with a compound described herein.
  • a compound described herein is administered with one or more targeted therapies.
  • Targeted therapy constitutes the use of agents specific for the deregulated proteins of cancer cells.
  • Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell.
  • Prominent examples are the tyrosine kinase inhibitors such as Axitinib, Bosutinib, Cediranib, dasatinib, erlotinib, imatinib, gefitinib, lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sorafenib, Sunitinib, and Vandetanib, and also cyclin-dependent kinase inhibitors such as Alvocidib and Seliciclib.
  • Monoclonal antibody therapy is another strategy in which the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells. Examples include the anti-HER2/neu antibody trastuzumab (HERCEPTIN®) typically used in breast cancer, and the anti-CD20 antibody rituximab and
  • Tositumomab typically used in a variety of B-cell malignancies.
  • Other exemplary anbibodies include Cetuximab, Panitumumab, Trastuzumab, Alemtuzumab, Bevacizumab, Edrecolomab, and Gemtuzumab.
  • Exemplary fusion proteins include Aflibercept and Denileukin diftitox.
  • the targeted therapy can be used in combination with a compound described herein.
  • Targeted therapy can also involve small peptides as "homing devices” which can bind to cell surface receptors or affected extracellular matrix surrounding the tumor. Radionuclides which are attached to these peptides (e.g., RGDs) eventually kill the cancer cell if the nuclide decays in the vicinity of the cell.
  • RGDs Radionuclides which are attached to these peptides
  • An example of such therapy includes BEXXAR®.
  • a compound described herein is administered with one or more immunotherapies.
  • Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor.
  • Contemporary methods for generating an immune response against tumors include intravesicular BCG immunotherapy for superficial bladder cancer, and use of interferons and other cytokines to induce an immune response in renal cell carcinoma and melanoma patients.
  • Allogeneic hematopoietic stem cell transplantation can be considered a form of immunotherapy, since the donor' s immune cells will often attack the tumor in a graft- versus -tumor effect.
  • the immunotherapy agents can be used in combination with a compound described herein.
  • a compound described herein is administered with one or more hormonal therapies.
  • the growth of some cancers can be inhibited by providing or blocking certain hormones.
  • hormone- sensitive tumors include certain types of breast and prostate cancers. Removing or blocking estrogen or testosterone is often an important additional treatment.
  • administration of hormone agonists, such as progestogens may be therapeutically beneficial.
  • the hormonal therapy agents can be used in combination with a compound described herein.
  • a compound or composition described herein can be used to treat or prevent obesity, e.g., in a human subject, e.g. a child or adult subject.
  • Obsity refers to a condition in which a subject has a body mass index of greater than or equal to 30.
  • Many compounds described herein can be used to treat or prevent an over- weight condition.
  • “Over-weight” refers to a condition in which a subject has a body mass index of greater or equal to 25.0.
  • the body mass index (BMI) and other definitions are according to the "NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults" (1998).
  • Treatment with the compound may be in an amount effective to alter the weight of the subject, e.g., by at least 2, 5, 7, 10, 12, 15, 20, 25, 30, 25, 40, 45, 50, or 55%.
  • Treatment with a compound may be in an amount effective to reduce the body mass index of the subject, e.g., to less than 30, 28, 27, 25, 22, 20, or 18.
  • the compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
  • a compound or composition described herein can be administered to treat obesity associated with Prader-Willi Syndrome (PWS).
  • PWS is a genetic disorder associated with obesity (e.g., morbid obesity).
  • a compound or composition described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • reduce cholesterol e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol
  • reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • compositions and routes of administration are provided.
  • compositions delineated herein include the compounds delineated herein (e.g., a compound described herein), as well as additional therapeutic agents if present, in amounts effective for achieving a modulation of disease or disease symptoms, including those described herein.
  • pharmaceutically acceptable carrier or adjuvant refers to a carrier or adjuvant that may be administered to a patient, together with a compound provided herewith, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions provided herewith include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-oc-tocopherol polyethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium
  • Cyclodextrins such as ⁇ -, ⁇ -, and ⁇ -cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3 -hydroxypropyl- ⁇ -cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein.
  • compositions provided herewith may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection.
  • the pharmaceutical compositions provided herewith may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer' s solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • surfactants such as Tweens or Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions provided herewith may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase is combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions provided herewith may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound provided herewith with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions provided herewith is useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds provided herewith include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier with suitable emulsifying agents.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions provided herewith may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included.
  • compositions provided herewith may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • compositions provided herewith comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds provided herewith. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds provided herewith in a single composition.
  • the compounds described herein can, for example, be administered by injection, intravenously, intraarterially, subdermally, intraperitoneally,
  • the methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect.
  • the pharmaceutical compositions provided herewith will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations contain from about 20% to about 80% active compound.
  • a maintenance dose of a compound, composition or combination provided herewith may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • a patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject is in need of activation of PKM2, and if the subject is determined to be in need of activation of PKM2, then administering to the subject a compound described herein.
  • a subject can be evaluated as being in need of activation of PKM2 using methods known in the art, e.g., by measuring the presence and/or activity of PKM2 in the patient.
  • the activity and/or level of PKM2 is evaluated in the cancer.
  • a patient receiving a compound described herein can be monitored, for example, for improvement in the condition and/or adverse effects. Improvement of a patient' s condition can be evaluated, for example, by monitoring the growth, absence of growth, or regression of the cancer (e.g., a tumor). In some embodiments, the patient is evaluated using a radiological assay or evaluation of hemolytic parameters.
  • the methods of treatment of this invention include a first step of identifying or selecting a patient who would benefit from modulation (e.g., activation) of PKM2 by determining the level of PKM2 activity in a patient or more particularly in an organ or cell of the patient (e.g., as opposed to merely being in need of treatment of the disorder itself, e.g., cancer).
  • the level of PKM2 would be compared to a control (e.g., the PKM2 activity of another patient not suffering from the disorder (e.g., cancer) or the PKM2 activity of the same patient taken at an earlier time) to determine if the current level of PKM2 activity warranted treatment with a compound of this invention.
  • a patient who has a level of PKM2 activity below that of a control would be a candidate for treatment with a compound of this invention.
  • the methods of treatment of this invention include the subsequent step of monitoring the level of PKM2 activity in a patient or more particularly in an organ or cell of the patient during the course of or following treatment with a compound of this invention to determine the efficacy of the treatment.
  • the level of PKM2 would be compared to a control (e.g., PKM2 activity of the same patient taken just prior to treatment) to determine if the PKM2 activity had been altered by the treatment, thus providing evidence of efficacy of the treatment.
  • a control e.g., PKM2 activity of the same patient taken just prior to treatment
  • an increase in PKM2 activity during the course of or following treatment is indicative that the treatment was effective.
  • R 1 is aryl or cyclopropyl
  • R c is methyl or CF 3
  • R 3 is alkyl and n is 0 or 1.
  • R 1 is cyclopropyl or aryl.
  • R d when present, is aryl; R 3 is methoxy or OCF 3 ; and q is 1, 2, 3, or 4.
  • R d Aryl (when present)
  • R 1 is cycloalkyl, heterocyclyl, aryl, or heteroaryl
  • R 3 is chloro, fluoro, CF 3 or
  • R 4 is alkyl or phenyl.
  • R 4 alkyl, phenyl
  • R 3 CI, F, CF 3 , OCF3
  • R 1 carbocyclyl, aryl, heterocyclyl, heteroaryl
  • R 1 is alkyl or aryl; and L is -(CR
  • R 1 Alkyl, Aryl
  • reaction mixture was then brought to room temperature and stirred for 6 h. After completion of the reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (3 x 25 ml). The organic layer was washed with water (3 x 10 ml), dried over anhydrous sodium sulfate, filtered and concentrated over the rotary evaporator to get the crude product.
  • the crude product was purified by column chromatography (60-120 silica gel, ethyl acetate:hexane, 4:6) to afford pure product LXI (0.3 gm, 60%) as an off-white solid.
  • reaction mixture was quenched by the addition of water (10 mL), diluted with diethyl ether (100 mL), washed with water (2 x 25 mL), brine (25 mL), dried over anhydrous sodium sulfate and concentrated in vacuo.
  • the crude product was purified by column chromatography (Silica gel, 60:120; ethyl acetate:hexane, 3:7) to afford product LXII in 65-72% yield.
  • R aryl, carbocyclyl
  • reaction mixture was cooled at room temperature and the residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 2:8) to afford the 3,8-dibenzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione in 40-45% yield.
  • R 1 is aryl or carbocyclyl; and L is -(CR c R c ) m -.
  • R aryl, carbocyclyl
  • R 1 is aryl, carbocyclyl, heterocyclyl or heteroaryl;
  • R 3 is OCF 3 or OCH 3 ;
  • R 4 is alkyl;
  • L is -C(O)- or -(CR c R c )-C(0)-;
  • n is 0 or 1; and
  • p is 0 or 1.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Quinoline Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pyridine Compounds (AREA)

Abstract

Compounds and compositions comprising compounds that modulate pyruvate kinase M2 (PKM2) are described herein. Also described herein are methods of using the compounds that modulate PKM2 in the treatment of cancer.

Description

Therapeutic Compounds and Compositions
CLAIM OF PRIORITY
This application claims priority from U.S.S.N. 61/428,030, filed December 29, 2010 which is incorporated herein by reference in its entirety.
BACKGROUND OF INVENTION
Cancer cells rely primarily on glycolysis to generate cellular energy and biochemical intermediates for biosynthesis of lipids and nucleotides, while the majority of "normal" cells in adult tissues utilize aerobic respiration. This fundamental difference in cellular metabolism between cancer cells and normal cells, termed the Warburg Effect, has been exploited for diagnostic purposes, but has not yet been exploited for therapeutic benefit.
Pyruvate kinase (PK) is a metabolic enzyme that converts phosphoenolpyruvate to pyruvate during glycolysis. Four PK isoforms exist in mammals: the L and R isoforms are expressed in liver and red blood cells, the Ml isoform is expressed in most adult tissues, and the M2 isoform is a splice variant of Ml expressed during embryonic development. All tumor cells exclusively express the embryonic M2 isoform. A well-known difference between the Ml and M2 isoforms of PK is that M2 is a low-activity enzyme that relies on allosteric activation by the upstream glycolytic intermediate, fructose- 1,6-bisphosphate (FBP), whereas Ml is a
constitutively active enzyme.
All tumor cells exclusively express the embryonic M2 isoform of pyruvate kinase, suggesting PKM2 as a potential target for cancer therapy. PKM2 is also expressed in adipose tissue and activated T-cells. Thus, the activation of PKM2 may be effective in the treatment of, e.g., obesity, diabetes, autoimmune conditions, and proliferation-dependent diseases, e.g., benign prostatic hyperplasia (BPH). Current inhibitors of pyruvate kinase are not selective, making it difficult to treat disease related to pyruvate kinase function.
Furthermore, phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form. Compounds that bind to PKM2 and lock the enzyme in the active confirmation will lead to the loss of allosteric control of PKM2 needed for shunting biochemical intermediates from glycolysis into biosynthesis of nucleotides and lipids. Thus, the activation of PKM2 (i.e., activators of PKM2) can also inhibit the growth and proliferation of cancer cells, activated immune cells, and fat cells.
There is a continuing need for novel treatments of diseases such as cancer, diabetes, obesity, autoimmune conditions, proliferation-dependent diseases (e.g., BPH), and other diseases related to the function of pyruvate kinase (e.g., PKM2).
SUMMARY OF INVENTION
Described herein are compounds of Formula I that activate pyruvate kinase M2 (PKM2) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000003_0001
W, X, Y and Z are each independently selected from CH or N;
Q and Q1 are independently selected from a bond or NRb;
A is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl;
L is a bond, -C(O)-, -(CRcRc)m-, -OC(O)-, -(CRcRc)m-OC(0)-, -(CRcRc)m-C(0)-, -NRbC(S)-, or -NRbC(0)- (wherein the point of the attachment to R1 is on the left-hand side);
R1 is selected from alkyl, carbocycle, aryl, heteroaryl, and heterocyclyl; each of which is substituted with 0-5 occurrences of Rd;
each R3 is independently selected from halo, haloalkyl, alkyl, hydroxyl and -ORa, or two adjacent R taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
each R4 is independently selected from halo, haloalkyl, alkyl, hydroxyl, =0, -ORa, and phenyl, or two R4 taken together with the carbon atoms to which they are attached form a carbocycle;
each Ra is independently selected from alkyl, acyl, hydroxyalkyl and haloalkyl;
each Rb is independently selected from hydrogen and alkyl; each Rc is independently selected from hydrogen, halo, alkyl, alkoxy and halo alkoxy or two Rc taken together with the carbon atoms to which they are attached form an optionally substituted carbocycle;
each Rd is independently selected from halo, haloalkyl, haloalkoxy, alkyl, alkynyl, nitro, cyano, hydroxyl, -C(0)Ra, -OC(0)Ra, -C(0)ORa, -SRa, -NRaRb and -ORa, or two Rd taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
n is 0, 1, or 2;
m is 1, 2 or 3;
h is 0, 1, 2;
g is 0, 1 or 2;
the sum of g + h is equal to or greater than 2; and
p is 0, 1 or 2; and provided that the compound of formula (I) is not
N-[3-[(3,5-dimethoxyphenyl)amino]-2-quinoxalinyl]-4-[(4-methyl-l- piperazinyl)carbonyl] - benzenesulfonamide;
N-[4-[[4-(2-furanylmethyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-2-oxo-N-[4-[[4-(2,2,2-trifluoroethyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-N-[4-[[4-(4-nitrophenyl)-l-piperazinyl]carbonyl]phenyl]-2-oxo-lH- benzimidazole-5-sulfonamide;
N-[4-[[4-(2-ethoxyphenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-2-oxo-N-[4-[[4-(3-thienylmethyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole-5-sulfonamide;
N-[4-[[4-(2,3-dimethylphenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-N-[4-[[4-(2-hydroxyphenyl)-l-piperazinyl]carbonyl]phenyl]-2-oxo-lH- benzimidazole-5-sulfonamide;
4-[4-[[(2,3-dihydro-2-oxo-lH-benzimidazol-5-yl)sulfonyl]amino]benzoyl]-l- piperazinecarboxylic acid ethyl ester; N-[4-[(4-acetyl-l-piperazinyl)carbonyl]phenyl]-2,3-dihydro-2-oxo-lH-benzimidazole-5- sulfonamide;
N-[4-[[4-(4-fluorophenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-2-oxo-N-[4-[(4-phenyl-l-piperazinyl)carbonyl]phenyl]-lH-benzimidazole-5- sulfonamide; or
2,3-dihydro-2-oxo-N-[4-[[4-(2-pyridinyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole-5-sulfonamide.
Also provided are pharmaceutical compositions comprising a compound of Formula I or a pharmaceutically acceptable salt thereof and the use of such compositions in methods of treating diseases and conditions that are related to reduced pyruvate kinase function (e.g., PKM2 function), including, e.g., cancer, diabetes, obesity, autoimmune disorders, and benign prostatic hyperplasia (BPH).
In another embodiments, provided is a method of modulating (e.g., increasing or decreasing) the level of PKM2 activity and/or glycolysis (e.g., modulating the endogenous ability of a cell in the patient to down regulate PKM2) in a patient in need thereof. The method comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby modulating (e.g., increasing or decreasing) the level of PKM2 activity and/or glycolysis in the patient. In certain embodiment, the method includes identifying or selecting a patient who would benefit from activation of PKM2. For example, the patient can be identified on the basis of the level of PKM2 activity in a cell of the patient for treatment of cancer associated with PKM2 function.
In another embodiment, provided is a method of inhibiting cell proliferation in a patient in need thereof. The method comprises the step of administering an effective amount of a compound described herein to the patient in need thereof, thereby inhibiting cell proliferation in the patient.
In another embodiment, the compound described herein is administered at a dosage and frequency sufficient to increase lactate production or oxidative phosphorylation.
DETAILED DESCRIPTION The details of construction and the arrangement of components set forth in the following description or illustrated in the drawings are not meant to be limiting. Embodiments can be practiced or carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of "including," "comprising," or "having," "containing", "involving", and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
Definitions
The term "halo" or "halogen" refers to any radical of fluorine, chlorine, bromine or iodine.
The term "alkyl" refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Q-C12 alkyl indicates that the group may have from 1 to 12 (inclusive) carbon atoms in it. In certain aspects, the term "alkyl" refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 6 carbon atoms. In other aspects, the term "alkyl" refers to a monovalent hydrocarbon chain that may be a straight chain or branched chain, containing 1 to 4 carbon atoms.
The term "haloalkyl" refers to an alkyl in which one or more hydrogen atoms are replaced by halo, and includes alkyl moieties in which all hydrogens have been replaced by halo (e.g., perfluoroalkyl).
The term "alkenyl" refers to a monovalent straight or branched hydrocarbon chain containing 2-12 carbon atoms and having one or more double bonds. Examples of alkenyl groups include, but are not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups. One of the double bond carbons may optionally be the point of attachment of the alkenyl substituent. In certain aspects, the term "alkenyl" refers to a monovalent straight or branched hydrocarbon chain containing 2-6 carbon atoms and having one or more double bonds. In other aspects, the term "alkenyl" refers to a monovalent straight or branched hydrocarbon chain containing 2-4 carbon atoms and having one or more double bonds.
The term "alkynyl" refers to a monovalent straight or branched hydrocarbon chain containing 2-12 carbon atoms and characterized in having one or more triple bonds. Examples of alkynyl groups include, but are not limited to, ethynyl, propargyl, and 3-hexynyl. One of the triple bond carbons may optionally be the point of attachment of the alkynyl substituent.
The terms "alkylamino" and "dialkylamino" refer to -NH(alkyl) and -NH(alkyl)2 radicals respectively.
The term "aralkylamino" refers to a -NH(aralkyl) radical.
The term "alkylaminoalkyl" refers to a (alkyl)NH-alkyl- radical.
The term "dialkylaminoalkyl" refers to a (alkyl)2N-alkyl- radical.
The term "mercapto" refers to an -SH radical.
The term "thioalkoxy" refers to an -S-alkyl radical.
The term "thioaryloxy" refers to an -S-aryl radical.
The term "alkoxy" refers to an -O-alkyl radical.
The term "aryl" refers to a monocyclic, bicyclic, or tricyclic aromatic hydrocarbon ring system. Examples of aryl moieties include, but are not limited to, phenyl, naphthyl, and anthracenyl.
The terms "arylalkyl" or "aralkyl" refer to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group. Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of "arylalkyl" or "aralkyl" include benzyl, 2-phenylethyl, 3-phenylpropyl, 9-fluorenyl, benzhydryl, and trityl groups.
The term "carbocyclyl" refers to a non-aromatic, monocyclic, bicyclic, or tricyclic hydrocarbon ring system. Carbocyclyl groups include fully saturated ring systems (e.g., cycloalkyls), and partially saturated ring systems.
The term "cycloalkyl" as employed herein includes saturated cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 3 to 12 carbons. Any ring atom can be substituted (e.g., by one or more substituents). Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclohexyl, methylcyclohexyl, adamantyl, and norbornyl.
The term "heteroaryl" refers to a fully aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms selected independently from N, O, or S if monocyclic, bicyclic, or tricyclic, respectively). The term "heterocyclyl" refers to a nonaromatic, 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively). The heteroatom may optionally be the point of attachment of the heterocyclyl substituent. Examples of heterocyclyl include, but are not limited to, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, pyrrolinyl, pyrimidinyl, and pyrrolidinyl.
Bicyclic and tricyclic ring systems containing one or more heteroatoms and both aromatic and non-aromatic rings are considered to be heterocyclyl groups according to the present definition. Such bicyclic or tricyclic ring systems may be alternately characterized as being an aryl or a heteroaryl fused to a carbocyclyl or heterocyclyl, particularly in those instances where the ring bound to the rest of the molecule is required to be aromatic.
The terms "heteroarylalkyl" and "heteroaralkyl", as used herein, refers to an alkyl group substituted with a heteroaryl group.
The term "heterocyclylalkyl", as used herein, refers to an alkyl group substituted with a heterocyclyl group.
The term "acyl" refers to an alkylcarbonyl, carbocyclecarbonyl, arylcarbonyl,
heterocyclylcarbonyl, or heteroarylcarbonyl substituent, any of which may be further substituted (e.g., by one or more substituents).
All ring systems (i.e, aryl, heteroaryl, carbocyclyl, cycloalkyl, heterocyclyl, etc.) or ring system portions of groups (e.g., the aryl portion of an aralkyl group) are optionally substituted at one or more substitutable carbon atoms with substituents including: halo, -C≡N, C C4 alkyl, =0, C3-C7 carbocyle (e.g., cycloalkyl), C1-C4 alkyl, -OH, -0-(d-C4 alkyl), -SH, -S-(C C4 alkyl), -(C1-C4 alkyl)-N(Rb')(Rb'), -N(Rb')(Rb ), -0-(C1-C4 alkyl)-N(Rb')(Rb'), -(C C4 alkyl) -0-(CrC4 alkyl)-N(Rb')(Rb'), -C(0)-0(Rb'), -OC(0)(Rb'), -0-C(0)-0(Rb'), -C(0)-N(Rb')(Rb'),
-N(Rb')-C(0)Rb', -N(Rb')C(0)N(Rb')(Rb'), -N(Rb')-S(0)i_2Rb', -S(0)i_2N(Rb')(Rb'), -N(Rb')S(0)i_ 2N(Rb')(Rb'), -(C1-C4 alkyl)-C(0)-N(Rb')(Rb'), -O-(heteroaryl), -O-(heterocycle), -O-phenyl, -heteroaryl, -heterocycle, and -phenyl, wherein:
each Rb is independently selected from hydrogen, -CrC4 alkyl, carbocycle, carbocyclylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocyclyl, or heterocyclylalkyl; or two Rb are taken together with the nitrogen atom to which they are bound to form a 4- to 8-membered saturated heterocycle optionally comprising one additional heteroatom selected from N, S, S(=0), S(=0)2, and O,
any alkyl substituent is optionally further substituted with one or more of -OH, -0-(Ci-C4 alkyl), halo, -NH2, -NH(C C4 alkyl), or -N(C C4 alkyl)2; and
any carbon atom on a phenyl, carbocycle (e.g., cycloalkyl), heteroaryl or heterocycle substituent is optionally further substituted with one or more of -(Ci-C4 alkyl), -(C C4 fluoroalkyl), -OH, -0-(C C4 alkyl), -0-(C C4 fluoroalkyl), halo, -NH2, -NH(CrC4 alkyl), or -N(C C4 alkyl)2.
All heterocyclyl ring systems (and any heterocyclyl substituents on any ring system) are optionally substituted on one or more any substitutable nitrogen atom with -Ci-C4 alkyl, oxo, fluoro-substituted C C4 alkyl, or acyl.
The term "substituted" refers to the replacement of a hydrogen atom by another group.
The term "oxo" refers to an oxygen atom, which forms a carbonyl when attached to carbon, an N-oxide when attached to nitrogen, and a sulfoxide or sulfone when attached to sulfur.
The term "selective" is meant at least 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, or 10-fold greater activation of PKM2 than PKM1.
The term "activator" as used herein means an agent that (measurably) increases the activity of PKM2 or causes PKM2 activity to increase to a level that is greater than PKM2's basal levels of activity. For example, the activator may mimic the effect caused by a natural ligand (e.g., FBP). The activator effect caused by a compound provided herein may be to the same, or to a greater, or to a lesser extent than the activating effect caused by a natural ligand, but the same type of effect is caused. A compound provided herein can be evaluated to determine if it is an activator by measuring either directly or indirectly the activity of the pyruvate kinase when subjected to said compound. The activity of PKM2 can be measured, for example, by monitoring the concentration of a substrate such as ATP or NADH, for example, as described herein.
Compounds Described herein are compounds and compositions that activate PKM2. Compounds that activate PKM2, can be used to treat disorders such as neoplastic disorders (e.g., cancer) or fat related disorders (e.g., obesity).
In one embodiment, provided is a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I) or a
pharmaceutically acceptable salt thereof:
Figure imgf000010_0001
(I)
wherein: W, X, Y and Z are each independently selected from CH
Q and Q1 are independently selected from a bond or NRb;
A is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl;
L is a bond, -C(O)-, -(CRcRc)m-, -OC(O)-, -(CRcRc)m-OC(0)-, -(CRcRc)m-C(0)-, - NRbC(S)-, or -NRbC(0)- (wherein the point of the attachment to R1 is on the left-hand side);
R1 is selected from alkyl, carbocycle, aryl, heteroaryl, and heterocyclyl; each of which is substituted with 0-5 occurrences of Rd;
each R3 is independently selected from halo, haloalkyl, alkyl, hydroxyl and -ORa, or two adjacent R taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
each R4 is independently selected from halo, haloalkyl, alkyl, hydroxyl, =0, -ORa and phenyl, or two R4 taken together with the carbon atoms to which they are attached form a bridged, fused or spyro-fused carbocycle, an aryl or a heteroaryl;
each Ra is independently selected from alkyl, acyl, hydroxyalkyl and haloalkyl;
each Rb is independently selected from hydrogen and alkyl;
each Rc is independently selected from hydrogen, halo, alkyl, alkoxy and halo alkoxy or two Rc taken together with the carbon atoms to which they are attached form an optionally substituted cycloalkyl;
each Rd is independently selected from halo, haloalkyl, haloalkoxy, alkyl, alkynyl, nitro, cyano, hydroxyl, -C(0)Ra, -OC(0)Ra, -C(0)ORa, -SRa, -NRaRb and -ORa, or two Rd taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
n is 0, 1, or 2;
m is 1, 2 or 3;
h is 0, 1, 2;
g is 0, 1 or 2;
the sum of g + h is equal to or greater than 2; and
p is 0, 1 or 2; and provided that the compound of formula (I) is not
N-[3-[(3,5-dimethoxyphenyl)amino]-2-quinoxalinyl]-4-[(4-methyl-l- piperazinyl)carbonyl] - benzenesulfonamide;
N-[4-[[4-(2-furanylmethyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-2-oxo-N-[4-[[4-(2,2,2-trifluoroethyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-N-[4-[[4-(4-nitrophenyl)-l-piperazinyl]carbonyl]phenyl]-2-oxo-lH- benzimidazole-5-sulfonamide;
N-[4-[[4-(2-ethoxyphenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-2-oxo-N-[4-[[4-(3-thienylmethyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole-5-sulfonamide;
N-[4-[[4-(2,3-dimethylphenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide;
2,3-dihydro-N-[4-[[4-(2-hydroxyphenyl)-l-piperazinyl]carbonyl]phenyl]-2-oxo-lH- benzimidazole-5-sulfonamide;
4-[4-[[(2,3-dihydro-2-oxo-lH-benzimidazol-5-yl)sulfonyl]amino]benzoyl]-l- piperazinecarboxylic acid ethyl ester;
N-[4-[(4-acetyl-l-piperazinyl)carbonyl]phenyl]-2,3-dihydro-2-oxo-lH-benzimidazole-5- sulfonamide;
N-[4-[[4-(4-fluorophenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole-5-sulfonamide; 2,3-dihydro-2-oxo-N-[4-[(4-phenyl-l-piperazinyl)carbonyl]phenyl]- lH-benzimidazole-5- sulfonamide; or
2,3-dihydro-2-oxo-N-[4-[[4-(2-pyridinyl)- l-piperazinyl]carbonyl]phenyl]- lH- benzimidazole-5-sulfonamide.
In certain embodiments of a compound of formula (I) or a pharmaceutically acceptable salt thereof p is 1 or 2. In one aspect of this embodiment, p is 2 and the compound has the formula la:
Figure imgf000012_0001
(la) or formula lb:
Figure imgf000012_0002
(lb). In an alternate aspect of this
embodiment, p is 1 or 2; and each R4 is independently selected from (5,)-alkyl, (R)-alkyl, (S)- phenyl, and (R)-phenyl. In an even more specific aspect of this embodiment, g is 1, h is 1 ; p is 1 or 2; and each R4 is independently selected from (^-methyl, (R)-methyl, (5,)-ethyl, (R)-ethyl, (S)- isopropyl, (R)-isopropyl, (^-phenyl, and (R)-phenyl. In still another alternate aspect, p is 2 and the two R4 taken together with the carbon atoms to which they are attached form a phenyl ring that is fused to the piperazine ring.
In certain embodiments of a compound of formula (I) or a pharmaceutically acceptable salt thereof n is 1 or 2.
In certain embodiments of a compound of formula (I) or a pharmaceutically acceptable
salt thereof, A is an optionally substituted bicyclic heteroaryl. In an aspect A is
Figure imgf000012_0003
In some embodiments, g is 1 or 2; h is 1 or 2; and g + h is 2 or 3. In one aspect of this embodiment g + h = 2. In an alternate aspect of this embodiment, g + h = 3.
In some embodiments, W, X, Y, Z and the carbons to which they are attached form a phenyl ring.
In some embodiments, W, X, Y, Z and the carbons to which they are attached form a pyridyl ring. In one aspect of this embodiment W, X and Y are CH and Z is N. In an alternate aspect X, Y and Z are CH and W is N.
In some embodiments, W, X, Y, Z and the carbon atoms to which they are attached form a pyrimidyl ring.
In some embodiments, W, X, Y, Z and the carbon atoms to which they are attached form a pyridazinyl ring.
In some embodiments the ring comprising W, X, Y and Z is unsubstituted (i.e., n is 0). In some embodiments, the ring comprising W, X, Y and Z is mono substituted (i.e., n is 1).
In some embodiments where n is 1, R is selected from fluoro, chloro methyl, ethyl, CF3, methoxy, and OCF .
In some embodiments, Q is NRb and Q1 is a bond. In some aspects of these
embodiments, Rb is methyl. In other aspects of these embodiments, Rb is hydrogen (H).
In some embodiments, L is a bond.
In some embodiments, L is -(CRcRc)m- and m is 1. In some aspects of these
embodiments, each Rc is hydrogen. In other aspects of these embodiments, one Rc is methyl and the other Rc is hydrogen. In some aspects of these embodiments, one Rc is -CF3 and one Rc is hydrogen. In some aspects of these embodiments, both Rc are methyl. In some aspects of these embodiments, two Rc taken together with the carbon to which they are attached form a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In some embodiments, L is ethyl or n-propyl.
In some embodiments, L is -C(O)-.
In some embodiments, L is -O-C(O)-.
In some embodiments, L is -(CRcRc)m-C(0)- and m is 1. In some aspects of these embodiments, each Rc is hydrogen. In some aspects of these embodiments, one Rc is methyl and one Rc is hydrogen. In some aspects of these embodiments, both Rc are methyl. In some embodiments, L is -(CRcRc)m-0-C(0)- and m is 1 or 2. In some aspects of these embodiments, each Rc is hydrogen.
In some embodiments, L is selected from bond, -C(O)-, -OC(O)-, -CH2-OC(0)-, - -OC(0)-, -C(CH3)2-C(0)-, -CH2-, -(CH2)2-, -(CH2)3-, -CH(CH3)-, -CH(CF3)-, -C(CH3)2-,
Figure imgf000014_0001
In some embodiments, R1 is selected from methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 1,2,3- thiadiazol-5-yl, l,2,3-thiadiazol-4-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, lH-imidazol-4-yl, lH-imidazol-2-yl, lH-pyrazol-3-yl, lH-pyrazol-4-yl, lH-pyrazol-5-yl, pyrazin-2-yl, oxazol-4-yl, isoxazol-5-yl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrofuran-4-yl, tetrahydro-2H- pyran-4-yl, tetrahydro-2H-pyran-3-yl, and tetrahydro-2H-pyran-2-yl.
In certain embodiments R1 is substituted with one or more substituents independently selected from fluoro, chloro, methyl, CF , and methoxy.
In certain embodiments, the compound of Formula I is selected from any one of the compounds set forth in the Examples, Table 1 or Table 2.
Table 1
Figure imgf000014_0002
Figure imgf000015_0001
Cmpd # Structure Cmpd# Structure
Figure imgf000017_0001
Figure imgf000018_0001
The compounds described herein can be made using a variety of synthetic techniques as set forth in the Examples. As can be appreciated by the skilled artisan, methods of synthesizing additional compounds of the formulae herein will be evident to those of ordinary skill in the art by appropriate modifications of the exemplified schemes. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser 's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.
The compounds provided herein may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included within the scope. Unless otherwise indicated when a compound is named or depicted by a structure without specifying the
stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound. The compounds provided herewith may also contain linkages (e.g., carbon-carbon bonds) or substituents that can restrict bond rotation, e.g. restriction resulting from the presence of a ring or double bond.
Accordingly, all cis/trans and E/Z isomers are expressly included.
The compounds provided herein (e.g. of Formula I) may also comprise one or more isotopic substitutions. For example, H may be in any isotopic form, including ]Η, 2H (D or deuterium), and 3H (T or tritium); C may be in any isotopic form, including 12C, 13C, and 14C; O may be in any isotopic form, including 160 and 180; and the like. The compounds provided herein may also be represented in multiple tautomeric forms, in such instances, expressly includes all tautomeric forms of the compounds described herein, even though only a single tautomeric form may be represented (e.g., alkylation of a ring system may result in alkylation at multiple sites; all such reaction products are expressly included). All such isomeric forms of such compounds are expressly included.
The compounds provided herein include the compounds themselves, as well as their salts and their prodrugs, if applicable. A salt, for example, can be formed between an anion and a positively charged substituent (e.g., amino) on a compound described herein. Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate. Likewise, a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein. Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion. Examples of prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing active compounds.
The compounds provided herein may be modified by appending appropriate functionalities to enhance selected biological properties, e.g., targeting to a particular tissue. Such modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
Methods of evaluatin2 compounds
The compounds described herein can be evaluated for ability to activate PKM2 by methods known in the art. Exemplary methods include contacting the compound with a cell-based assay which allows assessment of the ability to modulate (e.g., activate) PKM2. E.g., the candidate compound can be contacted with a cell and measuring the consumption of oxygen or production of lactate. A change in cellular phosphoenolpyruvate, a change in glycerol-phosphate, a change in ribose or deoxyribose, a change in lipid synthesis, or a change in glucose conversion to lipid or nucleic acids or amino acids or protein can also be used to evaluate a compound for its ability to modulate PKM2 (e.g., activate PKM2). The evaluation could also include measuring a change in pyruvate or a determination of an alteration in mitochondrial membrane potential, e.g., as measured by fluorescent potentiometric dyes.
PKM1 and PKM2 for use in the screening/testing method may be produced by any method known in the art for expression of recombinant proteins. For example, nucleic acids that encode the desired polypeptide may be introduced into various cell types or cell-free systems for expression. Eukaryotic (e.g., COS, HEK293T, CHO, and NIH cell lines) and prokaryotic (e.g., E. coli) expression systems may be generated in which a PKM sequence is introduced into a plasmid or other vector, which is then used to transform living cells. Constructs in which the PKM cDNA contains the entire open reading frame, or biologically active fragment thereof, are inserted in the correct orientation into an expression plasmid and may be used for protein expression. Prokaryotic and eukaryotic expression systems allow for the expression and recovery of fusion proteins in which the PKM protein is covalently linked to a tag molecule on either the amino terminal or carboxy terminal side, which facilitates identification and/or purification. Examples of tags that can be used include hexahistidine, HA, FLAG, and c-myc epitope tags. An enzymatic or chemical cleavage site can be engineered between the PKM protein and the tag molecule so that the tag can be removed following purification.
The activity of the PKM enzyme measured in the screening/testing assay may be measured by, e.g., monitoring the concentration of a substrate (e.g., ATP or NADH) present in the reaction mixture. Pyruvate, produced by the enzymatic activity of pyruvate kinase, is converted into lactate by lactate dehydrogenase, which requires the consumption of NADH (NADH→ NAD+). Thus, the activity of PKM2 can be indirectly measured by monitoring the consumption of NADH through, e.g., fluorescence assays. Additionally, the activity of the PKM2 enzyme can be directly monitored by measuring the production of ATP, as ATP is produced when phosphoenolpyruvate is converted to pyruvate. Methods for monitoring the amount of substrate in a reaction mixture include, e.g., absorbance, fluorescence, Raman scattering, phosphorescence, luminescence, luciferase assays, and radioactivity.
The screening procedure requires the presence of specific components in the reaction mixture. Components utilized in the assay include, e.g., a nucleoside diphosphate (e.g., ADP), phosphoenolpyruvate, NADH, lactate dehydrogenase, FBP, a reducing agent (e.g., dithiothreitol), a detergent (e.g., Brij 35), glycerol, and a solvent (e.g., DMSO). Exemplary reaction conditions are found in Table 2.
Table 2
Component of Reaction Condition Amount in Activation Assay
ADP 0.1-5.0 mM
Phosphoenolpyruvate 0.1-5.0 mM
NADH 10-1000 μΜ
Lactate dehydrogenase 0.1-10 units
Fructose- 1 ,6-bisphosphate 0
DTT 0.1-50 mM
Brij 35 0.01-1%
Glycerol 0.1-10%
Pyruvate Kinase M2 (used for screen) 1-100 pg
DMSO 1-10%
Compounds useful as PKM2 activators are those demonstrate specificity and activation of PKM2 enzyme in the absence of FBP to a level greater than that of 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99, or 100% in the presence of FBP. Furthermore, compounds can be evaluated in the presence or absence of a phosphotyrosine peptide. Phosphotyrosine peptide binding to PKM2 leads to a dissociation of FBP from PKM2 and conformational changes of PKM2 from an active, tetrameric form to an inactive form. Compounds that bind to PKM2 and lock the enzyme in the active confirmation even in the presence of a
phosphotyrosine peptide will lead to the loss of allosteric control of PKM2 needed for shunting the biochemical intermediates from glycolysis into biosynthesis of other intermediates. This, in turn, will lead to inhibition of growth of cancer cells, activated immune cells and fat cells.
Other methods for evaluating the present compounds also include an ex vivo assays as described below.
A PKM2 Ex- Vivo Assay.
Described herein is a method to measure the activity of PKM2 activators in living cells and tissue. One of ordinary skill in the art would recognize and understand that this method can be adapted to high throughput format, and can accommodate a variety of cell lines and growth conditions. In the assay, cells are treated with a compound described herein (i.e., a PKM2 activator). This compound is capable of entering the cell and binding to PKM2, inducing an activated conformation. The excess unbound compound is washed away with PBS, and the cells are lysed by snap-freezing on dry ice, followed by addition of a detergent-containing lysis buffer. The lysate, in which activated PKM2 remains intact, is removed and added to a chemical cocktail including the chemicals necessary to measure pyruvate kinase activity, in an assay that is coupled to the LDHa enzyme. The amount of pyruvate kinase activity that is measured is normalized to the total protein content in the lysate, and related to the concentration of PKM2 activator that was added to the cell. This allows an AC50 (concentration at which PKM2 is activated 50%) value to be derived. The total fold-increase in activity over mock- treated cells can also be calculated, and the "maximum level of activation" can be used to distinguish between compounds that fully activate PKM2 and compounds that can only partially activate PKM2.
In the case of measuring PKM2 activity from tissue (for example, in a cell tumor), animals harboring the tissue/tumor of interest are dosed with a compound. After a specified period of time in which exposure has been achieved in the target tissue/tumor of interest, the tissue/tumor is harvested from the animal, snap-frozen, and then lysed and homogenized. The amount of pyruvate kinase activity in this lysate can then be quantitated as described above.
Materials:
Lysis buffer*
20 mM Tris-HCl (pH 7.5)
150 mM NaCl
1 mM Na2EDTA
1 mM EGTA
1% Triton
2.5 mM sodium pyrophosphate
1 mM beta-glycerophosphate
1 mM Na3V04
1 μg/ml leupeptin
1 mM PMSF**
* This lysis buffer (without PMSF) is available from Cell Signaling Technology as a lOx stock (#9803) 1 mM PMSF is added fresh from a 100 mM stock solution made up in isopropanol. The stock solution can be stored at 4 degrees indefinitely.
Pyruvate kinase assay master mix (Same for PKM2 Activator Assay):
Table 3
Figure imgf000024_0001
Procedure:
On the first day (day 1) cells are normally cultured in RPMI-1640 (Lonza #12- 115°F) (with 25 mM Hepes, L-glutamine)/10 FBS. The cells are subsequently trypsinized and plated in RPMI-1640 (Lonza, #12-918F) (no phenol red,
supplemented with L-glutamine @300 mg/L (Sigma, #G8540))/10 FBS at the following densities in 96 well plates:
A549: 40k/well
100 uL final volume of media per well.
On the second day (Day 2), the cells should be 70-90% confluent. The cells are then treated with a compound described herein dissolved in media at final assay concentrations in a 96-well assay block (500 uL) (Costar, #3956). The final DMSO concentration is 0.1% (0.5 into 500 uL). Compound dilutions in DMSO are prepared so that the final DMSO concentration is constant at all compound concentrations. The media for the assay is RPMI-1640 (no phenol red, with L- glutamine @300 mg/L).
The media is then aspirated carefully from the cells using a multi-channel aspirator. 100 μL· of media w/compounds is added onto cells with a multichannel pipette. Each compound concentration is then assayed in triplicate (a duplicate assay is also sufficient).
The cells are treated for 1-4 hrs (this time is determined empirically compared to DMSO reference treatment). During the cell treatment, PBS (containing calcium and magnesium) and lysis buffer is cooled on ice.
The cells are lysed and the pyruvate kinase activity is assayed. The remaining media is aspirated and the cells are washed 2x with 100 uL ice-cold PBS. The PBS is removed, and the cell plate frozen on dry ice for 5 minutes. The cells are lysed in 50 μL· cold lysis buffer. Cells are subsequently kept on ice for 5 minutes, and then agitated on a plate shaker for 5 minutes (repeat 3x). Remove 10 μL· for protein quantitation (or use OD28o on entire plate).
In a fresh plate, 170 uL of pyruvate kinase assay master mix was added to each well (see end for recipe). 10 uL of cell lysate was then transferred into each well. The assay was initiated upon addition of 20 uL of ADP solution. The rates were then calculated against the initial rates to determine pyruvate kinase specific activity.
The concentration and type of detergent in the lysis buffer can be varied to accommodate the specific physicochemical properties of the specific PKM2 activator. For instance, the interaction between some PKM2 activators and PKM2 can be disrupted by higher detergent concentrations, but preserved when cells are lysed with lower detergent concentrations.
Methods of Treatment
In one embodiment, provided is a method for treating or preventing a disease, condition or disorder as described herein (e.g., treating) comprising administering a compound, a pharmaceutically acceptable salt of a compound or pharmaceutical composition comprising a compound described herein (e.g., a compound of formula (I), (I-a), (II), in Examples, Table 1 or Table 2).
The compounds and compositions described herein can be administered to cells in culture, e.g. in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below. As used herein, the term "treat" or "treatment" is defined as the application or administration of a compound, alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a disorder (e.g., a disorder as described herein), a symptom of a disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder, or one or more symptoms of the disorder.
As used herein, an amount of a compound effective to treat a disorder, or a "therapeutically effective amount" refers to an amount of the compound which is effective, upon single or multiple dose administration to a subject, in treating a cell, or in curing, alleviating, relieving or improving a subject with a disorder beyond that expected in the absence of such treatment.
As used herein, the term "prevent" is defined as the application or
administration of a compound, alone or in combination with, a second therapeutic agent to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a predisposition toward a disorder, with the purpose to prevent the occurrence of at least one symptom of the disorder or to delay onset of at least one symptom of the disorder).
As used herein, an amount of a compound effective to prevent a disorder, or a "a prophylactically effective amount" of the compound refers to an amount effective, upon single- or multiple-dose administration to the subject, in preventing or delaying the occurrence of the onset or recurrence of a disorder or a symptom of the disorder.
As used herein, the term "subject" is intended to include human and non-human animals. Exemplary human subjects include a human patient having a disorder, e.g., a disorder described herein or a normal subject. The term "non-human animals" includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals, e.g., sheep, dog, cat, cow, pig, etc. Neoplastic Disorders
A compound or composition described herein can be used to treat a neoplastic disorder. A "neoplastic disorder" is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth. Exemplary neoplastic disorders include: carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors. Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
The disclosed methods are useful in the prevention and treatment of cancer, including for example, solid tumors, soft tissue tumors, and metastases thereof. The disclosed methods are also useful in treating non-solid cancers. Exemplary solid tumors include malignancies (e.g., sarcomas, adenocarcinomas, and carcinomas) of the various organ systems, such as those of lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary (e.g., renal, urothelial, or testicular tumors) tracts, pharynx, prostate, and ovary. Exemplary adenocarcinomas include colorectal cancers, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, and cancer of the small intestine.
Without being bound by theory, applicants believe that altered PKM2 levels characterize a subset of all types of cancers, without regard to their cellular nature or location in the body. Thus, the compounds and methods of this invention are useful to treat any type of cancer that is characterized by altered PKM2 levels.
Chemotherapy
In some embodiments, a compound described herein is administered with one or morechemotherapies. Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. "Chemotherapy" usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g., with the duplication of DNA or the separation of newly formed chromosomes. Most forms of
chemotherapy target all rapidly dividing cells and are not specific for cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
Examples of chemotherapeutic agents used in cancer therapy include, for example, antimetabolites (e.g., folic acid, purine, and pyrimidine derivatives) and alkylating agents (e.g., nitrogen mustards, nitrosoureas, platinum, alkyl sulfonates, hydrazines, triazenes, aziridines, spindle poison, cytotoxic agents, toposimerase inhibitors and others). Exemplary agents include Aclarubicin, Actinomycin, Alitretinon, Altretamine, Aminopterin, Aminolevulinic acid, Amrubicin, Amsacrine, Anagrelide, Arsenic trioxide, Asparaginase, Atrasentan, Belotecan, Bexarotene, endamustine, Bleomycin, Bortezomib, Busulfan, Camptothecin, Capecitabine, Carboplatin, Carboquone, Carmofur, Carmustine, Celecoxib, Chlorambucil,
Chlormethine, Cisplatin, Cladribine, Clofarabine, Crisantaspase, Cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Daunorubicin, Decitabine, Demecolcine, Docetaxel, Doxorubicin, Efaproxiral, Elesclomol, Elsamitrucin, Enocitabine, Epirubicin, Estramustine, Etoglucid, Etoposide, Floxuridine, Fludarabine,
Fluorouracil (5FU), Fotemustine, Gemcitabine, Gliadel implants, Hydroxycarbamide, Hydroxyurea, Idarubicin, Ifosfamide, Irinotecan, Irofulven, Ixabepilone, Larotaxel, Leucovorin, Liposomal doxorubicin, Liposomal daunorubicin, Lonidamine,
Lomustine, Lucanthone, Mannosulfan, Masoprocol, Melphalan, Mercaptopurine, Mesna, Methotrexate, Methyl aminolevulinate, Mitobronitol, Mitoguazone, Mitotane, Mitomycin, Mitoxantrone, Nedaplatin, Nimustine, Oblimersen, Omacetaxine, Ortataxel, Oxaliplatin, Paclitaxel, Pegaspargase, Pemetrexed, Pentostatin, Pirarubicin, Pixantrone, Plicamycin, Porfimer sodium, Prednimustine, Procarbazine, Raltitrexed, Ranimustine, Rubitecan, Sapacitabine, Semustine, Sitimagene ceradenovec,
Satraplatin, Streptozocin, Talaporfin, Tegafur-uracil, Temoporfin, Temozolomide, Teniposide, Tesetaxel, Testolactone, Tetranitrate, Thiotepa, Tiazofurin, Tioguanine, Tipifarnib, Topotecan, Trabectedin, Triaziquone, Triethylenemelamine, Triplatin, Tretinoin, Treosulfan, Trofosfamide, Uramustine, Valrubicin, Verteporfin, Vinblastine, Vincristine, Vindesine, Vinflunine, Vinorelbine, Vorinostat, Zorubicin, and other cytostatic or cytotoxic agents described herein.
Because some drugs work better together than alone, two or more drugs are often given at the same time. Often, two or more chemotherapy agents are used as combination chemotherapy. In some embodiments, the chemotherapy agents (including combination chemotherapy) can be used in combination with a compound described herein.
Targeted therapy
In some embodiments, a compound described herein is administered with one or more targeted therapies. Targeted therapy constitutes the use of agents specific for the deregulated proteins of cancer cells. Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell. Prominent examples are the tyrosine kinase inhibitors such as Axitinib, Bosutinib, Cediranib, dasatinib, erlotinib, imatinib, gefitinib, lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sorafenib, Sunitinib, and Vandetanib, and also cyclin-dependent kinase inhibitors such as Alvocidib and Seliciclib. Monoclonal antibody therapy is another strategy in which the therapeutic agent is an antibody which specifically binds to a protein on the surface of the cancer cells. Examples include the anti-HER2/neu antibody trastuzumab (HERCEPTIN®) typically used in breast cancer, and the anti-CD20 antibody rituximab and
Tositumomab typically used in a variety of B-cell malignancies. Other exemplary anbibodies include Cetuximab, Panitumumab, Trastuzumab, Alemtuzumab, Bevacizumab, Edrecolomab, and Gemtuzumab. Exemplary fusion proteins include Aflibercept and Denileukin diftitox. In some embodiments, the targeted therapy can be used in combination with a compound described herein.
Targeted therapy can also involve small peptides as "homing devices" which can bind to cell surface receptors or affected extracellular matrix surrounding the tumor. Radionuclides which are attached to these peptides (e.g., RGDs) eventually kill the cancer cell if the nuclide decays in the vicinity of the cell. An example of such therapy includes BEXXAR®.
Immunotherapy In some embodiments, a compound described herein is administered with one or more immunotherapies. Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor. Contemporary methods for generating an immune response against tumors include intravesicular BCG immunotherapy for superficial bladder cancer, and use of interferons and other cytokines to induce an immune response in renal cell carcinoma and melanoma patients.
Allogeneic hematopoietic stem cell transplantation can be considered a form of immunotherapy, since the donor' s immune cells will often attack the tumor in a graft- versus -tumor effect. In some embodiments, the immunotherapy agents can be used in combination with a compound described herein.
Hormonal therapy
In some embodiments, a compound described herein is administered with one or more hormonal therapies. The growth of some cancers can be inhibited by providing or blocking certain hormones. Common examples of hormone- sensitive tumors include certain types of breast and prostate cancers. Removing or blocking estrogen or testosterone is often an important additional treatment. In certain cancers, administration of hormone agonists, such as progestogens may be therapeutically beneficial. In some embodiments, the hormonal therapy agents can be used in combination with a compound described herein.
Obesity and fat disorders
A compound or composition described herein can be used to treat or prevent obesity, e.g., in a human subject, e.g. a child or adult subject. "Obesity" refers to a condition in which a subject has a body mass index of greater than or equal to 30. Many compounds described herein can be used to treat or prevent an over- weight condition. "Over-weight" refers to a condition in which a subject has a body mass index of greater or equal to 25.0. The body mass index (BMI) and other definitions are according to the "NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults" (1998). Treatment with the compound may be in an amount effective to alter the weight of the subject, e.g., by at least 2, 5, 7, 10, 12, 15, 20, 25, 30, 25, 40, 45, 50, or 55%. Treatment with a compound may be in an amount effective to reduce the body mass index of the subject, e.g., to less than 30, 28, 27, 25, 22, 20, or 18. The compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
A compound or composition described herein can be administered to treat obesity associated with Prader-Willi Syndrome (PWS). PWS is a genetic disorder associated with obesity (e.g., morbid obesity).
A compound or composition described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
Compositions and routes of administration
The compositions delineated herein include the compounds delineated herein (e.g., a compound described herein), as well as additional therapeutic agents if present, in amounts effective for achieving a modulation of disease or disease symptoms, including those described herein.
The term "pharmaceutically acceptable carrier or adjuvant" refers to a carrier or adjuvant that may be administered to a patient, together with a compound provided herewith, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions provided herewith include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-oc-tocopherol polyethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium
carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as α-, β-, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3 -hydroxypropyl-β -cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein.
The pharmaceutical compositions provided herewith may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection. The pharmaceutical compositions provided herewith may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with
pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer' s solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions. Other commonly used surfactants such as Tweens or Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
The pharmaceutical compositions provided herewith may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase is combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
The pharmaceutical compositions provided herewith may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound provided herewith with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
Topical administration of the pharmaceutical compositions provided herewith is useful when the desired treatment involves areas or organs readily accessible by topical application. For application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier. Carriers for topical administration of the compounds provided herewith include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier with suitable emulsifying agents. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The pharmaceutical compositions provided herewith may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included.
The pharmaceutical compositions provided herewith may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
When the compositions provided herewith comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents, both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen. The additional agents may be administered separately, as part of a multiple dose regimen, from the compounds provided herewith. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds provided herewith in a single composition.
The compounds described herein can, for example, be administered by injection, intravenously, intraarterially, subdermally, intraperitoneally,
intramuscularly, or subcutaneously; or orally, buccally, nasally, transmucosally, topically, in an ophthalmic preparation, or by inhalation, with a dosage ranging from about 0.5 to about 100 mg/kg of body weight, alternatively dosages between 1 mg and 1000 mg/dose, every 4 to 120 hours, or according to the requirements of the particular drug. The methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect.
Typically, the pharmaceutical compositions provided herewith will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations contain from about 20% to about 80% active compound.
Lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician.
Upon improvement of a patient' s condition, a maintenance dose of a compound, composition or combination provided herewith may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
Patient selection and monitorin2
The compounds described herein activate PKM2. Accordingly, a patient and/or subject can be selected for treatment using a compound described herein by first evaluating the patient and/or subject to determine whether the subject is in need of activation of PKM2, and if the subject is determined to be in need of activation of PKM2, then administering to the subject a compound described herein.
A subject can be evaluated as being in need of activation of PKM2 using methods known in the art, e.g., by measuring the presence and/or activity of PKM2 in the patient. In some embodiments, the activity and/or level of PKM2 is evaluated in the cancer.
A patient receiving a compound described herein can be monitored, for example, for improvement in the condition and/or adverse effects. Improvement of a patient' s condition can be evaluated, for example, by monitoring the growth, absence of growth, or regression of the cancer (e.g., a tumor). In some embodiments, the patient is evaluated using a radiological assay or evaluation of hemolytic parameters.
In one embodiment, the methods of treatment of this invention include a first step of identifying or selecting a patient who would benefit from modulation (e.g., activation) of PKM2 by determining the level of PKM2 activity in a patient or more particularly in an organ or cell of the patient (e.g., as opposed to merely being in need of treatment of the disorder itself, e.g., cancer). The level of PKM2 would be compared to a control (e.g., the PKM2 activity of another patient not suffering from the disorder (e.g., cancer) or the PKM2 activity of the same patient taken at an earlier time) to determine if the current level of PKM2 activity warranted treatment with a compound of this invention. In one aspect, a patient who has a level of PKM2 activity below that of a control would be a candidate for treatment with a compound of this invention.
In another embodiment, the methods of treatment of this invention include the subsequent step of monitoring the level of PKM2 activity in a patient or more particularly in an organ or cell of the patient during the course of or following treatment with a compound of this invention to determine the efficacy of the treatment. The level of PKM2 would be compared to a control (e.g., PKM2 activity of the same patient taken just prior to treatment) to determine if the PKM2 activity had been altered by the treatment, thus providing evidence of efficacy of the treatment. In one aspect, an increase in PKM2 activity during the course of or following treatment is indicative that the treatment was effective.
EXAMPLES
In the synthesis examples set forth below, certain compounds have specified stereochemistry at one of more positions. These compounds were prepared using the indicated scheme either using the appropriate chirally pure reagents or were separated from a racemate produced by the indicated scheme using an appropriate chiral separation column, such as a Chiralpak AD-H column (250 x 4.6 mm) 5 μΜ column, eluting with 0.05% diethyl amine in hexane/isopropanol (75: 25 v/v) with a flow rate of 2 ml/min with absorbance monitored at 220 nm. The chiral HPLC elution conditions set forth above can be easily modified by those of skill in the art to optimize separation for various chiral compounds of this invention.
Example 1. Preparation of Compounds of Formula Ic:
Figure imgf000037_0001
wherein R1 is aryl or cyclopropyl; Rc is methyl or CF3; R3 is alkyl and n is 0 or 1.
Scheme 1
Figure imgf000038_0001
VII VIII V VI
Figure imgf000038_0002
Synthesis of Intermediate IV. To a stirred solution of aryl ketones II (17 mmol) in 10 ml of dichloromethane were added optionally substituted tert-butyl piperazine-l-carboxylate I (16 mmol), Et3N (48 mmol) and 1 M TiCl4 (8 mmol) at room temperature, followed by stirring the reaction mixture at room temperature for 18 h. To the reaction mixture was added a solution of NaBH3CN (48 mmol) in MeOH (5 ml) at room temperature, followed by stirring the reaction mixture at room temperature for 6 h. Ethyl acetate and saturated aqueous NaHCC>3 solution were added to the reaction mixture. The insoluble material obtained was filtered off using celite. The ethyl acetate layer was separated, washed with brine, dried over anhydrous MgS04 and filtered. The filtrate was concentrated in vacuum and the crude compound was taken for the next step without purification. The compound obtained from this step was dissolved in 10 ml of Methanolic HCl and stirred the reaction mixture for 2 h at room temperature. The mixture was concentrated under vacuum to leave intermediate IV as solid. The obtained solid was neutralized with base to get a free base which was used for the next step (45-60% over two steps).
Synthesis of ethyl 4-(quinoline-8-sulfonamido)benzoate (VII). To a solution of amine V (16 gm, 96.85 mmol) in a mixture (1:1) of DCM and pyridine, sulfonyl chloride VI (27.56 gm, 121.07 mmol) was added at room temperature under N2 atmosphere. The resulting mixture was allowed to stir for 16 hrs. After completion of reaction, the crude mixture was diluted with DCM, washed with water followed by IN HC1. The organic layer was then dried over Na2S04 and concentrated under reduced pressure to afford intermediate VII in 98% yields (34 gm).
Synthesis of 4-(quinoline-8-sulfonamido)benzoic acid (VIII). To a solution of sulfonamide VII (34 gm, 95.5 mmol) in THF and water (1:1), LiOH (20 gm, 47.66 mmol) was added and the resulting mixture was allowed to stir at 80°C overnight. After completion of reaction, the crude mixture was washed with EtOAc. The aqueous layer was acidified with citric acid and filtered. Thus obtained solid was washed with Et20 and azeotroped by toluene, under reduced pressure to afford acid VIII (30 gm, 95.8% yield) which was taken forward for the next step without further purification.
Synthesis of Compounds of Formula I According to Scheme 1. To a solution of acid VIII (1 mmol) in DMF (2 ml), PyBoP (Benzotriazole-l-yl-oxy-tris- (dimethylamino)-phosphonium hexafluorophosphate) (0.78 gm, 1.5 mmol) was added at 0°C and allowed to stir for 5 minutes. Then amine IV (1 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH-DCM, 2:8) to afford product IX in 40-66% yield.
The above procedure was used to produce the following compounds of Formula Ic using the appropriate aryl ketone II and the appropriate optionally substituted tert-butyl piperazine-l-carboxylate I.
(S)-N-(4-(4-(l-phenylethyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 387):
Figure imgf000040_0001
1H NMR (400 MHz, CDC13) δ: 1.3 (d, 3H), 2.2-2.4 (m, 4H), 3.2-3.4 (m, 2H), 3.5 (m, IH), 3.6-3.8 (m, 2H), 5.3 (s, IH), 7.0-7.4 (m, 8H), 7.5-7.65 (m, 2H), 8.0 (d, IH), 8.38 (m, 2H), 8.55 (s, IH), 9.0 (m, IH); HPLC Purity: 99.31%; Mass (M+1): 477.40.
N-(4-(4-(l-(3,5-difluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 331):
Figure imgf000040_0002
1H NMR (400 MHz, DMSOd6) δ: 1.3 (d, 3H), 2.2-2.4 (m, 4H), 3.2-3.5 (m, 2H), 3.6 (m, IH), 7.0 (m, 4H), 7.2-7.4 (m, 3H), 7.6 (m, 2H), 8.2-8.4 (m, 3H), 9.1 (m, IH), 10.2 (bs, IH); HPLC Purity: 91.96%; Mass (M+1): 537.10.
N-(4-(4-(l-(3-chloro-4-fluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000040_0003
1H NMR (400 MHz, DMSOd6) δ: 1.2 (d, 3H), 1.3 (m, IH), 2.2-2.5 (m, 6H), 3.1-3.4 (m, 2H), 7.0 (m, 4H), 7.2-7.4 (m, 3H), 7.8 (m, 2H), 8.2-8.4 (m, 3H), 9.1 (m, IH), 10.2 (bs, IH); HPLC Purity: 93.02%; Mass (M+1): 575.10.
N-(4-(4-(l-(2,3,4-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 403):
Figure imgf000041_0001
XH NMR (400 MHz, CDC13) δ: 1.2 (d, 3H), 1.4 (m, IH), 2.2-2.7 (m, 4H), 3.0- 3.6 (m, 4H), 7.0-7.25 (m,6H), 7.55-7.6 (m,2H), 8.2-8.25 (d, IH), 8.4 (m, IH), 8.5- 8.55 (d, IH), 9.1 (m, IH); HPLC Purity: 99.46%; Mass (M+l): 555.45.
N-(4-(4-(l-(2,3,6-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinol ):
Figure imgf000041_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.35 (d, 3H), 1.4 (m, IH), 2.2-2.7 (m, 4H), 3.0-3.6 (m, 4H), 7.0-7.25 (m,5H), 7.4 (m, IH), 7.78-7.8 (m, 2), 8.25-8.3 (d, IH), 8.4 (m, IH), 8.5-8.55 (d, IH), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 98.97%; Mass (M+l): 555.1.
N-(4-(4-(l-(2,6-difluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 357):
Figure imgf000041_0003
XH NMR (400 MHz, CDC13) δ: 1.3 (s, 3H), 2.2-2.5 (m, 4H), 3.0-3.4 (m, 2H), 3.5-3.8 (m, 2H), 7.0-7.2 (m, 6H), 7.4 (m, IH), 8.2-8.6 (m, 3H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, IH) 10.4 (s, IH); HPLC Purity: 98.29%; Mass (M+l): 537.20.
N-(4-(4-(l-(pyridin-3-yl)ethyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 370):
Figure imgf000042_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.3 (d, 3H), 2.2-2.2.4 (m, 4H), 3.2-3.6 (m, 4H), 3.5 (m, IH), 7.1 (m, 4H), 7.3 (m, IH), 7.7 (m, 3H), 8.3-8.5 (m, 5H), 9.0 (m, IH), 10.0 (s, IH); HPLC Purity: 98.12%; Mass (M+l): 502.40.
N-(4-(4-(l-(2,4,5-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quin
Figure imgf000042_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.4 (d, 3H), 2.2-2.4 (m, 2H), 3.0-3.8 (m, 4H), 3.90 (q, IH), 7.0-7.2 (m, 6H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 96.86%; Mass (M+l): 555.50.
N-(4-(4-(l-(2,3,5-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 396):
Figure imgf000042_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.25 (d, 3H), 2.2-2.4 (m, 4H), 3.1-3.7 (m, 4H), 3.90 (q, IH), 7.0-7.2 (m, 5H), 7.4 (m, IH), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 96.86%; Mass (M+l): 555.50.
N-(4-(4-(l-(2,4,6-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 397):
Figure imgf000043_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.4 (s, 3H), 2.0-2.4 (m, 4H), 3.0-3.6 (m, 2H), 3.90 (m, IH), 7.0-7.2 (m, 6H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 99.97%; Mass (M+l): 555.50.
N-(4-(4-(l-(3,4,5-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 398):
Figure imgf000043_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.4 (s, 3H), 2.0-2.4 (m, 4H), 3.0-3.6 (m, 4H), 3.90 (m, IH), 7.0-7.2 (m, 6H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 95.10%; Mass (M+l): 555.45.
N-(4-(4-(l-cyclopropylethyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compoun
Figure imgf000043_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.6 (d, 3H), 2.1-2.2 (m, 2H), 2.21-2.4 (m, 4H), 2.99-3.6 (m, 8H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH) 10.41 (bs, IH); HPLC Purity: 99.49%; Mass (M+l): 465.3.
(R)-N-(4-(4-(l-phenylethyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 388):
Figure imgf000044_0001
XH NMR (400 MHz, CDC13) δ: 1.3 (d, 3H), 2.2-2.4 (m, 4H), 3.2-3.8 (m, 4H), 3.5 (m, IH), 5.3 (s, IH), 7.0-7.4 (m, 8H), 7.55-7.65 (m, 2H), 8.0 (d, IH), 8.38-8.4 (m, 2H), 8.55 (s, IH), 9.0 (m, IH); HPLC Purity: 98.51%; Mass (M+l): 501.20.
N-(4-(4-(2,2,2-trifluoro-l-(4-fluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000044_0002
1H NMR (400 MHz, CDC13) δ: 2.2 (m, 4H), 3.2 (m, 2H), 3.7 (m, 2H), 4.7 (m, IH), 7.0 (m, 4H), 7.4-7.6 (m, 4H), 7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.0 (m, IH); HPLC Purity: 97.49%; Mass (M+l): 573.15.
N-(4-(4-(2,2,2-trifluoro-l-phenylethyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 358):
Figure imgf000044_0003
1H NMR (400 MHz, CDC13) δ: 2.2-2.5 (m, 2H), 3.0-3.4 (m, 2H), 3.5-3.8 (m, 4H), 4.6 (m, IH), 7.0 (m, 4H), 7.4 (m, 5H), 7.9 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH) 10.4 (s, IH); HPLC Purity: 97.65%; Mass (M+l): 555.15.
N-(4-(4-(2,4-dimethoxybenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 333):
Figure imgf000045_0001
XH NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 3.2-3.6 (m, 4H), 3.8 (m, 6H), 6.5 (m, 2H), 7.2 (m, 3H), 7.5 (m, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 98.57%; Mass (M+l): 631.60.
N-(4-((2R)-4-(l-(4-fluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)qui
Figure imgf000045_0002
1H NMR (400 MHz, DMSO-d6) δ: 0.9 (d, 3H), 1.0-1.12 (m, IH), 1.13-1.3 (d, 3H), 1.4 (m, IH), 1.99-2.1 (m, 2H), 2.8-3.6 (m, 4H), 7.0-7.2 (m, 5H), 7.22-7.4 (m, 2H), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.93%; Mass (M+l): 533.55.
N-(4-((2R)-4-(l-(3,5-difluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)qui
Figure imgf000045_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.1-1.21 (d, 6H), 1.82-2.1 (m, 2H), 2.6 (m, IH), 2.8-3.2 (m, 2H), 3.8-4.0 (m, 3H), 7.0-7.2 (m, 7H), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.95%; Mass (M+l): 551.3. N-(4-((2R)-4-(l-(4-chloro-3-fluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)qu
Figure imgf000046_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.21 (d, 6H), 1.23-1.3 (m, IH), 1.8-2.1 (m, 3H), 2.6-2.8 (m, 2H), 3.0-3.4 (m, 3H), 7.0-7.2 (m, 4H), 7.4-7.7 (m, 6H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.65%; Mass
(M+l): 567.3.
N-(4-((2R)-2-methyl-4-(l-(3,4,5-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quin
Figure imgf000046_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.21 (d, 6H), 1.8-2.1 (m, 2H), 2.4-2.45 (m, IH), 2.6-2.8 (m, 2H), 2.9-3.25 (m, 2H), 3.5-3.6 (m, IH), 7.0-7.2 (m, 6H), 7.4-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity:
99.18%; Mass (M+l): 569.5.
N-(4-((2R)-4-(l-(2,6-difluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)quino :
Figure imgf000046_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.24 (d, 3H), 1.8-2.0 (m, 2H), -2.8 (m, IH), 3.4-3.6 (m, 3H), 3.8-4.0 (m, 2H), 7.0-7.2 (m, 5H), 7.4-7.7 (m, 4H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.18%; Mass (M+1): 569.5.
N-(4-((2R)-4-(l-(2,4-difluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)quin
Figure imgf000047_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.15 (d, 3H), 1.8-2.0 (m, 2H), 2.6-3.0 (m, 4H), 3.4-3.6 (m, 2H), 7.0-7.4 (m, 5H), 7.41-7.7 (m, 3H), 8.0 (d, IH), 8.3- 8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.18%; Mass (M+1): 551.3
N-(4-((2S)-4-(l-(3,5-difluorophenyl)ethyl)-2-ethylpiperazine-l- carbonyl)phenyl)qu :
Figure imgf000047_0002
1H NMR (400 MHz, DMSO-d6) δ: 0.8 (t, 3H), 1.19 (d, 3H), 1.2 (m, IH), 1.6- 1.8 (m, 2H), 2.0-2.4 (m, 2H), 2.8-3.7 (m, 5H), 7.0-7.4 (m, 7H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 97.51%; Mass (M+1): 565.3
N-(4-((2S)-2-ethyl-4-(l-(3,4,5-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)qu :
Figure imgf000047_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.8 (t, 3H), 1.19 (d, 3H), 1.6-1.8 (m, 2H), 2.0-2.4 (m, 2H), 2.8-3.2 (m, 2H), 3.8-4.0 (m, 4H), 7.0-7.4 (m, 6H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.86%; Mass (M+l): 583.3
Example 2: Preparation of Compounds of Formula Id:
Figure imgf000048_0001
wherein R1 is cyclopropyl or aryl.
Scheme 2
Figure imgf000048_0002
XI XII XIII
H
Figure imgf000048_0003
XIV XV XVI
Figure imgf000048_0004
Cyclopropyl Synthesis of intermediate XL A solution of Aryl/Cycloalkyl methyl ketone X (1.6 mmol) in dry THF (10 ml) was cooled to -70°C in N2 atmosphere.
Methylmagnesium bromide (8 mmol) in THF was added slowly at -70°C and the reaction mixture stirred for 1 h under N2 atmosphere. The reaction mixture was quenched with saturated NH4C1 and diluted with Ethyl acetate (20 ml) and Brine (20 ml). The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 2:8) to afford product XI as an oil in 60-72% yield.
Synthesis of intermediate XII. To a solution of Gemdimethyl alcohol XI (1.1 mmol) in dry DCM (10 ml) was added sodium azide (3.5 mmol) at room temperature under N2 atmosphere. The reaction mixture was cooled to 0°C and was added a solution of TFA (3 mmol) in DCM (1 ml). The reaction bath was allowed to warm up to room temperature and stirred further for 1 h at room temperature. The white precipitate formed was filtered and concentrated the solvent to get the desired azide which was used for the next step without purification (Yield 50-55%).
Synthesis of intermediate XIII. To a stirred solution of azide (1 mmol) in MeOH (5 ml) under H2 atmosphere, was added 10% Pd/C (0.1 mmol) at room temperature. The reaction mixture stirred further for 3 h at room temperature and filtered through celite. The filtrate concentrated under reduced pressure to leave the amine which was used for the next step without purification (Yield 60%).
Synthesis of Intermediate XV. Tosyl Chloride (22 g, 0.114 moles, 3 eq) was dissolved in DCM (40 ml) at 0° C with stirring. To this solution was added a solution of diethanol amine XIV (4 g, 0.038 moles, 1 eq) and triethylamine (17 ml, 0.114 moles, 3 eq) in DCM (20 ml) at 0° C. Stirring was continued for overnight at room temperature after the addition was completed. The precipitate generated from the reaction was filtered and the solutions was washed with water, dilute HC1, saturated NaHCC>3 and brine in turn, and dried (Na2S04). The organic phase was concentrated under reduced pressure and purified by column chromatography (silica gel, 60-120 mess, 20% ethyl acetate in hexane) to give the intermediate XV as a white solid (9.8 g, 46%). ]H NMR (400 MHz, CDC13) 2.38 (s, 3H), 2.43 (s, 6 H), 3.32-3.36 (m, 4H), 4.05-4.16 (m, 4H), 7.21-7.38 (m, 6H), 7.61-7.81 (m, 6H). Synthesis of Intermediate XVI. To a solution of Tritosylate XV (1 g, 0.00176 moles, 1 eq) in 6 ml of DMF was added NaBr (0.93 g, 0.009 moles, 5 eq). The resulting suspension was stirred in an oil bath at 120° C for 4 h. After cooling to room temperature, the reaction mixture was concentrated to about 2 ml. The viscous milky product was poured into rapidly stirred mixture of ice- water (30 ml) and extracted with ethyl acetate (30 ml). The organic phase was dried (Na2S04) and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 10% ethyl acetate in hexane) to leave the product XVI as a pale yellow liquid (0.34 g, 51%) ]H NMR (400 MHz, CDC13) 2.41 (s, 3H), 3.44 (s, 8H), 7.38 (d, 4H), 7.76 (d, 4H)
Synthesis of Intermediate XVII. A mixture of dibromide XVI (0.150 g, 0.000389 moles, 1.1 eq) and amine XIII (0.000354 moles, 1 eq) and N, N- diisopropylethyl amine (0.15 ml) under nitrogen atmosphere was heated at 125° C for 20 h. The reaction was allowed to cool to room temperature. The reaction mixture is extracted with water (10 ml), ethyl acetate (20 ml) and the organic phase was dried (Na2S04) and concentrated under reduced pressure. The crude product was purified with column chromatography (silica gel, 60-120 mess, 20% ethyl acetate in hexane) to leave the product XVII as a pale yellow liquid (yield 55-60%)
Synthesis of Intermediate XVIII. To a mixture of compound N- tosylpiperazine derivative XVII (0.000398 moles, 1 eq) and 4-hydroxybenzoic acid (0.164g, 0.00119 moles, 3 eq) was added hydrogen bromide solution (33 wt % in acetic acid, 1.8 ml) at room temperature. The reaction mixture was stirred under nitrogen atmosphere for 2 days at room temperature. Water (10 ml) was slowly added to the reaction mixture and the reaction mixture was continuously stirred for 2 h. A white precipitate was formed which was removed by filtration. The filter cake was washed with water (2 x 10 ml). The combined acidic aqueous washes were washed with toluene (20 ml). The aqueous phase was then cooled to 0°C and basified with KOH pellets portion wise until pH > 10, and extracted with toluene (20 ml) and ethyl acetate (2 x20 ml). The combined organic phases were washed with brine, dried (Na2S04), and concentrated under pressure to give the product XVIII as a pale yellow liquid which is used for the next step (Yield 90%) Synthesis of Compounds of Formula Id. To a solution of acid VIII (1 mmol) in DMF (2 ml), PyBoP (Benzotriazole-l-yl-oxy-tris-(dimethylamino)- phosphonium hexafluorophosphate) (1.5 mmol) was added at 0°C and allowed to stir for 5 minutes. Then Gem dimethyl piperizine XVIII (1 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH-DCM, 2:8) to afford product XIX in 35-55% yield.
The above procedure was used to produce the following compounds of Formula Id using the appropriate methyl ketone X and acid VIII intermediates.
N-(4-(4-(2-(2-fluorophenyl)propan-2-yl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000051_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.22 (s, 6H), 2.2-2.4 (m, 4H), 2.99-3.2 (m, 2H), 3.4-3.6 (m, 2H), 7.0-7.2 (m, 5H), 7.22-7.4 (m, 3H), 7.6-7.8 (d, 2H), 8.25 (d, 1H), 8.4-8.5 (m, 2H), 9.1 (m, 1H), 10.4 (s, 1H); HPLC Purity: 96.19%; Mass (M+l): 535.05.
N-(4-(4-(2-phenylpropan-2-yl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 340):
Figure imgf000051_0002
1H NMR (400 MHz, DMSOd6) δ: 1.2 (s, 6H), 2.2-2.4 (m, 4H), 3.1-3.6 (m, 4H), 7.2 (m, 5), 7.5 (m, 3H), 7.7 (m, 2H), 8.3 (m, 2H), 8.6 (m IH), 9.0(m, IH);
HPLC Purity: 97.72%; Mass (M+l): 599.4.
N-(3-methoxy-4-(4-(2-phenylpropan-2-yl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 355):
Figure imgf000052_0001
XH NMR (400 MHz, CDC13) δ: 3.4-3.7 (s, 6H), 2.1-2.4 (m, 4H), 3.0 (m, 4H), 4.5 (s, 3H), 6.6-6.9 (m, 3H), 7.1-7.5 (m, 5H), 7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.0 (m, IH) 10.4 (s, IH); HPLC Purity: 95.72%; Mass (M+l): 545.3.
N-(2-methoxy-4-(4-(2-phenylpropan-2-yl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 356):
Figure imgf000052_0002
1H NMR (400 MHz, CDC13) δ: 1.3 (s, 6H), 2.2-2.5 (m, 4H), 3.2-3.7 (m, 7H), 6.8-7.1 (m, 2H), 7.0-7.4 (m, 6H), 7.6-7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, 2H); HPLC Purity: 94.22%; Mass (M+l): 544.66.
N-(4-(4-(2-cyclopropylpropan-2-yl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 438):
Figure imgf000052_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.2-0.37 (m, 4H), 0.8 (s, 6H), 3.0-3.8 (m, 8H), 7.0-7.2 (m, 4H), 7.5-7.8 (m, 2H), 8.0-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 94.80%; Mass (M+l): 479.4.
N-(4-(4-(2-methyl-2-phenylpropanoyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 359):
Figure imgf000053_0001
1H NMR (400 MHz, CDC13) δ: 1.2 (m, 6H), 3.0-3.6 (m, 8H), 7.0-7.4 (m, 7H), 7.6 (m, 2H), 7.9 (m, IH), 8.0-8.4 (m, 3H), 9.0 (m, IH); HPLC Purity: 99.85%; Mass (M+l): 627.2.
Example 3. Preparation of Compounds of Formula Ie:
Figure imgf000053_0002
wherein Rd, when present, is aryl; R3 is methoxy or OCF3; and q is 1, 2, 3, or 4.
Figure imgf000054_0001
3. NaHC03, pH 7 DIPEA,
XXIV 125°C, 20 h
Figure imgf000054_0002
XXVI
XXV
Figure imgf000054_0003
Rd = Aryl (when present) R3 = 2-OMe, 3-OMe, OCF3 q = 1 , 2, 3 or 4
Synthesis of Intermediate XXII. To a stirred solution NaH (21 mmol) in DMF (10 ml) at 0°C was added aryl acetonitrile XX (4.2 mmol) in DMF slowly and stirred for 15 minutes at the same temperature. Dibromoalkane (4.2 mmol, n = 2, 3, 4, 5) in DMF (5 ml) followed by sodium iodide was added to reaction mixture and was heated to 120°C for 4 h. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 1:9) to afford product XXII in 75-89% yield.
Synthesis of Intermediate XXIII. In a sealed tube a stirred solution of Nitrile XXII (3 mmol) in HC1 (aqueous, 6 ml) was heated for 24 h at 100°C. After completion of the reaction the reaction mixture was poured into ice water and extracted with ethyl acetate (20 ml). The organic layer was washed with brine, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 1:9) to afford product XXIII as a white solid in 50-65% yield.
Synthesis of Intermediate XXIV. Diphenylphosphoryl azide (0.85 ml, 0.00386 moles, 1.1 eq) was added to a solution of the acid XXIII (0.00346 moles, 1 eq) and triethylamine (1 ml, 0.00776 moles, 2.2 eq) in Toluene (12 ml) and the mixture was stirred at 90° C for 2 h. The mixture was cooled, diluted with ethyl acetate (15 ml) and washed with sodium carbonate (2 x 20 ml). The combined aqueous fractions were washed with brine (40 ml), dried (Na2S04) and the solvent was evaporated under reduced pressure. The residue was suspended in hydrochloric acid (5M, 2 ml) and the mixture was heated under reflux for 2 h. The mixture was cooled, the solvent was evaporated under reduced pressure and the residue was dried azetropically by evaporating toluene under reduced pressure to give the crude compound as a white solid. The solid was taken in ethyl acetate (20 ml), cooled and saturated solution of NaHCC>3 was added to achieve pH~7. The organic phase was dried (Na2S04), concentrated to leave the compound XXIV as pale yellow liquid.
Synthesis of Intermediate XXV. A mixture of dibromide XVI (0.150 g, 0.000389 moles, 1.1 eq) and amine XXIV (0.000354 moles, 1 eq) and N, N- diisopropylethyl amine (0.15 ml) under nitrogen atmosphere was heated at 125° C for 20 h. The reaction was allowed to cool to room temperature. The reaction mixture was extracted with water (10 ml), ethyl acetate (20 ml) and the organic phase was dried (Na2S04), and concentrated under pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 20% ethyl acetate in hexane) to leave the product XXV as a pale yellow liquid (yield 45-55%).
Synthesis of Intermediate XXVI. To a mixture of compound N- tosylpiperazine derivative XXV (0.000398 moles, 1 eq) and 4-hydroxybenzoic acid (0.164g, 0.00119 moles, 3eq) was added hydrogen bromide solution (33 wt % in acetic acid, 1.8 ml) at room temperature. The reaction mixture was stirred under nitrogen atmosphere for 2 days at room temperature. Water (10 ml) was slowly added to the reaction mixture and the reaction mixture was continuously stirred for 2 h. A white precipitate was formed which was removed by filtration. The filter cake was washed with water (2 x 10 ml). The combined acidic aqueous washes were washed with toluene (20 ml). The aqueous phase was then cooled to 0°C and basified with KOH pellets portion wise until pH > 10, and extracted with toluene (20 ml) and ethyl acetate (2 x20 ml). The combined organic phases were washed with brine, dried (Na2S04), and concentrated under pressure to give the product XXVI as a pale yellow liquid which is used for the next step (Yield 90%).
Synthesis of Compounds of Formula Ie. To a stirred solution of acid VIII (0.000315 moles, 1 eq) in DMF (5 ml), were added EDCI (0.066 g, 0.000346 moles, 1.1 eq), HOBt (0.047 g, 0.000346 moles, 1.1 eq) and DIPEA (0.13 ml, 0.00078 moles, 2.5 eq) at 0°C and stirred for 15 minutes. A solution of amine XXVI (0.000315 moles, 1 eq) was then added at 0°C and then the resulting mixture was allowed to stir at room temperature overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 mL). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 70% ethyl acetate in hexane) to give XXVIII in 40-45% yield.
The above procedure was used to produce the following compounds of Formula Ie using the appropriate aryl acetonitrile XX, dibromoalkane XXI, and acid VIII intermediates.
N-(4-(4-(l-phenylcyclopropyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound
Figure imgf000056_0001
1H NMR (400 MHz, CDC13) δ: 1.6 (m, 4H), 2.5 (m, 4H), 3.2 (m, 2H), 3.6 (m, 2H), 7.0 (m, 3H), 7.2 (m, 6H), 7.6 (m, 2H), 8.0 (m, 1H), 8.3 (m, 2H), 8.5 (s, 1H), 9.1 (m, 1H); HPLC Purity: 97.71%; Mass (M+l): 513.30. N-(4-(4-(l-(4-fluorophenyl)cyclopropyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 399):
Figure imgf000057_0001
1H NMR (400 MHz, CDC13) δ: 0.9-1.0 (m, 5H), 2.2-2.7 (m, 4H), 3.15-3.7 (m, 4H), 6.9-7.3 (m, 7H), 7.58-7.61 (m, 2H), 8.0 (d, IH), 8.2-8.4 (m, 2H), 8.5 (s, IH), 9.0 (s, IH); HPLC Purity: 99.60%; Mass (M+l): 531.45.
N-(2-methoxy-4-(4-(l-phenylcyclopropyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 352):
Figure imgf000057_0002
XH NMR (400 MHz, CDC13) δ: 0.9-1.0 (m, 4H), 2.5 (s, 3H), 3.2-3.7 (m, 8H), 6.5 (m, 2H), 7.2 (m, 6H), 7.7 (m, 3H), 8.0-8.4 (m, 3H), 9.0 (m, IH); HPLC Purity: 92.0%; Mass (M+l): 543.43.
N-(4-(4-(l-phenylcyclopropyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 353):
Figure imgf000057_0003
1H NMR (400 MHz, CDC13) δ: 0.9-1.0 (m, 4H), 2.5 (m, 4H), 3.2-3.7 (m, 5H), 6.8-7.1 (m, 4H), 7.5 (m, 3H), 7.7 (m, 3H), 8.0-8.4 (m, 3H), 9.0 (m, 2H); HPLC Purity: 96.83%; Mass (M+l): 597.34.
N-(3-methoxy-4-(4-(l-phenylcyclopropyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 393):
Figure imgf000058_0001
1H NMR (400 MHz, CDC13) δ: 0.89 (m, 2H), 0.90 (m, IH), 0.95 (m, 2H), 2.2-2.6 (m, 4H), 2.8-3.0 (m, 2H), 3.4 (s, 3H), 3.45-3.6 (m, 2H), 6.3 (m, IH), 6.7-6.9 (m, 2H), 7.0-7.2 (m, 4H), 7.55-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.4 (d, 2H), 8.5(s, IH), 9.0 (m, IH); HPLC Purity: 99.86%; Mass (M+l): 543.4.
N-(4-(4-(l-(4-fluorophenyl)cyclopropyl)piperazine-l-carbonyl)-2- methoxyphenyl)quinoli -8-sulfonamide (Compound 400):
Figure imgf000058_0002
1H NMR (400 MHz, CDC13) δ: 0.8 (m, 2H), 0.85 (m, IH), 1.0 (m, 2H), 2.2- 2.7 (m, 4H), 3.4 (s, 3H), 3.15-3.7 (m, 4H), 6.6-6.7 (m, 2H), 6.9-7.0 (m, 2H), 7.15-7.2 (m, 4H), 7.55-7.6(m, 3H), 8.0 (d, IH), 8.2 (d, IH), 8.4 (d, IH), 8.5 (s, IH), 9.0 (s, IH); HPLC Purity: 99.93%; Mass (M+l): 561.50.
N-(4-(4-(l-(4-fluorophenyl)cyclopropyl)piperazine-l-carbonyl)-3- methoxyphenyl)quinoli -8-sulfonamide (Compound 401):
Figure imgf000058_0003
1H NMR (400 MHz, CDC13) δ: 0.89-1.0 (m, 4H), 2.2-2.7 (m, 4H), 3.0-3.6 (m, 4H), 3.58 (s,3H), 6.3 (d,lH), 6.8-7.2 (m,6H), 7.55-7.6 (m,2H), 8.0 (d, IH), 8.2 (d, IH), 8.38 (d, IH), 8.5 (s, IH), 9.0 (s, IH); HPLC Purity: 99.68%; Mass (M+l): 561.45.
N-(4-(4-(l-(4-fluorophenyl)cyclopropyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 402):
Figure imgf000059_0001
1H NMR (400 MHz, CDC13) δ: 0.79 (m, 2H), 0.8 (m, 2H), 2.2-2.7 (m, 4H), 3.0-3.6 (m, 4H), 6.95-7.25 (m,5H), 7.55-7.6 (m,2H), 8.0 (d, IH), 7.9-7.95 (d, IH), 8.0-8.1 (d, IH), 8.38-8.4 (d, 2H), 9.1 (m, IH); HPLC Purity: 99.75%; Mass (M+1): 615.45.
N-(4-(4-(l-phenylcyclobutyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compou
Figure imgf000059_0002
XH NMR (400 MHz, DMSO-d6) δ: 0.92 (m, IH), 1.22 (m, 2H), 1.8-2.1 (m, 2H), 2.2-2.4 (m, 4H), 2.85-3.2 (m, IH), 3.4-3.6 (m, 4H), 7.0-7.2 (m, 4H), 7.22-7.4 (m, 5H), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.15%; Mass (M+l):527.55.
N-(4-(4-(l-phenylcyclopentyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compoun
Figure imgf000059_0003
XH NMR (400 MHz, CDC13) δ: 1.5-2.4 (m, 8H), 1.9-2.4 (m, 4H), 3.0-3.8 (m, 4H), 7.0-7.18 (m,4H), 7.19-7.3 (m, 4H), 7.5-7.6 (m, 2), 8.0 (d, IH), 8.2-8.4 (m, 2H), 8.45 (m, IH), 9.1 (m, IH); HPLC Purity: 97.95%; Mass (M+1): 541.55. N-(4-(4-(l-phenylcyclohexyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compou
Figure imgf000060_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.15-1.45 (m, 4H), 1.5-1.7 (m, 2H), 2.85- 2.2 (m, 8H), 3.0-3.5 (m, 4H), 7.0-7.2 (m, 4H), 7.2-7.4 (m, 5H), 7.6-7.8 (m, 2H), 8.2- 8.23 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.17%; Mass (M+l): 555.40.
Example 4: Prep
Figure imgf000060_0002
(If).
Scheme 4:
hanolic 0 °C-RT
Figure imgf000060_0003
Formula If Synthesis of Intermediate XXX. To a solution of acid VIII (6.09 mmol) in DMF, PyBoP (Benzotriazole- 1 -yl-oxy-tris-(dimethylamino)-phosphonium
hexafluorophosphate) (4.75 gm, 9.14 mmol) was added at 0°C and allowed to stir for 5 minutes. Then Boc protected piperizine XXIX (6.09 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH-DCM, 1:9) to afford product
XXVIII in 66% yield.
Synthesis of Intermediate XXXI. To a solution of MeOH- HC1 (10 ml) Boc protected amine XXX (4.03 mmol) was added and the resulting mixture was stirred for 1 nr. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product XXXI in 94.30% yield.
Synthesis of Compounds of Formula If. To a solution of amine XXXI (0.25 mmoles) and appropriate aldehyde (0.27 mmol) in DCM, acetic acid (0.2 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 min. Then NaBD4 (0.25 mmol) was added to reaction mixture and the resulting mixture was allowed to stir at 50°C for 2 nr. After completion of reaction, the crude mixture was diluted with DCM washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60- 120 mesh; MeOH-DCM, 2:8) to afford product XXXII in 45-55% yield.
The above procedure was used to produce the following compounds of Formula If using the appropriate aldehyde in the final step.
N-(4-(4-(4-fluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide-(D) (Compound 448):
Figure imgf000062_0001
XH NMR (400 MHz, DMSOd6) δ: 2.2-2.4 (m, 4H), 3.1-3.6 (m, 4H), 3.7 (m, IH), 7.2 (m, 6), 7.3 (m, 2H), 7.7 (m, 2H), 8.3 (m, 3H), 9.0(m, IH) 10.2 (bs, IH); HPLC Purity: 97.28%; Mass (M+l): 506.25
N-(4-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide-(D) (Compound 450):
Figure imgf000062_0002
XH NMR (400 MHz, DMSOd6) δ: 2.2-2.4 (m, 4H), 3.1-3.6 (m, 4H), 3.7 (m, IH), 7.2 (m, 7), 7.8 (m, 2H), 8.3 (m, 3H), 9.0(m, IH) 10.2 (bs, IH); HPLC Purity: 99.50%; Mass (M+l): 524.35
Example 5. Prep
Figure imgf000062_0003
Scheme 5:
Figure imgf000063_0001
XXXIII XXXIV XXXV
Figure imgf000063_0002
XXXVI XXXVII
XXXVII
Figure imgf000063_0003
Formula Ig
Synthesis of Intermediate XXXIV. To a stirred solution of optionally substituted ethyl benzoate XXXIII (0.38 g, 0.00204 moles) in dry THF (5 ml) was added L1AID4 at -78°C. The reaction mixture was stirred further for 2 h at -78°C and quenched with saturated solution of ammonium chloride. The crude mixture was diluted with ethyl acetate, washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 2:8) to afford product XXXIV in 60% yield.
Synthesis of Intermediate XXXV. To a stirred solution of compound
XXXIV (0.00204 moles) in dry DCM (10 ml) was added Et3N (0.75 ml, 0.0051 moles) at 0°C and stirred for 2 h. Mesyl chloride (0.16 ml, 0.00204 moles) was added to the reaction mixture and the mixture was stirred further for 2 h at room
temperature. The crude mixture was diluted with DCM and washed with water. The organic layer dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA- Hexane, 2:8) to afford product XXXV in 75% yield.
Synthesis of Intermeidate XXXVI. To a stirred solution of compound
XXXV (0.0013 moles) in dry DMF (10 ml) was added DIPEA (0.7 ml, 0.0039 moles) at room temperature and stirred for 2 h. Boc-Piperazine (0.24 gm, 0.0013 moles) was added to the reaction mixture and the mixture was stirred further for 3 h at room temperature. After completion of the reaction, the mixture was quenched with water and diluted with ethyl acetate. The organic layer dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 3:7) to afford product XXXVI in 70% yield.
Synthesis of Intermediate XXXVII. To a solution of MeOH HCl (10 ml) Boc protected amine XXXVI (4.03 mmol) was added and the resulting mixture was stirred for 1 h. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product XXXVII in 92% yield.
Synthesis of Compounds of Formula Ig. To a solution of unsubstituted acid VIII (6.09 mmol) in DMF, PyBoP (Benzotriazole-l-yl-oxy-tris-(dimethylamino)- phosphonium hexafluorophosphate) (4.75 gm, 9.14 mmol) was added at 0°C and allowed to stir for 5 minutes. Then Boc protected piperizine XXXVII (6.09 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH-DCM, 2:8) to afford product.
The following compound was produced by the above-described method using ethyl-3,5-difluorobenzoate as starting material.
N-(4-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide-(D2) (Compound 449):
Figure imgf000064_0001
1H NMR (400 MHz, CDC13) δ: 2.2-2.6 (m, 4H), 2.4-2.49 (m, 2H), 3.2-3.8 (m, 4H), 6.7-7.0 (m, 3H), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.1 (d, 1H), 8.2-8.6 (m, 3H), 9.0 (m, 1H), 10.46 (bs, 1H); HPLC Purity: 98.11%; Mass (M+l): 525.15.
Figure imgf000065_0001
Formula Ih
Synthesis of Intermediate XLII. Nitrogen was purged through a stirred solution of arylbromide (XLI, 2.15 mmol) in 1,4-dioxane (20 ml) at room temperature for 30 minutes. BINAP (0.134 gm, 0.215 mmol), palladium acetate (0.0096 g, 0.043 mmol) and cesium carbonate (1.40 gm, 4.3 mmol) were added to the reaction mixture and the nitrogen purging was continued for another 20 minutes and finally diamine (XL, 2.15 mmol) was added and stirred at 100 °C overnight under nitrogen atmosphere. After completion of the reaction (monitored by TLC), the reaction mixture was concentrated under vacuum. The residue was dissolved in water, extracted with ethyl acetate (3 x 50 ml). Combined organic extracts were washed with brine (20 ml), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was then purified by column chromatography (60- 120 silica gel) using 20 % ethyl acetate-hexane to afford compound XLII (40-60%).
Synthesis of Compounds of Formula Ih: To a stirred solution of the carboxylic acid (VIII, 0.61 mmol) in DMF at 0°C under nitrogen atmosphere, EDCI (0.129 gm, 0.671 mmol), HOBt (0.91 gm, 0.671 mmol) and DIPEA (0.31 ml, 1.83 mmol) were added and the resultant solution was stirred at room temperature for 30 min. Amine (XLII 0.61 mmol) was then added at 0°C and stirred overnight at room temperature. After completion of the reaction (monitored by TLC), the reaction mixture was poured into 1.0 M HC1 and extracted with EtOAc. The organic layer was washed with saturated NaHC03 solution, dried over NaS04 and filtered. The solvent was removed by rotary evaporation and the product was isolated by chromatography on silica gel (60-120 silica gel, 2% MeOH-DCM) or preparative HPLC to yield product (40-60%) as an off-white solid.
The following compounds were produced by the above-described method using the appropriate amine XL.
N-(4-(4-(2-methoxyphenyl)-2-methylpiperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 223):
Figure imgf000066_0001
*H NMR (400 MHz, DMSOd6) δ: 1.2 (d, 3H), 2.5-2.55 (m, 2H), 2.6-2.69 (m, 1H), 3.0- 3.2 (m, 4H), 3.8 (s, 3H), 6.8-7.0 (d, 4H), 7.1-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, 1H), 10.46 (bs, 1H); HPLC Purity: 99.04%; Mass (M+l): 517.40.
N-(4-((2R,6S)-4-(2-methoxyphenyl)-2,6-dimethylpiperazine-l- carbonyl)phenyl)quinoline-8-sulf
Figure imgf000066_0002
*H NMR (400 MHz, DMSOd6) δ: 1.2-1.4 (s, 6H), 2.4-2.49 (m, 2H), 2.6-2.69 (m, 2H), 3.0-3.2 (m, 2H), 3.8 (s, 3H), 4.2 (bs, 1H), 6.8-7.0 (d, 4H), 7.1-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.2- 8.6 (m, 3H), 9.0 (m, 1H), 10.46 (bs, 1H); HPLC Purity: 99.74%; Mass (M+l):531.40.
Example 7. Preparation of Compounds of Formula Ii:
Figure imgf000067_0001
(R4)' 0 (li),
wherein R1 is cycloalkyl, heterocyclyl, aryl, or heteroaryl; R3 is chloro, fluoro, CF3 or
OCF3; and R4 is alkyl or phenyl.
Scheme 7
Figure imgf000067_0002
R4 = alkyl, phenyl
R3 = CI, F, CF3, OCF3
STAB = Sodium tri-acetoxy borohydride
R1 = Cycloalkyl, Heterocyclyl, Aryl, Heteroaryl L = -(CRcRc)m- p = 0 or 1
Synthesis of Intermediate XLIV. To a solution of acid VIII (6.09 mmol) in DMF, PyBoP (Benzotriazole- 1 -yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate) (4.75 gm, 9.14 mmol) was added at 0°C and allowed to stir for 5 minutes. Then Boc protected piperazine/substituted piperizine I (1.13 gm, 6.09 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH- DCM, 2:8) to afford product XLIV in 66% yield. Synthesis of Intermediate XLV. To a solution of MeOH- HC1, Boc protected amine XLIV (4.03 mmol) was added and the resulting mixture was stirred for 1 nr. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product XLV (94.30% yield).
Synthesis of Compounds of Formula Ii. To a solution of amine XLV (0.25 mmol) and appropriate aldehyde (0.27 mmol) in DCM, acetic acid (0.2 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 minutes. Then sodium triacetoxyborohydride (STAB) (0.26 gm, 1.26 mmol) was added to reaction mixture and the resulting mixture was allowed to stir at 50°C for 1 h. After completion of reaction, the crude mixture was diluted with DCM washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH-DCM, 2:8) to afford product in 32-45% yield.
The following compounds were produced by the above-described method using the appropriate N-Boc protected piperazine I and acid VIII.
N-(4-(4-(4-fluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compou
Figure imgf000068_0001
1H NMR (400 MHz, CDC13) δ: 2.2-2.6 (m, 4H), 2.8 (s, 2H), 3.2-3.5 (m, 2H), 3.6-3.8 (m, 2H), 6.9-7.3 (m, 9H), 7.6 (m, 2H), 8.0 (m, 1H), 8.3 (m, 2H), 9.0(m, 1H); HPLC Purity: 98.15%; Mass (M+l): 503.76.
N-(4-(4-((lH-pyrazol-3-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 384):
Figure imgf000069_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 2H), 2.65 (s, 2H), 3.2-3.6 (m, 6H), 6.1 (s, IH), 7.0-7.2 (m, 4H), 7.4 (s, IH), 7.6-7.8 (m, 3H), 8.3 (d, IH), 8.4 (d, IH), 8.5 (d, IH), 9.0 (m, IH), 10.4 (s, IH), 12.6 (s, IH); HPLC Purity: 96.98%; Mass (M+l): 477.30.
N-(4-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 394):
Figure imgf000069_0002
1H NMR (400 MHz, CDC13) δ: 2.2-2.6 (m, 4H), 2.4-2.49 (m, 2H), 3.2-3.8 (m, 4H), 6.7-7.0 (m, 3H), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.1 (d, IH), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 98.11%; Mass (M+l): 525.15.
N-(4-(4-((lH-pyrazol-4-yl)methyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000069_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 2H), 2.6 (s, 2H), 3.2-3.6 (m, 6H), 6.1 (s, IH), 7.0-7.2 (m, 4H), 7.4 (s, IH), 7.6 (s, IH), 7.7 (m, 2H), 8.3 (d, IH), 8.4 (d, IH), 8.5 (d, IH), 9.0 (m, IH), 10.4 (s, IH), 12.7 (s, IH); HPLC Purity: 99.42%; Mass (M+l): 477.30. N-(4-(4-((lH-imidazol-4-yl)methyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000070_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.2-3.4 (s, 2H), 3.4-3.6 (m, 3H), 6.1 (s, IH), 6.9 (s, IH), 7.0-7.2 (m, 4H), 7.5 (m, 2H), 7.6-7.8 (m, 2H), 8.3 (d, 3H), 8.4 (d, IH), 8.5 (d, IH), 9.0 (m, IH), 10.45 (s, IH), 12.9 (s, IH); HPLC Purity: 99.31%; Mass (M+l): 477.40.
N-(3-fluoro-4-(4-(4-fluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline- 8-sulfonamide (Comp
Figure imgf000070_0002
XH NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.0-3.2 (m, 2H), 3.4 (s, 2H), 3.5-3.6 (m, 2H), 6.9-7.4 (m, 7H), 7.5-7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 98.69%; Mass (M+l): 523.3.
N-(4-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)-3- fluorophenyl)quinoline-8-sulfonamide (Compound 421):
Figure imgf000070_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.0-3.2 (m, 2H), 3.4 (s, 2H), 3.5-3.6 (m, 2H), 6.9-7.4 (m, 6H), 7.5-7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 95.95%; Mass (M+l): 541.3. N-(4-(4-(4-chloro-3-fluorobenzyl)piperazine-l-carbonyl)-3- fluorophenyl)quinoline- -sulfonamide (Compound 422):
Figure imgf000071_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.0-3.2 (m, 2H), 3.4 (s, 2H), 3.5-3.6 (m, 2H), 6.9-7.4 (m, 6H), 7.5-7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 92.56%; Mass (M+l): 557.6.
N-(3-fluoro-4-(4-(3,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000071_0002
XH NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.0-3.2 (m, 2H), 3.4 (s, 2H), 3.5-3.6 (m, 2H), 6.9-7.1 (m, 2H), 7.2-7.4 (m, 3H), 7.5-7.7 (m, 2H), 8.2-8.25 (d, IH), 8.4-8.6 (m, 2H), 9.1-9.2 (m, IH), 10.78 (s, IH); HPLC Purity: 98.93%; Mass (M+l): 559.5.
N-(4-(4-(cyclopropylmethyl)piperazine-l-carbonyl)-3- fluorophenyl)quinoline-8-sulfonamide (Compound 424):
Figure imgf000071_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.1-0.2 (m, 2H), 0.35-0.45 (m, 2H), 0.8- 0.9 (m, IH), 2.0-2.4 (m, 6H), 3.0-3.2 (m, 2H), 3.4-3.6 (m, 2H), 6.9-7.2 (m, 3H), 7.59- 7.7 (m, 2H), 8.2-8.25 (d, IH), 8.4-8.6 (m, 2H), 9.1-9.2 (m, IH), 10.78 (s, IH); HPLC Purity: 98.95%; Mass (M+l): 469.3. N-(3-fluoro-4-(4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 425):
Figure imgf000072_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.4-1.5 (m, IH), 1.8-2.0 (m, IH), 2.0-2.4 (m, 7H), 3.0-3.2 (m, 2H), 3.4-3.6 (m, 7H), 6.9-7.2 (m, 3H), 7.59-7.7 (m, 2H), 8.2-8.25 (d, IH), 8.4-8.6 (m, 2H), 9.1-9.2 (m, IH), 10.78 (s, IH); HPLC Purity: 99.36%; Mass (M+l): 499.3.
N-(3-chloro-4-(4-(3,5-difluorobenzyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000072_0002
XH NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 2H), 3.0 (s, 2H), 3.4-3.6 (m, 4H), 7.0-7.2 (m, 6H), 7.59-7.7 (m, 2H), 8.2-8.25 (d, IH), 8.4-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 99.12%; Mass (M+l): 557.45.
N-(3-chloro-4-(4-(cyclopropylmethyl)piperazine-l- carbonyl)phenyl)quinolin -8-sulfonamide (Compound 427):
Figure imgf000072_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.1-0.2 (m, 2H), 0.3-0.4 (m, 2H), 0.8-0.85 (m,lH), 2.2-2.4 (m, 6H), 3.0 (s, 2H), 3.5-3.6 (m, 2H), 7.0-7.2 (m, 3H), 7.59-7.7 (m, 2H), 8.2-8.25 (d, IH), 8.4-8.6 (m, 2H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 99.57%; Mass (M+l): 485.5. N-(3-chloro-4-(4-(cyclopentylmethyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 428):
Figure imgf000073_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (m, 2H), 1.4-1.6 (m, 6H), 2.2-2.4 (m, 7H), 3.0 (s, 2H), 3.5-3.6 (m, 2H), 7.0-7.2 (m, 3H), 7.59-7.7 (m, 2H), 8.2-8.6 (m, 2H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 99.40%; Mass (M+1): 513.45.
N-(3-chloro-4-(4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 429):
Figure imgf000073_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.2-1.5 (m, IH), 1.8-2.0 (m, IH), 2.0-2.4 (m, 8H), 3.0 (m, 2H), 3.5-3.8 (m, 5H), 7.0-7.2 (m, 3H), 7.59-7.7 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 95.96%; Mass (M+1): 515.45.
N-(3-chloro-4-(4-(4-fluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline- 8-sulfonamide (Comp
Figure imgf000073_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 2.9-3.2 (s, 2H), 3.4-3.7 (m, 4H), 7.0-7.2 (m, 5H), 7.3-7.4 (m, 2H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.5 (s, IH); HPLC Purity: 98.11%; Mass (M+1): 539.50.
N-(3-chloro-4-(4-(4-chloro-3-fluorobenzyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 432):
Figure imgf000074_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 2.9-3.2 (s, 2H), 3.4-3.7 (m, 4H), 7.0-7.2 (m, 5H), 7.3-7.4 (m, IH), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 97.27%; Mass (M+l): 573.45.
N-(3-chloro-4-(4-(2,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000074_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 2.9-3.2 (s, 2H), 3.4-3.7 (m, 4H), 7.0-7.2 (m, 3H), 7.3-7.4 (m, 2H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 99.17%; Mass (M+l): 575.45.
N-(3-chloro-4-(4-(3,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000074_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 2.9-3.2 (s, 2H), 3.4-3.7 (m, 4H), 7.0-7.2 (m, 5H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 99.55%; Mass (M+l): 575.50.
N-(3-fluoro-4-(4-((tetrahydro-2H-pyran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 435):
Figure imgf000075_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (m, IH), 1.3-1.56 (m, 5H), 1.6-1.7 (m, IH), 2.2-2.4 (m, 6H), 2.9-3.4 (m, 3H), 3.7-3.8 (m, IH), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.6 (s, IH); HPLC Purity: 95.44%; Mass (M+1): 513.3.
N-(3-fluoro-4-(4-((tetrahydro-2H-pyran-4-yl)methyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 436):
Figure imgf000075_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (m, 3H), 1.56-1.6 (m, 3H), 2.2-2.6 (m, 5H), 2.99-3.4 (m, 5H), 3.6-3.8 (m, 3H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 93.35%; Mass (M+1): 513.3.
N-(3-fluoro-4-(4-((tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 437):
Figure imgf000075_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.4-1.45 (m, IH), 1.7-2.0 (m, 3H), 2.2-2.6 (m, 6H), 2.99-3.2 (m, 2H), 3.4-4.0 (m, 5H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 97.02%; Mass (M+1): 499.3.
N-(3-chloro-4-(4-((tetrahydro-2H-pyran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 439):
Figure imgf000076_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.8 (m, 6H), 2.1-2.7 (m, 6H), 3.0-3.8 (m, 7H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH) 10.6 (s, IH); HPLC Purity: 95.14%; Mass (M+l): 529.4.
N-(3-chloro-4-(4-((tetrahydro-2H-pyran-4-yl)methyl)piperazine-l- carbonyl)phenyl)quino :
Figure imgf000076_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.8 (m, 4H), 2.1-2.5 (m, 7H), 2.7-3.85 (m, 8H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH) 10.6 (bs, IH); HPLC Purity: 96.39%; Mass (M+l): 529.4.
N-(3-chloro-4-(4-((tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000076_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.6-2.0 (m, 4H), 2.1-3.0 (m, 7H), 3.4-4.0 (m, 6H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH) 10.6 (bs, IH); HPLC Purity: 97.11%; Mass (M+l): 515.3.
N-(4-(4-((5-fluoropyridin-3-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 390):
Figure imgf000077_0001
XH NMR (400 MHz, DMSO-d6) δ: 2.3-2.4 (m, 4H), 2.8 (s, 2H), 3.4-3.6 (m, 4H), 7.2-7.4 (m, 2H), 7.5-7.8 (m, 4H), 8.2-8.6 (m, 5H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 97.9 ; Mass (M+1): 590.0.
N-(4-(4-(cyclopropylmethyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 301):
Figure imgf000077_0002
XH NMR (400 MHz, DMSO-d6) δ: 0.1 (m, 2H), 0.4 (m, 2H), 0.8 (m, IH), 2.2 (d, 2H), (2.4-2.6 (m, 4H), 3.2-3.6 (m, 3H), 7.3(d, 2H), 7.5 (d, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m,lH), 9.1 (m, IH), 10(bs, IH); HPLC Purity: 98.12 ; Mass (M+1): 535.0.
N-(4-(4-((tetrahydrofuran-3-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy) phenyl)quinoline-8-sulfonamide (Compound 302):
Figure imgf000077_0003
XH NMR (400 MHz, DMSO-d6) δ: 0.6 (m, 2H), 0.9 (m, 2H), 0.8 (m, IH), 2.5 (m, 6H), 3.0 (m, 2H), 3.6 (m, IH), 3.7(m, 4H), 7.2 (m, 2H), 7.5 (d, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m,lH), 9.1 (m, IH), 10(bs, IH); HPLC Purity: 97.93%; Mass (M+1): 565.0. N-(4-(4-phenethylpiperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 303):
Figure imgf000078_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.4-2.6 (m, 4H), 2.7 (m, 4H), 3.2 (m, 2H), 3.7 (m, 2H), 7.1-7.4 (m, 7H), 7.6 (s, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 99.67% ; Mass (M+l): 585.
N-(4-(4-((3-fluoropyridin-4-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 304):
Figure imgf000078_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.0 (m, 2H), 4.6 (m, 4H), 3.7 (m, 2H), 7.3(m, 2H), 7.5 (m, 2H), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, 2H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 97.7%; Mass (M+l): 590.
N-(4-(4-((4-methoxypyridin-3-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 305):
Figure imgf000078_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.4(m, 6H), 3.8 (s, 3H), 7.0(m, IH), 7.3(m, 2H), 7.5 (m, IH), 7.7 (m, 2H), 8.3 (m, 4H), 8.6 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 98.7% ;Mass (M+l): 602. N-(4-(4-(2,3-dichlorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 306):
Figure imgf000079_0001
XH NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.4 (m, 2H), 3.8 (m, 4H), 7.2-7.4 (m, 6H), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 99% ;Mass (M+l): 639.
N-(4-(4-((3-chloropyridin-4-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 307):
Figure imgf000079_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.2 (s, 2H), 3.6 (m, 4H), 7.3 (m, 2H), 7.6 (m, 2H), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, 2H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 99.24%; Mass (M+l): 606.
N-(4-(4-(2-fluoro-6-methoxybenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy) phenyl)quinoline-8-sulfonamide (Compound 308):
Figure imgf000079_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.6 (s, 3H), 3.7 (s, 2H), 3.8 (m, 4H), 6.8 (m, 2H), 7.2 (m, 3H), 7.5 (m, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 96.97%; Mass (M+l): 619. N-(2-(trifluoromethoxy)-4-(4-((3-(trifluoromethyl)pyridin-2- yl)methyl)piperazine-l-carbonyl) phen l )quinoline-8-sulfonamicle (Compound 309):
Figure imgf000080_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.2 (s, 2H), 3.6-3.8 (m, 4H), 7.2 (m, 2H), 7.5 (m, 2H), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 8.8 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 98.32%; Mass (M+l): 640.
N-(4-(4-(4-methoxybenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 310):
Figure imgf000080_0002
XH NMR (400 MHz, DMSO-d6) δ: 2.4 (m, 4H), 3.2(s, 2H), 3.6-3.8 (m, 4H), 7.2 (m, 2H), 7.5 (m, 2H), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 8.8 (m, IH), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 97.32%; Mass (M+l): 640.
N-(4-(4-(pyridin-4-ylmethyl)piperazine-l-carbonyl)-2- (trifluoromethyl)phenyl)quinoline-8-sulfonamide (Compound 328):
Figure imgf000080_0003
1H NMR (400 MHz, CDC13) δ: 2.2-2.8 (m, 4H), 2.9 (s, 2H), 3.2-3.7 (m, 4H), 7.2 (m, IH), 7.6 (m, 4H), 7.9 (m, IH), 8.1 (m, IH), 8.3 (m, 2H), 8.6 (m, 3H), 9.1 (m, IH); HPLC Purity: 99.67%; Mass (M+l): 556.2. N-(4-(4-((tetrahydrofuran-2-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethyl)phenyl)quinoline-8-sulfonamide (Compound 329):
Figure imgf000081_0001
XH NMR (400 MHz, CDC13) δ: 1.5 (m, IH), 1.8-2.0 (m, 3H), 2.3-2.6 (m, 6H), 3.6 (s, 2H), 3.8 (m, 4H), 4.0 (m, IH), 7.4 (m, IH), 7.6 (m, 3H), 7.8 (m, IH), 8.0 (m, IH), 8.3 (m, IH), 8.5 (m, IH), 9.1 (m, IH); HPLC Purity: 98.77%; Mass (M+1): 549.2.
N-(2-(trifluoromethoxy)-4-(4-(4-(trifluoromethyl)benzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000081_0002
1H NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 2.8 (s, 2H), 3.2-3.6 (m, 2H), 3.8 (m, 2H), 7.2 (m, 2H), 7.5 (m, 3H), 7.8 (m, 4H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 99.88%; Mass (M+1): 639.25.
N-(4-(4-(2-fluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 335):
Figure imgf000081_0003
1H NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 2.8 (s, 2H), 3.2-3.6 (m, 2H), 3.8 (m, 2H), 7.2 (m, 6H), 7.5 (m, IH), 7.8 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 99.63%; Mass (M+1): 589.35. N-(4-(4-(cyclopentylmethyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 336):
Figure imgf000082_0001
1H NMR (400 MHz, DMSOd6) δ: 1.0 (m, 2H), 1.5 (m, 4H), 1.6 (m, 2H), 2.0 (m, IH), 2.3 (s, 2H), 2.2-2.5 (m, 4H), 3.2-3.6 (m, 4H), 7.2 (m, 2H), 7.5 (m, IH), 7.7 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 99.92%; Mass (M+l): 563.40.
N-(4-(4-(4-fluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 337):
Figure imgf000082_0002
XH NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 3.1 (s, 2H), 3.2-3.6 (m, 4H), 7.0 (m, 2H), 7.1 (m, 3.0), 7.5 (m, IH), 7.7 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 97.29%; Mass (M+l): 589.40.
N-(4-(4-(4-chlorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 338):
Figure imgf000082_0003
1H NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 3.1 (s, 2H), 3.2-3.6 (m, 4H), 7.3 (m, 5.0), 7.5 (m, IH), 7.7 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 99.95%; Mass (M+l): 605.35.
N-(4-(4-(4-chloro-2-fluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 339):
Figure imgf000083_0001
XH NMR (400 MHz, DMSOd6) δ: 2.2-2.5 (m, 4H), 3.1 (s, 2H), 3.2-3.6 (m, 4H), 7.3 (m, 5.0), 7.5 (m, IH), 7.7 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0(m, IH), 10.0 (bs, IH); HPLC Purity: 99.20%; Mass (M+l): 623.25.
N-(2-(trifluoromethoxy)-4-(4-((4-(trifluoromethyl)pyridin-3- yl)methyl)piperazine-l-arbonyl)phenyl)quinoline-8-sulfonamide (Compound 366):
Figure imgf000083_0002
XH NMR (400 MHz, DMSO-d6) δ: 2.2 (s, 4H), 3.2-3.6 (m, 4H), 3.9 (m, 2H), 7.2 (m, 2H), 7.5 (m, IH), 7.8 (m, 3H), 8.3 (m, 2H), 8.6-9.1 (m, 4H), 10.0 (s, IH); HPLC Purity: 99.76%; Mass (M+l): 640.40.
N-(4-(4-((5-chloropyridin-3-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 367):
Figure imgf000084_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.6 (m, 4H), 2.8 (s, 2H), 3.2-3.6 (m, 4H), 7.0-7.8 (m, 6H), 8.3-8.6 (m, 5H), 9.0 (m, IH), 10.0 (s, IH); HPLC Purity: 99.85%; Mass (M+l): 606.30.
N-(4-(4-((2-methoxypyridin-3-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 368):
Figure imgf000084_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.6 (m, 4H), 2.8 (s, 2H), 3.2-3.6 (m, 4H), 7.0-7.8 (m, 7H), 8.3-8.6 (m, 4H), 9.0 (m, IH), 10.0 (s, IH); HPLC Purity: 99.38%; Mass (M+l): 602.40.
N-(4-(4-(2,4-difluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 369):
Figure imgf000084_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 3.5 (s, 2H), 3.2-3.4 (m, 4H), 7.0 (m, IH), 7.2-7.3 (m, 3H), 7.4-7.6 (m, 2H), 7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.0 (m, IH), 10.0 (s, IH); HPLC Purity: 99.17%; Mass (M+l): 607.30.
N-(4-(4-((3-methoxypyridin-2-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 373):
Figure imgf000085_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.3-2.5 (m, 4H), 3.1-3.45 (s, 2H), 3.5-3.6 (m, 4H), 7.2-7.6 (m, 5H), 7.7 (m, 2H), 8.1 (m, IH), 8.3 (m, 2H), 8.6 (d, IH), 9.0 (m, IH), 9.90 (s, IH); HPLC Purity: 97.46%; Mass (M+l): 402.30.
N-(4-(4-(2,4-dichlorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 374):
Figure imgf000085_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.3-2.5 (m, 4H), 3.1-3.40 (m, 4H), 3.5-3.6 (s, 2H), 7.2-7.8 (m, 7H), 8.3 (m, 2H), 8.6 (d, IH), 9.0 (m, IH), 9.90 (bs, IH); HPLC Purity: 99.16%; Mass (M+l): 640.40.
N-(4-(4-(2,3-difluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 375):
Figure imgf000085_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.5 (m, 2H), 2.8 (s, 2H), 3.1-3.8 (m, 6H), 7.2-7.4 (m, 5H), 7.58 (m, IH), 7.75 (m, 2H), 8.3 (m, 2H), 8.6 (d, IH), 9.0 (m, IH), 9.90 (bs, IH); HPLC Purity: 98.91%; Mass (M+l): 607.30.
N-(4-(4-(3-chloro-4-fluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 376):
Figure imgf000086_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.5 (m, 4H), 2.69 (s, 2H), 3.2-3.8 (m, 4H), 7.2-7.2 (m, 4H), 7.58 (m, 2H), 7.75 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, IH), 9.90 (bs, IH); HPLC Purity: 95.94%; Mass (M+1): 623.25.
N-(4-(4-(3-fluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 377):
Figure imgf000086_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.5 (m, 4H), 2.7 (s, 2H), 3.2-3.8 (m, 4H), 7.2-7.4 (m, 6H), 7.58 (m, IH), 7.75 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, IH), 9.90 (bs, IH); HPLC Purity: 98.81%; Mass (M+1): 589.35.
N-(4-(4-(3,4-difluorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 378):
Figure imgf000086_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.5 (m, 4H), 2.8 (s, 2H), 3.2-3.8 (m, 4H), 7.18-7.2 (m, 2H), 7.2-7.4 (m, 3H), 7.58 (m, IH), 7.75 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, IH), 10.0 (bs, IH); HPLC Purity: 99.27%; Mass (M+1): 607.35.
N-(4-(4-(3-chlorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 379):
Figure imgf000087_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.5 (m, 4H), 3.2-3.8 (m, 6H), 7.18-7.4 (m, 6H), 7.58 (m, IH), 7.75 (m, 2H), 8.3 (m, 2H), 8.6 (m, IH), 9.0 (m, IH), 10.0 (bs, IH); HPLC Purity: 97.70%; Mass (M+l): 607.25
N-(4-(4-((lH-imidazol-2-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 380):
Figure imgf000087_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 4H), 2.67 (s, 2H), 3.2-3.6 (m, 4H), 6.9 (s, IH), 7.2-7.58 (m, 2H), 7.75 (m, 4H), 8.3 (m, 2H), 8.6 (d, IH), 9.0 (m, IH); HPLC Purity: 98.55%; Mass (M+l): 561.10.
N-(4-(4-((lH-imidazol-4-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 381):
Figure imgf000087_0003
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 2H), 2.8 (s, 2H), 3.2-3.6 (m, 6H), 6.9 (s, IH), 7.2-7.58 (m, 2H), 7.75 (m, 4H), 8.3 (m, 3H), 9.0 (m, IH); HPLC Purity: 99.39%; Mass (M+l): 561.10.
N-(4-(4-((lH-pyrazol-5-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 382):
Figure imgf000088_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.4 (m, 2H), 2.6 (s, 2H), 3.2-3.6 (m, 6H), 6.1 (s, IH), 7.2-7.75 (m, 6H), 8.3 (m, 3H), 8.6 (m, IH), 9.0 (m, IH) ; HPLC Purity: 96.98%; Mass (M+1): 561.10.
N-(4-(4-((lH-pyrazol-5-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 389):
Figure imgf000088_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.7 (m, 2H), 2.3 (m, 6H), 2.5 (m, 2H), 3.2-3.8 (m, 2H), 7.1-7.3 (m, 7H), 7.55 (d, IH), 7.78 (m, 2H), 8.38 (m, 2H), 8.6 (d, IH), 9.0 (m, IH), 9.9 (bs, IH); HPLC Purity: 89.93%; Mass (M+1): 599.35.
N-(4-(4-((5-fluoropyridin-2-yl)methyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 390):
Figure imgf000088_0003
XH NMR (400 MHz, DMSO-d6) δ: 2.2-2.6 (m, 2H), 2.8 (s, 2H), 3.2-3.8 (m, 6H), 7.2-7.3 (m, 2H), 7.55 (m, 2H), 7.78 (m, 3H), 8.38 (m, 2H), 8.6 (d, 2H), 9.0 (m, IH), 9.9 (bs, IH); HPLC Purity: 96.54%; Mass (M+1): 590.35.
N-(4-(4-(3,5-dichlorobenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 391):
Figure imgf000089_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.6 (m, 2H), 2.8 (s, 2H), 3.2-3.8 (m, 6H), 7.2-7.3 (m, 4H), 7.55 (m, 2H), 7.78 (m, 2H), 8.38 (m, 2H), 8.6 (d, IH), 9.0 (m, IH); HPLC Purity: 96.54%; Mass (M+l): 639.15.
N-(4-(4-(2,6-dimethoxybenzyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 392):
Figure imgf000089_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2-2.6 (m, 2H), 2.8 (s, 2H), 3.0-3.5 (m, 6H), 3.8 (s, 6H) 6.67 (m, 2H), 7.2-7.3 (m, 3H), 7.55 (m, IH), 7.78 (m, 2H), 8.38 (m, 2H), 8.6 (d, IH), 9.0 (m, IH); HPLC Purity: 99.67%; Mass (M+l): 631.20.
(R)-N-(4-(4-(cyclopropylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinol ):
Figure imgf000089_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.12 (m, 2H), 0.4 (m, 2H), 0.9 (m, IH), 2.0 (s, 2H), 2.1-2.2 (d, 2H), 2.6-3.2 (m, 2H), 4.0 (bs, IH), 7.0-7.2 (m, 4H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 99.84%; Mass (M+l): 465.05.
(R)-N-(4-(4-(cyclopentylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 114):
Figure imgf000090_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (m, 5H), 1.4-1.5 (m, 4H), 1.59-1.6 (m, 2H), 1.9 (s, 2H), 2.4 (d, 3H), 2.6-2.8 (m, 2H), 4.0 (bs, IH), 7.0-7.2 (m, 4H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 99.94%; Mass (M+l): 493.10.
N-(4-((2R)-2-methyl-4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000090_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.19 (m, 4H), 1.45-1.5 (m, IH), 1.7-2.0 (m, 3H), 2.0-2.2 (m, 2H), 2.35-2.4 (m, IH), 2.6-2.8 (m, 2H), 3.0 (bs, IH), 3.59-3.7 (m, 4H), 7.0-7.15 (m, 4H), 7.6-7.69 (m, 2H), 8.2-8.6 (m, 3H), 9.0 (m, IH), 10.46 (bs, IH); HPLC Purity: 98.25%; Mass (M+l): 495.10.
(R)-N-(4-(2-methyl-4-(2,3,4-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu :
Figure imgf000090_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.16 (s, 3H), 1.9-2.1 (m, 2H), 2.5-2.7 (m, 2H), 3.0-3.13 (m, IH), 3.5 (s, 2H), 4.1 (m, 2H), 7.1-7.4 (m, 6H), 7.7-7.8 (m, 2H), 8.3- 8.6 (m, 3H), 9.0 (s, IH), 10.6-10.7 (bs, IH); HPLC Purity: 99.83%; Mass (M+l): 555.35. (R)-N-(4-(4-(3,5-difluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu :
Figure imgf000091_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.21 (q, IH), 1.9-2.1 (m, 2H), 2.5-2.8 (m, 3H), 3.0-3.2 (s, 2H), 3.4-3.6 (m, 2H), 4.0 (bs, IH), 7.1-7.4 (m, 7H), 7.6- 7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.0 (s, IH), 10.6-10.7 (bs, IH); HPLC Purity: 99.64%; Mass (M+l): 537.35.
(R)-N-(4-(4-(2,3-dimethoxybenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu
Figure imgf000091_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.18-1.95 (d, 3H), 2.0 (m, IH), 1.8-2.1 (m, 2H), 2.45-2.8 (s, 2H), 2.99-3.2 (m, IH), 3.25-3.5 (m, 4H), 3.7 (s, 3H), 3.8 (s, 3H), 6.82-7.19 (m, 7H), 7.6-7.8 (m, 2H), 8.2-8.23 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 98.21%; Mass (M+l): 561.40.
(R)-N-(4-(4-(4-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu
Figure imgf000091_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.18-1.95 (d, 3H), 2.0 (m, IH), 2.1 (s, 2H), 2.45-2.8 (m, IH), 3.0-3.2 (m, IH), 3.8-4.0 (m, 4H), 7.0-7.2 (m, 6H), 7.22-7.4 (m, 2H), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.51%; Mass (M+1): 519.35.
(R)-N-(4-(2-methyl-4-(2,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu :
Figure imgf000092_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.9-2.15 (m, 2H), 2.6-2.8 (m, 2H), 3.0-3.2 (m, 2H), 3.4 (s, 2H), 4.0 (bs, IH), 7.4-7.5 (m, 2H), 7.6-7.8 (m, 2H), 8.3- 8.6 (m, 3H), 9.0 (s, IH), 10.4 (bs, IH); HPLC Purity: 99.97%; Mass (M+1): 555.25.
(R)-N-(4-(4-(4-chloro-3-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quino ):
Figure imgf000092_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.23 (m, IH), 1.5 (m, IH), 1.6-1.7 (m, 2H), 1.89-2.0 (s, 2H), 2.7-3.2 (m, 4H), 3.8-4.85 (m, 6H), 7.0-7.2 (m, 4H), 7.5-7.6 (m, 2H), 7.69-7.8 (m, 2H), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.96%; Mass (M+1): 553.25.
(R)-N-(4-(2-methyl-4-(2,3,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000092_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.12 (d, 3H), 1.2 (m, IH), 1.8-2.1 (m, 2H), 2.45-2.8 (s, 2H), 2.85-3.2 (m, IH), 3.8-4.0 (m, 3H), 7.0-7.2 (m, 5H), 7.22-7.4 (m, IH), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.18%; Mass (M+l): 555.25.
(S)-N-(4-(2-ethyl-4-(4-fluorobenzyl)piperazine-l- carbonyl)phenyl)qui
Figure imgf000093_0001
XH NMR (400 MHz, DMSO-d6) δ: 0.8 (d, 2H), 1.59-2.0 (m, 2H), 2.3-2.5 (s, 2H), 3.2-3.6 (m, 4H), 7.0-7.4 (m, 8H), 7.56-7.8 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 2H), 9.1 (m, IH) 10.4 (s, IH); HPLC Purity: 99.88%; Mass (M+l): 533.1.
(S)-N-(4-(4-(3,5-difluorobenzyl)-2-ethylpiperazine-l- carbonyl)phenyl)qui
Figure imgf000093_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.1-1.21 (d, 6H), 1.82-2.1 (m, 2H), 2.6 (m, IH), 2.8-3.2 (s, 2H), 3.8-4.0 (m, 3H), 7.0-7.2 (m, 7H), 7.6-7.8 (d, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.95%; Mass (M+l): 551.3.
(S)-N-(4-(2-methyl-4-(2,3,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qui
Figure imgf000093_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.21 (d, 3H), 1.23 (m, IH), 1.82-2.1 (m, 2H), 2.5-2.8 (s, 2H), 3.0-3.2 (m, 2H), 3.4-3.6 (m, 2H), 7.0-7.2 (m, 5H), 7.4-7.45 (m, IH), 7.6-7.8 (m, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.60%; Mass (M+l): 555.3.
(S)-N-(4-(4-(4-chloro-3-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qui
Figure imgf000094_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.96 (d, 3H), 1.23 (m, IH), 1.82-2.1 (m, 2H), 2.5-2.8 (s, 2H), 3.0-3.2 (m, 2H), 3.4-3.6 (m, 2H), 7.0-7.2 (m, 5H), 7.3 (d, IH), 7.4-7.45 (m, IH), 7.6-7.8 (m, 2H), 8.25 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.98%; Mass (M+l): 553.3.
N-(4-((2S)-2-methyl-4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl)quino :
Figure imgf000094_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.96 (d, 3H), 1.23 (m, IH), 1.5 (m, IH), 1.8-2.0 (m, 3H), 2.15-2.44 (s, 2H), 2.8-3.1 (m, 4H), 3.6-3.8 (m, 4H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.25-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 92.25%; Mass (M+l): 495.35.
(S)-N-(4-(4-(cyclopentylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 140):
Figure imgf000095_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.23 (m, 3H), 1.45-1.6 (m, 4H), 1.61-1.8 (m, 3H), 1.86-2.2 (m, 5H), 2.6-3.2 (s, 2H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.25-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.35%; Mass (M + Na): 515.15.
(S)-N-(4-(2-methyl-4-(2,3,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000095_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.15 (d, 3H), 1.23 (m, IH), 1.86-2.2 (m, 2H), 2.6-3.2 (s, 3H), 7.0-7.2 (m, 5H), 7.4-7.8 (m, 3H), 8.25-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.77%; Mass (M+l): 555.05.
(S)-N-(4-(4-(3,5-difluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu
Figure imgf000095_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.86-2.2 (m, 2H), 2.6-3.2 (m, IH), 3.0-3.2 (s, 2H), 3.3-4.0 (m, 4H), 7.0-7.2 (m, 6H), 7.6-7.8 (m, 2H), 8.25-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.80%; Mass (M+l): 537.30.
(S)-N-(4-(4-(2-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 143):
Figure imgf000096_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.23 (m, IH), 1.86-2.2 (m, 2H), 2.6-3.2 (s, 2H), 3.0-3.2 (m, 2H), 3.3-3.6 (m, 2H), 7.0-7.2 (m, 6H), 7.3-7.4 (m, 2H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 98.56%; Mass (M+l): 519.10.
(S)-N-(4-(4-(cyclopropylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 144):
Figure imgf000096_0002
XH NMR (400 MHz, DMSO-d6) δ: 0.12-0.2 (d, 2H), 0.2-0.24 (m, 2H), 0.8- 0.86 (m, IH), 1.1 (d, 3H), 1.23 (m, IH), 1.8-2.2 (m, 4H), 2.7-3.2 (s, 2H), 3.6-4.0 (m, 2H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.91%; Mass (M+l): 465.35.
(S)-N-(4-(2-methyl-4-(2,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000096_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.23 (m, IH), 1.8-2.2 (s, 2H), 2.7-3.2 (m, 4H), 3.4-3.6 (m, 2H), 7.0-7.2 (m, 4H), 7.4-7.6 (m, IH), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.12%; Mass (M+l): 465.35. (S)-N-(4-(4-(2,3-dimethoxybenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 146):
Figure imgf000097_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.23 (m, IH), 1.8-2.2 (s, 2H), 2.6-3.2 (m, 3H), 3.3-3.5 (m, 3H), 3.7 (s, 3H), 3.8 (s, 3H), 7.0-7.2 (m, 7H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 97.84%; Mass (M+l): 561.40.
N-(4-((2R)-2-methyl-4-((tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinol ):
Figure imgf000097_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.5 (m, IH), 1.76-2.2 (m, 5H), 2.3-2.4 (s, 2H), 2.8-3.2 (m, 4H), 3.6-4.0 (m, 4H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 97.78%; Mass (M+l): 495.40.
N-(4-(4-(2-methoxybenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinol ):
Figure imgf000097_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 2.5 (m, IH), 3.0-3.4 (m, 4H), 3.7 (s, 3H), 3.8-4.0 (s, 2H), 6.8-7.2 (m, 8H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.69%; Mass (M+l): 517.35
N-(4-(4-(2-methoxybenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu
Figure imgf000098_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.1 (d, 3H), 1.8-2.2 (s, 2H), 2.6-3.2 (m, 3H), 3.8-4.0 (m, 4H), 7.0-7.2 (m, 5H), 7.36-7.4 (m, IH), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.69%; Mass (M+l): 555.0.
(R)-N-(4-(2-methyl-4-(3,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000098_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.8-2.2 (m, 2H), 2.6-2.8 (s, 2H), 3.0-3.2 (m, IH), 3.6-3.86 (m, 4H), 7.0-7.3 (m, 6H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.97%; Mass (M+l): 554.95.
(R)-N-(4-(4-(2-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 150):
Figure imgf000099_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.8-2.2 (m, 2H), 2.6-2.85 (s, 2H), 3.0-3.2 (m, IH), 3.8-4.0 (m, 4H), 7.0-7.5 (m, 8H), 7.7-7.8 (m, 2H), 8.23 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.67%; Mass (M+l): 519.05.
(R)-N-(4-(4-(cyclohexylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 151):
Figure imgf000099_0002
1H NMR (400 MHz, CDC13) δ: 0.8-0.9 (d, 3H), 1.0-1.4 (m, 8H), 1.5-1.6 (m, 2H), 1.61-1.8 (m, 9H), 2.0-2.1 (4H), 2.6-2.85 (s, 2H), 3.0-3.2 (m, IH), 4.0-4.1 (m, IH), 7.0-7.4 (m, 4H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.85%; Mass (M+l): 507.40.
(S)-N-(4-(4-(4-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)qu :
Figure imgf000099_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.21-1.3(m, IH), 1.8-2.1 (m, 2H), 2.6-2.85 (s, 2H), 3.0-3.2 (m, IH), 3.4-3.5 (m, 3H), 7.0-7.4 (m, 8H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 97.31%; Mass (M+l): 519.35. (S)-N-(4-(4-(cyclohexylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quino ):
Figure imgf000100_0001
1H NMR (400 MHz, DMSO-d6) δ: 0.8-0.9 (m, 2H), 1.21-1.3(m, 6H), 1.4- 1.42 (m, IH), 1.6-1.8 (m, 6H), 1.96-2.12 (m, 3H), 2.5-2.6 (s, 2H), 3.6-3.8 (m, 2H), 7.0-7.2 (m, 4H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.20%; Mass (M+l): 507.15.
(S)-N-(4-(2-methyl-4-(2,3,4-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000100_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.21-1.3(d, 3H), 1.8-2.1 (m, 2H), 2.5-2.; (s, 2H), 3.0-3.4 (m, IH), 3.8-4.0 (m, 4H), 7.0-7.2 (m, 6H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.99%; Mass (M+l): 555.35.
(S)-N-(4-(2-methyl-4-(3,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000100_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.21-1.3(d, 3H), 1.8-2.1 (m, 3H), 2.6-2.; (s, 2H), 3.0-3.4 (m, IH), 3.4-3.5 (m, 2H), 3.8-4.0 (m, IH), 7.0-7.2 (m, 6H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.65%; Mass (M+l): 575.05.
N-(4-((2S)-2-methyl-4-((tetrahydro-2H-pyran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 156):
Figure imgf000101_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.21 (d, 3H), 1.23-1.3 (m, IH), 1.31-1.4 (m, 3H), 1.59-1.6(m, 2H), 1.8-2.1 (m, 4H), 2.1-2.2 (s, 2H), 2.6-2.8 (m, 2H), 3.0-3.4 (m, 3H), 3.8-4.0 (m, IH), 7.0-7.2 (m, 4H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 94.56%; Mass (M+l): 509.05.
N-(4-((2R)-2-methyl-4-((tetrahydro-2H-pyran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quino ):
Figure imgf000101_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.4 (m, 7H), 1.56-1.6 (m, 2H), 1.6- 1.65 (m, 2H), 2.0-2.4 (s, 2H), 2.6-3.4 (m, 5H), 3.8-3.9 (m, 3H), 7.0-7.4 (m, 4H), 7.6- 7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 97.32%; Mass (M+l): 509.15.
(S)-N-(4-(2-methyl-4-(2,4,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)qu :
Figure imgf000101_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.8-2.1 (m, 2H), 2.6-2.7 (s, 2H), 2.9-3.2 (m, IH), 3.6-4.0 (m, 4H), 7.0-7.2 (m, 6H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 96.52%; Mass (M+1): 544.7.
(S)-N-(3-chloro-4-(4-(4-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 184):
Figure imgf000102_0001
1H NMR (400 MHz, CDCI3) δ: 1.0 (d, 3H), 2.0 (m, IH), 2.8-3.2 (m, 4H), 3.5- 3.6 (m, 2H), 4.2 (m, IH), 7.0-7.4 (m, 6H), 7.6-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.81%; Mass (M+1): 553.2.
(S)-N-(3-chloro-4-(4-(4-chloro-3-fluorobenzyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 185):
Figure imgf000102_0002
1H NMR (400 MHz, CDC13) δ: 1.0 (d, 3H), 1.99 (m, IH), 1.8-2.2 (m, 2H), 2.6-3.6 (m, 4H), 4.2 (m, IH), 4.6 (s, IH), 7.0-7.6 (m, 6H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 99.85%; Mass (M+1): 587.1.
(S)-N-(3-chloro-4-(4-(cyclopentylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 186):
Figure imgf000102_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.8 (m, IH), 1.2-1.4 (m, 7H), 1.5-1.6 (m, 6H), 1.8-2.0 (m, 3H), 2.0-2.2 (m, 2H), 4.2 (m, IH), 4.6 (m, IH), 7.0-7.2 (m, 3H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 99.85%; Mass (M+l):527.6.
N-(3-chloro-4-((2S)-2-methyl-4-((tetrahydrofuran-3-yl)methyl)piperazine- l-carbonyl)phenyl)quinoline-8-sulfonamide (Compound 187):
Figure imgf000103_0001
1H NMR (400 MHz, DMSO-d6) δ: 0.8 (m, IH), 1.0-1.4 (m, 4H), 1.5-1.6 (m, IH), 1.8-2.0 (m, 2H), 2.0-2.4 (m, 3H), 2.8-3.0 (m, 2H), 3.3-3.4 (m, IH), 3.6-3.7 (m, 3H), 4.2 (m, IH), 4.6 (m, IH), 7.0-7.2 (m, 3H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 94.53%; Mass (M+1): 529.55.
(S)-N-(3-chloro-4-(2-methyl-4-(2,4,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 188):
Figure imgf000103_0002
1H NMR (400 MHz, DMSO-d6) δ: 0.8 (m, IH), 1.0-1.3 (m, 4H), 1.8-2.0 (m, 2H), 2.6-2.8 (m, IH), 3.4 (s, 2H), 4.0-4.1 (m, IH), 4.55-4.6 (m, IH), 7.0-7.2 (m, 5H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 99.17%; Mass (M+1): 589.55.
N-(4-((2R)-2-methyl-4-(l-(2,3,4-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 189):
Figure imgf000103_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.3 (d, 6H), 1.8-2.0 (m, 2H), 2.6-3.0 (m, 4H), 3.6-3.8 (m, 2H), 7.0-7.4 (m, 6H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 97.92%; Mass (M+l): 569.3.
N-(4-((2R)-2-methyl-4-(l-(2,3,6-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 190):
Figure imgf000104_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.3 (d, 3H), 1.4 (m, 2H), 1.8-2.0 (m, 2H), 2.6-2.6 (m, 2H), 3.0-3.4 (s, 2H), 4.0 (m, IH), 7.0-7.2 (m, 5H), 7.3-7.4 (m, IH), 7.61- 7.8 (m, 3H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 99.36%; Mass (M+l): 569.3.
N-(4-((2R)-4-(l-(2-chloro-4-fluorophenyl)ethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 191):
Figure imgf000104_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.3 (d, 6H), 1.8-2.0 (m, 3H), 2.2-2.4 (m, IH), 2.8-3.2 (m, 3H), 3.6-4.8 (m, IH), 7.0-7.6 (m, 6H), 7.61-7.8 (m, 3H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.6 (s, IH); HPLC Purity: 94.03%; Mass (M+l): 567.5.
N-(4-((2S)-2-methyl-4-(l-(2,3,6-trifluorophenyl)ethyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 192):
Figure imgf000104_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.3 (d, 6H), 1.4-1.6 (m, 3H), 1.8-2.2 (m, 2H), 2.8-3.2 (m, 4H), 3.6-4.8 (m, 2H), 7.0-7.5 (m, 6H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 98.39%; Mass (M+l): 569.55.
(S)-N-(4-(4-(cyclobutylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 207):
Figure imgf000105_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 3H), 1.5-1.6 (m, 2H), 1.8-1.85 (m, 3H), 2.0-2.1 (m, 3H), 2.2-2.4 (m, 4H), 2.6-2.99 (m, 4H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.5 (s, IH); HPLC Purity: 99.03%; Mass (M+l): 479.3.
N-(4-((2S)-2-methyl-4-((tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 225):
Figure imgf000105_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.6 (m, 6H), 1.8-2.4 (m, 6H), 2.6-3.0 (m, 3H), 3.4-3.8 (m, 4H), 7.0-7.2 (m, 4H), 7.5-7.8 (m, 2H), 8.0-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 99.37%; Mass (M+l): 495.10
N-(4-((2S)-2R-methyl-4-((tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 211):
Figure imgf000105_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.2-1.8 (m, 6H), 2.0-2.4 (m, 3H), 2.6-3.0 (m, 3H), 3.4-4.0 (m, 4H), 7.0-7.2 (m, 4H), 7.5-7.8 (m, 2H), 8.0-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 96.80%; Mass (M+l): 495.20.
N-(4-((R)-2-methyl-4-(((R)-tetrahydrofuran-2-yl)methyl)piperazine-l- carbonyl)phenyl)quinolin -8-sulfonamide (Compound 212).
1H NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.2-1.8 (m, 6H), 2.0-2.4 (m, 3H), 2.6-3.0 (m, 3H), 3.4-4.0 (m, 4H), 7.0-7.2 (m, 4H), 7.5-7.8 (m, 2H), 8.0-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 99.80%; Mass (M+l): 495.20.
(R)- and (S)-N-(4-(4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 444 and 445).
Figure imgf000106_0002
1H-NMR (400 MHz, DMSO-d6) δ: 1.5 (m, IH), 1.9 (m, IH), 2.2-2.4 (m, 7H), 3.3 (m, 5H), 3.56-3.8 (m, 3H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.3 (d, IH), 8.4 (d, IH), 8.5 (d, IH), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 99%; RT 31.15min and 36.57min; Mass (M+l): 481.20.
Example 8. Preparation of Compounds of Formula Ij:
COOEt
Figure imgf000107_0001
L XLIX
Figure imgf000107_0002
W, Y, Z = N or CH
STAB = Sodium tri-acetoxy borohydride |_ = -(CRcRc)m-
R1 = carbocyclyl, aryl, heterocyclyl, heteroaryl
Synthesis of Intermediate LI. To a solution of appropriate amine L (9.6 mmol) in a mixture (1:1) of DCM and pyridine, sulfonyl chloride XLIX (12.1 mmol) was added at room temperature under N2 atmosphere. The resulting mixture was allowed to stir for 16 h. After completion of reaction, the crude mixture was diluted with DCM, washed with water followed by IN HCl. The organic layer was then dried over Na2S04 and concentrated under reduced pressure to afford product LI in 78% yield.
Synthesis of Intermediate LIL To a solution of sulfonamide LI (9.5 mmol) in THF and water (1:1), LiOH (4.7 mmol) was added and the resulting mixture was allowed to stir at 80°C overnight. After completion of reaction, the crude mixture was washed with EtOAc. The aqueous layer was acidified with citric acid and filtered. Thus obtained solid was washed with Et20 and azeotroped by toluene, under reduced pressure to afford acid LII (75% yield) which was taken forward for the next step without further purification.
Synthesis of Intermediate LIII. To a solution of acid LII (6.09 mmol) in DMF, PyBoP (Benzotriazole- 1 -yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate) (4.75 gm, 9.14 mmol) was added at 0°C and allowed to stir for 5 minutes. Then Boc protected piperazine/substituted piperizine XXIX (1.13 gm, 6.09 mmol) was added to the reaction mixture at the same temperature under N2 atmosphere and stirred overnight at room temperature. After completion of reaction, mixture was diluted with water and extracted with EtOAc. The organic layer was washed with water, dried over Na2S04, and evaporated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH- DCM, 1:9) to afford product LIII in 56% yield.
Synthesis of Intermediate LIV. To a solution of MeOH HCl, Boc protected amine LIII (4.03 mmol) was added and the resulting mixture was stirred for 1 h. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product LIV (84% yield).
Synthesis of Compounds of Formula Ij. To a solution of amine LIV (0.25 mmoles) and appropriate aldehyde (0.27 mmol) in DCM, acetic acid (0.2 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 minutes. Then STAB (0.26 gm, 1.26 mmol) was added to reaction mixture and the resulting mixture was allowed to stir at 50°C for 2 nr. After completion of reaction, the crude mixture was diluted with DCM washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column
chromatography (silica gel, 60-120 mesh; MeOH-DCM, 2:8) to afford product in 22- 45% yield.
The following compounds were prepared according to the above methods using the appropriate amine L and the appropriate aldehyde.
N-(5-(4-(cyclopropylmethyl)piperazine-l-carbonyl)pyridin-2-yl)quinoline- 8-sulfonamide (XIV- 1) (
Figure imgf000108_0001
1H NMR (400 MHz, DMSO-d6) δ: 0.1-0.15 (m, 2H), 0.4-0.6 (m, 2H), 08- 0.85 (m, IH), 2.2-2.3 (m, 2H), 2.4-2.8 (m, 4H), 3.6-3.8 (m, 4H), 3.99-4.0 (m, 2H), 7.5-7.7 (m, 4H), 8.3-8.5 (m, 4H), 9.1 (m, IH); HPLC Purity: 99.67%; Mass (M+1): 452.5.
N-(5-(4-(4-fluorobenzyl)piperazine-l-carbonyl)pyridin-2-yl)quinoline-8- sulfonamide (Compound
Figure imgf000109_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.3 (s, 2H), 2.35-2.4 (m, 4H), 3.4-3.6 (m, 4H), 7.0-7.4 (m, 4H), 7.6-7.8 (m, 3H), 8.0 (m, IH), 8.3-8.5 (m, 3H), 8.9-9.0 (m, IH); HPLC Purity: 99.86%; Mass (M+1): 506.4.
N-(5-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)pyridin-2-yl)quinoline- 8-sulfonamide (Comp
Figure imgf000109_0002
1H NMR (400 MHz, CDC13) δ: 2.35-2.7 (m, 4H), 3.4-3.59 (m, 4H), 3.6-3.8 (s, 2H), 6.6-7.0 (m, 3H), 7.5-7.7 (m, 4H), 8.3-8.5 (m, 4H), 8.9-9.0 (m, IH); HPLC Purity: 93.78%; Mass (M+1): 524.5.
N-(6-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)pyridin-3-yl)quinoline- 8-sulfonamide (Comp
Figure imgf000109_0003
1H NMR (400 MHz, CDC13) δ: 2.35-2.7 (m, 4H), 3.4-3.59 (m, 4H), 3.6-3.8 (s, 2H), 6.6-7.0 (m, 4H), 7.4-7.8 (m, 3H), 8.0-8.4 (m, 4H), 8.9-9.0 (m, IH); HPLC Purity: 96.0%; Mass (M+l): 524.3.
N-(6-(4-(cyclopropylmethyl)piperazine-l-carbonyl)pyridin-3-yl)quinoline- 8-sulfonamide (Compoun
Figure imgf000110_0001
1H NMR (400 MHz, CDC13) δ: 0.9-1.0 (m, 2H), 1.2-1.4 (m, 4H), 1.6-1.8 (m, 3H), 3.4-3.59 (m, IH), 3.9-4.3 (m, 5H), 7.2-7.75 (m, 9H), 8.2-8.4 (m, IH); HPLC Purity: 99.35%; Mass (M+l):452.3.
N-(6-(4-(4-fluorobenzyl)piperazine-l-carbonyl)pyridin-3-yl)quinoline-8- sulfonamide (Compound 416):
Figure imgf000110_0002
1H NMR (400 MHz, CDC13) δ: 2.2-2.7 (m, 4H), 3.2-3.8 (m, 8H), 7.0-7.5 (m, 4H), 7.56-7.8 (m, 3H), 8.2-8.4 (m, 4H), 8.6-8.8 (m, IH), 9.1-9.2 (m, IH); HPLC Purity: 99.85%; Mass (M+l):506.3.
N-(5-(4-(cyclopropylmethyl)piperazine-l-carbonyl)pyrazin-2- yl)quinoline-8-sulfonamide (Compound 451)
Figure imgf000110_0003
1H NMR (400 MHz, DMSO-d6) δ: 0.2 (m, 2H), 0.6 (m, 2H), 0.8-1.0 (m, IH), 1.2 (s, 2H), 2.5-2.8 (m, 4H), 3.1-3.8 (m, 4H), 7.6-7.8 (m, 2H), 8.2 (m, IH), 8.2-8.6 (m, 4H), 9.0(m, IH); HPLC Purity: 94.0%; Mass (M+l): 453.25.
N-(4-(4-(3,5-difluorobenzoyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 345):
Figure imgf000111_0001
1H NMR (400 MHz, DMSO-d6) δ: 2.5-2.8 (m, 4H), 3.1-3.8 (m, 6H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 4H), 9.0(m, IH); HPLC Purity: 97.74%; Mass (M+l): 537.40.
N-(5-(4-(4-fluorobenzyl)piperazine-l-carbonyl)pyrazin-2-yl)quinoline-8- sulfonamide (Compo
Figure imgf000111_0002
XH NMR (400 MHz, CDC13) δ: 2.5-2.8 (m, 6H), 3.1-3.6 (m, 4H), 3.5-3.8 (s, 2H), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 3H), 8.2-8.6 (m, 4H), 8.8-8.85 (m, IH), 9.0(m, IH); HPLC Purity: 92.85%; Mass (M+l): 507.30.
Example 9. Preparation of Compounds of Formula Ik:
Figure imgf000111_0003
(Ik), wherein L is -(CRcRc)m-; and R is alkyl, carbocyclyl or aryl.
Scheme 9:
Figure imgf000112_0001
Synthesis of V -(4-nitrobenzoyl)-2,6-dimethylpiperazine (LVI). To a stirred solution of 2,6-di-methylpiperazine (LV, 5.0 g, 43.8 mmol) in dry THF (50 mL), maintained at room temperature under an argon atmosphere, was added a solution of 2.5 M n-BuLi in THF (38.54 mL, 96.36 mmol). After the mixture was stirred for 30 min at room temperature, trimethylsilyl chloride (5.5 mL, 43.8 mmol) was added and the reaction mixture stirred for 1 h before the addition of 4-nitrobenzoyl chloride (7.8 gm, 42.05 mmol). After 10 min, the reaction mixture was quenched with MeOH and the solvents were evaporated in vacuo. The residue was purified by silica gel column chromatography to provide product LVI (10.37 gm, 90% yield):
N -alkylation of V -(4-nitrobenzoyl)-2,6-dimethylpiperazine (LVII). To a solution of amine LVI (0.5 gm, 1.9 mmol) and appropriate aldehyde (2.28 mmol) in dichloroethane, acetic acid (0.2 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 minutes. Then sodium triacetoxyborohydride (1.2 gm, 5.7 mmol) was added to the reaction mixture and the resulting mixture was allowed to stir at room temperature over night. After completion of reaction, the crude mixture was concentrated, diluted with DCM washed with water, dried over Na2SC>4, concentrated under reduced pressure and purified by column chromatography (silica gel, 60-120 mesh) to afford product LVII in 50-60% yield.
I l l Reduction of N -alkyl-jV -(4-nitrobenzoyl)-2,6-dimethylpiperazine (LVIII). To a solution of nitro compound (LVII, 1.10 mmol) in 15 ml of ethanol and ethyl acetate (1:1), SnCl2 (0.418 gm, 2.2 mmol) was added and the mixture was stirred at 60 °C for overnight. The mixture was quenched by the addition of 10 ml of saturated solution of NaHCC>3 and extracted with ethyl acetate (2x25 mL). The combined organic layers were washed with brine solution, dried over anhydrous Na2S04 and concentrated under reduced pressure to afford amine product LVIII in 50-55% yield.
Synthesis of Compounds of Formula Ik. To a solution of amine (LVIII, 0.55 mmol) in a 5 mL mixture (1: 1) of DCM and pyridine, 8-quinoline sulfonyl chloride (0.14 gm, 0.61 mmol) was added at room temperature under N2 atmosphere. The resulting mixture was allowed to stir for overnight. After completion of reaction, the crude mixture was diluted with DCM, washed with water followed by IN HC1. The organic layer was then dried over Na2S04, concentrated under reduced pressure to afford product in 50-65% yields.
The following compounds were produced by the above-described method using the appropriate aldehyde.
N-(5-(4-(4-fluorobenzyl)piperazine-l-carbonyl)pyrazin-2-yl)quinoline-8- sulfonamide (Compound
Figure imgf000113_0001
XH NMR (400 MHz, DMSO-d6) δ: 0.1-0.15 (m, 2H), 0.3-0.4 (m, 2H), 0.8-0.9 (m, 1H), 1.1-1.4 (d, 6H), 1.99-2.3 (m, 4H), 2.4-3.0 (m, 2H), 3.8-4.2 (d, 2H), 7.0-7.2 (m, 4H), 7.61-7.8 (m, 2H), 8.0 (d, 1H), 8.3-8.6 (m, 2H), 9.1 (m, 1H), 10.4 (s, 1H); HPLC Purity: 99.46%; Mass (M+l): 479.50.
N-(4-(2,6-dimethyl-4-((tetrahydrofuran-3-yl)methyl)piperazine-l- carbonyl)phenyl) quinoline-8-sulfonamide (Compound 204):
Figure imgf000114_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.3 (d, 6H), 1.4-1.6 (m, 2H), 1.9-2.67 (m, 8H), 3.6-3.8 (m, 3H), 3.99-4.0 (m, 2H), 7.0-7.27 (m, 4H), 7.6-7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.92%; Mass (M+l): 509.5.
N-(4-(4-(cyclohexylmethyl)-2,6-dimethylpiperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 198):
Figure imgf000114_0002
1H NMR (400 MHz, DMSO-d6) δ: 0.9-1.0 (m, 2H), 1.1-1.4 (m, 11H), 1.45- 1.5 (m, 2H), 1.55-1.75 (m, 5H), 1.8-2.1 (m, 4H), 2.2-2.7 (m, 2H), 3.99-4.0 (m, 3H), 7.0-7.2 (m, 4H), 7.55-7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.53%; Mass (M+l): 521.60.
N-(4-(4-(4-fluorobenzyl)-2,6-dimethylpiperazine-l- carbonyl)phenyl)quino ):
Figure imgf000114_0003
1H NMR (400 MHz, CDC13) δ: 0.1-0.15 (m, 2H), 1.0-1.4 (d, 6H), 1.99-2.3 (m, 2H), 2.4-2.8 (m, 2H), 3.3-3.6 (s, 2H), 4.19-4.2 (m, IH), 6.9-7.2 (m, 6H), 7.23-7.4 (m, 2H), 7.55-7.7 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH); HPLC Purity: 99.02%; Mass (M+l): 533.55.
N-(4-(4-(3,5-difluorobenzyl)-2,6-dimethylpiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 194):
Figure imgf000115_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 6H), 1.3 (m, 2H), 2.0-2.4 (m, 2H), 2.4-2.6 (s, 2H), 3.2-3.6 (s, 2H), 7.0-7.5 (m, 7H), 7.61-7.8 (m, 2H), 8.0 (d, IH), 8.3-8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 98.34%; Mass (M+1): 551.55.
N-(4-(4-(4-chloro-3-fluorobenzyl)-2,6-dimethylpiperazine-l- carbonyl)phenyl)quino ):
Figure imgf000115_0002
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.4 (d, 6H), 2.0-2.2 (m, 2H), 2.4-2.6 (m, 2H), 3.5 (s, 2H), 3.9-4.0 (m, 2H), 7.23-7.4 (m, 6H), 7.55-7.8 (m, 3H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 95.27%; Mass (M+1): 567.50.
N-(4-(2,6-dimethyl-4-(2,3,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quin :
Figure imgf000115_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (d, 6H), 1.1-1.4 (m, 2H), 2.0-2.2 (m, 2H), 2.4-2.6 (m, 2H), 3.6 (s, 2H), 7.0-7.2 (m, 4H), 7.55-7.8 (m, 3H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.82%; Mass (M+1): 569.55.
N-(4-(2,6-dimethyl-4-(2,3,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 200):
Figure imgf000116_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (d, 6H), 1.25-1.4 (m, 4H), 2.0-2.2 (m, 2H), 2.4-2.6 (m, 2H), 3.6 (s, 2H), 7.0-7.2 (m, 4H), 7.4-7.5 (m, IH), 7.6-7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 99.92%; Mass (M+l):569.55.
N-(4-(2,6-dimethyl-4-(3,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quin :
Figure imgf000116_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (d, 6H), 1.3-1.4 (m, 2H), 2.0-2.2 (m, 2H), 2.4-2.6 (m, IH), 3.6 (s, 2H), 3.99-4.0 (m, IH), 7.0-7.4 (m, 6H), 7.6-7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.13%; Mass (M+l): 569.5.
N-(4-(2,6-dimethyl-4-(2,4,6-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quinol ):
Figure imgf000116_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (d, 6H), 1.3-1.4 (m, IH), 2.0-2.2 (m, 3H), 3.6 (s, 2H), 3.99-4.0 (m, 2H), 7.0-7.27 (m, 6H), 7.6-7.8 (m, 2H), 8.3-8.6 (m, 3H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.21%; Mass (M+l): 569.6.
N-(4-(2,6-dimethyl-4-(2,4,5-trifluorobenzyl)piperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 203):
Figure imgf000117_0001
XH NMR (400 MHz, DMSO-d6) δ: 1.0-1.4 (d, 6H), 2.0-2.2 (m, 2H), 2.5-2.7 (m, 2H), 3.6 (s, 2H), 3.99-4.0 (m, 2H), 7.0-7.27 (m, 6H), 7.4-7.8 (m, 4H), 8.3-8.6 (m, 3H), 9.1 (m, 1H), 10.4 (s, 1H); HPLC Purity: 98.34%; Mass (M+l): 569.5.
Example 10: Preparation of Compounds of Formula II:
Figure imgf000117_0002
wherein R1 is alkyl or aryl; and L is -(CR
Scheme 10:
Figure imgf000117_0003
R1 = Alkyl, Aryl
L = -(CRcRc)m-
Figure imgf000117_0004
Synthesis of 4-(4-nitrobenzoyl)piperazin-2-one (LXI). EDCI (0.394 gm, 2.05 mmol) and HOBT (0.276 gm, 2.05 mmol) were added to a stirred solution of the 4- nitrobenzoic acid (LX, 0.253 gm, 2.05 mmol) in anhydrous DMF. The temperature of the mixture was reduced to 0 °C, at which time DIPEA (1.14 ml, 6.15 mmol) was added under nitrogen atmosphere and the resultant solution (or suspension) was stirred at room temperature for 30 min. 2-Piperazinone (LIX, 2.05 mmol) was then added at 0 °C. The reaction mixture was then brought to room temperature and stirred for 6 h. After completion of the reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (3 x 25 ml). The organic layer was washed with water (3 x 10 ml), dried over anhydrous sodium sulfate, filtered and concentrated over the rotary evaporator to get the crude product. The crude product was purified by column chromatography (60-120 silica gel, ethyl acetate:hexane, 4:6) to afford pure product LXI (0.3 gm, 60%) as an off-white solid.
Synthesis of intermediate LXII. A solution of 4-(4-nitrobenzoyl)piperazin- 2-one (LXI) (0.1 gm, 0.4 mmol) in anhydrous DMF was cooled to 0 °C and added sodium hydride (0.02 gm, 0.48 mmol) under nitrogen atmosphere. The mixture was then stirred at room temperature for 30 min. Then the mixture was added appropriate alkyl bromide (R^-L-Br) (0.4 mmol) at 0 °C and stirred at room temperature for 24 h. After completion of reaction, the reaction mixture was quenched by the addition of water (10 mL), diluted with diethyl ether (100 mL), washed with water (2 x 25 mL), brine (25 mL), dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by column chromatography (Silica gel, 60:120; ethyl acetate:hexane, 3:7) to afford product LXII in 65-72% yield.
Synthesis of intermediate LXIIL To a solution of compound LXII (0.77 mmol) in 15 mL of methanol, iron powder (0.215 gm, 3.85 mmol) and concentrated hydrochloric acid (0.2 mL) were added. The mixture was then heated to 60 °C and stirred for 6 h. After completion of reaction, evaporated the solvent, the residue was added 10 mL of saturated sodium bicarbonate solution and extracted with ethyl acetate (3 x 25 mL). The combined organic layers were washed with water (10 mL), brine (lOmL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford amine LXIII in 60-70% yield.
Synthesis of compounds of Formula II. To a stirred solution of amine LXIII (0.26 mmol) in 5 mL of 1:1 mixture of pyridine and dichloromethane at 0 °C was added 8-quinoline sulfonylchloride (XLIX; 0.066 gm, 0.29 mmol). The mixture was allowed to stir for 6 h at room temperature. After completion of reaction, the mixture was concentrated under reduced pressure, residue dissolved in dichloromethane (50 mL), washed with dilute HC1 (10 mL), water (10 mL), brine (10 mL) and
concentrated. The crude product was purified by column chromatography (Silica gel, 60-120; 2% MeOH-DCM) to afford pure product as an off-white solid in 55-60% yields.
The following compounds were prepared according to the above methods using the appropriate alkyl bromide.
N-(4-(4-(cyclopropylmethyl)-3-oxopiperazine-l- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 215):
Figure imgf000119_0001
XH NMR (400 MHz, DMSO-d6) δ: 0.2-0.4 (m, 2H), 0.45-0.8 (m, 2H), 1.0 (m, IH), 2.6-2.8 (s, 2H), 3.2-3.4 (m, 2H), 3.5-4.0 (m, 4H), 7.2-7.4 (m, 4H), 7.4-7.6 (m, 2H), 8.0-8.4 (m, 3H), 8.79-8.8 (m, IH), 10.5 (s, IH); HPLC Purity: 94.48%; Mass (M+l): 465.2.
N-(4-(4-(3,5-difluorobenzyl)-3-oxopiperazine-l- carbonyl)phenyl)quino :
Figure imgf000119_0002
1H NMR (400 MHz, DMSO-d6) δ: 3.2-4.0 (m, 6H), 4.5 (s, 2H), 7.0-7.4 (m, 7H), 7.4-7.6 (m, 2H), 8.0-8.4 (m, 3H), 8.79-8.8 (m, IH), 10.5 (s, IH); HPLC Purity: 97.06%; Mass (M+l): 537.45.
Example 11: Preparation of a Compound of Formula Im:
Figure imgf000120_0001
Scheme 11
Figure imgf000120_0002
_ H2/Pd-C, , -
Boc2Q, PCM ^NBoc MeOH ( NBoc R -L-CHO, STAB rV^NBoc MeOH-HCI NH.HCI 90% 85% *" HN\^ ACOH-DCM* R^L^N- ^ 2 h, rt " RV
LXXI
R = aryl, carbocyclyl
LXXI L = -(CRcRc)m-
Figure imgf000120_0003
Formula Im
Synthesis of (2R,5S)-ethyl l-benzyl-5-(benzylcarbamoyl)pyrrolidine-2- carboxylate (LXV). To a stirred solution of Diethyl meso-2,5-dibromoadipate
(LXIV, 0.00069 mol, 250 mg) in toluene (5 mL) was added benzylamine (0.0021 mol, 0.234 mL) and the reaction mixture was heated at 85 °C for 16 h. After
completion of the reaction (checked by TLC), the reaction mixture was cooled and the formed solid was filtered. The filtrate was concentrated under reduced pressure to leave the product as pale yellow liquid. The residue was purified by column
chromatography (silica gel, 60-120 mesh; EA-Hexane, 2:8) to afford Diethyl
pyrrolidine-2,5-carboxylate in 72% yield.
To a stirred solution of Diethyl pyrrolidine-2,5-carboxylate (0.000327 mol, 100 mg) in xylene (5 mL) was added benzylamine (0.000327 mol, 0.035 mL) under nitrogen atmosphere and heated under reflux for 18 h. After completion of the reaction (checked by TLC), the reaction mixture was cooled and concentrated under reduced pressure to leave the product as yellow liquid. The residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 4:6) to afford the product LXV in 50% yield.
Synthesis of 3-benzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione (LXVI). Ethyl l-benzyl-5-(benzylcarbamoyl)pyrrolidine-2-carboxylate (LXV, 0.00122 mol, 450 mg) was heated under stirring at 210-220 °C for 3 h under atmospheric pressure and the formed ethyl alcohol was collected. After completion of the reaction (checked by TLC), the reaction mixture was cooled at room temperature and the residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 2:8) to afford the 3,8-dibenzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione in 40-45% yield.
To a stirred solution of 3,8-dibenzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione (0.00025 mol, 80 mg) in MeOH (2 mL) containing few drops of HC1 was
hydrogenated with 10% Pd-C (8 mg) for 4 h at room temperature. After completion of the reaction (checked by TLC), the reaction mixture was filtered through celite and the filtrate was concentrated under reduced pressure. The crude residue was purified by column chromatography (silica gel, 60-120 mesh; EA-Hexane, 6:4) to afford the 3- benzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione (LXVI) in 80% yield.
Synthesis of 3-benzyl-3,8-diazabicyclo[3.2.1]octane (LXVII). A solution of 3-benzyl-3,8-diazabicyclo[3.2.1]octane-2,4-dione LXVI (0.00108 mol, 250 mg) in dry ether (2 mL) was added to a stirred suspension of L1AIH4 (122 mg, 0.00315 mol) in dry ether (8 mL) at 0 °C under nitrogen atmosphere. The reaction bath was allowed to return at room temperature and stirring was continued for 30 h. After completion of the reaction (checked by TLC), the reaction mixture was quenched with chilled water and then stirred for lh. The reaction mixture was diluted with ether (20 mL) and the organic layer was washed with water, dried over Na2SC>4, concentrated under reduced pressure. The crude residue was purified by column chromatography (silica gel, 60- 120 mesh; EA-Hexane, 1:1) to afford the product LXVII in 52% yield.
Synthesis of tert-butyl 3-benzyl-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (LXVIII). To a stirred solution of compound LXVII (0.00108 mol, 220 mg) in DCM (10 mL) was added Boc20 (0.00108 mol, 237 mg) and the reaction mixture was stirred for 16 h at room temperature. The progress of the reaction was monitored by TLC. The reaction mixture was diluted with DCM (30 mL) and washed with water. The organic layer was dried over Na2S04 and concentrated under reduced pressure to give the crude product LXVII which was used for the next step without further purification.
Synthesis of tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (LXIX). To a stirred solution of compound LXVIII (0.00028 mol, 85 mg) in MeOH (5 mL) was hydrogenated with 10% Pd-C (15 mg) for 4 h at room temperature. After completion of the reaction (checked by TLC), the reaction mixture was filtered through celite and the filtrate was concentrated under reduced pressure. The crude residue was purified by column chromatography (silica gel, 60-120 mesh; MeOH- DCM, 1:9) to afford Compound LXIX in 85% yield.
Synthesis of intermediate LXX. To a solution of amine LXIX (0.00023 mol) and appropriate aldehyde (0.00023 mol) in DCM (5 mL), acetic acid (0.1 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 min. Then STAB (0.100 gm, 0.00047 mol) was added to reaction mixture and the resulting mixture was allowed to stir at room temperature for 16 h. After completion of reaction, the crude mixture was diluted with DCM washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EtOAc-Hexane, 2:8) to afford product LXX in 70-75% yield.
Synthesis of intermediate LXXL To a solution of MeOH- HC1 (5 mL), Boc protected amine LX (1.03 mmol) was added and the resulting mixture was stirred for 1 hr. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product LXXI as free base (94.30% yield).
Synthesis of compounds of Formula Im. To a stirred solution of acid VIII (0.00021 mol, 1 eq) in DMF (5 mL), EDCI (0.048 g, 0.00024 mol, 1.1 eq), HOBt (0.038 g, 0.00024 mol, 1.1 eq) and DIPEA (0.15 mL, 0.00078 mol, 2.5 eq) were added at 0°C and stirred for 15 minutes. A solution of amine LXXI (0.00021 mol, 1 eq) was then added at 0°C and then the resulting mixture was allowed to stir at room temperature for overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 mL). The combined organic layer was dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 70% ethyl acetate in hexane) to give compound in 49-55% yield.
The above-described method was used to prepare the following compounds using the appropriate aldehyde (R^-L-CHO) and the appropriate acid VIII.
(2R,5S)-ethyl l-benzyl-5-(benzylcarbamoyl)pyrrolidine-2-carboxylate (Compound 213):
Figure imgf000123_0001
1H NMR (400 MHz, CDC13) δ: 1.6-1.9 (m, 4H), 2.2-2.4 (m, 2H), 2.6-2.8 (m, 2H), 3.5-3.6 (m, 2H), 3.9 (s, IH), 4.6 (s, IH), 7.0 (d, IH), 7.2-7.3 (m, 6H), 7.5-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 3H), 9.1 (d, IH); HPLC Purity: 91.41%; Mass (M+1): 513.33.
N-(4-(3-(4-fluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-8- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 226):
Figure imgf000123_0002
1H NMR (400 MHz, CDC13) δ: 1.6-1.9 (m, 4H), 2.2-2.4 (m, 2H), 2.6-2.8 (m, 2H), 3.5-3.6 (m, 2H), 3.9 (s, IH), 4.7 (s, IH), 6.9-7.1 (m, 4H), 7.2-7.3 (m, 3H), 7.5- 7.6 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 3H), 9.1 (d, IH); HPLC Purity: 96.11%; Mass (M+1): 531.25. N-(4-(3-(3,5-difluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-8- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 227):
Figure imgf000124_0001
1H NMR (400 MHz, CDC13) δ: 1.6-1.9 (m, 4H), 2.2-2.4 (m, 2H), 2.6-2.8 (m, 2H), 3.5-3.6 (m, 2H), 3.9 (s, IH), 4.7 (s, IH), 6.6 (m, IH), 6.8 (d, 2H), 6.9-7.1 (m, 2H), 7.2-7.3 (m, IH), 7.5-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 3H), 9.1 (d, IH); HPLC Purity: 94.31%; Mass (M+l): 549.23.
N-(4-(3-(cyclopropylmethyl)-3,8-diazabicyclo[3.2.1]octane-8- carbonyl)phenyl)qui
Figure imgf000124_0002
1H NMR (400 MHz, CDC13) δ: 0.2-0.3 (m, 2H), 0.4-0.5 (m, 2H), 0.8-0.9 (m, IH), 1.6-1.9 (m, 4H), 2.1-2.4 (m, 4H), 2.6-2.8 (m, 2H), 3.9 (s, IH), 4.7 (s, IH), 7.0- 7.1 (m, 2H), 7.2-7.3 (m, IH), 7.6-7.7 (m, IH), 8.0 (d, IH), 8.2-8.6 (m, 3H), 9.1 (d, IH); HPLC Purity: 99.28%; Mass (M+l): 477.41.
Example 12. Preparation of Compounds of Formula In:
Figure imgf000124_0003
wherein R1 is aryl or carbocyclyl; and L is -(CRcRc)m-. Scheme 12
Figure imgf000125_0001
R = aryl, carbocyclyl
L = -(CR3R3)m-
Synthesis of tert-butyl 3-(4-(quinoline-8-sulfonamido)benzoyl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (LXXII). To a stirred solution of acid VIII (0.001179 mol, 1 eq) in DMF (5 mL), EDCI (0.248 g, 0.00129 mol, 1.1 eq), HOBt (0.198 g, 0.00129 mol, 1.1 eq) and DIPEA (0.30 g, 0.00235 mol, 2 eq) were added at 0°C and stirred for 15 minutes. A solution of amine LXIX from Example 11 (0.00117 mol, 1 eq) was then added at 0°C and then the resulting mixture was allowed to stir at room temperature for overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 mL). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 70% ethyl acetate in hexane) to give LXXII in 57% yield.
Synthesis of jV-(4-(3,8-diazabicyclo[3.2.1]octane-3- carbonyl)phenyl)quinoline-8-sulfonamide (LXXIII). To a solution of MeOH- HC1 (5 mL), Boc protected amine LXXII (1 mmol) was added and the resulting mixture was stirred for 2 hr. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product LXXIII as free base (92% yield). Synthesis of compounds of Formula In. To a solution of amine LXXIII (0.118 mmol) and appropriate aldehyde (0.118 mmol) in DCM (5 mL), acetic acid (0.1 mL) was added at room temperature and the resulting mixture was allowed to stir for 30 min. Then STAB (0.050 gm, 0.236 mol) was added to reaction mixture and the resulting mixture was allowed to stir at room temperature for 16 h. After completion of reaction, the crude mixture was diluted with DCM washed with water, dried over Na2S04 and concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, 60-120 mesh; EtOAc-Hexane, 2:1) to afford product in 25-45% yield.
The following compounds were made by the above-described method using the appropriate aldehyde (R^-L-CHO) and the appropriate acid VIII.
N-(4-(3-(4-fluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-8- carbonyl)phenyl)qu
Figure imgf000126_0001
1H NMR (400 MHz, CDC13) δ: 1.7 (br s, 1H), 1.9 (br s, 2H), 3.0 (br s, 2H), 3.2 (br s, 2H), 3.5 (s, 2H), 3.8 (d, 1H), 4.3 (d, 1H), 7.0-7.2 (m, 4H), 7.3-7.4 (m, 4H), 7.6-7.7 (m, 2H), 8.0 (d, 1H), 8.2-8.5 (m, 3H), 9.1 (d, 1H); HPLC Purity: 99.85%; Mass (M+l): 512.62.
N-(4-(8-(4-fluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-3- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 219):
Figure imgf000126_0002
1H NMR (400 MHz, CDC13) δ: 1.7 (br s, IH), 1.9 (br s, 2H), 3.0 (br s, 2H), 3.2 (br s, 2H), 3.5 (s, 2H), 3.8 (d, IH), 4.3 (d, IH), 6.9-7.2 (m, 5H), 7.2-7.3 (m, 2H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 3H), 9.1 (d, IH); HPLC Purity: 99.07%; Mass (M+l): 530.62.
N-(4-(8-(3,5-difluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-3- carbonyl)phenyl)quinoline-8-sulfonamide (Compound 218):
Figure imgf000127_0001
1H NMR (400 MHz, CDC13) δ: 1.7 (br s, IH), 1.9 (br s, 2H), 3.0 (br s, 2H), 3.2 (br s, 2H), 3.5 (s, 2H), 3.8 (d, IH), 4.3 (d, IH), 6.7 (t, IH), 6.8-6.9 (m, 2H), 7.0- 7.2 (m, 3H), 7.5-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.5 (m, 3H), 9.1 (d, IH); HPLC Purity: 94.61%; Mass (M+l): 548.60.
N-(4-(8-(cyclopropylmethyl)-3,8-diazabicyclo[3.2.1]octane-3- carbonyl)phenyl)qui
Figure imgf000127_0002
XH NMR (400 MHz, CDC13) δ: 0.2-0.3 (m, 2H), 0.4-0.5 (m, 2H), 0.8-0.9 (m, IH), 1.6-1.9 (m, 4H), 2.1-2.3 (m, 2H), 3.0 (d, IH), 3.2-3.5 (m, 4H), 4.4 (d, IH), 7.0- 7.2 (m, 3H), 7.5-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.6 (m, 3H), 9.1 (d, IH); HPLC Purity: 99.37%; Mass (M+l): 477.59. Example 13. Preparation of Compounds of Formula Io:
Figure imgf000128_0001
wherein R1 is aryl, carbocyclyl, heterocyclyl or heteroaryl; R3 is OCF3 or OCH3; R4 is alkyl; L is -C(O)- or -(CRcRc)-C(0)-; n is 0 or 1; and p is 0 or 1.
Scheme 13:
Figure imgf000128_0002
Formula Io
Synthesis of intermediate LXXV. To a stirred solution of substituted amine LXXIV (30.3 mmol) under nitrogen atmosphere was added pyridine (50 ml) at 0°C and stirred for 10 min. Quinoline-8-sulfonyl chloride VI (8.94 gm, 39.4 mmol) was then added to the reaction mixture at the same temperature. The resulting mixture was stirred for 16 h at room temperature. After completion of the reaction, the solvent was removed under reduced pressure. The traces of pyridine were removed by co-distillation with toluene. Diethylether was added to the resulting residue, and the solid product was filtered out and air-dried. The resulting crude product (74 ) was taken to the next step without further purification.
Synthesis of intermediate LXXVI. To a stirred solution of acid LXXV
(0.000315 moles) in DMF (5 ml), were added EDCI (0.066 g, 0.000346 moles), HOBt (0.047 g, 0.000346 moles) and DIPEA (0.13 ml, 0.00078 moles) at 0°C and stirred for 15 minutes. A solution of amine I (0.000315 moles) was then added at 0°C and then the resulting mixture was allowed to stir at room temperature overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 ml). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 70% ethyl acetate in hexane) to give
LXXVI in 65-70% yield.
Synthesis of intermediate LXXVII. To a solution of MeOH HCl (10 ml), Boc protected amine LXXVI (4.03 mmol) was added and the resulting mixture was stirred for 2 nr. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product LXXVII in 92% yield.
General procedure for Syntheses of Compounds of Formula Io. To a stirred solution of aryl/heteroaryl acid (0.000315 moles) in DMF (5 ml), were added EDCI (0.066 g, 0.000346 moles), HOBt (0.047 g, 0.000346 moles) and DIPEA (0.13 ml, 0.00078 moles) at 0°C and stirred for 15 minutes. A solution of amine LXXVII (0.000315 moles) was then added at 0°C and then the resulting mixture was allowed to stir at room temperature overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 ml). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, MeOH-DCM, 2:8) to give a compound of Formula Io in 35-50% yield.
The following compounds of Formula Io were made by the above-described method using the appropriate acid (R1-C(0)OH) and the appropriate Boc-protected amine I.
N-(4-(4-(l,2,3-thiadiazole-5-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide( VI-1) Compound 313:
Figure imgf000129_0001
1H NMR (400 MHz, CDC13) δ: 3.2-3.5 (m, 8H), 7.2 (m, 4H), 7.6 (m, 2H), 8.3 (m, 2H), 8.8 (m, 2H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 99.55%; Mass (M+l): 509.2.
N-(4-(4-(3-fluoroisonicotinoyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compoun
Figure imgf000130_0001
XH NMR (400 MHz, CDC13) δ: 3.2-3.8 (m, 8H), 7.0 (m, 4H), 7.3 (m, IH), 7.6 (m, 2H), 8.1 (m, IH), 8.3 (m, 2H), 8.6 (m, 3H), 9.1 (m, IH); HPLC Purity: 98.06%; Mass (M+l): 520.30.
N-(4-(4-(3,5-difluorobenzyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide-(D) (Compound 342):
Figure imgf000130_0002
XH NMR (400 MHz, CDC13) δ: 3.0-3.8 (m, 8H), 6.9-7.3 (m, 6H), 7.6 (m, 2H), 7.7-7.9 (m, 2H), 8.0 (m, IH), 8.3 (m, IH), 8.6 (m, IH), 9.0(m, IH); HPLC Purity: 99.30%; Mass (M+l): 525.20.
N-(4-(4-(5-methylpyrazine-2-carbonyl)piperazine-l- carbonyl)phenyl)qu
Figure imgf000130_0003
1H NMR (400 MHz, CDC13) δ: 2.6 (s, 3H), 3.2-3.9 (m, 8H), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.0 (m, IH), 8.3 (m, 3H), 8.6 (s, IH), 8.9 (m, IH), 9.0(m, IH); HPLC Purity: 99.74%; Mass (M+1): 517.2.
N-(4-(4-(oxazole-4-carbonyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compou
Figure imgf000131_0001
1H NMR (400 MHz, CDC13) δ: 3.2-3.9 (m, 8H), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.3-8.5 (m, 4H), 9.0(m, IH), 10.5 (s, IH); HPLC Purity: 95.63%; Mass (M+1): 492.15.
N-(4-(4-(thiazole-5-carbonyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compou
Figure imgf000131_0002
1H NMR (400 MHz, CDC13) δ: 3.2-3.9 (m, 8H), 7.0-7.2 (m, 4H), 7.6 (m 8.0 (m, 2H), 8.3 (m, 2H), 8.6 (s, IH), 9.0(m, IH), 10.5 (s, IH); HPLC Purity:
97.14%; Mass (M+1): 508.2.
N-(4-(4-(lH-imidazole-4-carbonyl)piperazine-l- carbonyl)phenyl)quin :
Figure imgf000131_0003
1H NMR (400 MHz, CDC13) δ: 3.2-3.9 (m, 8H), 7.0-7.2 (m, 4H), 7.6 (m, 4H), 8.0 (m, IH), 8.3 (m, 2H), 8.6 (s, IH), 9.0(m, IH); HPLC Purity: 99.40%; Mass (M+l): 491.2.
N-(4-(4-(lH-imidazole-2-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 354):
Figure imgf000132_0001
1H NMR (400 MHz, CDC13) δ: 3.2-3.7 (m, 6H), 4.3-4.8 (m, 2H), 7.0-7.1 (m, 5H), 7.6 (m, 2H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 8.6 (m, IH), 9.0 (m, 2H) 10.3 (s, IH); HPLC Purity: 99.22%; Mass (M+l): 491.2.
N-(4-(4-(isoxazole-5-carbonyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound
Figure imgf000132_0002
1H NMR (400 MHz, CDC13) δ: 3.2-3.8 (m, 8H), 6.8-7.3 (m, 5H), 7.6 (m, 2H), 8.0 (m, IH), 8.3-8.4 (m, 3H), 9.0 (m, IH), 10.4 (m, IH); HPLC Purity: 99.30%; Mass (M+l): 492.2.
N-(4-(4-(lH-pyrazole-3-carbonyl)piperazine-l-carbonyl)phenyl)quinoline- 8-sulfonamide (Compo
Figure imgf000132_0003
1H NMR (400 MHz, CDC13) δ: 3.2-3.9 (m, 8H), 7.0-7.2 (m, 4H), 7.6 (m, 3H), 8.0 (m, IH), 8.3 (m, 2H), 8.6 (s, IH), 9.0(m, IH); HPLC Purity: 99.97%; Mass (M+l): 491.2.
N-(4-(4-(thiazole-2-carbonyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound
Figure imgf000133_0001
XH NMR (400 MHz, CDCI3) δ: 3.4-3.8 (m, 8H), 7.1-7.2 (m, 4H), 7.6 (m, 2H), 8.1-8.4 (m, 4H), 8.6 (m, IH), 8.9 (m, IH), 9.0 (m, IH); HPLC Purity: 97.89%; Mass (M+l): 508.30.
N-(4-(4-(tetrahydro-2H-pyran-4-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 417):
1H NMR (400 MHz, DMSOd6) δ: 1.2-1.6 (m, 8H), 2.8-3.0 (m, 2H), 3.4-3.7 (m, 4H), 3.75-3.8 (m, 2H), 7.0-7.2 (m, 4H), 7.56-7.8 (m, 2H), 8.2-8.4 (m, 3H), 9.0-9.2 (m, IH), 10.45 (s, IH); HPLC Purity: 96.68%; Mass (M + Na): 531.2.
N-(4-(4-(tetrahydrofuran-3-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 418):
Figure imgf000133_0003
1H NMR (400 MHz, CDC13) δ: 1.95-2.2 (m, 2H), 3.2-3.6 (m, 8H), 3.65-3.8 (m, 5H), 7.0-7.5 (m, 4H), 7.56-7.8 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.5 (m, IH); HPLC Purity: 99.65%; Mass (M+1): 495.2.
N-(4-(4-(cyclobutylmethyl)piperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound
Figure imgf000134_0001
XH NMR (400 MHz, CDC13) δ: 1.6-2.1 (m, 6H), 2.2-2.8 (m, 7H), 3.2-3.8 (m, 4H), 7.0-7.3 (m, 4H), 7.5-7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH); HPLC Purity: 95.52%; Mass (M+1): 465.2.
N-(4-(4-(tetrahydro-2H-pyran-2-carbonyl)piperazine-l- carbonyl)phenyl)quinol :
Figure imgf000134_0002
1H NMR (400 MHz, CDC13) δ: 1.2-1.6 (m, 5H), 1.6-1.8 (m, IH), 3.0-3.2 (m, 3H), 3.4-3.6 (m, 6H), 3.8-4.2 (m, 2H), 7.0-7.4 (m, 4H), 7.6-7.8 (m, 2H), 8.0-8.6 (m, 3H), 9.1-9.2 (m, IH), 10.5 (s, IH); HPLC Purity: 97.96%; Mass (M+1): 509.2.
N-(4-(4-(tetrahydrofuran-2-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 443):
Figure imgf000134_0003
1H NMR (400 MHz, DMSOd6) δ: 1.2-1.4 (m, 3H), 1.6-2.1 (m, 4H), 2.9-3.3 (m, 6H), 3.4-3.6 (m, 2H), 7.0-7.25 (m, 4H), 7.6-7.8 (m, 2H), 8.2-8.6 (m, 3H), 9.1-9.2 (m, IH) 10.5 (bs, IH); HPLC Purity: 97.44%; Mass (M+l): 484.25.
(R)-N-(4-(4-(cyclobutylmethyl)-2-methylpiperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 206):
Figure imgf000135_0001
XH NMR (400 MHz, CDC13) δ: 1.2 (d, 3H), 1.6-2.1 (m, 8H), 2.2-2.8 (m, 5H), 4.0-4.1 (m, 3H), 7.0-7.2 (m, 4H), 7.5-7.7 (m, 2H), 8.2-8.4 (m, 3H), 9.1-9.2 (m, IH), 10.4 (s, IH); HPLC Purity: 96.13%; Mass (M+l): 479.15.
N-(4-(4-(2,3-difluorobenzoyl)piperazine-l-carbonyl)-2- methoxyphenyl)quinoli -8-sulfonamide (Compound 318):
Figure imgf000135_0002
1H NMR (400 MHz, CDC13) δ: 3.3 (s, 3H), 3.2-3.8 (m, 8H), 6.8 (m, 2H), 7.2 (m, 2H), 7.6 (m, 3H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 96.96%; Mass (M+l): 567.30.
N-(4-(4-(3,4-difluorobenzoyl)piperazine-l-carbonyl)-2- methoxyphenyl)quinoli -8-sulfonamide (Compound 319):
Figure imgf000135_0003
1H NMR (400 MHz, CDC13) δ: 3.4 (s, 3H), 3.5-3.8 (m, 8H), 6.8 (m, 2H), 7.2 (m, 2H), 7.6 (m, 3H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 8.8 (m, IH), 9.1 (m, IH); HPLC Purity: 95.87% ; Mass (M+l): 567.30.
N-(4-(4-(2-fluoro-3-methoxybenzoyl)piperazine-l-carbonyl)-2- methoxyphenyl)quinoli -8-sulfonamide (Compound 320):
Figure imgf000136_0001
XH NMR (400 MHz, CDC13) δ: 3.2 (s, 3H), 3.4 (s, 3H), 3.6-3.8 (m, 8H), 6.8- 7.0 (m, 5H), 7.6 (m, 3H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 8.8 (m, IH), 9.1 (m, IH); HPLC Purity: 95.65% ; Mass (M+l): 579.40.
N-(4-(4-(l,2,3-thiadiazole-4-carbonyl)piperazine-l-carbonyl)-2- methoxyphenyl)quinolin -8-sulfonamide (Compound 321):
Figure imgf000136_0002
1H NMR (400 MHz, CDC13) δ: 3.4 (s, 3H), 3.6-3.8 (m, 8H), 6.8 (m, 2H), 7.6 (m, 3H), 8.0 (m, IH), 8.2 (m, IH), 8.4 (m, IH), 8.9 (s, IH), 9.1 (m, IH), 9.2 (m, IH) ; HPLC Purity: 98.30% ; Mass (M+l): 539.25.
N-(2-methoxy-4-(4-(thiazole-4-carbonyl)piperazine-l- carbonyl)phenyl)quinoli -8-sulfonamide (Compound 322):
Figure imgf000136_0003
1H NMR (400 MHz, CDC13) δ: 3.4 (s, 3H), 3.6-3.8 (m, 8H), 6.8 (m, 2H), 7.6 (m, 4H), 8.0 (m, 2H), 8.2 (m, IH), 8.4 (m, IH), 8.7 (m, IH), 8.9 (s, IH), 9.1 (m, IH); HPLC Purity: 96.49%; Mass (M+1): 538.10.
N-(4-(4-nicotinoylpiperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 323):
Figure imgf000137_0001
1H NMR (400 MHz, CDC13) δ: 3.4-3.8 (m, 8H), 7.2 (m, 2H), 7.4 (m, IH), 7.6 (m, 2H), 7.8 (m, IH), 8.0 (m, IH), 8.1 (m, IH), 8.2 (m, IH), 8.4 (m, 2H), 8.6 (m, IH), 9.1 (m, IH); HPLC Purity: 99.38% ; Mass (M+1): 586.27.
N-(4-(4-(thiazole-4-carbonyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 324):
Figure imgf000137_0002
1H NMR (400 MHz, CDC13) δ: 3.4-3.8 (m, 8H), 7.2 (m, 2H), 7.5 (m, 2H), 8.0 (m, 3H), 8.4 (m, 2H), 8.8 (m, IH), 9.1 (m, IH); HPLC Purity: 99.30%; Mass (M+1): 592.15.
N-(4-(4-(5-methylpyrazine-2-carbonyl)piperazine-l-carbonyl)-2- trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 325):
Figure imgf000137_0003
1H NMR (400 MHz, CDC13) δ: 2.6 (s, 3H), 3.5-3.8 (m, 3H), 7.2 (m, 2H), 7.6 (m, 2H), 8.0 (m, 2H), 8.4 (m, 3H), 8.9 (m, IH), 9.1 (m, IH); HPLC Purity: 97.67%; Mass (M+1): 601.30.
N-(4-(4-(3,5-difluorobenzoyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 326):
Figure imgf000138_0001
1H NMR (400 MHz, CDC13) δ: 3.3-3.8 (m, 8H), 6.9 (m, 3H), 7.2 (m, 2H), 7.6 (m, 2H), 8.0 (m, 2H), 8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.48%; Mass (M+1): 621.25
N-(4-(4-(3,5-dimethylbenzoyl)piperazine-l-carbonyl)-2- (trifluoromethoxy)phenyl)quinoline-8-sulfonamide (Compound 327):
Figure imgf000138_0002
XH NMR (400 MHz, CDC13) δ: 3.5-4.2 (m, 8H), 7.2 (m, 3H), 7.6 (m, 2H), 7.9 (m, IH), 8.1 (m, IH), 8.3 (m, IH), 8.4 (m, IH), 9.1 (m, 2H); HPLC Purity: 96.80%; Mass (M+1): 576.25.
Example 14. Prep
Figure imgf000138_0003
wherein R1 is alkyl, cycloalkyl, aryl, or heteroaryl; R3 is OCH3, or OCF3; R4 is alkyl; X and Y are independently selected from CH and N; p is 0 or 1; and n is 0 or 1.
Scheme 14
Figure imgf000139_0001
1) R1-OH, LXXXII
Figure imgf000139_0002
LXXXI Formula Ip
Synthesis of intermediate LXXIX. To a stirred solution of appropriately substituted amine LXXVIII (30.3 mmol) under nitrogen atmosphere was added pyridine (50 ml) at 0°C and stirred for 10 min. Quinoline-8-sulfonyl chloride VI (8.94 gm, 39.4 mmol) was then added to the reaction mixture at the same temperature. The resulting mixture was stirred for 16 h at room temperature. After completion of the reaction, the solvent was removed under reduced pressure. The traces of pyridine were removed by co-distillation with toluene. Diethyl ether was added to the resulting residue, and the solid product was filtered out and air-dried. The resulting crude product LXXIX (74 ) was taken to the next step without further purification.
Synthesis of intermediate LXXX. To a stirred solution of acid LXXIX
(0.000315 moles) in DMF (5 ml), were added EDCI (0.066 g, 0.000346 moles), HOBt (0.047 g, 0.000346 moles) and DIPEA (0.13 ml, 0.00078 moles) at 0°C and stirred for 15 minutes. A solution of amine I (0.000315 moles) was then added at 0°C and the resulting mixture was allowed to stir at room temperature overnight. After completion of the reaction, water (20 mL) was added and extracted with ethyl acetate (2x30 ml). The combined organic layer was dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, 70% ethyl acetate in hexane) to give LXXX in 45-55% yield.
Synthesis of intermediate LXXXI. To a solution of MeOH HCl (12 ml), Boc protected amine LXXX (4.03 mmol) was added and the resulting mixture was stirred for 2 h. After completion of reaction, solvent was removed under reduced pressure, washed with water followed by addition of NaHCC>3 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford product LXXXI in 94% yield.
General procedure for Syntheses of Compounds of Formula Ip. To a stirred solution of Triphosgene (1.7 g, 57 mmol) in dry THF (15 ml) was added alcohol LXXXII (39 mmol) at 0°C under nitrogen atmosphere and reaction mixture was stirred further for 15 minutes at room temperature. DIPEA (2.5 ml, 0.014 moles) was added slowly to the reaction mixture and stirred for further 30 minutes. The reaction mixture was filtered and filtrate was concentrated under reduced pressure to leave the crude chloroformate which was used for the next step.
To a stirred solution of amine IX (24.3 mmol) in dry DCM (10 ml) was added DIPEA (0.1 ml, 0.007 moles) at 0°C under nitrogen atmosphere. The crude chlorofomate (29.2 mmol) was added to the reaction mixture and stirred further for 30 minutes at room temperature. After completion of the reaction, water (10 mL) was added and extracted with DCM (2x30 ml). The combined organic layer was dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by column chromatography (silica gel, 60-120 mess, MeOH-DCM, 1: 9) to give a compound of Formula Ip in 50-60% yield.
The following compounds of Formula Ip were made by the above-described method using the appropriate acid LXXVIII, the appropriate alcohol LXXXII and the appropriate Boc-protected amine I.
Pyridin-2-yl 4-(3-methoxy-4-(quinoline-8-sulfonamido)benzoyl)piperazine-l- carboxylate (XI-3) (Compound 315):
Figure imgf000141_0001
XH NMR (400 MHz, CDC13) δ: 3.4-3.7 (m, 8H), 6.9 (m, 2H), 7.2 (m, 2H), 7.6 (m, 3H), 7.9 (m, IH), 8.1 (m, IH), 8.3 (m, IH), 8.5 (m, IH), 9.1 (m, IH); HPLC Purity: 97.17% ; Mass (M+l): 548.20.
(S)-Tetrahydrofuran-3-yl4-(4-(quinoline-8-sulfonamido)-3- (trifluoromethoxy)benzoyl)piperazine-l-carboxylate (Compound 343):
Figure imgf000141_0002
1H NMR (400 MHz, DMSO-d6) δ: 2.2 (m, 2H), 3.2-3.7 (m, 4H), 3.9 (m, 2H), 5.1 (m, IH), 7.2 (m, 2H), 7.6 (m, 2H), 7.8 (m, IH), 8.1 (m, IH), 8.4 (m, 2H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 98.45% ;Mass (M+l): 595.3.
2-cyclopentylethyl4-(4-(quinoline-8-sulfonamido)-3- (trifluoromethoxy)benzoyl)piperazine-l -carboxylate (Compound 312):
Figure imgf000141_0003
XH NMR (400 MHz, CDC13) δ: 1.2 (m, 6H), 1.7 (m, 6H), 3.2-3.5 (m, 8H), 4.6 (m, IH), 7.2 (m, 2H), 7.6 (m, 2H), 8.1 (m, 2H), 8.4 (m, 2H), 9.1 (m, IH), 10.0 (bs, IH); HPLC Purity: 99.18% ;Mass (M+l): 621.4.
tetrahydro-2H-pyran-4-yl4-(4-(quinoline-8-sulfonamido)-3- (trifluoromethoxy)benzoyl)piperazine-l -carboxylate (Compound 314):
Figure imgf000142_0001
XH NMR (400 MHz, CDC13) δ: 1.6 (m, 3H), 2.1 (m, IH), 3.3-3.6 (m, 10H), 3.9 (m, 2H), 4.8 (m, IH), 7.2 (m, 2H), 7.6 (m, 2H), 7.9 (m, IH), 8.1 (m, IH), 8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.45% ; Mass (M+1): 609.4.
(tetrahydrofuran-2-yl)methyl4-(4-(quinoline-8-sulfonamido)-3- (trifluoromethoxy)benzoyl) piperazine-l-carboxylate (Compound 316):
Figure imgf000142_0002
XH NMR (400 MHz, CDC13) δ: 1.6 (m, IH), 1.9-2.0 (m, 3H), 3.4-3.7 (m, 8H), 3.8 (m, 2H), 4.2 (m, 3H), 6.9 (m, 2H), 7.2 (m, 2H), 7.6 (m, 3H), 7.9 (m, IH), 8.1 (m, IH), 8.3 (m, IH), 8.5 (m, IH), 9.1 (m, IH); HPLC Purity: 96% ; Mass (M+1):
609.30.
(R)-tetrahydrofuran-3-yl4-(4-(quinoline-8-sulfonamido)-3- (trifluoromethoxy)benzoyl)piperazine-l -carboxylate (Compound 311):
Figure imgf000142_0003
1H NMR (400 MHz, DMSOd6) δ: 2.2 (m, 2H), 3.2-3.7 (m, 8H), 4.0 (m, 4H), 5.2 (m, IH), 7.2 (m, 2), 7.8 (m, 2H), 8.0 (m, 2H), 8.4 (m, 2H), 9.0(m, IH); HPLC Purity: 99.63%; Mass (M+1): 595.35. Pyridin-2-yl4-(4-(quinoline-8-sulfonamido)-3-(trifluoromethoxy) benzoyl)piperazine-l-carboxylate (Compound 344):
XH NMR (400 MHz, CDC13) δ: 3.0-3.8 (m, 8H), 7.0 (m, 6H), 7.6 (m, 2H), 7.7-7.9 (m, 2H), 8.0 (m, IH), 8.4 (m, 3H), 9.0(m, IH); HPLC Purity: 94.84%; Mass (M+l): 601.0.
(S)-Ethyl 3-methyl-4-(4-(quinoline-8-sulfonamido)benzoyl)piperazine-l- carboxylate (Compound
Figure imgf000143_0002
1H NMR (400 MHz, CDC13) δ: 1.2 (t, 3H), 1.6 (q, 2H), 3.0-3.4 (m, 3H), 3.8- 4.2 (m, 4H), 5.0 (m, IH), 7.0-7.3 (m, 4H), 7.6 (m, 2H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 8.6 (s, IH), 9.0 (m, IH); HPLC Purity: 99.05%; Mass (M+l): 483.2.
(S)-isopropyl 3-methyl-4-(4-(quinoline-8-sulfonamido)benzoyl)piperazine- 1-carboxylate (Compoun
Figure imgf000143_0003
XH NMR (400 MHz, CDC13) δ: 1.2 (d, 6H), 1.4 (m, IH), 2.8-3.2 (m, 2H), 3.8- 4.2 (m, 4H), 5.0 (m, IH), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 8.6 (s, IH), 9.0 (m, IH); HPLC Purity: 99.77%; Mass (M+l): 497.3.
(R)-Ethyl 3-methyl-4-(4-(quinoline-8-sulfonamido)benzoyl)piperazine-l- carboxylate (Compound 109):
Figure imgf000144_0001
XH NMR (400 MHz, CDC13) δ: 1.2 (t, 2H), 1.4 (q, 2H), 2.2 (d, 3H), 3.4-3.8 (m, 7H), 7.0-7.2 (m, 4H), 7.6 (m, 2H), 8.1 (m, IH), 8.2-8.4 (m, 2H), 8.6 (m, IH), 9.0 (m, IH); HPLC Purity: 99.23%; Mass (M+l): 483.20.
(R)-Isopropyl 3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 110):
Figure imgf000144_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.2 (d, 9H), 1.25 (m, IH), 2.8-3.2 (m, 4H), 3.8-4.2 (m, 2H), 4.9 (m, IH), 7.0-7.2 (m, 4H), 7.55-7.6 (m, 2H), 8.0 (d, IH), 8.38 (d, 2H), 8.5(s, IH), 9.0 (m, IH); HPLC Purity: 99.23%; Mass (M+l): 497.40.
(S)-Cyclopropylmethyl3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 111):
Figure imgf000144_0003
1H NMR (400 MHz, CDC13) δ: 0.1 (m, IH), 0.2 (m, 2H), 0.5 (m, 2H), 1.2 (s, 3H), 2.6-3.2 (m, 4H), 2.8-3.0 (m, 3H), 3.8-4.2 (m, 6H), 7.0-7.2 (m, 4H), 7.55-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.4 (d, 2H), 8.5(s, IH), 9.0 (m, IH); HPLC Purity: 99.02%; Mass (M+l): 509.3.
(R)-Cyclopropylmethyl 3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 112):
Figure imgf000145_0001
1H NMR (400 MHz, CDC13) δ: 0.3 (m, 2H), 0.5 (m, 2H), 1.2 (d, 3H), 1.3 (m, IH), 2.8-3.2 (m, 4H), 3.8-4.2 (m, 5H), 7.0-7.2 (m, 4H), 7.55-7.6 (m, IH), 8.0 (d, IH), 8.2-8.4 (d, 2H), 8.57(s, IH), 9.0 (m, IH); HPLC Purity: 92.31%; Mass (M+l): 509.3.
(R)-2-Cyclohexylethyl 3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 121):
Figure imgf000145_0002
1H NMR (400 MHz, CDC13) δ: 0.85 (m, 2H), 1.15-1.2 (m, 10H), 1.4-1.56 (m, 2H), 1.59-1.67 (m, IH), 1.8 (d, 3H), 2.7-3.2 (m, 4H), 3.8-4.2 (m, 2H), 7.19-7.3 (m, 4H), 7.5-7.6 (m, 2), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 97.73%; Mass (M+l): 565.25.
(R)-2-Cyclopentylethyl-3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 122):
Figure imgf000145_0003
1H NMR (400 MHz, CDC13) δ: 1.2-1.4 (m, 10H), 1.4-1.56 (m, 5H), 2.0 (m, IH), 2.7-3.2 (m, 3H), 3.8-4.2 (m, 2H), 7.0-7.2 (m, 4H), 7.5-7.6 (m, 2), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.96%; Mass (M+l): 551.23.
(R)-Cyclohexyl 3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 123):
Figure imgf000146_0001
XH NMR (400 MHz, CDC13) δ: 1.2 (d, 3H), 1.23-1.4 (m, 8H), 1.5 (m, IH), 1.6 (m, 2H), 1.89 (m, 2H), 2.7-3.2 (m, 3H), 3.8-4.2 (m, 3H), 7.0-7.2 (m, 4H), 7.5-7.6 (m, 2), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.72%; Mass (M+l): 537.50.
(R)-Tetrahydro-2H-pyran-4-yl-3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 124):
Figure imgf000146_0002
1H NMR (400 MHz, CDC13) δ: 1.2 (d, 3H), 1.23 (m, IH), 1.5 (m, IH), 1.6- 1.7 (m, 2H), 1.89-2.0 (m, 2H), 2.7-3.2 (m, 4H), 3.8-4.85 (m, 6H), 7.0-7.2 (m, 4H), 7.5-7.6 (m, 2H), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH); HPLC Purity: 98.83%; Mass (M+l): 539.30.
(R)-((R)-tetrahydrofuran-3-yl)3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 126):
Figure imgf000146_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.23 (m, IH), 1.8-2.2 (m, 2H), 2.7-3.2 (m, 3H), 3.6-4.0 (m, 6H), 5.17 (m, IH), 7.0-7.2 (m, 3H), 7.6-7.8 (m, 2H), 8.0 (d, IH), 8.2-8.4 (m, 2H), 9.1 (m, IH), 10.45 (s, IH); HPLC Purity: 99.52%; Mass (M+l): 525.45. (R)-((R)-tetrahydro-2H-pyran-3-yl)3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 127)(R):
Figure imgf000147_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.23 (m, IH), 1.75-1.78 (m, 3H), 2.85-2.9 (m, 3H), 3.41-3.79 (m, 6H), 3.89-3.9 (m, IH), 4.44 (m, IH), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.4 (d, IH), 8.5 (d, IH), 9.1 (m, IH), 10.45 (s, IH); HPLC Purity: 99.67%; Mass (M+1): 539.1.
(3R)-(Tetrahydrofuran-2-yl)methyl3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 128):
Figure imgf000147_0002
1H NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.53 (m, IH), 1.57-2.0 (m, 3H), 2.8-3.1 (m, 3H), 3.6-4.1 (m, 9H), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH), 10.45 (s, IH); HPLC Purity: 99.80%; Mass (M+1): 539.1.
(R)-Cyclopentyl3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 129):
Figure imgf000147_0003
XH NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.38 (m, IH), 1.39-1.8 (m, 8H), 2.6-3.0 (m, 3H), 3.5-3.8 (m, 3H), 4.95 (m, IH), 7.0-7.2 (m, 4H), 7.6-7.8 (m, 2H), 8.23 (d, IH), 8.4-8.5 (m, 2H), 9.1 (m, IH); HPLC Purity: 99.68%; Mass (M+1): 523.50.
(R)-((S)-tetrahydrofuran-3-yl)3-methyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 129)(S):
Figure imgf000148_0001
1H NMR (400 MHz, DMSO-d6) δ: 1.0 (d, 3H), 1.8 (m, IH), 2.0-2.2 (m, IH), 2.8-3.3 (m, 3H), 4.0-4.2 (m, 8H), 7.0-7.2 (m, 4H), 7.6-7.7 (m, 2H), 8.0 (d, IH), 8.3- 8.6 (m, 2H), 9.1 (m, IH), 10.4 (s, IH); HPLC Purity: 96.16%; Mass (M+1): 525.45.
Ethyl 4-(5-(quinoline-8-sulfonamido)picolinoyl)piperazine-l-carboxylate (Compound 447):
Figure imgf000148_0002
XH NMR (400 MHz, CDC13) δ: 1.2 (t, 3H), 3.2-3.6 (m, 8H), 4.0-4.2 (q, 2H), 7.4-7.8 (m, 4H), 8.0-8.6 (m, 4H), 9.1-9.2 (m, IH); HPLC Purity: 97.7%; Mass (M+1): 470.2.
Ethyl 4-(6-(quinoline-8-sulfonamido)nicotinoyl)piperazine-l-carboxylate (Compound 446):
Figure imgf000148_0003
1H NMR (400 MHz, DMSO-d6) δ: 1.0-1.2 (t, 3H), 2.2-2.4 (m, 2H), 3.2-3.35 (q, 2H), 3.4-3.6 (m, 4H), 3.99-4.0 (m, 2H), 7.5-7.7 (m, 4H), 8.3-8.5 (m, 4H), 9.1 (m, IH); HPLC Purity: 99.89%; Mass (M+1): 470.4. Example 15: Preparation of Compound 104 (racemic):
Scheme 15:
Figure imgf000149_0001
Synthesis of intermediate LXXXIII. The product LXXXIII was prepared by following similar method used for the preparation of intermediate LXXVI (Scheme 13) using carboxylic acid VIII (0.226 gm, 0.69 mmol) and cis-2,6-dimethylpiperazine XL (0.079 gm, 0.69 mmol). Crude product was purified by column chromatography (60-120 silica gel, 2% MeOH-DCM) to get 0.146 gm of pure product LXXXIII in 50% yields.
Synthesis of N-(4-(3,5-dimethylpiperazine-l-carbonyl)phenyl)quinoline-8- sulfonamide (Compound 104) (racemic). To a solution of amine LXXXIII (0.125 gm, 0.29 mmol) and ethyldiisopropylamine in dichloromethane was added picolyl chloride (XC, 0.045 gm, 0.32 mmol) at 0°C. The reaction mixture was allowed to warm to room temperature and stirred for 5 h. After completion of reaction, the mixture was diluted with dichloromethane, washed with water (2 x 10 ml), brine (10 ml), dried over anhydrous Na2S04 and concentrated. The crude material was then purified by column chromatography (60-120 silica gel, 2% MeOH-DCM) to get 0.039 gm of pure Compound 104 (racemic) in 25% yield.
1H NMR (400 MHz, CDC13) δ: 1.2 (s, 6H), 3.0 (m, IH), 3.6 (m, 2H), 4.3 (m, 2H), 7.0 (m, 4H), 7.4-7.6 (m, 4H), 8.0 (m, IH), 8.2 (m, IH), 8.4 (m, 3H), 9.0(m, IH); HPLC Purity: 98.75%; Mass (M+l): 530.3. Example 16: Preparation of Compound 116 (racemic)
Scheme 16:
Figure imgf000150_0001
ompoun
Synthesis of Intermediate XCII. The product XCII was prepared by following similar method used for the preparation of intermediate LXXVI (Scheme 13) using picolinic acid XCI (0.092 gm, 0.75 mmol) and cis-2,6-dimethylpiperazine XL (0.086 gm, 0.75 mmol). The crude material was purified by column
chromatography (60-120 silica gel, 2% MeOH-DCM) to get 0.126 gm of pure product XCII in 77% yield.
Synthesis of Compound 116 (racemic). Compound 116 was prepared by following similar method used for the preparation of a compound of Formula Io (Scheme 13) using carboxylic acid VIII (0.164 gm, 0.50 mmol) and amine XCII (0.110 gm, 0.50 mmol). Crude product was purified by column chromatography (60- 120 silica gel, 2% MeOH-DCM) to get 0.066 gm of pure Compound 116 (racemic) in 25% yields.
1H NMR (400 MHz, CDC13) δ: 2.8-3.0 (s, 6H), 3.35-3.4 (m, 2H), 4.0 (m, 2H), 4.5-4.66 (m, IH), 7.0-7.2 (m, 4H), 7.3-7.8 (m, 6H), 8.0-8.1 (m, IH), 8.2-8.6 (m, 4H), 9.0 (m, IH); HPLC Purity: 93.09%; Mass (M+l): 530.45.
Example 17: Preparation of a Compound of Formula Iq:
Figure imgf000150_0002
Scheme 17:
Figure imgf000151_0001
X III Formula Iq
Synthesis of Intermediate XCII. EDCI (0.09 gm, 0.46 mmol) and HOBt (0.062 gm, 0.46 mmol) were added to a stirred solution of the carboxylic acid (VIII, 0.151 gm, 0.46 mmol) in anhydrous DMF. The temperature of the mixture was reduced to 0°C, at which time DIPEA (0.24 mL, 1.38 mmol) was added under nitrogen atmosphere and the resultant solution (or suspension) was stirred at room temperature for 30 min. Amine XL (VI, 0.46 mmol) was then added at 0°C. The reaction mixture was then brought to room temperature and stirred for 12 h. After completion of the reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (2 x 25 ml). The organic layer was washed with water (2 x 10 ml), dried over anhydrous sodium sulfate, filtered and concentrated under vaccum to get the crude product. The obtained crude product was purified by column
chromatography (60-120 silica gel, 2% MeOH-DCM) to get pure product XCIII as an off-white solid in 40-50% yields.
Synthesis of Compounds of Formula Iq. To a solution of amine XI (0.102 gm, 0.24 mmol) and triethyl amine (0.66 mmol) in 5 mL of dichloromethane, appropriate chloroformate (0.26 mmol) was added at 0°C and allowed to stir at room temperature for 1-2 h. After completion of reaction, the mixture was diluted with dichloromethane (25 mL), washed with water (2 x 10 mL), brine (10 mL), dried over anhydrous Na2S04 and concentrated. The crude product was purified by column chromatography (60-120 silica gel, 2% MeOH-DCM) to afford a Compound of Formula Iq as an off-white solid in 30-40% yields.
The following compounds were made according to the above procedure using the appropriate chloroformate.
(2S,6R)-ethyl 2,6-dimethyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (XXIV-1) (Compound 117):
Figure imgf000152_0001
1H NMR (400 MHz, CDC13) δ: 1.2 (t, 3H), 1.3-1.4 (d, 6H), 3.0-3.4 (m, 2H), 4.0-4.4 (m, 6H), 7.0-7.3 (m, 4H), 7.6 (m, 2H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 9.0 (m, IH); HPLC Purity: 98.96%; Mass (M+l): 497.3.
(2S,6R)-isopropyl 2,6-dimethyl-4-(4-(quinoline-8- sulfonamido)benzoyl)pip 106):
Figure imgf000152_0002
1H NMR (400 MHz, CDC13) δ: 1.2 (s, 6H), 3.0-3.4 (m, 3H), 4.0-4.4 (m, 3H), 4.9 (m, IH), 7.0-7.3 (m, 4H), 7.6 (m, 2H), 8.0 (m, IH), 8.2-8.4 (m, 2H), 9.0 (m, IH); HPLC Purity: 99.96%; Mass (M+l): 511.4.
(2S,6R)-isobutyl2,6-dimethyl-4-(4-(quinoline-8- sulfonamido)benzoyl)piperazine-l-carboxylate (Compound 105):
Figure imgf000152_0003
1H NMR (400 MHz, CDC13) δ: 3.2-3.8 (m, 8H), 6.8-7.3 (m, 5H), 7.6 (m, 2H), 8.0 (m, IH), 8.3-8.4 (m, 3H), 9.0 (m, IH), 10.4 (m, IH); HPLC Purity: 99.45%; Mass (M+l): 492.2.
Example 18. PKM2 Assay.
Procedure:
• PKM2 stock enzyme solution was diluted in Reaction Buffer • 2 iL of compound was added into each well first, and then 180 μL· of the Reaction Mix was added.
• Reaction mixture with compound (without ADP) was incubated for 30
minutes at 4°C.
• Plates were re-equilibrated to room temperature prior to adding 20 \lL ADP to initiate the reaction.
• Reaction progress was measured as changes in absorbance at 340 nm
wavelength at room temperature (25 °C)
Reaction Mix: PKM2 (50 ng/well), ADP (0.7 mM), PEP (0.15 mM), NADH (180 μΜ), LDH (2 units) in Reaction Buffer
Reaction Buffer: 100 mM KC1, 50 mM Tris pH 7.5, 5 mM MgC12, 1 mM DTT, 0.03% BSA.
Representative compounds of the invention were tested for their ability to activate PKM2 usiong the above assay. For simplicity, the activation activity of these compounds is represented as an AC50 in Tables 4 and 5 and throughout the application. As shown in Tables 4 and 5, "A" refers to an activator of PKM2 with an EC50 < 100 nM. "B" refers to an activator of PKM2 with an EC50 between 100 nM and 500 nM. "C" refers to an activator of PKM2 with an EC50 between 500 nM and 1000 nM. "D" refers to an activator of PKM2 with an EC50 between 1 μΜ and 20 μΜ. "E" refers a compound whose activation of PKM2 was not measurable. "N/C" refers to AC50 data not available.
Table 4
Compound # AC50 Compound # AC50 Com ound # AC
131 A 199 A
119 A 186 A
118 A 190 A
150 A 123 A
149 A 111 A
206 A 221 A
151 A 212 A
132 A 155 A
112 A 165 A
213 A 108 A
129 A 160 A
148 A 211 A
125 A 128 A
133 A 227 B
135 A 228 B
223 A 202 B
120 A 124 B
114 A 109 B
113 A 136 B
110 A 218 B
216 A 164 B
217 A 207 B
226 A 134 B
183 A 102 B
161 A 195 B
143 A 191 B
145 A 141 B
147 A 157 B
167 A 219 B
196 A 201 B
163 A 115 B
152 A 138 B
137 A 153 B
189 A 126 B
194 A 103 B
140 A 168 B
222 A 154 B
122 A 178 B
Figure imgf000154_0001
142 A 127 B
130 A 121 B
200 A 192 B
220 A 173 B
144 A 106 B
203 A 181 B Table 5
Figure imgf000155_0001
Figure imgf000156_0001
ı55 Having thus described several aspects of several embodiments, it is to be appreciated various alterations, modifications, and improvements will readily occur to those skilled in the art. Such alterations, modifications, and improvements are intended to be part of this disclosure, and are intended to be within the spirit and scope of the invention. Accordingly, the foregoing description and drawings are by way of example only.

Claims

What is claimed is:
1. A com ound of formula (I):
Figure imgf000158_0001
(I), or a pharmaceutically acceptable salt thereof wherein:
W, X, Y and Z are each independently selected from CH or N;
Q and Q1 are independently selected from a bond or NRb;
A is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl;
L is a bond, -C(0)-, -(CRcRc)m-, -OC(O)-, -(CRcRc)m-OC(0)-, -(CRcRc)m- C(O)-, -NRbC(S)-, or -NRbC(0)- (wherein the point of the attachment to R1 is on the left-hand side);
R1 is selected from alkyl, carbocycle, aryl, heteroaryl, and heterocyclyl; each of which is substituted with 0-5 occurrences of Rd;
each R3 is independently selected from halo, haloalkyl, alkyl, hydroxyl and -ORa, or two adjacent R3 taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
each R4 is independently selected from halo, haloalkyl, alkyl, hydroxyl, =0, -ORa and phenyl, or two R4 taken together with the carbon atoms to which they are attached form a bridged, fused or spyro-fused carbocycle, an aryl or a heteroaryl; each Ra is independently selected from alkyl, acyl, hydroxyalkyl and haloalkyl;
each Rb is independently selected from hydrogen and alkyl;
each Rc is independently selected from hydrogen, halo, alkyl, alkoxy and halo alkoxy or two Rc taken together with the carbon atoms to which they are attached form an optionally substituted cycloalkyl; each Rd is independently selected from halo, haloalkyl, haloalkoxy, alkyl, alkynyl, nitro, cyano, hydroxyl, -C(0)Ra, -OC(0)Ra, -C(0)ORa, -SRa, -NRaRb and - ORa, or two Rd taken together with the carbon atoms to which they are attached form an optionally substituted heterocyclyl;
n is 0, 1, or 2;
m is 1 , 2 or 3 ;
h is 0, 1, 2;
g is 0, 1 or 2;
the sum of g + h is equal to or greater than 2; and
p is 0, 1 or 2; and provided that the compound of formula (I) is not
N-[3-[(3,5-dimethoxyphenyl)amino]-2-quinoxalinyl]-4-[(4-methyl-l- piperazinyl)carbonyl]- benzenesulfonamide;
N-[4-[[4-(2-furanylmethyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2- oxo- 1 H-benzimidazole-5 - sulfonamide;
2,3-dihydro-2-oxo-N-[4-[[4-(2,2,2-trifluoroethyl)-l- piperazinyl] carbonyljphenyl] - 1 H-benzimidazole- 5 - sulfonamide ;
2,3-dihydro-N-[4-[[4-(4-nitrophenyl)-l-piperazinyl]carbonyl]phenyl]-2-oxo- 1 H-benzimidazole-5 - sulfonamide ;
N- [4- [ [4-(2-ethoxyphenyl)- 1 -piperazinyl]carbonyl]phenyl] -2,3 -dihydro-2-oxo- 1 H-benzimidazole-5 - sulfonamide ;
2,3-dihydro-2-oxo-N-[4-[[4-(3-thienylmethyl)-l- piperazinyl] carbonyljphenyl] - 1 H-benzimidazole- 5 - sulfonamide ;
N-[4-[[4-(2,3-dimethylphenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2- oxo- 1 H-benzimidazole-5 - sulfonamide;
2,3-dihydro-N- [4- [ [4-(2-hydroxyphenyl)- 1 -piperazinyl]carbonyl]phenyl] -2- oxo- 1 H-benzimidazole-5 - sulfonamide;
4- [4- [ [(2,3 -dihydro-2-oxo- lH-benzimidazol-5 -yl)sulfonyl] aminojbenzoyl] - 1 - piperazinecarboxylic acid ethyl ester;
N-[4-[(4-acetyl-l-piperazinyl)carbonyl]phenyl]-2,3-dihydro-2-oxo-lH- benzimidazole- 5 - sulfonamide ; N-[4-[[4-(4-fluorophenyl)-l-piperazinyl]carbonyl]phenyl]-2,3-dihydro-2-oxo- 1 H-benzimidazole-5 - sulfonamide ;
2,3-dihydro-2-oxo-N-[4-[(4-phenyl-l-piperazinyl)carbonyl]phenyl]-lH- benzimidazole- 5 -sulfonamide; or
2,3-dihydro-2-oxo-N-[4-[[4-(2-pyridinyl)-l-piperazinyl]carbonyl]phenyl]-lH- benzimidazole- 5 - sulfonamide.
2. The compound of claim 1 , wherein In certain embodiments of a compound of formula (I) or a pharmaceutically acceptable salt thereof p is 1 or 2.
3. The compound of claim 2, wherein p is 2 and the compound has the formula la:
Figure imgf000160_0001
(la), or formula lb
Figure imgf000160_0002
(lb), wherein R1, L, R3, W,
X, Y, Z, Q, Q1, A, and n are as defined in claim 1.
4. The compound of claim 2, wherein:
p is 1 or 2; and
each R4 is independently selected from alkyl, phenyl, (5)-alkyl, (tf)-alkyl, (5")-phenyl, and (tf)-phenyl.
5. The compound of claim 4, wherein:
g is 1 ;
h is 1 ; and
each R4 is independently selected from methyl, (S)-methyl, (R)-methyl, ethyl, (S)-ethyl, (R)-ethyl, isopropyl, (5)-isopropyl, (/?)-isopropyl, phenyl, (S)- phenyl, and (R)-phenyl.
6. The compound of any one of claims 1-5, wherein A is
Figure imgf000161_0001
.
7. The compound of any one of claims 1-6, wherein W, X, Y, Z and the carbons to which they are attached form a phenyl ring.
8. The compound of any one of claims 1-7, wherein:
n is 1 ; and
R3 is selected from fluoro, chlororo, methyl, ethyl, CF3, methoxy, and OCF3.
9. The compound of any one of claims 1-8, wherein:
Q is NH; and
Q1 is a bond.
10. The compound of any one of claims 1-9, wherein L is selected from a bond,
-C(0)-, -OC(O)-, -CH2-OC(0)-, -(CH2)2-OC(0)-, -C(CH3)2-C(0)-, -CH2-, - -, -(CH2)3-, -CH(CH3)-, -CH(CF3)-, -C(CH3)2-, -CHD-, -CD2-,
Figure imgf000161_0002
11. The compound of any one of claims 1- 10, wherein R is selected from methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, l,2,3-thiadiazol-5-yl, 1,2,3-thiadiazol- 4-yl, thiazol-4-yl, thiazol-5-yl, lH-imidazol-4-yl, lH-imidazol-2-yl, 1H- pyrazol-3-yl, lH-pyrazol-4-yl, lH-pyrazol-5-yl, pyrazin-2-yl, oxazol-4-yl, isoxazol-5-yl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydro-2H- pyran-4-yl, tetrahydro-2H-pyran-3-yl, and tetrahydro-2H-pyran-2-yl.
12. The compound of claim 1, wherein the compound is selected from a
compound of Tables 1-2 and 3-4.
13. The compound of claim 12, wherein the compound is selected from any one of Compounds 108, 110, 111, 112, 113, 114, 118, 119, 120, 122, 123, 125, 128, 129, 130, 131, 132, 133, 135, 137, 140, 142, 143, 144, 145, 147, 148, 149, 150, 151, 152, 155, 160, 161, 163, 165, 167, 183, 186, 189, 190, 194, 196, 199, 200, 203, 206, 211, 212, 213, 216, 217, 220, 221, 222, and 223.
14. A pharmaceutical composition comprising a compound of a claim 1, and a pharmaceutically acceptable carrier.
15. A method of activating PKM2 activity in a subject in need thereof, comprising the step of administering to the subject a pharmaceutical composition of claim 14.
16. A method of treating a cancer associated with reduced PKM2 activity in a subject in need thereof, the method comprising administering to a subject a pharmaceutical composition of claim 14.
17. A composition comprising a compound of a claim 1, and a pharmaceutically acceptable carrier for use in treating a cancer associated with reduced PKM2 activity.
PCT/US2011/067752 2010-12-29 2011-12-29 Therapeutic compounds and compositions WO2012092442A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2013547663A JP5989664B2 (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositions
MX2013007541A MX342274B (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositions.
AU2011352085A AU2011352085B2 (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositions
EP11811286.1A EP2658543B1 (en) 2010-12-29 2011-12-29 Pyruvate kinase m2 (pkm2) activators for use in therapy
BR112013016811A BR112013016811A2 (en) 2010-12-29 2011-12-29 therapeutic compounds and compositions
CN201180068645.3A CN103491960B (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositionss
CA2823401A CA2823401C (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201061428030P 2010-12-29 2010-12-29
US61/428,030 2010-12-29

Publications (1)

Publication Number Publication Date
WO2012092442A1 true WO2012092442A1 (en) 2012-07-05

Family

ID=45509717

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/067752 WO2012092442A1 (en) 2010-12-29 2011-12-29 Therapeutic compounds and compositions

Country Status (11)

Country Link
US (2) US8889667B2 (en)
EP (1) EP2658543B1 (en)
JP (2) JP5989664B2 (en)
CN (1) CN103491960B (en)
AR (1) AR084748A1 (en)
AU (1) AU2011352085B2 (en)
BR (1) BR112013016811A2 (en)
CA (1) CA2823401C (en)
MX (1) MX342274B (en)
TW (1) TWI549947B (en)
WO (1) WO2012092442A1 (en)

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014018851A1 (en) 2012-07-26 2014-01-30 Joslin Diabetes Center, Inc. Predicting and treating diabetic complications
JP2014513134A (en) * 2011-05-03 2014-05-29 アジオス ファーマシューティカルズ, インコーポレイテッド How to use pyruvate kinase activator
US8742119B2 (en) 2009-04-06 2014-06-03 Agios Pharmaceuticals, Inc. Pyruvate kinase M2 modulators, therapeutic compositions and related methods of use
US8785450B2 (en) 2009-06-29 2014-07-22 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US8889667B2 (en) 2010-12-29 2014-11-18 Agios Pharmaceuticals, Inc Therapeutic compounds and compositions
US20150025068A1 (en) * 2011-12-22 2015-01-22 Kancera Ab Bisarylsulfonamides useful in the treatment of inflammation and cancer
CN104817490A (en) * 2015-05-13 2015-08-05 北京大学 Novel aminodithioformate compounds, and preparation method and application thereof
US9115086B2 (en) 2009-06-29 2015-08-25 Agios Pharmaceuticals, Inc. Therapeutic compositions and related methods of use
US9193701B2 (en) 2011-05-03 2015-11-24 Agios Pharmaceuticals, Inc Pyruvate kinase activators for use in therapy
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
JP2016501189A (en) * 2012-11-08 2016-01-18 アジオス ファーマシューティカルズ, インコーポレイテッド Therapeutic compounds and compositions and their use as PKM2 modulators
US9328077B2 (en) 2010-12-21 2016-05-03 Agios Pharmaceuticals, Inc Bicyclic PKM2 activators
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9850277B2 (en) 2012-01-19 2017-12-26 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US9982309B2 (en) 2009-10-21 2018-05-29 Agios Pharmaceuticals, Inc. Method for treating cell proliferation related disorders
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10028961B2 (en) 2013-07-11 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
WO2019075367A1 (en) 2017-10-13 2019-04-18 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
WO2020177668A1 (en) 2019-03-02 2020-09-10 察略盛医药科技(上海)有限公司 Pyrazolo[1,5-a]pyridine derivatives, preparation method therefor and use thereof
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies
US11001588B2 (en) 2018-09-19 2021-05-11 Forma Therapeutics, Inc. Activating pyruvate kinase R and mutants thereof
US11014927B2 (en) 2017-03-20 2021-05-25 Forma Therapeutics, Inc. Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US11040036B2 (en) 2017-08-15 2021-06-22 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
US11071725B2 (en) 2018-09-19 2021-07-27 Forma Therapeutics, Inc. Activating pyruvate kinase R
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US11566030B2 (en) 2021-02-08 2023-01-31 Global Blood Therapeutics, Inc. Substituted 2,6-dihydropyrrolo[3,4-c]pyrazoles as pyruvate kinase activators
WO2023116774A1 (en) 2021-12-21 2023-06-29 赛诺哈勃药业(成都)有限公司 Compound containing bis(azanylylidene) sulfonyl structure and use thereof in medicine
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
US11844758B2 (en) 2013-07-11 2023-12-19 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030207882A1 (en) * 1996-02-02 2003-11-06 Zeneca Limited Aminoheterocyclic derivatives as antithrombotic or anticoagulant agents
WO2007023186A1 (en) * 2005-08-26 2007-03-01 Laboratoires Serono S.A. Pyrazine derivatives and use as pi3k inhibitors
WO2010105243A1 (en) * 2009-03-13 2010-09-16 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
WO2011002817A1 (en) * 2009-06-29 2011-01-06 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions

Family Cites Families (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE510151A (en) 1949-07-23
GB935538A (en) 1959-04-06 1963-08-28 Stop Motion Devices Corp Stop-motion head for use on knitting machines
GB1274436A (en) 1970-06-09 1972-05-17 Wolfen Filmfab Veb Process for the sensitization of photographic silver chloride and silver chlorobromide emulsions that may contain colour couplers
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
DE2948434A1 (en) 1979-12-01 1981-06-11 Hoechst Ag, 6000 Frankfurt 1-PIPERIDINSULFONYL UREA AND METHOD FOR THE PRODUCTION THEREOF
US4474599A (en) 1982-07-14 1984-10-02 The Dow Chemical Company 1-(Pyridyl)-1H-1,2,3-triazole derivatives, and use as herbicidal agents
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
GB8325370D0 (en) 1983-09-22 1983-10-26 Fujisawa Pharmaceutical Co Benzoxazoline and benzothiazoline derivatives
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
JPS61129129A (en) 1984-11-28 1986-06-17 Kureha Chem Ind Co Ltd Antitumor agent
FI855180A (en) 1985-01-18 1986-07-19 Nissan Chemical Ind Ltd PYRAZOLESULFONAMIDDERIVAT, FOERFARANDE FOER DESS FRAMSTAELLANDE OCH DET INNEHAOLLANDE OGRAESGIFT.
EP0246749A3 (en) 1986-05-17 1988-08-31 AgrEvo UK Limited Triazole herbicides
JPS6339875A (en) 1986-08-05 1988-02-20 Nissin Food Prod Co Ltd Pyrimidine derivative
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US4775762A (en) 1987-05-11 1988-10-04 The Dow Chemical Company Novel (1H-1,2,3-triazol-1-yl)pyridines
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
DE3813885A1 (en) 1988-04-20 1989-11-02 Schering Ag 1-CHLORPYRIMIDINYL-1H-1,2,4-TRIAZOL-3-SULPHONIC ACID AMIDE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS AGENT WITH HERBICIDES, FUNGICIDES AND PLANT GROWTH REGULATING EFFECT
DE3813886A1 (en) 1988-04-20 1989-11-02 Schering Ag 1-Triazinyl-1H-1,2,4-triazole-3-sulphonamides, processes for their preparation, and their use as herbicides, fungicides and plant growth regulators
US5220028A (en) 1988-10-27 1993-06-15 Nissan Chemical Industries, Ltd. Halogeno-4-methylpyrazoles
KR910021381A (en) 1990-02-20 1991-12-20 모리 히데오 4-tert-butylimidazole derivatives, and preparation method and use thereof
CA2036148A1 (en) 1990-06-29 1991-12-30 Hiroki Tomioka A 1-phenylimidazole derivative and its production and use
CZ129493A3 (en) 1990-12-31 1994-03-16 Monsanto Co Elimination of pesticides influence on plants
US5252590A (en) 1991-06-28 1993-10-12 Sumitomo Chemical Company, Limited 1-pyridylimidazole derivative
IT1252567B (en) 1991-12-20 1995-06-19 Italfarmaco Spa DERIVATIVES OF 5-ISOKINOLINSULPHONAMIDS INHIBITORS OF PROTEIN-KINASES
JPH0625177A (en) 1992-07-09 1994-02-01 Nissan Chem Ind Ltd Pyrazole derivative and herbicide
JP3409165B2 (en) 1993-04-28 2003-05-26 株式会社林原生物化学研究所 Hair restorer and its production method
JP3719612B2 (en) 1993-06-14 2005-11-24 塩野義製薬株式会社 Urea derivatives containing heterocycles
IL115420A0 (en) 1994-09-26 1995-12-31 Zeneca Ltd Aminoheterocyclic derivatives
EP0804428B1 (en) 1995-01-20 2007-12-26 G.D. Searle LLC. Bis-sulfonamide hydroxyethylamino retroviral protease inhibitors
IL117580A0 (en) 1995-03-29 1996-07-23 Merck & Co Inc Inhibitors of farnesyl-protein transferase and pharmaceutical compositions containing them
FR2735127B1 (en) 1995-06-09 1997-08-22 Pf Medicament NEW HETEROAROMATIC PIPERAZINES USEFUL AS MEDICAMENTS.
ES2100129B1 (en) 1995-10-11 1998-02-16 Medichem Sa NEW POLYCLIC AMINOPYRIDINE COMPOUNDS ACETYLCHOLINESTERASE INHIBITORS, PROCEDURE FOR THE PREPARATION AND USE.
DE19541146A1 (en) 1995-10-25 1997-04-30 Schering Ag Imidazole derivatives and their use as nitric oxide synthase inhibitors
AU1608397A (en) * 1996-02-02 1997-08-22 Zeneca Limited Heterocyclic compounds useful as pharmaceutical agents
FR2744449B1 (en) 1996-02-02 1998-04-24 Pf Medicament NOVEL AROMATIC PIPERAZINES DERIVED FROM SUBSTITUTED CYCLOAZANES, AS WELL AS THEIR PREPARATION METHOD, PHARMACEUTICAL COMPOSITIONS AND THEIR USE AS MEDICAMENTS
US5834485A (en) 1996-05-20 1998-11-10 Chiroscience Limited Quinoline sulfonamides and their therapeutic use
US5843485A (en) 1996-06-28 1998-12-01 Incoe Corporation Valve-gate bushing for gas-assisted injection molding
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
DE19702988A1 (en) 1997-01-28 1998-07-30 Hoechst Ag Isoxazole and crotonic acid amide derivatives and their use as pharmaceuticals and diagnostics
EP0970072A1 (en) 1997-03-11 2000-01-12 E.I. Dupont De Nemours And Company Heteroaryl azole herbicides
US7863444B2 (en) 1997-03-19 2011-01-04 Abbott Laboratories 4-aminopyrrolopyrimidines as kinase inhibitors
DE19743435A1 (en) 1997-10-01 1999-04-08 Merck Patent Gmbh Benzamidine derivatives
US6106849A (en) 1998-01-21 2000-08-22 Dragoco Gerberding & Co. Ag Water soluble dry foam personal care product
EP1109544A4 (en) 1998-09-01 2004-10-27 Bristol Myers Squibb Co Potassium channel inhibitors and method
DE19841985A1 (en) 1998-09-03 2000-03-09 Schering Ag New heterocyclic alkanesulfonic and alkane carboxylic acid derivatives are VEGF receptor blockers useful in treatment of e.g. psoriasis, rheumatoid arthritis, stroke, tumors and endometriosis
HUP0200355A3 (en) 1998-09-18 2004-07-28 Abbott Gmbh & Co Kg 4-aminopyrrolopyrimidines as kinase inhibitors, pharmaceutical compositions containing them and their use
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
US6492408B1 (en) 1999-07-21 2002-12-10 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
MX226123B (en) 1999-09-17 2005-02-07 Millennium Pharm Inc BENZAMIDES AND RELATED INHIBITORS OF FACTOR Xa.
RU2002110295A (en) 1999-09-17 2003-12-10 Милленниум Фармасьютикалс, Инк. (Us) Factor Xa Inhibitors
ES2254385T3 (en) 2000-02-29 2006-06-16 Millennium Pharmaceuticals, Inc. RELATED BENZAMIDS AND INHIBITORS OF FACTOR XA.
US20010037689A1 (en) 2000-03-08 2001-11-08 Krouth Terrance F. Hydraulic actuator piston measurement apparatus and method
FR2817349B1 (en) 2000-11-28 2003-06-20 Centre Nat Rech Scient NEW METHOD FOR SCREENING MODULATORS OF THE BACTERIAL TRANSCRIPTION
JP2002193710A (en) 2000-12-25 2002-07-10 Kumiai Chem Ind Co Ltd Pyrimidine or triazine derivative and fungicide for agriculture and horticulture
DE10164711A1 (en) 2001-03-13 2002-10-17 Schebo Biotech Ag Use of sugar phosphates, sugar phosphate analogs, amino acids and / or amino acid analogs for modulating the glycolysis-enzyme complex, the malate aspartate shuttle and / or the transaminases
WO2002088101A2 (en) 2001-04-27 2002-11-07 Vertex Pharmaceuticals Incorporated Inhibitors of bace
US20040198979A1 (en) 2001-05-07 2004-10-07 Dashyant Dhanak Sulfonamides
DE60228233D1 (en) 2001-05-23 2008-09-25 Max Planck Gesellschaft PYRUVATE KINASE AS A NEW TARGET MOLECULE
US6967212B2 (en) 2001-05-30 2005-11-22 Bristol-Myers Squibb Company Substituted azole acid derivatives useful as antidiabetic and antiobesity agents and method
KR100889718B1 (en) 2001-06-11 2009-03-23 바이오비트럼 에이비(피유비엘) Substituted sulfonamide compounds, process for their use as medicament for the treatment of cns disorders, obesity and type ? diabetes
JP4083397B2 (en) 2001-06-18 2008-04-30 株式会社ルネサステクノロジ Semiconductor integrated circuit device
US7288554B2 (en) 2001-08-15 2007-10-30 E.I. Du Pont De Nemours And Company Ortho-substituted aryl amides for controlling invertebrate pests
JP2003081937A (en) 2001-09-07 2003-03-19 Bayer Ag Benzenesulfonamide derivative
US7169788B2 (en) 2001-10-30 2007-01-30 Merck & Co., Inc. Tyrosine kinase inhibitors
EP1472248A1 (en) 2002-01-17 2004-11-03 Eli Lilly And Company Modulators of acetylcholine receptors
AU2003217870A1 (en) 2002-03-01 2003-09-16 Pintex Pharmaceuticals, Inc. Pini-modulating compounds and methods of use thereof
US7816363B2 (en) 2002-03-13 2010-10-19 Janssen Pharmaceutica, N.V. Inhibitors of histone deacetylase
DK1485365T3 (en) 2002-03-13 2008-09-01 Janssen Pharmaceutica Nv Sulfonyl derivatives as novel inhibitors of histane deacetylase
EP1501514B1 (en) 2002-05-03 2012-12-19 Exelixis, Inc. Protein kinase modulators and methods of use
GB0215775D0 (en) 2002-07-06 2002-08-14 Astex Technology Ltd Pharmaceutical compounds
DE60334386D1 (en) 2002-08-09 2010-11-11 Merck Sharp & Dohme tyrosine kinase inhibitors
AU2003274373A1 (en) 2002-10-24 2004-05-13 Sterix Limited Inhibitors of 11-beta-hydroxy steroid dehydrogenase type 1 and type 2
WO2004074438A2 (en) 2003-02-14 2004-09-02 Smithkline Beecham Corporation Ccr8 antagonists
WO2004073619A2 (en) 2003-02-14 2004-09-02 Smithkline Beecham Corporation Ccr8 antagonists
CN100361992C (en) * 2003-02-19 2008-01-16 霍夫曼-拉罗奇有限公司 Sulfonamide substituted xanthine derivatives for use as PEPCK inhibitors
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
DE602004027171D1 (en) 2003-04-11 2010-06-24 High Point Pharmaceuticals Llc Compounds with activity on 11Beta hydroxasteroid dehydrogenase
WO2004110375A2 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Combination therapy for the treatment of diabetes
US6818631B1 (en) 2003-08-15 2004-11-16 Nippon Soda Co. Ltd. Fungicidal pyrimidine derivatives
US20080051414A1 (en) 2003-10-14 2008-02-28 Arizona Board Of Regents On Behalf Of The University Of Arizona Protein Kinase Inhibitors
US20050170316A1 (en) 2004-01-29 2005-08-04 Russell Bruce M. Toothbrush for detecting the presence of plaque
US7585850B2 (en) 2004-02-10 2009-09-08 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
JP2007523202A (en) 2004-02-24 2007-08-16 ビオアクソン・テラプティーク・インコーポレーテッド 4-substituted piperidine derivatives
WO2005117591A2 (en) 2004-05-28 2005-12-15 Andrx Labs Llc Novel pharmaceutical formulation containing a biguanide and an angiotensin antagonist
GB0412526D0 (en) 2004-06-05 2004-07-14 Leuven K U Res & Dev Type 2 diabetes
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
MX2007003319A (en) 2004-09-20 2007-06-05 Xenon Pharmaceuticals Inc Pyridazine derivatives for inhibiting human stearoyl-coa- desaturase.
WO2006033628A1 (en) 2004-09-24 2006-03-30 Astrazeneca Ab Benzimidazole derivatives, compositions containing them, preparation thereof and uses thereof
WO2006038594A1 (en) 2004-10-04 2006-04-13 Ono Pharmaceutical Co., Ltd. N-type calcium channel inhibitor
JP2008517054A (en) 2004-10-20 2008-05-22 スミスクライン・ビーチャム・コーポレイション IL-8 receptor antagonist
WO2006052546A2 (en) 2004-11-04 2006-05-18 Neurogen Corporation Pyrazolylmethyl heteroaryl derivatives
SE0402762D0 (en) 2004-11-11 2004-11-11 Astrazeneca Ab Indazole sulphonamide derivatives
WO2006055880A2 (en) 2004-11-16 2006-05-26 Genzyme Corporation Diagnostic pkm2 methods and compositions
JP2008526723A (en) 2004-12-30 2008-07-24 アステックス、セラピューティックス、リミテッド Pyrazole derivatives that modulate the activity of CDK, GSK and Aurora kinase
NZ561613A (en) 2005-02-18 2010-12-24 Astrazeneca Ab Antibacterial piperidine derivatives
WO2006117762A2 (en) 2005-05-03 2006-11-09 Ranbaxy Laboratories Limited Antimicrobial agents
WO2006122546A1 (en) 2005-05-18 2006-11-23 Forschungsverbund Berlin E.V. Non-peptidic inhibitors of akap/pka interaction
GB0513702D0 (en) 2005-07-04 2005-08-10 Sterix Ltd Compound
CN101258135A (en) 2005-07-05 2008-09-03 阿斯利康(瑞典)有限公司 New compounds, process for their preparation, intermediates, pharmaceutical compositions and their use in the treatment of 5-HT6 mediated disorders such as alzheimer desease, cognitive disorders, cogn
FI20055644A0 (en) 2005-12-02 2005-12-02 Nokia Corp group Communications
CA2633379A1 (en) 2005-12-22 2007-07-05 Merck & Co., Inc. Substituted piperidines as calcium channel blockers
JP2009530231A (en) 2006-01-20 2009-08-27 スミスクライン ビーチャム コーポレーション Use of sulfonamide derivatives in the treatment of metabolic and nervous system diseases
JP2007238458A (en) 2006-03-06 2007-09-20 D Western Therapeutics Institute Inc Novel isoquinoline derivative and medicine containing the same
JP2009533325A (en) 2006-03-17 2009-09-17 アストラゼネカ・アクチエボラーグ Novel tetralins as 5-HT6 modulators
AU2007231594A1 (en) 2006-03-23 2007-10-04 Amgen Inc. 1-phenylsulfonyl-diaza heterocyclic amide compounds and their uses as modulators of hydroxsteroid dehydrogenases
CA2659956C (en) 2006-08-04 2016-01-05 Lewis C. Cantley Inhibitors of pyruvate kinase and methods of treating disease
EP2056828A4 (en) 2006-08-21 2010-06-23 Merck Sharp & Dohme Sulfonylated piperazines as cannabinoid-1 receptor modulators
EP2050745B1 (en) 2006-09-01 2012-03-21 Otsuka AgriTechno Co., Ltd N-pyridylpiperidine compound, method for producing the same, and pest control agent
ES2435430T3 (en) 2006-10-16 2013-12-19 Thesan Pharmaceuticals, Inc. Therapeutic pyrazolyl thienopyridines
CA2669117A1 (en) 2006-10-26 2008-05-02 Gary A. Flynn Aquaporin modulators and methods of using them for the treatment of edema and fluid imbalance
HUP0600810A3 (en) 2006-10-27 2008-09-29 Richter Gedeon Nyrt New sulfonamide derivatives as bradykinin antagonists, process and intermediates for their preparation and pharmaceutical compositions containing them
NZ578180A (en) 2006-12-08 2012-02-24 Millennium Pharm Inc Unit dose formulations and methods of treating thrombosis with an oral factor xa inhibitor
WO2008130718A1 (en) 2007-04-23 2008-10-30 Atherogenics, Inc. Sulfonamide containing compounds for treatment of inflammatory disorders
KR101538391B1 (en) 2007-07-18 2015-07-21 얀센 파마슈티카 엔.브이. Sulfonamides as trpm8 modulators
US8299057B2 (en) 2007-07-20 2012-10-30 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
WO2009025781A1 (en) 2007-08-16 2009-02-26 Beth Israel Deaconess Medical Center Activators of pyruvate kinase m2 and methods of treating disease
CL2008003178A1 (en) 2007-10-26 2009-06-05 Syngenta Participations Ag Imidazole derived compounds; its preparation process; fungicidal composition comprising said compounds; use to control or prevent infestation of plants, harvested food crops, seeds, or non-living materials; and method to control or prevent infestation by phytopathogenic organisms.
EP2053045A1 (en) 2007-10-26 2009-04-29 Syngenta Participations AG Novel imidazole derivatives
US8642660B2 (en) 2007-12-21 2014-02-04 The University Of Rochester Method for altering the lifespan of eukaryotic organisms
ES2620634T3 (en) 2008-10-09 2017-06-29 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Human Pyruvate Kinase Activators
WO2010118063A2 (en) 2009-04-06 2010-10-14 Agios Pharmaceuticals, Inc. Therapeutic compositions and related methods of use
WO2010129596A1 (en) 2009-05-04 2010-11-11 Agios Pharmaceuticals, Inc. Pmk2 modulators for use in the treatment of cancer
WO2010130638A1 (en) 2009-05-14 2010-11-18 Evotec Ag Sulfonamide compounds, pharmaceutical compositions and uses thereof
US20120189670A1 (en) 2009-09-14 2012-07-26 Kirkpatrick D Lynn Pharmaceutical compositions and formulations including inhibitors of the pleckstrin homology domain and methods for using same
US9062076B2 (en) 2009-10-22 2015-06-23 Fibrotech Therapeutics Pty Ltd Fused ring analogues of anti-fibrotic agents
ES2642109T3 (en) 2009-12-09 2017-11-15 Agios Pharmaceuticals, Inc. Therapeutically active compounds for use in the treatment of cancer characterized by having an IDH mutation
JP2013525438A (en) * 2010-04-29 2013-06-20 アメリカ合衆国 Human pyruvate kinase activator
KR20130087002A (en) 2010-06-04 2013-08-05 알바니 몰레큘라 리써치, 인크. Glycine transporter-1 inhibitors, methods of making them, and uses thereof
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
TWI549947B (en) 2010-12-29 2016-09-21 阿吉歐斯製藥公司 Therapeutic compounds and compositions
EP2704720B1 (en) * 2011-05-03 2019-08-07 Agios Pharmaceuticals, Inc. Pyruvate kinase r activators for use in therapy

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030207882A1 (en) * 1996-02-02 2003-11-06 Zeneca Limited Aminoheterocyclic derivatives as antithrombotic or anticoagulant agents
WO2007023186A1 (en) * 2005-08-26 2007-03-01 Laboratoires Serono S.A. Pyrazine derivatives and use as pi3k inhibitors
WO2010105243A1 (en) * 2009-03-13 2010-09-16 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
WO2011002817A1 (en) * 2009-06-29 2011-01-06 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
L. FIESER; M. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS

Cited By (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10610125B2 (en) 2009-03-13 2020-04-07 Agios Pharmaceuticals, Inc. Methods and compositions for cell-proliferation-related disorders
US9657004B2 (en) 2009-04-06 2017-05-23 Agios Pharmaceuticals, Inc Pyruvate kinase M2 modulators, therapeutic compositions and related methods of use
US9938259B2 (en) 2009-04-06 2018-04-10 Agios Pharmaceuticals, Inc. Therapeutic compositions and related methods of use
US8742119B2 (en) 2009-04-06 2014-06-03 Agios Pharmaceuticals, Inc. Pyruvate kinase M2 modulators, therapeutic compositions and related methods of use
US11866411B2 (en) 2009-06-29 2024-01-09 Agios Pharmaceutical, Inc. Therapeutic compounds and compositions
US9115086B2 (en) 2009-06-29 2015-08-25 Agios Pharmaceuticals, Inc. Therapeutic compositions and related methods of use
US8785450B2 (en) 2009-06-29 2014-07-22 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10029987B2 (en) 2009-06-29 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
USRE49582E1 (en) 2009-06-29 2023-07-18 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10988448B2 (en) 2009-06-29 2021-04-27 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US10711314B2 (en) 2009-10-21 2020-07-14 Agios Pharmaceuticals, Inc. Methods for diagnosing IDH-mutant cell proliferation disorders
US9982309B2 (en) 2009-10-21 2018-05-29 Agios Pharmaceuticals, Inc. Method for treating cell proliferation related disorders
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
US9328077B2 (en) 2010-12-21 2016-05-03 Agios Pharmaceuticals, Inc Bicyclic PKM2 activators
US10087169B2 (en) 2010-12-21 2018-10-02 Agios Pharmaceuticals, Inc. Bicyclic PKM2 activators
US9199968B2 (en) 2010-12-29 2015-12-01 Agios Pharmaceuticals, Inc. Therapeutic compounds and compositions
US8889667B2 (en) 2010-12-29 2014-11-18 Agios Pharmaceuticals, Inc Therapeutic compounds and compositions
US9980961B2 (en) 2011-05-03 2018-05-29 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
EP2704720B1 (en) * 2011-05-03 2019-08-07 Agios Pharmaceuticals, Inc. Pyruvate kinase r activators for use in therapy
JP2014513134A (en) * 2011-05-03 2014-05-29 アジオス ファーマシューティカルズ, インコーポレイテッド How to use pyruvate kinase activator
US10632114B2 (en) 2011-05-03 2020-04-28 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US9193701B2 (en) 2011-05-03 2015-11-24 Agios Pharmaceuticals, Inc Pyruvate kinase activators for use in therapy
US9404081B2 (en) 2011-05-03 2016-08-02 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US11793806B2 (en) 2011-05-03 2023-10-24 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in therapy
US9682080B2 (en) 2011-05-03 2017-06-20 Agios Pharmaceuticals, Inc Pyruvate kinase activators for use in therapy
US9662327B2 (en) 2011-06-17 2017-05-30 Agios Pharmaceuticals, Inc Phenyl and pyridinyl substituted piperidines and piperazines as inhibitors of IDH1 mutants and their use in treating cancer
US9856279B2 (en) 2011-06-17 2018-01-02 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US9718809B2 (en) * 2011-12-22 2017-08-01 Kancera Ab Bisarylsulfonamides useful in the treatment of inflammation and cancer
US20150025068A1 (en) * 2011-12-22 2015-01-22 Kancera Ab Bisarylsulfonamides useful in the treatment of inflammation and cancer
US9850277B2 (en) 2012-01-19 2017-12-26 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US10717764B2 (en) 2012-01-19 2020-07-21 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10640534B2 (en) 2012-01-19 2020-05-05 Agios Pharmaceuticals, Inc. Therapeutically active compositions and their methods of use
US11667673B2 (en) 2012-01-19 2023-06-06 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US9921221B2 (en) 2012-07-26 2018-03-20 Joslin Diabetes Center, Inc. Predicting and treating diabetic complications
WO2014018851A1 (en) 2012-07-26 2014-01-30 Joslin Diabetes Center, Inc. Predicting and treating diabetic complications
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
JP2016501189A (en) * 2012-11-08 2016-01-18 アジオス ファーマシューティカルズ, インコーポレイテッド Therapeutic compounds and compositions and their use as PKM2 modulators
US10457655B2 (en) 2013-03-13 2019-10-29 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10472342B2 (en) 2013-03-13 2019-11-12 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10450286B2 (en) 2013-03-13 2019-10-22 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10995078B2 (en) 2013-03-13 2021-05-04 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10800750B2 (en) 2013-03-13 2020-10-13 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10376510B2 (en) 2013-07-11 2019-08-13 Agios Pharmaceuticals, Inc. 2,4- or 4,6-diaminopyrimidine compounds as IDH2 mutants inhibitors for the treatment of cancer
US11844758B2 (en) 2013-07-11 2023-12-19 Servier Pharmaceuticals Llc Therapeutically active compounds and their methods of use
US10172864B2 (en) 2013-07-11 2019-01-08 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10028961B2 (en) 2013-07-11 2018-07-24 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10017495B2 (en) 2013-07-11 2018-07-10 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10946023B2 (en) 2013-07-11 2021-03-16 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US10689414B2 (en) 2013-07-25 2020-06-23 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US11021515B2 (en) 2013-07-25 2021-06-01 Agios Pharmaceuticals, Inc. Therapeutically active compounds and their methods of use
US9968595B2 (en) 2014-03-14 2018-05-15 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US10449184B2 (en) 2014-03-14 2019-10-22 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
US11504361B2 (en) 2014-03-14 2022-11-22 Servier Pharmaceuticals Llc Pharmaceutical compositions of therapeutically active compounds
US10799490B2 (en) 2014-03-14 2020-10-13 Agios Pharmaceuticals, Inc. Pharmaceutical compositions of therapeutically active compounds
CN104817490A (en) * 2015-05-13 2015-08-05 北京大学 Novel aminodithioformate compounds, and preparation method and application thereof
CN104817490B (en) * 2015-05-13 2017-10-13 北京大学 Dithiocarbamates compound and preparation method and application
US11234976B2 (en) 2015-06-11 2022-02-01 Agios Pharmaceuticals, Inc. Methods of using pyruvate kinase activators
US11419859B2 (en) 2015-10-15 2022-08-23 Servier Pharmaceuticals Llc Combination therapy for treating malignancies
US10653710B2 (en) 2015-10-15 2020-05-19 Agios Pharmaceuticals, Inc. Combination therapy for treating malignancies
US11649242B2 (en) 2017-03-20 2023-05-16 Forma Therapeutics, Inc. Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US12071440B2 (en) 2017-03-20 2024-08-27 Novo Nordisk Health Care Ag Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US11396513B2 (en) 2017-03-20 2022-07-26 Forma Therapeutics, Inc. Compositions for activating pyruvate kinase
US11014927B2 (en) 2017-03-20 2021-05-25 Forma Therapeutics, Inc. Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US11590132B2 (en) 2017-08-15 2023-02-28 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
US11957680B2 (en) 2017-08-15 2024-04-16 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
US11872225B2 (en) 2017-08-15 2024-01-16 Agios Pharmaceuticals, Inc. Pyruvate kinase modulators and use thereof
US11464775B2 (en) 2017-08-15 2022-10-11 Agios Pharmaceuticals, Inc. Pyruvate kinase modulators and use thereof
US11364240B2 (en) 2017-08-15 2022-06-21 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
US11040036B2 (en) 2017-08-15 2021-06-22 Agios Pharmaceuticals, Inc. Pyruvate kinase activators for use in treating blood disorders
WO2019075367A1 (en) 2017-10-13 2019-04-18 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
US10980788B2 (en) 2018-06-08 2021-04-20 Agios Pharmaceuticals, Inc. Therapy for treating malignancies
US11844787B2 (en) 2018-09-19 2023-12-19 Novo Nordisk Health Care Ag Activating pyruvate kinase R
US11071725B2 (en) 2018-09-19 2021-07-27 Forma Therapeutics, Inc. Activating pyruvate kinase R
US11001588B2 (en) 2018-09-19 2021-05-11 Forma Therapeutics, Inc. Activating pyruvate kinase R and mutants thereof
US11980611B2 (en) 2018-09-19 2024-05-14 Novo Nordisk Health Care Ag Treating sickle cell disease with a pyruvate kinase R activating compound
US12053458B2 (en) 2018-09-19 2024-08-06 Novo Nordisk Health Care Ag Treating sickle cell disease with a pyruvate kinase R activating compound
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11299484B2 (en) 2018-10-10 2022-04-12 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11267805B2 (en) 2018-10-29 2022-03-08 Forma Therapeutics, Inc. Solid forms of (4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl) piperazine-1-yl)(1-hydroxycyclopropyl)methanone
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
WO2020177668A1 (en) 2019-03-02 2020-09-10 察略盛医药科技(上海)有限公司 Pyrazolo[1,5-a]pyridine derivatives, preparation method therefor and use thereof
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
US11566030B2 (en) 2021-02-08 2023-01-31 Global Blood Therapeutics, Inc. Substituted 2,6-dihydropyrrolo[3,4-c]pyrazoles as pyruvate kinase activators
WO2023116774A1 (en) 2021-12-21 2023-06-29 赛诺哈勃药业(成都)有限公司 Compound containing bis(azanylylidene) sulfonyl structure and use thereof in medicine

Also Published As

Publication number Publication date
EP2658543A1 (en) 2013-11-06
JP2016104814A (en) 2016-06-09
TWI549947B (en) 2016-09-21
EP2658543B1 (en) 2019-02-20
MX2013007541A (en) 2014-07-09
US9199968B2 (en) 2015-12-01
AU2011352085A1 (en) 2013-07-18
CN103491960B (en) 2017-03-29
CN103491960A (en) 2014-01-01
CA2823401C (en) 2019-02-26
JP5989664B2 (en) 2016-09-07
US8889667B2 (en) 2014-11-18
MX342274B (en) 2016-09-21
AR084748A1 (en) 2013-06-05
JP2014505048A (en) 2014-02-27
US20150183760A1 (en) 2015-07-02
US20120172349A1 (en) 2012-07-05
AU2011352085B2 (en) 2017-04-27
TW201305131A (en) 2013-02-01
CA2823401A1 (en) 2012-07-05
BR112013016811A2 (en) 2016-09-27

Similar Documents

Publication Publication Date Title
AU2011352085B2 (en) Therapeutic compounds and compositions
USRE49582E1 (en) Therapeutic compounds and compositions
AU2010234526B2 (en) Pyruvate kinase M2 modulators, therapeutic compositions and related methods of use
EP2448581A1 (en) Therapeutic compositions and related methods of use
WO2012083246A1 (en) Novel n- (4- (azetidine - 1 - carbonyl) phenyl) - (hetero - ) arylsulfonamide derivatives as pyruvate kinase m2 (pmk2) modulators

Legal Events

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

Ref document number: 11811286

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2823401

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/007541

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2013547663

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011811286

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2011352085

Country of ref document: AU

Date of ref document: 20111229

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013016811

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013016811

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130628