US20070178134A1 - Pyridines and uses thereof - Google Patents

Pyridines and uses thereof Download PDF

Info

Publication number
US20070178134A1
US20070178134A1 US11/724,459 US72445907A US2007178134A1 US 20070178134 A1 US20070178134 A1 US 20070178134A1 US 72445907 A US72445907 A US 72445907A US 2007178134 A1 US2007178134 A1 US 2007178134A1
Authority
US
United States
Prior art keywords
compound
salt
alkyl
alkoxy
lpaat
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/724,459
Inventor
Feng Hong
J. Klein
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cell Therapeutics Inc
Original Assignee
Cell Therapeutics 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 Cell Therapeutics Inc filed Critical Cell Therapeutics Inc
Priority to US11/724,459 priority Critical patent/US20070178134A1/en
Publication of US20070178134A1 publication Critical patent/US20070178134A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/28Materials for coating prostheses
    • A61L27/34Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates

Definitions

  • the invention is in the field of organic and medicinal chemistry.
  • the invention relates to pyridines and uses thereof, such as.inhibiting the activity of lysophosphatidic acid acyltransferase ⁇ (LPAAT- ⁇ ) activity and/or inhibiting the proliferation of a cell (e.g., tumor cell).
  • LPAAT- ⁇ lysophosphatidic acid acyltransferase ⁇
  • Lysophosphatidic acid acyltransferase catalyzes the acylation of lysophosphatidic acid (LPA) to phosphatidic acid.
  • LPA is the simplest glycerophospholipid, consisting of a glycerol molecule, a phosphate group, and a fatty acyl chain.
  • LPAAT adds a second fatty acyl chain to LPA, producing phosphatidic acid (PA).
  • PA is the precursor molecule for certain phosphoglycerides, such as phosphatidylinositol, and diacylglycerols, which are necessary for the production of other phosphoglycerides, such as phosphatidylcholine, and for triacylglycerols, which are essential biological fuel molecules.
  • phosphoglycerides such as phosphatidylinositol
  • diacylglycerols which are necessary for the production of other phosphoglycerides, such as phosphatidylcholine, and for triacylglycerols, which are essential biological fuel molecules.
  • LPA has been added to the list of intercellular lipid messenger molecules.
  • LPA interacts with G protein-coupled receptors, coupling to various independent effector pathways including inhibition of adenylate cyclase, stimulation of phospholipase C, activation of MAP kinases, and activation of the small GTP-binding proteins Ras and Rho. Moolenaar, J. Biol. Chem. 28:1294 (1995).
  • the physiological effects of LPA have not been fully characterized as yet. However, one of the physiological effects that is known is that LPA promotes the growth and invasion of tumor cells.
  • LPA LPA-induced MM1 tumor cells to invade cultured mesothelial cell monolayers. Imamura et al., Biochem. Biophys. Res. Comm. 193:497 (1993).
  • PA is also a messenger molecule.
  • PA is a key messenger in a common signaling pathway activated by proinflammatory mediators such as interleukin-1 ⁇ , tumor necrosis factor ⁇ , platelet activating factor, and lipid A. Bursten et al., Am. J. Physiol. 262:C328 (1992); Bursten et al., J. Biol. Chem. 255:20732 (1991); Kester, J. Cell Physiol. 156:317 (1993). PA has been implicated in mitogenesis of several cell lines [English, Cell Signal 8:341 (1996)].
  • PA level has been found to be increased in either ras or fps transformed cell lines compared to the parental Rat2 fibroblast cell line [Martin et al., Oncogene 14:1571 (1997)].
  • Activation of Raf-1 an essential component of the MAPK signaling cascade, by extracellular signals is initiated by association with intracellular membranes. Recruitment of Raf-1 to membranes has been reported to be mediated by direct association with phosphatidic acid [Rizzo et al., J. Biol. Chem. 275:23911-8 (2000)].
  • LPAAT as an enzyme that regulates PA content in cells, may play a role in cancer, and may also mediate inflammatory responses to various proinflammatory agents.
  • LPAAT exists in a LPAAT- ⁇ form and a LPAAT- ⁇ form.
  • Northern blot analysis shows that LPAAT- ⁇ is expressed in all human tissues tested with the highest expression level found in skeletal muscle (West et al., DNA Cell Biol. 16:691 (1997)).
  • the uniformity of LPAAT- ⁇ expression has also been found in additional tissues such as prostate, testis, ovary, small intestine, and colon (Stamps et al., Biochem. J. 326:455 (1997)) as well as in mouse tissues (Kume et al., Biochem. Biophys. Res. Commun. 237:663 (1997)).
  • LPAAT- ⁇ demonstrates a distinct tissue distribution of mRNA expression. West et al., DNA Cell Biol. 16:691 (1997). LPAAT- ⁇ is most highly expressed in liver and heart tissues. LPAAT- ⁇ is also expressed at moderate levels in pancreas, lung, skeletal muscle, kidney, spleen, and bone marrow; and at low levels in thymus, brain and placenta. This differential pattern of LPAAT- ⁇ expression has been confirmed independently (Eberhardt et al., J. Biol. Chem.
  • LPAAT- ⁇ can also be detected in myeloid cell lines THP-1, HL-60, and U937 with the mRNA levels remaining the same with or without phorbal-ester treatment.
  • the size difference between human LPAAT- ⁇ and LPAAT- ⁇ mRNA is consistent with the sequence data, in which LPAAT- ⁇ has a longer 3′-UTR.
  • the differential tissue expression pattern LPAAT- ⁇ and LPAAT- ⁇ mRNA would suggest these two genes are regulated differently and are likely to have independent functions. Therefore, a desirable feature in compounds that inhibit LPAAT activity is that they are specific in inhibiting one isoform of the enzyme over the other (i.e., LPAAT- ⁇ over LPAAT- ⁇ ).
  • LPAAT- ⁇ mRNA has been found to be elevated in tumor tissues (e.g., uterus, fallopian tube, and ovary), as compared to its expression in the corresponding normal tissues. However, no significant difference was found in LPAAT- ⁇ mRNA level between the various tumor tissues and the normal adjacent tissues. In two of the tumor tissues (fallopian tube and ovary) where LPAAT- ⁇ mRNA was elevated, PAP2- ⁇ mRNA expression was found to be suppressed, as it was also in tumors of the colon, rectum, and breast. Thus, LPAAT- ⁇ (rather than LPAAT- ⁇ ) appears to be a relevant target for inhibition.
  • tumor tissues e.g., uterus, fallopian tube, and ovary
  • the present invention provides a variety of compounds and uses thereof. More specifically, the compounds of the present invention are pyridines that possess aromatic substituents which are directly or indirectly attached to two non-adjacent carbons of the pyridine ring.
  • the compounds are generally of the formula: where R 1 —R 6 are hydrogen or non-hydrogen substituents, Q is a heteroatom or heteroatom attached to one or more methylene groups, and one of X, Y and Z is N with the other two being CH or a substituted C.
  • R 1 —R 6 are hydrogen or non-hydrogen substituents
  • Q is a heteroatom or heteroatom attached to one or more methylene groups
  • one of X, Y and Z is N with the other two being CH or a substituted C.
  • X, Y and Z are N, CH or CR where R is alkyl, alkoxy, Cl, Br, NH 2 , NHR′ or NR′R′′ where R′ and R′′ independently are alkyl;
  • Q is NR, RN—(CH 2 ) n , (CH 2 ) n ,—NR, O, O—(CH 2 ) n , (CH 2 ) n —O, S, S—(CH 2 ) n or (CH 2 ) n —S, where n is 1-10 and R is H or alkyl;
  • R 1 is H, OH, alkyl, alkoxy, Cl, F, Br, CR 3 where R 3 is Cl 3 , F 3 or Br 3 , NH 2 , NHR or NRR′ where R and R′ independently are alkyl;
  • R 2 is H, OH, alkyl, alkoxy, Cl, F, Br or CR 3 where R 3 is Cl 3 , F 3 or Br 3 ;
  • R 3 is H, alkyl, alkoxy, Cl, CCl 3 , NH 2 , NHR or NRR′ where R and R′ independently are alkyl or acyl;
  • R 4 , R 5 and R 6 are independently H, OH, alkyl, alkenyl, alkynyl, alkoxy, (CH 2 ) n —OR where R is H or alkyl and n is 1-10, Cl, F, Br, CR 3 where R 3 is Cl 3 , F 3 or Br 3 , acyl, heterocycle, N + ( ⁇ O)O ⁇ , C ⁇ N, N 3 , SH, SR or S( ⁇ O) 2 R where R is alkyl, NH 2 , NHR or NRR′ where R and R′ independently are alkyl, or R 4 and R 5 or R 5 and R 6 are taken together with the benzene ring to form a heterocycle;
  • a compound or salt thereof as described above may be combined with a pharmaceutical carrier or diluent to form a pharmaceutical composition of the present invention.
  • a compound, salt thereof or pharmaceutical composition of the present invention may be used in one or more methods.
  • the activity of LPAAT- ⁇ may be reduced by the step comprising contacting LPAAT- ⁇ with a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to reduce LPAAT- ⁇ activity.
  • the proliferation of a cell in which the activity of LPAAT- ⁇ is required for the proliferation of the cell may be inhibited by the step comprising contacting LPAAT- ⁇ with a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to inhibit the proliferation of the cell.
  • the treatment of a cancer in which LPAAT- ⁇ activity is associated may be effected by the step comprising administering to an animal in need a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to treat the cancer.
  • a coated medical device for inhibiting the proliferation of a cell in which the activity of LPAAT- ⁇ is required for the proliferation of the cell comprising a medical device coated with a compound, salt thereof or pharmaceutical composition of the present invention.
  • alkyl refers to straight- or branched-chain hydrocarbons having from 1 to 10 carbon atoms and more preferably 1 to 8 carbon atoms which include, by way of example, methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl and the like.
  • the alkyl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably one or more independently selected from alkyl, cycloalkyl, heteroalicyclic, aryl, heteroaryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, substituted imino and substituted amino.
  • Alkenyl includes monovalent hydrocarbon radicals having straight, cyclic, or branched moieties, and combinations thereof which comprise at least one carbon-carbon double bond.
  • the alkenyl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably one or more independently selected from alkyl, acyl, cycloalkyl, heteroalicyclic, aryl, haloalkyl, alkoxy and substituted amino.
  • Alkoxy refers to the group “—O-alkyl” which includes, by way of example, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, t-butoxy and the like.
  • Substituted amino denotes the group —NRR, wherein each R group is independently selected from hydrogen, acyl, alkyl, cycloalkyl, aryl, or the R groups can be joined together with the nitrogen to form a heterocyclic ring (e.g., piperidine, piperazine, or a morpholine ring).
  • a heterocyclic ring e.g., piperidine, piperazine, or a morpholine ring.
  • Substituted imino denotes the group ⁇ NR, wherein R is preferably selected from hydrogen, hydroxy, alkyl and acyl.
  • Aryl refers to an unsaturated aromatic carbocyclic group of 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl).
  • the aryl group may be unsubstituted or substituted; in the latter case, the substituent or substituents preferably are selected independently from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, nitro, and substituted amino.
  • Heterocycle includes “heteroaryl” and “heteroalicyclic”. Examples of heterocycles include oxazole, piperidine, piperazine and morpholine.
  • Heteroaryl is a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected preferably from nitrogen, oxygen and sulfur and, in addition, having a completely conjugated ⁇ -electron system.
  • exemplary heteroaryl groups are pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrimidine, quinoline, isoquinoline, purine and carbazole.
  • the heteroaryl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably one or more independently selected from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, nitro and substituted amino.
  • Cycloalkyl encompasses cyclic alkyl groups that contain between 3 and 8 carbon atoms and have a single cyclic ring, illustrated by cyclopropyl, cyclobutyl, cyclopentyl, and cyclooctyl.
  • the cycloalkyl ring may be substituted or unsubstituted.
  • a substituted cycloalkyl ring carries one or more substituent groups, independently selected preferably from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, vitro, and substituted amino.
  • Heteroalicyclic refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected preferably from nitrogen, oxygen and sulfur.
  • the rings may also have one or more double bonds. However, the rings do not have a completely conjugated ⁇ -electron system.
  • the heteroalicyclic ring may be substituted or unsubstituted. When substituted, the substituted group(s) preferably are selected independently from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, vitro, and substituted amino.
  • Halogen or “halo” refers to fluoro, chloro, bromo, iodo.
  • “Acyl” group refers to the C(O)—R′′ group, where R′′ is selected preferably from hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, aryl optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups, heteroaryl (bonded through a ring carbon) optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups and heteroalicyclic (bonded through a ring carbon) optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups.
  • Acyl groups include aldehydes, ketones, acids, acid halides, esters and amides. Preferred acyl groups are carboxy groups, e.g., acids and esters. Esters include amino acid ester derivatives.
  • the acyl group may be attached to a compound's backbone at either end of the acyl group, i.e., via the C or the R′′. Where the acyl group is attached via the R′′, then C will bear another substituent, such as hydrogen or alkyl.
  • physiologically acceptable salt refers to those salts that retain the biological effectiveness and properties of the particular compound. Physiologically acceptable salts are often useful because they may have improved stability and/or solubility in pharmaceutical compositions over the free base form of the compound.
  • a physiologically acceptable salt may be obtained by reaction of a free base with an inorganic acid such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, sulfuric acid, and perchloric acid and the like, or with an organic acid such as acetic acid, oxalic acid, malic acid, maleic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, succinic acid or malonic acid, and the like.
  • a physiologically acceptable salt may also be obtained by reaction of a free acid with a base such as sodium, potassium or lithium hydroxide, bicarbonate or carbonate, and the like.
  • the present invention provides pyridines, physiologically acceptable salts thereof and uses thereof.
  • the pyridines possess aromatic substituents that are directly or indirectly attached to two non-adjacent carbons of the pyridine ring.
  • the compounds are generally of the formula: where R 1 —R 6 are hydrogen or non-hydrogen substituents, Q is a heteroatom or heteroatom attached to one or more methylene groups, and two of X, Y and Z are N with the third being CH or a substituted C. The requirement that only one of X, Y and Z is N is consistent with the compounds including a pyridine ring.
  • Preferred embodiments include the following selections for the general formula above. Preferred embodiments include where X, Y and Z are N, CH or CR. R of CR is alkyl, alkoxy, halo (preferably Cl or Br), NH 2 , NHR′ or NR′R′′ where R′ and R′′ independently are alkyl. Particularly preferred is where X or Y is N.
  • Preferred embodiments include where Q is a heteroatom (preferably N, O or S) and may be attached to one or more methylene groups to provide additional spacing between the pyridine ring and the phenyl ring possessing R 4 , R 5 and/or R 6 .
  • Q may be NR where R is H or alkyl. Where there are one or more methylene groups, the heteroatom may be oriented such that it is attached directly to the pyridine ring or attached directly to the phenyl ring possessing R 4 , R 5 and/or R 6 .
  • Q may be RN—(CH 2 )n, (CH 2 ) n —NR, O—(CH 2 ) n , (CH 2 ) n —O, S—(CH 2 ) n or (CH 2 ) n —S, where n is typically 1-10 and R is H or alkyl. Particularly preferred is where Q is NH.
  • R 1 is H, OH, alkyl, alkoxy, halogen (preferably Cl, F or Br), CR 3 , NH 2 , NHR or NRR′.
  • R 3 of CR 3 is (halo) 3 , preferably Cl 3 , F 3 or Br 3 .
  • R and R′ of NHR and NRR′ are independently alkyl.
  • the term “independently,” as used throughout, refers to independent selection of a group, but does not exclude the possibility that two groups are identical.
  • the alkyl group of R and R′ of NRR′ may be the same or different. Particularly preferred is where R 1 is alkyl, alkoxy or Cl.
  • R 2 is H, OH, alkyl, alkoxy, halogen (preferably Cl, F or Br), or CR 3 .
  • R 3 of CR 3 is (halo) 3 , preferably Cl 3 , F 3 or Br 3 . Particularly preferred is where R 2 is Cl or Br.
  • R 3 is H, alkyl, alkoxy, halogen (preferably Cl), CR 3 , NH 2 , NHR or NRR′.
  • R 3 of CR 3 is (halo) 3 , preferably Cl 3 .
  • R and R′ of NHR and NRR′ are independently alkyl or acyl. Particularly preferred is where R 3 is alkyl or NH 2 .
  • R 4 , R 5 and R 6 are independently H, OH, alkyl, alkenyl, alkynyl, alkoxy, (CH 2 ) n —OR, halogen (preferably Cl, F or Br), CR 3 , acyl, heterocycle, N + ( ⁇ O)O ⁇ , C—N, N 3 , SH, SR, S( ⁇ O) 2 R, NH 2 , NHR or NRR′.
  • R of (CH 2 ) n —OR is H or alkyl, and n is typically 1-10, with CH 2 —OH and (CH 2 ) 2 —OH preferred.
  • R 3 of CR 3 is (halo) 3 , preferably Cl 3 , F 3 or Br 3 .
  • a preferred heterocycle is oxazol.
  • a preferred acyl is phenone (so forms benzophenone when taken with the benzene ring to which it is attached) or ester, such as an amino acid ester derivative.
  • R of SR and S( ⁇ O) 2 R is alkyl.
  • R and R′ of NHR and NRR′ are independently alkyl. Particularly preferred is where R 4 or R 5 or R 6 is Cl, Br, (CH 2 ) 2 —OH, N + ( ⁇ O)O ⁇ , C ⁇ N, or C( ⁇ O)R wherein R is alkyl or alkoxy. Also preferred is where R 4 or R 5 or R 6 is a non-polar substituent, e.g., alkyl.
  • R 4 and R 5 may be taken together with the benzene ring to form a heterocycle.
  • a preferred heterocycle is indazolyl, benzotriazolyl, indolyl, benzothiazolyl, benzimidazolyl or benzodioxolyl. Particularly preferred is where R 4 and R 5 (or R 5 and R 6 ) are taken together with the benzene ring to form indazole.
  • solubility and/or bioavailability of one or more of the compounds of the present invention may be accomplished, for example, by the addition of one or more substituents to the compound.
  • substituents such as hydroxyl groups
  • Other substituents for enhancing solubility and/or bioavailability include amino acids (e.g., polyglutamate or polylysine), di-peptides, polymers (e.g., PEG or POG), monocarboxylic acids (e.g., hemi-succinate), and esters. Any group that enhances solubility and/or bioavailability of a compound of the present invention may be used, provided that the group does not significantly impair the relevant biological property of the compound.
  • a compound or physiologically acceptable salt thereof
  • the term “compound” encompasses a prodrug form of the parent compound.
  • “Prodrug” herein refers to a chemical substance that is converted into the parent compound in vivo. Prodrugs often are useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent compound.
  • prodrug a parent compound of the present invention which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility.
  • the ester is then metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water solubility is beneficial.
  • Such a prodrug is generally inactive (or less active) until converted to the active form.
  • compositions of the compounds and the physiologically acceptable salts thereof are preferred embodiments of this invention.
  • Pharmaceutical compositions of the compounds of the present invention i.e., compounds and salts thereof as described above
  • compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers or diluents. Proper formulation is generally dependent upon the route of administration chosen.
  • the pyridines of the present invention may be formulated such that the formulation comprises a single pyridine or a mixture of two or more pyridines described herein.
  • one or more pyridines may be formulated with one or more other agents which are active for a general or specific disease, disorder or condition.
  • the compounds of the invention may be formulated as sterile aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with physiologically acceptable carriers well known in the art.
  • physiologically acceptable carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be made with the use of a solid carrier or diluent, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable carriers or diluents are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with a filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the embodiments of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro
  • the compounds may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include sterile aqueous solutions of the active compounds in water soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation (see, for example, U.S. Pat. No. 5,702,717 for a biodegradable depot for the delivery of a drug).
  • Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • the pharmaceutical compositions herein also may comprise suitable solid or gel phase carriers or diluents. Examples of such carriers or diluents include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • the compounds of the invention may be provided as physiologically acceptable salts wherein the claimed compound may form the negatively or the positively charged species.
  • salts in which the compound forms the positively charged moiety include, without limitation, quaternary ammonium (defined elsewhere herein), salts such as the hydrochloride, sulfate, carbonate, lactate, tartarate, maleate, succinate, etc. formed by the reaction of an amino group with the appropriate acid.
  • LPAAT- ⁇ appears to play a role in various cellular pathways that have a connection to various diseases, disorders or conditions.
  • the disclosure of the present invention shows unexpectedly that the pyridines set forth above inhibit the activity of LPAAT- ⁇ . This surprising inhibition is also specific for LPAAT- ⁇ , as the compounds tested showed weak to no inhibitory activity for LPAAT- ⁇ . In particular, none of the compounds tested had an IC 50 of less than 40 ⁇ M for LPAAT- ⁇ . In one use of the compounds of the present invention, the activity of LPAAT- ⁇ is reduced.
  • the method comprises contacting LPAAT- ⁇ with a compound or salt thereof or composition of the present invention in an amount effective to reduce the LPAAT- ⁇ activity.
  • the LPAAT- ⁇ to be contacted may reside in a cell-free preparation or in intact cells, including cells within an animal.
  • animal refers to any animal, including humans and other primates, rodents (e.g., mice, rats, and guinea pigs), lagamorphs (e.g., rabbits), bovines (e.g., cattle), ovines (e.g., sheep), caprines (e.g., goats), porcines (e.g., swine), equines (e.g., horses), canines (e.g., dogs), felines (e.g., cats), domestic fowl (e.g., chickens, turkeys, ducks, geese, other gallinaceous birds, etc), as well as feral or wild animals, including such animals as ungulates (e.g., deer), bear, fish, lagamorphs, rodents, birds, etc.
  • rodents e.g., mice, rats, and guinea pigs
  • lagamorphs e.g., rabbits
  • bovines e.g
  • the proliferation of a cell is inhibited.
  • the method comprises contacting the cell with a compound or salt thereof or composition of the present invention in an amount effective to inhibit the proliferation of the cell.
  • the cell to be contacted may be in vitro or in vivo in an animal.
  • An example of a cell whose proliferation it is desirable to inhibit is a tumor cell.
  • the term “inhibiting” refers to both total inhibition and partial inhibition (i.e., the inhibition need not be 100%).
  • a cancer in which LPAAT activity is associated
  • the method comprises administering to an animal in need, a compound or salt thereof or composition of the present invention in an amount effective to treat the cancer.
  • the term “treating a cancer” refers to any of a variety of positive effects from the treatment, including preventing the spread of a tumor, arresting tumor growth at a primary site, eradicating the tumor, relieving a symptom associated with the cancer, or prolonging the survival time of the animal treated.
  • treating a cancer may have the effect of (1) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth, (4) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the cancer, and/or (5) prolonging the survival time of the recipient.
  • treatment further includes preventing tumor occurrence or recurrence.
  • the method may further comprise inclusion of one or more other agents for treating a cancer.
  • the method may be used in conjunction with one or more other cancer therapies, such as radiation, surgery or other chemotherapy.
  • Suitable routes of administration may include, without limitation, oral, rectal, transmucosal or intestinal administration or intramuscular, subcutaneous, intramedullary, intrathecal, direct intraventricular, intravenous, intraperitoneal or intranasal injections.
  • a targeted drug delivery system for example, in a liposome coated with tumor-specific antibody.
  • the liposomes will be targeted to and taken up selectively by the tumor.
  • compositions suitable for use in the methods of the present invention are compounds and compositions wherein the active ingredients are contained in an amount effective to achieve its intended purpose. Determination of an effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the effective amount or dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of LPAAT- ⁇ activity). Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD 50 and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain LPAAT- ⁇ inhibitory effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90% inhibition of LPAAT- ⁇ using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • the amount of compound or composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • An exemplary systemic daily dosage is about 5 to about 200 mg/kg of body weight. Normally, from about 10 to about 100 mg/kg of body weight of the compounds of the present invention, in one or more dosages per day, is effective to obtain the desired results.
  • One of ordinary skill in the art can determine the optimal dosages and concentrations of the compounds of the preferred embodiments of the present invention with only routine experimentation.
  • the compounds of the present invention when used are substantially pure and preferably sterile.
  • the phrase “substantially pure” encompasses compounds created by chemical synthesis or compounds substantially free of chemicals which may accompany the compounds in the natural state, as evidenced by thin layer chromatography (TLC) or high performance liquid chromatography (HPLC).
  • a compound or salt thereof of the present invention, or pharmaceutical composition of either, may be used to coat a medical device.
  • a variety of medical devices such as a stent, may be coated.
  • the medical device may be composed of a bioadsorbable and biodegradable material. Due to the anti-proliferative properties of the compounds of the present invention, a stent or other medical device that is coated with such a compound or salt thereof or pharmaceutical composition of either may be used for inhibiting the proliferation of a cell.
  • the coated medical devices of the present invention may be used in a variety of ways. A preferred use is to inhibit the proliferation of tumor cells.
  • N-[6-(5-chloro-2-methoxy-phenyl)-4-p-tolylamino-pyridin-2-yl]-acetamide (15 mg, 0.039) prepared according to Example 5 in ethanol (4 ml) was added hydrazine (0.01 ml). After heating under reflux for 3 hours, the mixture was concentrated under reduced pressure. The residue was treated with water (40 ml) and extracted with ethyl acetate (50 ml). The organic extract was dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (6 mg, 45% yield).
  • Chelidamic acid monohydrate (1.0 g, 5.0 mmol) was treated with pyridine (100 ml) and concentrated under vacuum and then treated with toluene (100 ml) and concentrated under vacuum. The residue was treated with dichloromethane (30 ml), N,N-dimethylformamide (0.3 ml) and a solution of oxalyl chloride in dichloromethane (2 M, 13.7 ml). After stirring for 1 hour, volatiles were evaporated under reduced pressure to give crude 4-chloro-pyridine-2,6-dicarbonyl dichloride.
  • Example 12 To a solution of the title compound provided by Example 12 (25 mg, 0.068 mmol) in tetrahydofuran (5 ml) and methanol (10 ml) was added sodium borohydride (20 mg, 0.53 mmol). After stirring for 1.5 hours, the mixture was treated with water (10 ml) and hydrochloric acid (1 M, 0.6 ml). The mixture was concentrated to a volume of 8 ml and the solid was collected by filtration. The solid was treated with aqueous sodium carbonate solution (10%, 10 ml) and extracted with ethyl acetate (3 ⁇ 30 ml). The combined extracts were dried over sodium sulfate and concentrated under reduced pressure.
  • the full-length human LPAAT- ⁇ cDNA was amplified by PCR from the DNA template pCE9.LPAAT- ⁇ (West et al., DNA Cell Biol. 16:691-701 (1997)) using the primers 5′-TGATATCCGA AGAAGATCTT ATGGAGCTGT GGCCGTGTC-3′ (olpb1F; SEQ ID NO:1) and 5′-CAGGCTCTAG ACTACTGGGC CGGCTGCAC-3′ (olpb1R; SEQ ID NO:2).
  • the ⁇ 870 bp fragment generated was reamplified by PCR using the primers 5′ CCTACGTCG ACATGGAACA AAAATTGATA TCCGAAGAAG ATC-3′ (olpb2F; SEQ ID NO:3) and 5′-CAGGCTCTAG ACTACTGGGC CGGCTGCAC-3′ (olpb1R; SEQ ID NO:2).
  • the ⁇ 890 bp fragment generated was then cleaved with Sal I and Xba I for insertion into pFastBacTM HTc vector (Life Technologies, Gaithersberg, Md.) between the Sal I and Xba I sites for the generation of the plasmid pFB.LPAAT- ⁇ .
  • This plasmid was then transformed into E. coli DH10BacTM (Life Technologies, Gaithersberg, Md.) for the generation of recombinant Bacmid DNA for transfection into HighFive (Invitrogen, San Diego, Calif.) or SF9 insect cells for the production of recombinant Baculovirus stocks using the protocol described in the Bac-to-Bac® Baculovirus Expression System (Life Technologies, Gaithersberg, Md.), a eukaryotic expression system for generating recombinant baculovirus through site-specific transposition in E. coli .
  • Viral stocks harvested from the transfected cells can then be used to infect fresh insect cells for the subsequent expression of LPAAT- ⁇ fusion protein with a poly-histidine tag and a mycepitope near its N-terminus.
  • the membrane fraction from these Sf9 cells would be the source of LPAAT enzyme.
  • Sf9 cell pellets ( ⁇ 10 8 cells) were thawed and resuspended in 1-2 ml of buffer A (20 mM Hepes, pH 7.5, 1 mM DTT, 1 mM EDTA, 20% w/v glycerol, 1 mM Benzamidine, 1 ⁇ g/ml soybean trypsin inhibitor (SBTI), 1 ⁇ g/ml pepstatin A) w/o DTT but with 1 mM Pefabloc.
  • LPAAT- ⁇ catalyzes the transfer of an acyl group from a donor such as acyl-CoA to LPA.
  • the transfer of the acyl group from acyl-CoA to LPA leads to the release of free CoA, which can be reacted with the thiol reagent, 5,5 ′-dithiobis(2-nitrobenzoic acid) (DTNB).
  • DTNB 5,5 ′-dithiobis(2-nitrobenzoic acid)
  • the reaction between DTNB and the free sulfhydryl group from CoA generates a yellow-colored product, 3-carboxylato-4-nitrothiophenolate (CNP), that absorbs at 413 nm.
  • CNP 3-carboxylato-4-nitrothiophenolate
  • LPAAT- ⁇ derived from Sf9 cell membrane overexpressing LPAAT- ⁇ were resuspended in HEPES saline buffer (20 mM HEPES pH 7.5, 150 mM NaCl), 1 mg/ml BSA and 72 ⁇ l aliquots were distributed into 96-well microtiter plates. 8 ⁇ l of compound of interest at 200 ⁇ M dissolved in 100% DMSO was added into each well. 20 ⁇ l of 1 mM 18:1-CoA and 1 ⁇ M sn-1-18:1 lysoPA was then added to each well to initiate the reaction and allowed to run at room temperature for 25 min.
  • a secondary assay for LPAAT activity in cell extracts based on either the conversion of fluorescent NBD-LPA to NBD-PA (West, et al., DNA Cell Biol. 6:691-701, 1997) or [ 14 C]LPA to [ 14 C]PA using TLC analysis was used to screen compounds that showed >50% inhibition of LPAAT activity in the primary colorimetric assay.
  • the radiometric assay was carried out in Sf9 cell membrane overexpressing LPAAT- ⁇ resuspended in HEPES-saline buffer, pH 7.5, 1 mg/ml BSA, lmM EDTA and 200 ⁇ M [ 14 C]18:1-CoA and 200 ⁇ M sn-1-18:1 lysoPA. The samples were incubated 7 min at 37° C., extracted into organic solvent (CHCl 3 /CH 3 OH/HCl at 33/66/1), before loading onto TLC plates.
  • organic solvent CHCl 3 /CH 3 OH/HCl at 33/66/1
  • this LPAAT assay is a modification of the acyltransferase assay published previously (Hollenback and Glomset, Biochemistry 37:363-376 (1999)).
  • the basic assay in a total volume of 50 ⁇ l, employs a solution of substrates and the protein sample. Total assay volume, as well as the volume of each solution, can be changed to fit an experiment. In addition, other compounds, ex inhibitors and activators, can be included in the assay as well.
  • Appropriate amounts of each solution are added the to a 12 ⁇ 75 mm borosilicate glass test tube and dry the solvent under N 2 or Ar.
  • An appropriate volume of the solution prepared in 2a is added to the lysoPA and 14 C-labeled acyl-CoA.
  • the lipids are resuspended by sonication for 15 sec in a bath sonicator.
  • the resulting suspension is then incubated (with occasional gentle vortexing) for about 10 minutes at room temp.
  • the sn-1-16:0 lysoPA may require brief warming of the solvent to solubilize it.
  • concentration of lysoPA and 14 C-labeled acyl-CoA can vary, but typically the final lysoPA concentration ranges between 0 and 400 ⁇ M and the 14 C-labeled acyl-CoA specific activity ranges between 0.5 and 2 Ci/mol.
  • Protein sample varies from experiment-to-experiment.
  • the assay is performed by mixing the components in 12 ⁇ 75 mm borosilicate glass test tubes (the order of addition does not matter unless indicated) and incubating at 37° C. for 5 to 10 minutes such that the assay within the linear range for time and protein.
  • the sample is centrifuged for 3 minutes at 1000 ⁇ g, the upper (aqueous/methanol) phase is aspirated and the lower phase is dried under nitrogen.
  • cpm standard curve To be able to convert pixel volume (determined by the Storm phosphor imager, see step 8b) into cpm, cpm standard curve must be generated on the plate. 14 C-labeled oleate dilutions in chloroform are made for this purpose. Four stocks (50 cpm/ ⁇ l to 800 cpm/ ⁇ l) are made and 2 ⁇ l of a different concentration are spotted in each corner of the plate (where previously there was no radioactivity).
  • the plates are stained with primuline and scanned with the Storm (blue chemilluminescence mode).
  • PA and lysoPA bands are easily detected in this system because of the carrier added in step 5.
  • PA and lysoPA have respective Rf's of about 0.63 and 0.21.
  • the plates are then wrapped in saran wrap and exposed to a freshly phosphor screen overnight (longer exposures can also be done to increase the

Abstract

The invention relates to pyridines and uses thereof, including to inhibit lysophosphatidic acid acyltransferase β (LPAAT-β) activity and/or proliferation of cells such as tumor cells.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 11/056,777 filed Feb. 11, 2005 and allowed; which application is a continuation of U.S. patent application Ser. No. 10/667,916 filed Sep. 22, 2003 and issued as U.S. Pat. No. 6,875,781; which application claims the benefit of U.S. Provisional Application No. 60/460,782 filed Apr. 04, 2003, which applications are incorporated by reference herein in their entirety.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The invention is in the field of organic and medicinal chemistry. In particular, the invention relates to pyridines and uses thereof, such as.inhibiting the activity of lysophosphatidic acid acyltransferase β (LPAAT-β) activity and/or inhibiting the proliferation of a cell (e.g., tumor cell).
  • 2. Description of the Related Art
  • Lysophosphatidic acid acyltransferase (LPAAT) catalyzes the acylation of lysophosphatidic acid (LPA) to phosphatidic acid. LPA is the simplest glycerophospholipid, consisting of a glycerol molecule, a phosphate group, and a fatty acyl chain. LPAAT adds a second fatty acyl chain to LPA, producing phosphatidic acid (PA). PA is the precursor molecule for certain phosphoglycerides, such as phosphatidylinositol, and diacylglycerols, which are necessary for the production of other phosphoglycerides, such as phosphatidylcholine, and for triacylglycerols, which are essential biological fuel molecules.
  • In addition to being a crucial precursor molecule in biosynthetic reactions, LPA has been added to the list of intercellular lipid messenger molecules. LPA interacts with G protein-coupled receptors, coupling to various independent effector pathways including inhibition of adenylate cyclase, stimulation of phospholipase C, activation of MAP kinases, and activation of the small GTP-binding proteins Ras and Rho. Moolenaar, J. Biol. Chem. 28:1294 (1995). The physiological effects of LPA have not been fully characterized as yet. However, one of the physiological effects that is known is that LPA promotes the growth and invasion of tumor cells. It has been shown that the addition of LPA to ovarian or breast cancer cell lines induces cell proliferation, increases intracellular calcium levels, and activates MAP kinase. Xu et al., Biochem. J. 309:933 (1995). In addition, LPA has been shown to induce MM1 tumor cells to invade cultured mesothelial cell monolayers. Imamura et al., Biochem. Biophys. Res. Comm. 193:497 (1993).
  • Like LPA, PA is also a messenger molecule. PA is a key messenger in a common signaling pathway activated by proinflammatory mediators such as interleukin-1β, tumor necrosis factor α, platelet activating factor, and lipid A. Bursten et al., Am. J. Physiol. 262:C328 (1992); Bursten et al., J. Biol. Chem. 255:20732 (1991); Kester, J. Cell Physiol. 156:317 (1993). PA has been implicated in mitogenesis of several cell lines [English, Cell Signal 8:341 (1996)]. PA level has been found to be increased in either ras or fps transformed cell lines compared to the parental Rat2 fibroblast cell line [Martin et al., Oncogene 14:1571 (1997)]. Activation of Raf-1, an essential component of the MAPK signaling cascade, by extracellular signals is initiated by association with intracellular membranes. Recruitment of Raf-1 to membranes has been reported to be mediated by direct association with phosphatidic acid [Rizzo et al., J. Biol. Chem. 275:23911-8 (2000)]. Thus, LPAAT, as an enzyme that regulates PA content in cells, may play a role in cancer, and may also mediate inflammatory responses to various proinflammatory agents.
  • LPAAT exists in a LPAAT-α form and a LPAAT-β form. Northern blot analysis shows that LPAAT-α is expressed in all human tissues tested with the highest expression level found in skeletal muscle (West et al., DNA Cell Biol. 16:691 (1997)). The uniformity of LPAAT-α expression has also been found in additional tissues such as prostate, testis, ovary, small intestine, and colon (Stamps et al., Biochem. J. 326:455 (1997)) as well as in mouse tissues (Kume et al., Biochem. Biophys. Res. Commun. 237:663 (1997)). A 2 kb and a 1.3 kb forms, possibly due to alternative utilization of polyadenylation signals at the 3′-UTR, have been found in murine LPAAT-α mRNA (Kume et al., Biochem. Biophys. Res. Commun 237:663 (1997)), whereas only one major human LPAAT-α mRNA of 2 kb in size has been detected by Northern analysis. West et al., DNA Cell Biol. 16:691 (1997); Stamps et al., Biochem. J. 326:455 (1997).
  • In contrast, LPAAT-β demonstrates a distinct tissue distribution of mRNA expression. West et al., DNA Cell Biol. 16:691 (1997). LPAAT-β is most highly expressed in liver and heart tissues. LPAAT-β is also expressed at moderate levels in pancreas, lung, skeletal muscle, kidney, spleen, and bone marrow; and at low levels in thymus, brain and placenta. This differential pattern of LPAAT-β expression has been confirmed independently (Eberhardt et al., J. Biol. Chem. 272:20299 (1997)) with the only discrepancy being that high level, instead of moderate level, of LPAAT-β has been detected in pancreas, possibly due to slight lot variations in commercial RNA blots (Clontech, Palo Alto, Calif.). In addition, moderate LPAAT-β expression has been found in prostate, testis, ovary, small intestine, and colon with the small intestine containing relatively higher amounts. Eberhardt et al., J. Biol. Chem. 272:20299 (1997). Within various brain sections, high expression has been found in the subthalamic nucleus and spinal cord; and least in the cerebellum, caudate nucleus, corpus callosum, and hippocampus. LPAAT-β can also be detected in myeloid cell lines THP-1, HL-60, and U937 with the mRNA levels remaining the same with or without phorbal-ester treatment. The size difference between human LPAAT-α and LPAAT-β mRNA is consistent with the sequence data, in which LPAAT-α has a longer 3′-UTR. The differential tissue expression pattern LPAAT-α and LPAAT-β mRNA would suggest these two genes are regulated differently and are likely to have independent functions. Therefore, a desirable feature in compounds that inhibit LPAAT activity is that they are specific in inhibiting one isoform of the enzyme over the other (i.e., LPAAT-β over LPAAT-α).
  • LPAAT-β mRNA has been found to be elevated in tumor tissues (e.g., uterus, fallopian tube, and ovary), as compared to its expression in the corresponding normal tissues. However, no significant difference was found in LPAAT-α mRNA level between the various tumor tissues and the normal adjacent tissues. In two of the tumor tissues (fallopian tube and ovary) where LPAAT-α mRNA was elevated, PAP2-α mRNA expression was found to be suppressed, as it was also in tumors of the colon, rectum, and breast. Thus, LPAAT-β (rather than LPAAT-α) appears to be a relevant target for inhibition.
  • There is a need in the art for improved compositions and methods. The present invention fills this need, and further provides other related advantages.
  • BRIEF SUMMARY OF THE INVENTION
  • Briefly stated, the present invention provides a variety of compounds and uses thereof. More specifically, the compounds of the present invention are pyridines that possess aromatic substituents which are directly or indirectly attached to two non-adjacent carbons of the pyridine ring. The compounds are generally of the formula:
    Figure US20070178134A1-20070802-C00001

    where R1—R6 are hydrogen or non-hydrogen substituents, Q is a heteroatom or heteroatom attached to one or more methylene groups, and one of X, Y and Z is N with the other two being CH or a substituted C. In preferred embodiments:
  • X, Y and Z are N, CH or CR where R is alkyl, alkoxy, Cl, Br, NH2, NHR′ or NR′R″ where R′ and R″ independently are alkyl;
  • Q is NR, RN—(CH2)n, (CH2)n,—NR, O, O—(CH2)n, (CH2)n—O, S, S—(CH2)n or (CH2)n—S, where n is 1-10 and R is H or alkyl;
  • R1 is H, OH, alkyl, alkoxy, Cl, F, Br, CR3 where R3 is Cl3, F3 or Br3, NH2, NHR or NRR′ where R and R′ independently are alkyl;
  • R2 is H, OH, alkyl, alkoxy, Cl, F, Br or CR3 where R3 is Cl3, F3 or Br3;
  • R3 is H, alkyl, alkoxy, Cl, CCl3, NH2, NHR or NRR′ where R and R′ independently are alkyl or acyl;
  • R4, R5 and R6 are independently H, OH, alkyl, alkenyl, alkynyl, alkoxy, (CH2)n—OR where R is H or alkyl and n is 1-10, Cl, F, Br, CR3 where R3 is Cl3, F3 or Br3, acyl, heterocycle, N+(═O)O, C≡N, N3, SH, SR or S(═O)2R where R is alkyl, NH2, NHR or NRR′ where R and R′ independently are alkyl, or R4 and R5 or R5 and R6 are taken together with the benzene ring to form a heterocycle;
  • and with the proviso that one of X, Y and Z is N.
  • A compound or salt thereof as described above may be combined with a pharmaceutical carrier or diluent to form a pharmaceutical composition of the present invention.
  • A compound, salt thereof or pharmaceutical composition of the present invention may be used in one or more methods. In one method, the activity of LPAAT-β may be reduced by the step comprising contacting LPAAT-β with a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to reduce LPAAT-β activity. In another method, the proliferation of a cell in which the activity of LPAAT-β is required for the proliferation of the cell may be inhibited by the step comprising contacting LPAAT-β with a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to inhibit the proliferation of the cell. In a further method, the treatment of a cancer in which LPAAT-β activity is associated may be effected by the step comprising administering to an animal in need a compound, salt thereof or pharmaceutical composition of the present invention in an amount effective to treat the cancer.
  • Also provided is a coated medical device for inhibiting the proliferation of a cell in which the activity of LPAAT-β is required for the proliferation of the cell comprising a medical device coated with a compound, salt thereof or pharmaceutical composition of the present invention.
  • These and other aspects of the present invention will become evident upon reference to the following detailed description.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Prior to setting forth the invention, it may be helpful to an understanding thereof to set forth definitions of certain terms to be used hereinafter.
  • In the present description, the term “alkyl” refers to straight- or branched-chain hydrocarbons having from 1 to 10 carbon atoms and more preferably 1 to 8 carbon atoms which include, by way of example, methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl and the like. The alkyl group may be substituted or unsubstituted. When substituted, the substituted group(s) is preferably one or more independently selected from alkyl, cycloalkyl, heteroalicyclic, aryl, heteroaryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, substituted imino and substituted amino.
  • “Alkenyl” includes monovalent hydrocarbon radicals having straight, cyclic, or branched moieties, and combinations thereof which comprise at least one carbon-carbon double bond. The alkenyl group may be substituted or unsubstituted. When substituted, the substituted group(s) is preferably one or more independently selected from alkyl, acyl, cycloalkyl, heteroalicyclic, aryl, haloalkyl, alkoxy and substituted amino. “Alkoxy” refers to the group “—O-alkyl” which includes, by way of example, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, t-butoxy and the like. It further refers to the group “—O-alkyl-W-alkyl” where W is O or N; for example, —O—(CH2)n—W—(CH2)m where n and m are independently 1-10.
  • “Substituted amino” denotes the group —NRR, wherein each R group is independently selected from hydrogen, acyl, alkyl, cycloalkyl, aryl, or the R groups can be joined together with the nitrogen to form a heterocyclic ring (e.g., piperidine, piperazine, or a morpholine ring).
  • “Substituted imino” denotes the group ═NR, wherein R is preferably selected from hydrogen, hydroxy, alkyl and acyl.
  • “Aryl” refers to an unsaturated aromatic carbocyclic group of 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl). The aryl group may be unsubstituted or substituted; in the latter case, the substituent or substituents preferably are selected independently from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, nitro, and substituted amino.
  • “Heterocycle” includes “heteroaryl” and “heteroalicyclic”. Examples of heterocycles include oxazole, piperidine, piperazine and morpholine.
  • “Heteroaryl” is a monocyclic or fused ring (i.e., rings which share an adjacent pair of atoms) group having in the ring(s) one or more atoms selected preferably from nitrogen, oxygen and sulfur and, in addition, having a completely conjugated π-electron system. Exemplary heteroaryl groups are pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrimidine, quinoline, isoquinoline, purine and carbazole. The heteroaryl group may be substituted or unsubstituted. When substituted, the substituted group(s) is preferably one or more independently selected from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, nitro and substituted amino.
  • “Cycloalkyl” encompasses cyclic alkyl groups that contain between 3 and 8 carbon atoms and have a single cyclic ring, illustrated by cyclopropyl, cyclobutyl, cyclopentyl, and cyclooctyl. The cycloalkyl ring may be substituted or unsubstituted. Again, a substituted cycloalkyl ring carries one or more substituent groups, independently selected preferably from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, vitro, and substituted amino.
  • “Heteroalicyclic” refers to a monocyclic or fused ring group having in the ring(s) one or more atoms selected preferably from nitrogen, oxygen and sulfur. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated π-electron system. The heteroalicyclic ring may be substituted or unsubstituted. When substituted, the substituted group(s) preferably are selected independently from alkyl, aryl, haloalkyl, halo, hydroxy, alkoxy, mercapto, cyano, sulfonamidyl, aminosulfonyl, acyl, acyloxy, vitro, and substituted amino.
  • “Halogen” or “halo” refers to fluoro, chloro, bromo, iodo.
  • “Acyl” group refers to the C(O)—R″ group, where R″ is selected preferably from hydrogen, hydroxy, alkyl, haloalkyl, cycloalkyl, aryl optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups, heteroaryl (bonded through a ring carbon) optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups and heteroalicyclic (bonded through a ring carbon) optionally substituted with one or more alkyl, haloalkyl, alkoxy, halo and substituted amino groups. Acyl groups include aldehydes, ketones, acids, acid halides, esters and amides. Preferred acyl groups are carboxy groups, e.g., acids and esters. Esters include amino acid ester derivatives. The acyl group may be attached to a compound's backbone at either end of the acyl group, i.e., via the C or the R″. Where the acyl group is attached via the R″, then C will bear another substituent, such as hydrogen or alkyl.
  • The phrase “physiologically acceptable salt” refers to those salts that retain the biological effectiveness and properties of the particular compound. Physiologically acceptable salts are often useful because they may have improved stability and/or solubility in pharmaceutical compositions over the free base form of the compound. A physiologically acceptable salt may be obtained by reaction of a free base with an inorganic acid such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, sulfuric acid, and perchloric acid and the like, or with an organic acid such as acetic acid, oxalic acid, malic acid, maleic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, succinic acid or malonic acid, and the like. A physiologically acceptable salt may also be obtained by reaction of a free acid with a base such as sodium, potassium or lithium hydroxide, bicarbonate or carbonate, and the like.
  • As noted above, the present invention provides pyridines, physiologically acceptable salts thereof and uses thereof. The pyridines possess aromatic substituents that are directly or indirectly attached to two non-adjacent carbons of the pyridine ring. The compounds are generally of the formula:
    Figure US20070178134A1-20070802-C00002

    where R1—R6 are hydrogen or non-hydrogen substituents, Q is a heteroatom or heteroatom attached to one or more methylene groups, and two of X, Y and Z are N with the third being CH or a substituted C. The requirement that only one of X, Y and Z is N is consistent with the compounds including a pyridine ring.
  • Preferred embodiments include the following selections for the general formula above. Preferred embodiments include where X, Y and Z are N, CH or CR. R of CR is alkyl, alkoxy, halo (preferably Cl or Br), NH2, NHR′ or NR′R″ where R′ and R″ independently are alkyl. Particularly preferred is where X or Y is N.
  • Preferred embodiments include where Q is a heteroatom (preferably N, O or S) and may be attached to one or more methylene groups to provide additional spacing between the pyridine ring and the phenyl ring possessing R4, R5 and/or R6. Q may be NR where R is H or alkyl. Where there are one or more methylene groups, the heteroatom may be oriented such that it is attached directly to the pyridine ring or attached directly to the phenyl ring possessing R4, R5 and/or R6. For example, Q may be RN—(CH2)n, (CH2)n—NR, O—(CH2)n, (CH2)n—O, S—(CH2)n or (CH2)n—S, where n is typically 1-10 and R is H or alkyl. Particularly preferred is where Q is NH.
  • Preferred embodiments include where R1 is H, OH, alkyl, alkoxy, halogen (preferably Cl, F or Br), CR3, NH2, NHR or NRR′. R3 of CR3 is (halo)3, preferably Cl3, F3 or Br3. R and R′ of NHR and NRR′ are independently alkyl. The term “independently,” as used throughout, refers to independent selection of a group, but does not exclude the possibility that two groups are identical. For example, the alkyl group of R and R′ of NRR′ may be the same or different. Particularly preferred is where R1 is alkyl, alkoxy or Cl.
  • Preferred embodiments include where R2 is H, OH, alkyl, alkoxy, halogen (preferably Cl, F or Br), or CR3. R3 of CR3 is (halo)3, preferably Cl3, F3 or Br3. Particularly preferred is where R2 is Cl or Br.
  • Preferred embodiments include where R3 is H, alkyl, alkoxy, halogen (preferably Cl), CR3, NH2, NHR or NRR′. R3 of CR3 is (halo)3, preferably Cl3. R and R′ of NHR and NRR′ are independently alkyl or acyl. Particularly preferred is where R3 is alkyl or NH2.
  • Preferred embodiments include where R4, R5 and R6 are independently H, OH, alkyl, alkenyl, alkynyl, alkoxy, (CH2)n—OR, halogen (preferably Cl, F or Br), CR3, acyl, heterocycle, N+(═O)O, C—N, N3, SH, SR, S(═O)2R, NH2, NHR or NRR′. R of (CH2)n—OR is H or alkyl, and n is typically 1-10, with CH2—OH and (CH2)2—OH preferred. R3 of CR3 is (halo)3, preferably Cl3, F3 or Br3. A preferred heterocycle is oxazol. A preferred acyl is phenone (so forms benzophenone when taken with the benzene ring to which it is attached) or ester, such as an amino acid ester derivative. R of SR and S(═O)2R is alkyl. R and R′ of NHR and NRR′ are independently alkyl. Particularly preferred is where R4 or R5 or R6 is Cl, Br, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy. Also preferred is where R4 or R5 or R6 is a non-polar substituent, e.g., alkyl. Alternatively, R4 and R5 (or R5 and R6) may be taken together with the benzene ring to form a heterocycle. A preferred heterocycle is indazolyl, benzotriazolyl, indolyl, benzothiazolyl, benzimidazolyl or benzodioxolyl. Particularly preferred is where R4 and R5 (or R5 and R6) are taken together with the benzene ring to form indazole.
  • Particularly preferred compounds of the present invention are shown in Table 1 of Example 15 below, and physiologically acceptable salts thereof.
  • It may be advantageous for certain uses to enhance the solubility and/or bioavailability of one or more of the compounds of the present invention. This may be accomplished, for example, by the addition of one or more substituents to the compound. For example, the addition of hydrophilic groups, such as hydroxyl groups, may be advantageous. Other substituents for enhancing solubility and/or bioavailability include amino acids (e.g., polyglutamate or polylysine), di-peptides, polymers (e.g., PEG or POG), monocarboxylic acids (e.g., hemi-succinate), and esters. Any group that enhances solubility and/or bioavailability of a compound of the present invention may be used, provided that the group does not significantly impair the relevant biological property of the compound.
  • It may be advantageous for certain uses to prepare a compound (or physiologically acceptable salt thereof) as a “prodrug.” As used herein, the term “compound” encompasses a prodrug form of the parent compound. “Prodrug” herein refers to a chemical substance that is converted into the parent compound in vivo. Prodrugs often are useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent compound. An example of a prodrug would be a parent compound of the present invention which is administered as an ester (the “prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility. The ester is then metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water solubility is beneficial. Such a prodrug is generally inactive (or less active) until converted to the active form.
  • Pharmaceutical compositions of the compounds and the physiologically acceptable salts thereof are preferred embodiments of this invention. Pharmaceutical compositions of the compounds of the present invention (i.e., compounds and salts thereof as described above) may be manufactured by processes well known in the art; e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions may be formulated in a conventional manner using one or more physiologically acceptable carriers or diluents. Proper formulation is generally dependent upon the route of administration chosen. The pyridines of the present invention may be formulated such that the formulation comprises a single pyridine or a mixture of two or more pyridines described herein. Alternatively, one or more pyridines may be formulated with one or more other agents which are active for a general or specific disease, disorder or condition.
  • For injection, the compounds of the invention may be formulated as sterile aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • For oral administration, the compounds can be formulated readily by combining the active compounds with physiologically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be made with the use of a solid carrier or diluent, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable carriers or diluents are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Pharmaceutical compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with a filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • For administration by inhalation, the compounds for use according to the embodiments of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • The compounds may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical compositions for parenteral administration include sterile aqueous solutions of the active compounds in water soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • In addition to the formulations described previously, the compounds may also be formulated as a depot preparation (see, for example, U.S. Pat. No. 5,702,717 for a biodegradable depot for the delivery of a drug). Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. The pharmaceutical compositions herein also may comprise suitable solid or gel phase carriers or diluents. Examples of such carriers or diluents include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • The compounds of the invention may be provided as physiologically acceptable salts wherein the claimed compound may form the negatively or the positively charged species. Examples of salts in which the compound forms the positively charged moiety include, without limitation, quaternary ammonium (defined elsewhere herein), salts such as the hydrochloride, sulfate, carbonate, lactate, tartarate, maleate, succinate, etc. formed by the reaction of an amino group with the appropriate acid.
  • As noted above, LPAAT-β appears to play a role in various cellular pathways that have a connection to various diseases, disorders or conditions. The disclosure of the present invention shows unexpectedly that the pyridines set forth above inhibit the activity of LPAAT-β. This surprising inhibition is also specific for LPAAT-β, as the compounds tested showed weak to no inhibitory activity for LPAAT-α. In particular, none of the compounds tested had an IC50 of less than 40 μM for LPAAT-α. In one use of the compounds of the present invention, the activity of LPAAT-β is reduced.
  • The method comprises contacting LPAAT-β with a compound or salt thereof or composition of the present invention in an amount effective to reduce the LPAAT-β activity. The LPAAT-β to be contacted may reside in a cell-free preparation or in intact cells, including cells within an animal.
  • In the context of the present invention, the term “animal” refers to any animal, including humans and other primates, rodents (e.g., mice, rats, and guinea pigs), lagamorphs (e.g., rabbits), bovines (e.g., cattle), ovines (e.g., sheep), caprines (e.g., goats), porcines (e.g., swine), equines (e.g., horses), canines (e.g., dogs), felines (e.g., cats), domestic fowl (e.g., chickens, turkeys, ducks, geese, other gallinaceous birds, etc), as well as feral or wild animals, including such animals as ungulates (e.g., deer), bear, fish, lagamorphs, rodents, birds, etc. It is not intended that the term be limited to a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are encompassed by the term. A preferred animal within the present invention is a mammal, with humans particularly preferred.
  • In another use of the compounds of the present invention, the proliferation of a cell (in which the activity of LPAAT-β is required for the proliferation of the cell) is inhibited. The method comprises contacting the cell with a compound or salt thereof or composition of the present invention in an amount effective to inhibit the proliferation of the cell. The cell to be contacted may be in vitro or in vivo in an animal. An example of a cell whose proliferation it is desirable to inhibit is a tumor cell. However, there are other diseases, disorders and conditions with cell types other than tumor cells for which it may be desirable to inhibit proliferation of the cell. In the context of the present invention, the term “inhibiting” refers to both total inhibition and partial inhibition (i.e., the inhibition need not be 100%).
  • In another use of the compounds of the present invention, a cancer (in which LPAAT activity is associated) is treated. The method comprises administering to an animal in need, a compound or salt thereof or composition of the present invention in an amount effective to treat the cancer. In the context.of the present invention, the term “treating a cancer” refers to any of a variety of positive effects from the treatment, including preventing the spread of a tumor, arresting tumor growth at a primary site, eradicating the tumor, relieving a symptom associated with the cancer, or prolonging the survival time of the animal treated. For example, as used herein, treating a cancer may have the effect of (1) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth, (4) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the cancer, and/or (5) prolonging the survival time of the recipient. In addition, treatment further includes preventing tumor occurrence or recurrence. The method may further comprise inclusion of one or more other agents for treating a cancer. Alternatively, the method may be used in conjunction with one or more other cancer therapies, such as radiation, surgery or other chemotherapy.
  • Suitable routes of administration may include, without limitation, oral, rectal, transmucosal or intestinal administration or intramuscular, subcutaneous, intramedullary, intrathecal, direct intraventricular, intravenous, intraperitoneal or intranasal injections.
  • Alternately, one may administer the compound or composition in a local rather than systemic manner, for example, via injection of the compound or composition directly into a solid tumor, often in a depot or sustained release formulation.
  • Furthermore, one may administer the compound or composition in a targeted drug delivery system, for example, in a liposome coated with tumor-specific antibody. The liposomes will be targeted to and taken up selectively by the tumor.
  • Compounds and compositions suitable for use in the methods of the present invention are compounds and compositions wherein the active ingredients are contained in an amount effective to achieve its intended purpose. Determination of an effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • For any compound or composition used in the methods of the invention, the effective amount or dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of LPAAT-β activity). Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (see, e.g., Fingl, et al., in “The Pharmacological Basis of Therapeutics,” (1975), Chapter 1, pp. 1).
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain LPAAT-β inhibitory effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90% inhibition of LPAAT-β using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • The amount of compound or composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. An exemplary systemic daily dosage is about 5 to about 200 mg/kg of body weight. Normally, from about 10 to about 100 mg/kg of body weight of the compounds of the present invention, in one or more dosages per day, is effective to obtain the desired results. One of ordinary skill in the art can determine the optimal dosages and concentrations of the compounds of the preferred embodiments of the present invention with only routine experimentation.
  • The compounds of the present invention when used are substantially pure and preferably sterile. The phrase “substantially pure” encompasses compounds created by chemical synthesis or compounds substantially free of chemicals which may accompany the compounds in the natural state, as evidenced by thin layer chromatography (TLC) or high performance liquid chromatography (HPLC).
  • A compound or salt thereof of the present invention, or pharmaceutical composition of either, may be used to coat a medical device. A variety of medical devices, such as a stent, may be coated. The medical device may be composed of a bioadsorbable and biodegradable material. Due to the anti-proliferative properties of the compounds of the present invention, a stent or other medical device that is coated with such a compound or salt thereof or pharmaceutical composition of either may be used for inhibiting the proliferation of a cell. The coated medical devices of the present invention may be used in a variety of ways. A preferred use is to inhibit the proliferation of tumor cells.
  • The following examples are offered by way of illustration and not by way of limitation.
  • EXAMPLES Example 1 [12-Chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl]-p-tolyl-amine
  • To a solution of 4-amino-2,6-dichloropyridine (1.0 g, 6.1 mmol), 5-chloro-2-methoxy-phenylboronic acid (1.4 g, 7.3 mmol) and palladium acetate (137 mg, 0.61 mmol) in ethylene glycol dimethyl ether (50 ml), degassed with argon, was added a solution of cesium fluoride (3.8 g, 24.4 mmol) in water (10 ml) followed by triphenylphosphine (320 mg, 1.22 mmol). After stirring at 85° C. for 12 hours, the mixture was filtered through a pad of celite under suction. The filtrate was dried over sodium sulfate and purified by flash chromatography on silica gel eluting with ethyl acetate-hexane (1:4 followed by 2:5) to provide 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-amine (720 mg, 44% yield). 1H NMR (acetone-d6) δ 3.91 (s, 3H, CH3), 5.92 (s, 2H, NH2), 6.60 (d, 1H, J=1.8 Hz, Ar), 7.14 (d, 1H, J=8.8 Hz, Ar), 7.33 (d, 1H, J=1.9 Hz, Ar), 7.37 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.90 (d, 1H, J=2.8 Hz, Ar).
  • A mixture of 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-amine (715 mg, 2.65 mmol), p-tolylboronic acid (722 mg, 5.3 mmol), copper (II) acetate (962 mg, 5.3 mmol), triethylamine (1.5 ml, 10.6 mmol) and molecular sieves (4 angstrom) in dichloromethane (40 ml) was stirred under oxygen atmosphere for 60 hours. The mixture was filtered through a pad of celite under suction. The filtrate was purified by flash chromatography eluting with ethyl acetate-hexane (1:3 followed by 2:5) to provide the title compound (680 mg, 72% yield). 1H NMR (DMSO-d6) δ 2.32 (s, 3H, CH3), 3.91 (s, 3H, CH3), 6.73 (d, 1H, J=1.9 Hz, Ar), 7.14 (d, 2H, J=8.4 Hz, Ar), 7.18 (d, 1H, J=8.9 Hz, Ar), 7.22 (d, 2H, J=8.2 Hz, Ar), 7.44-7.48 (m, 2H, Ar), 7.75 (d, 1H, J=2.8 Hz, Ar), 9.1 (s, 1H, NH).
  • Example 2 [4-Cloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-2-yl]-p-tolyl-amine
  • To a mixture of 2,4,6-trichloropyridine (50 mg, 0.27 mmol), 5-chloro-2-methoxy-phenylboronic acid (54 mg, 0.28 mmol), cesium fluoride (100 mg, 0.66 mmol) in ethylene glycol dimethyl ether (10 ml), degassed with argon, was added tetrakis(triphenylphosphine)palladium(0) (30 mg, 0.026 mmol). The mixture was heated under reflux for 2 hours and filtered through a pad of celite under suction. The filtrate was concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography eluting with ethyl acetate-hexane (1:9) to provide 2,4-dichloro-6-(5-chloro-2-methoxy-phenyl)-pyridine (25 mg, 32% yield). 1H NMR (acetone-d6) δ 3.95 (s, 3H, CH3), 7.24 (d, 1H, J=8.9 Hz, Ar), 7.49 (dd, 1H, J=8.9 Hz, J=2.8 Hz, Ar), 7.56 (d, 1H, J=2.8 Hz, Ar), 7.94 (d, 1H, J=2.8 Hz, Ar), 8.10 (d, 1H, J=1.6 Hz, Ar).
  • To a mixture of 2,4-dichloro-6-(5-chloro-2-methoxy-phenyl)-pyridine (25 mg, 0.087 mmol), p-tolylamine (10.2 mg, 0.1 mmol), sodium tert-butoxide (26 mg, 0.26 mmol), and tetrahydrofuran (15 ml), degassed with argon, was added palladium acetate (15 mg, 0.066 mmol) followed by 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl (30 mg, 0.048 mmol). The mixture was stirred at 70° C. for 5 hours and filtered through a pad of celite under suction. The filtrate was concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography eluting with ethyl acetate-hexane (1:9) to provide the title compound (6.6 mg, 21% yield). 1H NMR (acetone-d6) δ 2.32 (s, 3H, CH3), 3.95 (s, 3H, CH3), 6.83-6.84 (m, 1H, Ar), 7.15 (d, 2H, J=8.2 Hz, Ar), 7.20 (d, 1H, J=8.9 Hz, Ar), 7.43 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.46 (d, 1H, J=1.5 Hz, Ar), 7.57-7.60 (m, 2H, Ar), 8.10 (d, 1H, J=2.7 Hz, Ar), 8.43 (s, 1H, NH).
  • Example 3 6-(5-Chloro-2-methoxy-phenyl)-N*2*-p-tolyl-pyridine-2,4-diamine
  • A mixture of 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-amine, prepared in Example 1, (120 mg, 0.45 mmol) and p-tolylamine (1.0 g, 9.3 mmol) was heated at 190° C. for 45 minutes. After cooling to room temperature, the mixture was treated with hydrochloric acid (1 M, 50 ml). The mixture was extracted with ethyl acetate (50 ml). The organic extract was dried over magnesium sulfate and purified by flash chromatography on silica gel column eluting with ethyl acetate-hexane (1:4 followed by 1:1) to give the title compound (120 mg, 79% yield). 1H NMR (acetone-d6) δ 2.27 (s, 3H, CH3), 3.89 (s, 3H, CH3), 5.30 (d, 2H, J=6.4 Hz, NH2), 6.11 (d, 1H, J=1.7 Hz, Ar), 6.89 (d, 1H, J=1.7 Hz, Ar), 7.07 (d, 2H, J=8.3 Hz, Ar), 7.10 (d, 1H, J=8.8 Hz, Ar), 7.32 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.51-7.54 (m, 2H, Ar), 7.68 (s, 1H, NH), 7.99 (d, 1H, J=2.8 Hz, Ar).
  • Example 4 6-(5-Chloro-2-methoxy-phenyl)-N*2*-(4-chloro-phenyl)-pyridine-2,4-diamine
  • Following the method described in Example 3, 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-amine and 4-chloro-phenylamine, provided the title compound (22% yield). 1H NMR (acetone-d6) δ 3.90 (s, 3H, CH3), 5.39 (s, 2H, NH2), 6.90 (d, 1H, J=1.7 Hz, Ar), 7.12 (d, 1H, J=8.8 Hz, Ar), 7.24 (d, 2H, J=8.9 Hz, Ar), 7.34 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.73 (d, 2H, J=8.9 Hz, Ar), 7.91 (d, 1H, J=2.8 Hz, Ar), 8.09 (s, 1H, NH).
  • Example 5 N-[6-(5-chloro-2-methoxy-phenyl)-4-p-tolylamino-pyridin-2-yl]-acetamide
  • A mixture of [2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl]-p-tolyl-amine (60 mg, 0.167 mmol) prepared according to Example 1, copper powder (100 mg, 1.54 mmol) and acetamide (2.0 g, 33.9 mmol) was heated at 180° C. for 3 hours. The mixture was purified by flash chromatography on silica gel eluting with ethyl acetate-hexane (1:1 followed by 3:2) to provide title compound (15 mg, 23% yield). 1H NMR (acetone-d6) δ 2.19 (s, 3H, CH3), 2.32 (s, 3H, CH3), 3.91 (s, 3H, CH3), 7.12 (d, 1H, J=8.8 Hz, Ar), 7.19-7.25 (m, 4H, Ar), 7.35 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.47-7.48 (m, 1H, Ar), 7.86 (s, 1H, NH), 7.89 (d, 1H, J=2.8 Hz, Ar), 8.05 (s, 1H, NH).
  • Example 6 6-(5-Chloro-2-methoxy-phenyl)-N*4*-p-tolyl-pyridine-2,4-diamine
  • To a solution of N-[6-(5-chloro-2-methoxy-phenyl)-4-p-tolylamino-pyridin-2-yl]-acetamide (15 mg, 0.039) prepared according to Example 5 in ethanol (4 ml) was added hydrazine (0.01 ml). After heating under reflux for 3 hours, the mixture was concentrated under reduced pressure. The residue was treated with water (40 ml) and extracted with ethyl acetate (50 ml). The organic extract was dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (6 mg, 45% yield). 1H NMR (acetone-d6) δ 2.31 (s, 3H, CH3), 3.90 (s, 3H, CH3), 5.40 (s, 2H, NH2), 6.16 (t, 1H, J=2.0 Hz, Ar), 7.04 (bs, 1H, Ar), 7.09 (t, 1H, J=8.8 Hz, Ar), 7.17 (bs, 4H, Ar), 7.32 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.72 (s, 1H, NH), 7.93 (d, 1H, J=2.8 Hz, Ar).
  • Example 7 6-(5-Chloro-2-methoxy-phenyl)-N*4*-(4-chloro-phenyl)-pyridine-2,4-diamine
  • Following the method described in Example 1, 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-amine and 4-chloro-phenylboronic acid gave 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-(4-chloro-phenyl)-amine in 56% yield. 1H NMR (acetone-d6) δ 3.92(s, 3H, CH3), 6.86 (d, 1H, J=8.9 Hz, Ar), 6.90 (t, 1H, J=1.9 Hz, Ar) 7.18 (d, 1H, J=8.9 Hz, Ar), 7.20 (d, 1H, J=8.9 Hz, Ar), 7.36 (d, 2H, J=8.9 Hz, Ar), 7.41 (dd, 1H, J=8.8 Hz, J=2.8 Hz, Ar), 7.46 (d, 2H, J=8.9 Hz, Ar), 7.72 (t, 1H, J=1.9 Hz, Ar), 7.95 (d, 1H, J=2.8 Hz, Ar).
  • Following the method described in Example 6, 2-chloro-6-(5-chloro-2-methoxy-phenyl)-pyridin-4-yl-(4-chloro-phenyl)-amine provided the title compound (17% yield for 2 steps). 1H NMR (acetone-d6) δ 3.90 (s, 3H, CH3), 5.27 (s, 2H, NH2), 6.22 (t, 1H, J=1.9 Hz, Ar), 7.09-7.12 (m, 2H, Ar), 7.26-7.36 (m, 5H, Ar), 7.86 (s, 1H, NH), 7.96 d, 1H, J=2.8 Hz, Ar).
  • Example 8 4-(5-Chloro-2-methoxy-phenyl)-N-(4-chloro-phenyl)-pyridine-2,6-diamine
  • Chelidamic acid monohydrate (1.0 g, 5.0 mmol) was treated with pyridine (100 ml) and concentrated under vacuum and then treated with toluene (100 ml) and concentrated under vacuum. The residue was treated with dichloromethane (30 ml), N,N-dimethylformamide (0.3 ml) and a solution of oxalyl chloride in dichloromethane (2 M, 13.7 ml). After stirring for 1 hour, volatiles were evaporated under reduced pressure to give crude 4-chloro-pyridine-2,6-dicarbonyl dichloride.
  • To a mixture of crude 4-chloro-pyridine-2,6-dicarbonyl dichloride, dichloromethane (100 ml), and ethanol (5 ml), cooled in an ice bath, was added a solution of pyridine (6 ml) and ethanol (5 ml). The cooling bath was removed and the mixture was stirred for 5 hours. Volatiles were evaporated under reduced pressure. The residue was treated with a mixture of hydrochloric acid (1M, 30 ml) and saturated aqueous sodium chloride solution (50 ml) and extracted with dichloromethane (200 ml). The organic extract was dried over magnesium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate-hexane (1:4 followed by 1:3) to provide 4-chloro-pyridine-2,6-dicarboxylic acid diethyl ester (800 mg, 62% yield). 1H NMR (DMSO-d6) δ 1.36 (t, 6H, J=7.1 Hz CH3), 3.33 (s, 3H, CH3), 4.40 (q, 4H, J=7.1 Hz, CH2), 8.31 (s, 2H, Ar).
  • To a solution of 4-chloro-pyridine-2,6-dicarboxylic acid diethyl ester (700 mg, 2.8 mmol), 5-chloro-2-methoxy-phenylboronic acid (630 mg, 3.4 mmol), palladium acetate (105 mg, 0.47 mmol) in ethylene glycol dimethyl ether (50 ml), degassed with argon, was added a solution of sodium carbonate (868 mg, 8.2 mmol) in water (5 ml) followed by triphenylphosphine (220 mg, 0.84 mmol). The mixture was stirred at 80° C. for 3 hours and filtered through a pad of celite under suction. The filtrate was purified by flash chromatography on silica gel eluting with ethyl acetate-hexane (1:3 followed by 1:2) to give 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarboxylic acid diethyl ester (755 mg, 74% yield) as a white solid. 1H NMR (acetone-d6) δ 1.42 (t, 6H, J=7.1 Hz CH3), 3.92 (s, 3H, CH3), 4.45 (q, 4H, J=7.1 Hz, CH2), 7.27 (d, 1H, J=8.9 Hz, Ar), 7.52 (dd, 1H, J=8.8 Hz, J=2.7 Hz, Ar), 7.56 (d, 1H, J=2.7 Hz, Ar), 8.43 (s, 2H, Ar).
  • To a solution of 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarboxylic acid diethyl ester (700 mg, 2.7 mmol) in ethanol (40 ml) was added hydrazine (1.0 ml) and the mixture was heated under reflux for 3 hours. After cooling to 10° C., the solid was filtered and dried under vacuum to give 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarbonyl dihydrazide (618 mg, 100% yield). 1H NMR (DMSO-d6) δ 3.92 (s, 3H, CH3), 4.68 (s, 4H, NH2), 7.24 (d, 1H, J=8.6 Hz, Ar), 7.53-7.56 (m, 2H, Ar), 8.20 (s, 2H, Ar), 10.72 (s, 2H, NH).
  • To a solution of 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarbonyl dihydrazide (600 mg, 2.61 mmol) in hydrochloric acid (0.5 M, 60 ml), cooled in an ice bath, was added a solution of sodium nitrite (1.7 g, 24.6 mmol) in water (5 ml). After stirring for 3 hours, the solid was filtered and dried under vacuum to give 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarbonyl diazide (600 mg, 91% yield) as a beige powder. 1H NMR (acetone-d6) δ 3.94 (s, 3H, CH3), 7.28 (d, 1H, J=8.9 Hz, Ar), 7.55 (dd, 1H, J=8.8 Hz, J=2.7 Hz, Ar), 7.62 (d, 1H, J=2.7 Hz, Ar), 8.54 (s, 2H, Ar).
  • To a solution of 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-dicarbonyl diazide (310 mg, 1.23 mmol) in toluene (20 ml) was added tert-butanol (1 ml). The mixture was stirred at 100° C. for 30 minutes and volatiles were evaporated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetatehexane (1:3) to provide [6-tert-butoxycarbonylamino-4-(5-chloro-2-methoxy-phenyl)-pyridin-2-yl]-carbamic acid tert-butyl ester (220 mg, 52% yield). 1H NMR (DMSO-d6) δ 1.47 (s, 18H, CH3), 3.79 (s, 3H, CH3), 7.18 (d, 1H, J=8.9 Hz, Ar), 7.30 (d, 1H, J=2.6 Hz, Ar), 7.47-7.49 (m, 3H, Ar), 9.41 (s, 2H, NH).
  • To a solution of [6-tert-butoxycarbonylamino-4-(5-chloro-2-methoxy-phenyl)-pyridin-2-yl]-carbamic acid tert-butyl ester (220 mg, 0.49 mmol) in dichloromethane (8 ml) was added trifluoroacetic acid (6 ml). After stirring for 1 hour, volatiles were evaporated under reduced pressure. Treatment with toluene (15 ml) and evaporation of volatiles under reduced pressure provided 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt (180 mg, 100% yield).
  • To a mixture of 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt (90 mg, 0.25 mmol), 4-chloro-phenylboronic acid (45 mg, 0.29 mmol), copper (II) acetate (218 mg, 1.2 mmol), 4A molecular sieves (5 g) in dichloromethane (20 ml) was added triethylamine (0.3 ml). The mixture was stirred under an oxygen atmosphere for 40 minutes and filtered through a pad of celite under suction. After evaporation of volatiles under reduced pressure, the residue was purified by flash chromatography on silica gel eluting with ethyl acetate-hexane (3:5 followed by 1:1 followed by 5:3) to provide the title compound (30 mg, 33% yield). 1H NMR (acetone-d6) δ 3.84 (s, 3H, CH3), 5.33 (d, 2H, J=8.8 Hz, NH2), 6.16 (d, 1H, J=1.1 Hz, Ar), 6.27 (t, 1H, J=1.1 Hz, Ar), 7.12 (d, 1H, J=8.8 Hz, Ar), 7.23 (d, 2H, J=8.9 Hz, Ar), 7.30 (d, 1H, J=2.7 Hz, Ar), 7.36 (dd, 1H, J=8.8 Hz, J=2.7 Hz, Ar), 7.76-7.79 (m, 2H, Ar), 8.05 (s, 1H, NH).
  • Example 9 4-(5-Chloro-2-methoxy-phenyl)-4-p-tolyl-pyridine-2,6-diamine
  • Following the method described in Example 8, 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt and p-tolyboronic acid provided the title compound (11% yield). 1H NMR (acetone-d6) δ 2.26 (s, 3H, CH3), 3.83 (s, 3H, CH3), 5.24 (bs, 2H, NH2), 6.10 (d, 1H, J=1.1 Hz, Ar), 6.26 (t, 1H, J=1.1 Hz, Ar), 7.06 (d, 2H, J=8.5 Hz, Ar), 7.08 (d, 1H, J=8.8 Hz, Ar), 7.29 (d, 1H, J=2.7 Hz, Ar), 7.35 (dd, 1H, J=9.0 Hz, J=3.3 Hz, Ar), 7.53-7.56 (m, 2H, Ar), 7.70 (s, 1H, NH).
  • Example 10 4-(5-Chloro-2-ethoxy-phenyl)-4-p-tolyl-pyridine-2,6-diamine
  • Following the method described in Example 8, (5-chloro-ethoxy-phenyl)-phenylboronic acid and 4-chloro-pyridine-2,6-dicarboxylic acid diethyl ester gave 4-(5-chloro-2-ethoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt (4% yield for 5 steps).
  • Following the method described in Example 8, 4-(5-chloro-2-ethoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt and p-tolylboronic acid provided the title compound (17% yield). 1H NMR (acetone-d6) δ 1.35 (t, 3H, J=7.0 Hz, CH3), 2.27 (s, 3H, CH3), 4.08 (q, 2H, J=7.0 Hz, CH2), 5.24 (bs, 2H, NH2), 6.14 (d, 1H, J=1.0 Hz, Ar), 6.32 (t, 1H, J=0.9 Hz, Ar), 7.06-7.10 (m, 3H, Ar), 7.29-7.34 (m, 2H, Ar), 7.51-7.54 (m, 2H, Ar), 7.67 (s, 1H, NH).
  • Example 11 4-(5-Chloro-2-ethoxy-phenyl)-N-(4-chloro-phenyl)-pyridine-2,6-diamine
  • Following the method described in Example 8, 4-(5-chloro-2-ethoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt and 4-chloro-phenylboronic acid provided the title compound (30% yield). 1H NMR (acetone-d6) δ 1.35 (t, 3H, J=7.0 Hz, CH3), 4.08 (q, 2H, J=7.0 Hz, CH2), 5.32 (bs, 2H, NH2), 6.21 (d, 1H, J=1.2 Hz, Ar), 6.32 (t, 1H, J=1.1 Hz, Ar), 7.09 (d, 2H, J=8.6 Hz, Ar), 7.23 (d, 2H, J=8.9 Hz, Ar), 7.31-7.78 (m, 2H, Ar), 8.04 (s, 1H, NH).
  • Example 12 4-16-Amino-4-(5-chloro-2-ethoxy-phenyl)-pyridin-2-ylaminol-benzaldehyde
  • Following the method described in Example 8, 4-(5-chloro-2-ethoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt and 4-formyl-phenylboronic acid provided the title compound (22% yield). 1H NMR (acetone-d6) δ 1.36 (t, 3H, J=6.9 Hz, CH3), 4.11 (q, 2H, J=7.0 Hz, CH2), 5.51 (bs, 2H, NH2), 6.33 (d, 1H, J=1.1 Hz, Ar), 6.44 (d, 1H, J=1.0 Hz, Ar), 7.12 (d, 1H, J=8.5 Hz, Ar), 7.31-7.37 (m, 2H, Ar), 7.78 (d, 2H, J=8.8 Hz, Ar), 7.88-7.97 (m, 2H, Ar), 8.57 (s, 1H, NH), 9.84 (s, 1H, CH).
  • Example 13 {4-[6-Amino-4-(5-chloro-2-ethoxy-phenyl)-pyridin-2-ylaminol-phenyl]}-methanol
  • To a solution of the title compound provided by Example 12 (25 mg, 0.068 mmol) in tetrahydofuran (5 ml) and methanol (10 ml) was added sodium borohydride (20 mg, 0.53 mmol). After stirring for 1.5 hours, the mixture was treated with water (10 ml) and hydrochloric acid (1 M, 0.6 ml). The mixture was concentrated to a volume of 8 ml and the solid was collected by filtration. The solid was treated with aqueous sodium carbonate solution (10%, 10 ml) and extracted with ethyl acetate (3×30 ml). The combined extracts were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative thin layer chromatography eluting with methanol-dichloromethane (5:95) to give the title compound (10 mg, 39% yield). 1H NMR (acetone-d6) δ 1.36 (t, 3H, J=6.9 Hz, CH3), 3.95 (bs, 1H, OH), 4.09 (q, 2H, J=7.0 Hz, CH2), 4.56 (s, 2H, CH2), 5.28 (bs, 2H, NH2), 6.16 (d, 1H, J=1.1 Hz, Ar), 6.34 (d, 1H, J=1.1 Hz, Ar), 7.09 (d, 1H, J=8.5 Hz, Ar), 7.24 (d, 2H, J=8.8 Hz, Ar), 7.30-7.34 (m, 2H, Ar), 7.60-7.63 (m, 2H, Ar), 7.80 (s, 1H, NH).
  • Example 14 4-[6-Amino-4-(5-chloro-2-methoxy-phenyl)-pyridin-2-ylamino]-benzaldehyde
  • Following the method described in Example 8, 4-(5-chloro-2-methoxy-phenyl)-pyridine-2,6-diamine trifluoroacetic acid salt and 4-formyl-phenylboronic acid provided the title compound (22% yield). 1H NMR (acetone-d6) δ 3.85 (s, CH3), 5.51 (bs, 2H, NH2), 6.27 (d, 1H, J=1.1 Hz, Ar), 6.38 (d, 1H, J=1.1 Hz, Ar), 7.14 (d, 1H, J=8.8 Hz, Ar), 7.32 (d, 1H, J=2.7 Hz, Ar), 7.38 (dd, 1H, J=8.8 Hz, J=2.7 Hz, Ar), 7.78 (d, 2H, J=8.7 Hz, Ar), 7.96 (d, 2H, J=8.7 Hz, Ar), 8.59 (s, 1H, NH), 9.84 (s, 1H, CH).
  • Example 15 LPAAT-β Assay
  • A. Production of Recombinant LPAAT-β for Assays
  • For the construction of Baculovirus expression vectors, the full-length human LPAAT-β cDNA was amplified by PCR from the DNA template pCE9.LPAAT-β (West et al., DNA Cell Biol. 16:691-701 (1997)) using the primers 5′-TGATATCCGA AGAAGATCTT ATGGAGCTGT GGCCGTGTC-3′ (olpb1F; SEQ ID NO:1) and 5′-CAGGCTCTAG ACTACTGGGC CGGCTGCAC-3′ (olpb1R; SEQ ID NO:2). The ˜870 bp fragment generated was reamplified by PCR using the primers 5′ CCTACGTCG ACATGGAACA AAAATTGATA TCCGAAGAAG ATC-3′ (olpb2F; SEQ ID NO:3) and 5′-CAGGCTCTAG ACTACTGGGC CGGCTGCAC-3′ (olpb1R; SEQ ID NO:2). The ˜890 bp fragment generated was then cleaved with Sal I and Xba I for insertion into pFastBac™ HTc vector (Life Technologies, Gaithersberg, Md.) between the Sal I and Xba I sites for the generation of the plasmid pFB.LPAAT-β. This plasmid was then transformed into E. coli DH10Bac™ (Life Technologies, Gaithersberg, Md.) for the generation of recombinant Bacmid DNA for transfection into HighFive (Invitrogen, San Diego, Calif.) or SF9 insect cells for the production of recombinant Baculovirus stocks using the protocol described in the Bac-to-Bac® Baculovirus Expression System (Life Technologies, Gaithersberg, Md.), a eukaryotic expression system for generating recombinant baculovirus through site-specific transposition in E. coli. Viral stocks harvested from the transfected cells can then be used to infect fresh insect cells for the subsequent expression of LPAAT-β fusion protein with a poly-histidine tag and a mycepitope near its N-terminus. The membrane fraction from these Sf9 cells would be the source of LPAAT enzyme.
  • B. Preparation of Cell Membranesfrom Sf9 Cells
  • For the preparation of membranes from Sf9 Cells, all steps are performed on ice or at 4° C. Sf9 cell pellets (˜108 cells) were thawed and resuspended in 1-2 ml of buffer A (20 mM Hepes, pH 7.5, 1 mM DTT, 1 mM EDTA, 20% w/v glycerol, 1 mM Benzamidine, 1 μg/ml soybean trypsin inhibitor (SBTI), 1 μg/ml pepstatin A) w/o DTT but with 1 mM Pefabloc. The cells were lysed by sonication using a Branson Sonifier at output=2, duty cycle=2, 10 pulses each at 10s. with the tip of small sonicator probe submerged but not touching the walls. DTT was then added to 1 mM from a 1 M stock. The samples were centrifuged at 1500 rpm for 5 min. The low speed supernatant was saved and centrifuged (TLA 100.3 rotor, polycarbonate tubes, 2 ml/tube or 1.5 ml/tube minimum) at 100000×g for 1 hr. The high speed pellet was resuspend in Buffer A with a probe sonicator (10 pulses @ (output #2 and duty cycle 20%) as a source of LPAAT enzyme.
  • C. Assay of LPAA T-β Activity
  • LPAAT-β catalyzes the transfer of an acyl group from a donor such as acyl-CoA to LPA. The transfer of the acyl group from acyl-CoA to LPA leads to the release of free CoA, which can be reacted with the thiol reagent, 5,5 ′-dithiobis(2-nitrobenzoic acid) (DTNB). The reaction between DTNB and the free sulfhydryl group from CoA generates a yellow-colored product, 3-carboxylato-4-nitrothiophenolate (CNP), that absorbs at 413 nm. LPAAT-β derived from Sf9 cell membrane overexpressing LPAAT-β were resuspended in HEPES saline buffer (20 mM HEPES pH 7.5, 150 mM NaCl), 1 mg/ml BSA and 72 μl aliquots were distributed into 96-well microtiter plates. 8 μl of compound of interest at 200 μM dissolved in 100% DMSO was added into each well. 20 μl of 1 mM 18:1-CoA and 1 μM sn-1-18:1 lysoPA was then added to each well to initiate the reaction and allowed to run at room temperature for 25 min. 100 μl of 1 mM DTNB in 100% ethanol was then added to each well to quench the reaction and for color development. The absorbance at 405 nm, measured using a spectrophotometer plate reader, is proportional to the activity of LPAAT-β in the sample. This colorimetric assay was used for the high throughput screening of LPAAT inhibitors. Compounds that showed >50% inhibition of the change in absorbance at 405 nm compared to control were selected for a secondary assay.
  • A secondary assay for LPAAT activity in cell extracts based on either the conversion of fluorescent NBD-LPA to NBD-PA (West, et al., DNA Cell Biol. 6:691-701, 1997) or [14C]LPA to [14C]PA using TLC analysis was used to screen compounds that showed >50% inhibition of LPAAT activity in the primary colorimetric assay. The radiometric assay was carried out in Sf9 cell membrane overexpressing LPAAT-β resuspended in HEPES-saline buffer, pH 7.5, 1 mg/ml BSA, lmM EDTA and 200 μM [14C]18:1-CoA and 200 μM sn-1-18:1 lysoPA. The samples were incubated 7 min at 37° C., extracted into organic solvent (CHCl3/CH3OH/HCl at 33/66/1), before loading onto TLC plates. A more detailed protocol for the radiometric assay is described below:
  • Specifically, this LPAAT assay is a modification of the acyltransferase assay published previously (Hollenback and Glomset, Biochemistry 37:363-376 (1999)).
  • 1. The basic assay, in a total volume of 50 μl, employs a solution of substrates and the protein sample. Total assay volume, as well as the volume of each solution, can be changed to fit an experiment. In addition, other compounds, ex inhibitors and activators, can be included in the assay as well.
  • 2. To prepare the solution of substrates:
  • a. Stocks of Hepes (pH 7.5), NaCl, EDTA, BSA and acyl-CoA (from Serdery or Sigma) are mixed with water to make the appropriate concentration of each compound. This can be varied from assay-to-assay, but the final reaction mix is about 50 mM Hepes, 100 mM NaCl, 1 mM EDTA, 1 mg/ml BSA and 0-400 μM acyl-CoA.
  • b. The lysoPA (from Avanti) is typically stored in chloroform and the 14C-labeled acyl-CoA (from Amersham) is typically stored in water/ethanol=1:1. Appropriate amounts of each solution are added the to a 12×75 mm borosilicate glass test tube and dry the solvent under N2 or Ar. An appropriate volume of the solution prepared in 2a is added to the lysoPA and 14C-labeled acyl-CoA. The lipids are resuspended by sonication for 15 sec in a bath sonicator. The resulting suspension is then incubated (with occasional gentle vortexing) for about 10 minutes at room temp. The sn-1-16:0 lysoPA may require brief warming of the solvent to solubilize it. The concentration of lysoPA and 14C-labeled acyl-CoA can vary, but typically the final lysoPA concentration ranges between 0 and 400 μM and the 14C-labeled acyl-CoA specific activity ranges between 0.5 and 2 Ci/mol.
  • 3. Protein sample: varies from experiment-to-experiment.
  • 4. The assay is performed by mixing the components in 12×75 mm borosilicate glass test tubes (the order of addition does not matter unless indicated) and incubating at 37° C. for 5 to 10 minutes such that the assay within the linear range for time and protein.
  • 5. The reaction is quenched by adding 1.3 ml of chloroform/methanol/HCl=48/51/0.7 and vortexing. 10 μl of carrier solution is then added (3 mg/ml each PA, ex. 16:0-18:1, and lysoPA, ex sn-1-18:1, in chloroform). Two phases are formed by adding 0.3 ml of water to each tube and vortexing.
  • 6. The sample is centrifuged for 3 minutes at 1000×g, the upper (aqueous/methanol) phase is aspirated and the lower phase is dried under nitrogen.
  • 7. Thin layer chromatography:
  • a. The dried samples are resuspended in 50 μl of chloroform and a 15 μl aliquot is immediately spotted on an Analtech silica gel 60 HP-TLC plate (10×20 cm).
  • b. Plates are developed in chloroform/methanol/acetic acid/water=85/12.5/12.5/3 (takes about 15 min) and dried.
  • c. To be able to convert pixel volume (determined by the Storm phosphor imager, see step 8b) into cpm, cpm standard curve must be generated on the plate. 14C-labeled oleate dilutions in chloroform are made for this purpose. Four stocks (50 cpm/μl to 800 cpm/μl) are made and 2 μl of a different concentration are spotted in each corner of the plate (where previously there was no radioactivity).
  • d. For quality control purposes, the plates are stained with primuline and scanned with the Storm (blue chemilluminescence mode).
  • The PA and lysoPA bands are easily detected in this system because of the carrier added in step 5. PA and lysoPA have respective Rf's of about 0.63 and 0.21.
  • 8. Quantitating activity:
  • a. The plates are then wrapped in saran wrap and exposed to a freshly phosphor screen overnight (longer exposures can also be done to increase the
  • b. The screens are scanned (Phosphorimager mode), and LPAAT is determined by quantifying the pixels in the band comigrating with PA standard the standard curve generated from the cpm standards that were spotted in step 7c.
    TABLE 1
    LPAAT-β
    cell-free assay
    Compound (IC50, μM) Compound Name
    1
    Figure US20070178134A1-20070802-C00003
    0.2 [2-Chloro-6-(5-chloro-2-methoxy- phenyl)-pyridin-4-yl]-p-tolyl-amine
    2
    Figure US20070178134A1-20070802-C00004
    >40 [4-Cloro-6-(5-chloro-2-methoxy- phenyl)-pyridin-2-yl]-p-tolyl-amine
    3
    Figure US20070178134A1-20070802-C00005
    29 6-(5-Chloro-2-methoxy-phenyl)- N*2*-p-tolyl-pyridine-2,4-diamine
    4
    Figure US20070178134A1-20070802-C00006
    6.5 6-(5-Chloro-2-methoxy-phenyl)- N*2*-(4-chloro-phenyl)-pyridine- 2,4-diamine
    5
    Figure US20070178134A1-20070802-C00007
    34 N-[6-(5-chloro-2-methoxy-phenyl)- 4-p-tolylamino-pyridin-2-yl]- acetamide
    6
    Figure US20070178134A1-20070802-C00008
    35 6-(5-Chloro-2-methoxy-phenyl)- N*4*-p-tolyl-pyridine-2,4-diamine
    7
    Figure US20070178134A1-20070802-C00009
    18 6-(5-Chloro-2-methoxy-phenyl)- N*4*-(4-chloro-phenyl)-pyridine- 2,4-diamine
    8
    Figure US20070178134A1-20070802-C00010
    0.04 4-(5-Chloro-2-methoxy-phenyl)-N- (4-chloro-phenyl)-pyridine-2,6- diamine
    9
    Figure US20070178134A1-20070802-C00011
    0.24 4-(5-Chloro-2-methoxy-phenyl)-4- p-tolyl-pyridine-2,6-diamine
    10
    Figure US20070178134A1-20070802-C00012
    0.49 4-(5-Chloro-2-ethoxy-phenyl)-4-p- tolyl-pyridine-2,6-diamine
    11
    Figure US20070178134A1-20070802-C00013
    0.05 4-(5-Chloro-2-ethoxy-phenyl)-N-(4- chloro-phenyl)-pyridine-2,6-diamine
    12
    Figure US20070178134A1-20070802-C00014
    0.05 4-[6-Amino-4-(5-chloro-2-ethoxy- phenyl)-pyridin-2-ylamino]- benzaldehyde
    13
    Figure US20070178134A1-20070802-C00015
    0.1 {4-[6-Amino-4-(5-chloro-2-ethoxy- phenyl)-pyridin-2-ylamino]- phenyl}-methanol
    14
    Figure US20070178134A1-20070802-C00016
    0.26 4-[6-Amino-4-(5-chloro-2-methoxy- phenyl)-pyridin-2-ylamino]- benzaldehyde
  • All of the above U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, are incorporated herein by reference, in their entirety.
  • From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention.

Claims (59)

1. A compound or physiologically acceptable salt thereof, wherein the compound has the formula:
Figure US20070178134A1-20070802-C00017
wherein:
X, Y and Z are N, CH or CR where R is alkyl, alkoxy, Cl, Br, NH2, NHR′ or NR′R″ where R′ and R″ independently are alkyl;
Q is NR, RN—(CH2)n, (CH2)n—NR, O, O—(CH2)n, (CH2)n—O, S, S—(CH2)n, or (CH2)n—S, where n is 1-10 and R is H or alkyl;
R1 is H, OH, alkyl, alkoxy, Cl, F, Br, CR3 where R3 is Cl3, F3 or Br3, NH2, NHR or NRR′ where R and R′ independently are alkyl;
R2 is H, OH, alkyl, alkoxy, Cl, F, Br or CR3 where R3 is Cl3, F3 or Br3;
R3 is H, alkyl, alkoxy, Cl, CCl3, NH2, NHR or NRR′ where R and R′ independently are alkyl or acyl;
R4, R5, and R are independently H, OH, alkyl, alkenyl, alkynyl, alkoxy, (CH2)n—OR where R is H or alkyl and n is 1-10, Cl, F, Br, CR3 where R3 is Cl3, F3 or Br3 heterocycle, N+(═O)O, C≡N, N3, SH, SR or S(═O)2R where R is alkyl, NH2, NHR or NRR′ where R and R′ independently are alkyl, or R4 and R5 or R5 and R6 are taken together with the benzene ring to form a heterocycle;
and with the proviso that one of X, Y and Z is N.
2. The compound or salt thereof of claim 1 wherein X or Y of the compound or salt thereof is N.
3. The compound or salt thereof of claim 1 wherein Q of the compound or salt thereof is NH.
4. The compound or salt thereof of claim 1 wherein R4 or R5 of the compound or salt thereof is acyl.
5. The compound or salt thereof of claim 1 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
6. The compound or salt thereof of claim 1 wherein R2 of the compound or salt thereof is Cl or Br.
7. The compound or salt thereof of claim 1 wherein R3 of the compound or salt thereof is alkyl or NH2.
8. The compound or salt thereof of claim 1 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
9. The compound or salt thereof of claim 1 wherein the compound is any one of compounds 1-14 of Table 1, or physiologically acceptable salts thereof.
10. A pharmaceutical composition comprising a compound or salt thereof according to claim 1 in combination with a pharmaceutically acceptable carrier or diluent.
11. The pharmaceutical composition of claim 10 wherein X or Y of the compound or salt thereof is N.
12. The pharmaceutical composition of claim 10 wherein Q of the compound or salt thereof is NH.
13. The pharmaceutical composition of claim 10 wherein R4 or R5 of the compound or salt thereof is acyl.
14. The pharmaceutical composition of claim 10 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
15. The pharmaceutical composition of claim 10 wherein R2 of the compound or salt thereof is Cl or Br.
16. The pharmaceutical composition of claim 10 wherein R3 of the compound or salt thereof is alkyl or NH2.
17. The pharmaceutical composition of claim 10 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
18. The pharmaceutical composition of claim 10 wherein the compound is any one of compounds 1-14 of Table 1, or physiologically acceptable salts thereof.
19. A method for reducing the activity of lysophosphatidic acid acyltransferase β (LPAAT-β) comprising contacting LPAAT-β with a compound or salt thereof according to claim 1 or a composition according to claim 10 in an amount effective to reduce LPAAT-β activity.
20. The method of claim 19 wherein the LPAAT-β resides in an animal.
21. The method of claim 20 wherein the animal is a mammal.
22. The method of claim 19 wherein X or Y of the compound or salt thereof is N.
23. The method of claim 19 wherein Q of the compound or salt thereof is NH.
24. The method of claim 19 wherein R4 or R5 of the compound or salt thereof is acyl.
25. The method of claim 19 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
26. The method of claim 19 wherein R2 of the compound or salt thereof is Cl or Br.
27. The method of claim 19 wherein R3 of the compound or salt thereof is alkyl or NH2.
28. The method of claim 19 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
29. The method of claim 19 wherein the compound is any one of compounds 1-14 of Table 1, or physiologically acceptable salts thereof.
30. A method for inhibiting the proliferation of a cell in which the activity of lysophosphatidic acid acyltransferase β (LPAAT-β) is required for the proliferation of the cell comprising contacting the cell with a compound or salt thereof according to claim 1 or a composition according to claim 10 in an amount effective to inhibit the proliferation of the cell.
31. The method of claim 30 wherein the cell resides in an animal.
32. The method of claim 31 wherein the animal is a mammal.
33. The method of claim 30 wherein X or Y of the compound or salt thereof is N.
34. The method of claim 30 wherein Q of the compound or salt thereof is NH.
35. The method of claim 30 wherein R4 or R5 of the compound or salt thereof is acyl.
36. The method of claim 30 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
37. The method of claim 30 wherein R2 of the compound or salt thereof is Cl or Br.
38. The method of claim 30 wherein R3 of the compound or salt thereof is alkyl or NH2.
39. The method of claim 30 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
40. The method of claim 30 wherein the compound is any one of compounds 1-14 of Table I, or physiologically acceptable salts thereof.
41. A method for treating a cancer in which lysophosphatidic acid acyltransferase β (LPAAT-β) activity is associated comprising administering to an animal in need, a compound or salt thereof according to claim 1 or a composition according to claim 10 in an amount effective to treat the cancer.
42. The method of claim 41 wherein the animal is a mammal.
43. The method of claim 41 wherein X or Y of the compound or salt thereof is N.
44. The method of claim 41 wherein Q of the compound or salt thereof is NH.
45. The method of claim 41 wherein R4 or R5 of the compound or salt thereof is acyl.
46. The method of claim 41 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
47. The method of claim 41 wherein R2 of the compound or salt thereof is Cl or Br.
48. The method of claim 41 wherein R3 of the compound or salt thereof is alkyl or NH2.
49. The method of claim 41 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
50. The method of claim 41 wherein the compound is any one of compounds 1-14 of Table 1, or physiologically acceptable salts thereof.
51. A coated medical device for inhibiting the proliferation of a cell in which the activity of lysophosphatidic acid acyltransferase β (LPAAT-β) is required for the proliferation of the cell comprising a medical device coated with a compound or salt thereof according to claim 1 or a composition according to claim 10.
52. The device of claim 51 wherein X or Y of the compound or salt thereof is N.
53. The device of claim 51 wherein Q of the compound or salt thereof is NH.
54. The device of claim 51 wherein R4 or R5 of the compound or salt thereof is acyl.
55. The device of claim 51 wherein R1 of the compound or salt thereof is alkyl, alkoxy or Cl.
56. The device of claim 51 wherein R2 of the compound or salt thereof is Cl or Br.
57. The device of claim 51 wherein R3 of the compound or salt thereof is alkyl or NH2.
58. The device of claim 51 wherein R4 or R5 of the compound or salt thereof is alkyl, Cl, Br, CF3, CH2—OH, (CH2)2—OH, N+(═O)O, C≡N, or C(═O)R wherein R is alkyl or alkoxy, or R4 and R5 are taken together with the benzene ring to form indazole.
59. The device of claim 51 wherein the compound is any one of compounds 1-14 of Table 1, or physiologically acceptable salts thereof.
US11/724,459 2003-04-04 2007-03-14 Pyridines and uses thereof Abandoned US20070178134A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/724,459 US20070178134A1 (en) 2003-04-04 2007-03-14 Pyridines and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US46078203P 2003-04-04 2003-04-04
US10/667,916 US6875781B2 (en) 2003-04-04 2003-09-22 Pyridines and uses thereof
US11/056,777 US7208507B2 (en) 2003-04-04 2005-02-11 Pyridines and uses thereof
US11/724,459 US20070178134A1 (en) 2003-04-04 2007-03-14 Pyridines and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/056,777 Continuation US7208507B2 (en) 2003-04-04 2005-02-11 Pyridines and uses thereof

Publications (1)

Publication Number Publication Date
US20070178134A1 true US20070178134A1 (en) 2007-08-02

Family

ID=33101484

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/667,916 Expired - Fee Related US6875781B2 (en) 2003-04-04 2003-09-22 Pyridines and uses thereof
US11/056,777 Expired - Fee Related US7208507B2 (en) 2003-04-04 2005-02-11 Pyridines and uses thereof
US11/724,459 Abandoned US20070178134A1 (en) 2003-04-04 2007-03-14 Pyridines and uses thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/667,916 Expired - Fee Related US6875781B2 (en) 2003-04-04 2003-09-22 Pyridines and uses thereof
US11/056,777 Expired - Fee Related US7208507B2 (en) 2003-04-04 2005-02-11 Pyridines and uses thereof

Country Status (1)

Country Link
US (3) US6875781B2 (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050148600A1 (en) * 2003-09-02 2005-07-07 Cell Therapeutics, Inc. Methods for the treatment of obesity
EP1805142A4 (en) * 2004-09-23 2009-06-10 Reddy Us Therapeutics Inc Novel pyridine compounds, process for their preparation and compositions containing them
CL2007002007A1 (en) * 2006-07-11 2008-08-22 Janssen Pharmaceutica Nv COMPOUNDS DERIVED FROM BENZOFURO AND BENZOTIENOPIRIMIDINAS, HISTAMINE RECEIVER MODULATORS H4; PREPARATION PROCEDURE; AND ITS USE TO TREAT INFLAMMATORY, ALLERGIC, DERMATOLOGICAL, AUTOIMMUNE, LYMPHATIC AND IMMU DISEASES
WO2008079945A2 (en) * 2006-12-20 2008-07-03 University Of South Florida Rock inhibitors and uses thereof
WO2009023059A2 (en) 2007-06-01 2009-02-19 The Trustees Of Princeton University Treatment of viral infections by modulation of host cell metabolic pathways
LT3459942T (en) 2012-04-24 2021-05-10 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
DK3527563T3 (en) 2013-03-12 2021-12-06 Vertex Pharma DNA-PK INHIBITORS
LT3057953T (en) 2013-10-17 2018-11-26 Vertex Pharmaceuticals Incorporated Co-crystals of (s)-n-methyl-8-(1-((2`-methyl-[4,5`-bipyrimidin]-6-yl)amino)propan-2-yl)quinoline-4-carboxamide and deuterated derivatives thereof as dna-pk inhibitors
DK3089971T3 (en) * 2014-01-01 2020-08-31 Medivation Tech Llc Compounds and methods of use
CN107382840B (en) * 2016-05-16 2020-09-01 四川大学 Pyridine compound and application thereof as IDH function mutation mutant inhibitor drug
MX2019003317A (en) 2016-09-27 2019-08-05 Vertex Pharma Method for treating cancer using a combination of dna-damaging agents and dna-pk inhibitors.

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3119823A (en) * 1962-09-10 1964-01-28 Us Vitamin Pharm Corp Process for preparation of o-hydroxyphenyl-triazines
US3933814A (en) * 1972-09-20 1976-01-20 Bayer Aktiengesellschaft 1-(4-Phenoxyphenyl)-1,3,5-triazines
US3948893A (en) * 1973-03-20 1976-04-06 Bayer Aktiengesellschaft 1-Phenyl-substituted 1,3,5-triazines
US3966725A (en) * 1974-03-21 1976-06-29 Bayer Aktiengesellschaft 1-(4-Phenoxy-phenyl)-1,3,5-triazines
US4219552A (en) * 1977-04-27 1980-08-26 Bayer Aktiengesellschaft 1-(4-Phenoxy-phenyl)-1,3,5-triazines, their use as growth promoters
US4499269A (en) * 1982-04-30 1985-02-12 Fisons Plc Fused 1,3,5-triazines
US5102927A (en) * 1982-09-30 1992-04-07 Ciba-Geigy Corporation Polyolefines which have been stabilized with bis-(polyalkylpiperidinylamino 1,3,5-triazines or tris-(polyalkylpiperidinylamino)-1,3-5-triazines
US5103014A (en) * 1987-09-30 1992-04-07 American Home Products Corporation Certain 3,3'-[[[(2-phenyl-4-thiazolyl)methoxy]phenyl]methylene]dithiobis-propanoic acid derivatives
US5260362A (en) * 1982-09-30 1993-11-09 Ciba-Geigy Corporation Polyolefines which have been stabilized with bis-(polyalkypiperidiny(amino)-1,3,5-triazines or tris-(polyalkylpiperidinyl(amino)-1,3,5-triazines
US5545836A (en) * 1993-09-28 1996-08-13 Ciba-Geigy Corporation Process for the preparation of 1,3,5-triazines
US5702717A (en) * 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
US5856331A (en) * 1993-11-12 1999-01-05 Cell Therapeutics, Inc. Method for preventing tissue injury from hypoxia
US6004985A (en) * 1996-10-09 1999-12-21 Berlex Laboratories, Inc. Thio acid derived monocylic N-heterocyclics as anticoagulants
US6150362A (en) * 1997-12-12 2000-11-21 Henkin; Jack Triazine angiogenesis inhibitors
US6150360A (en) * 1998-03-27 2000-11-21 Janssen Pharmaceutica N.V. Trisubstituted 1,3,5-Triazine Derivatives
US6193960B1 (en) * 1996-07-08 2001-02-27 Ciba Specialty Chemicals Corporation Triazine derivatives

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1321624A (en) 1962-02-14 1963-03-22 Ici Ltd Process for the production of amino-s-triazines and particularly their polymers
DE1200314B (en) 1963-10-26 1965-09-09 Bayer Ag Process for the preparation of substituted s-triazines
JPS5123199B2 (en) 1971-08-18 1976-07-15
JPS5017073B2 (en) 1971-08-18 1975-06-18
JPS4969688A (en) 1972-11-02 1974-07-05
JPH03190814A (en) 1989-12-21 1991-08-20 Daikyo Rubber Seiko:Kk Cosmetic for preventing sunburn
SG46445A1 (en) 1990-01-26 1998-02-20 Immunomedics Inc Vaccines against cancer and infectious diseases
WO2000025780A1 (en) 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
DE60039059D1 (en) 1999-10-07 2008-07-10 Amgen Inc TRIAZINE KINASE INHIBITORS
WO2002022584A1 (en) * 2000-09-11 2002-03-21 Merck & Co., Inc. Thrombin inhibitors
WO2002036578A2 (en) 2000-10-31 2002-05-10 Lynn Bonham Triazine derivatives as lpaat-b inhibitors and uses thereof

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3119823A (en) * 1962-09-10 1964-01-28 Us Vitamin Pharm Corp Process for preparation of o-hydroxyphenyl-triazines
US3933814A (en) * 1972-09-20 1976-01-20 Bayer Aktiengesellschaft 1-(4-Phenoxyphenyl)-1,3,5-triazines
US3948893A (en) * 1973-03-20 1976-04-06 Bayer Aktiengesellschaft 1-Phenyl-substituted 1,3,5-triazines
US3966725A (en) * 1974-03-21 1976-06-29 Bayer Aktiengesellschaft 1-(4-Phenoxy-phenyl)-1,3,5-triazines
US4219552A (en) * 1977-04-27 1980-08-26 Bayer Aktiengesellschaft 1-(4-Phenoxy-phenyl)-1,3,5-triazines, their use as growth promoters
US4499269A (en) * 1982-04-30 1985-02-12 Fisons Plc Fused 1,3,5-triazines
US5260362A (en) * 1982-09-30 1993-11-09 Ciba-Geigy Corporation Polyolefines which have been stabilized with bis-(polyalkypiperidiny(amino)-1,3,5-triazines or tris-(polyalkylpiperidinyl(amino)-1,3,5-triazines
US5102927A (en) * 1982-09-30 1992-04-07 Ciba-Geigy Corporation Polyolefines which have been stabilized with bis-(polyalkylpiperidinylamino 1,3,5-triazines or tris-(polyalkylpiperidinylamino)-1,3-5-triazines
US5103014A (en) * 1987-09-30 1992-04-07 American Home Products Corporation Certain 3,3'-[[[(2-phenyl-4-thiazolyl)methoxy]phenyl]methylene]dithiobis-propanoic acid derivatives
US5545836A (en) * 1993-09-28 1996-08-13 Ciba-Geigy Corporation Process for the preparation of 1,3,5-triazines
US5856331A (en) * 1993-11-12 1999-01-05 Cell Therapeutics, Inc. Method for preventing tissue injury from hypoxia
US5702717A (en) * 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
US6193960B1 (en) * 1996-07-08 2001-02-27 Ciba Specialty Chemicals Corporation Triazine derivatives
US6004985A (en) * 1996-10-09 1999-12-21 Berlex Laboratories, Inc. Thio acid derived monocylic N-heterocyclics as anticoagulants
US6150382A (en) * 1996-10-09 2000-11-21 Berlex Laboratories, Inc. Thio acid derived monocyclic n-heterocyclics as anticoagulants
US6166014A (en) * 1996-10-09 2000-12-26 Berlex Laboratories, Inc. Thio acid derived monocyclic N-heterocyclics as anticoagulants
US6150362A (en) * 1997-12-12 2000-11-21 Henkin; Jack Triazine angiogenesis inhibitors
US6288228B1 (en) * 1997-12-12 2001-09-11 Abbott Laboratories Triazine angiogenesis inhibitors
US6150360A (en) * 1998-03-27 2000-11-21 Janssen Pharmaceutica N.V. Trisubstituted 1,3,5-Triazine Derivatives

Also Published As

Publication number Publication date
US6875781B2 (en) 2005-04-05
US7208507B2 (en) 2007-04-24
US20040198728A1 (en) 2004-10-07
US20050182102A1 (en) 2005-08-18

Similar Documents

Publication Publication Date Title
US7208507B2 (en) Pyridines and uses thereof
US7618968B2 (en) Aryl triazines as LPAAT-β inhibitors and uses thereof
US7419984B2 (en) Pyrimidines and uses thereof
ES2289349T3 (en) DERIVATIVES OF HETEROARIL-PYRIMIDINE AS JAK INHIBITORS.
JP5188988B2 (en) Benzazole derivatives, compositions, and methods of use as Aurora kinase inhibitors
CN102015606B (en) IRE-1a inhibitors
JP5562256B2 (en) Novel compounds, their use and production
Wissner et al. 2-(Quinazolin-4-ylamino)-[1, 4] benzoquinones as covalent-binding, irreversible inhibitors of the kinase domain of vascular endothelial growth factor receptor-2
KR19990045726A (en) Selective β3 Adrenaline Agonists
CN104761585A (en) Substituted aminopyrimidine working as inhibitor of EGFR epidermal growth factor receptor
HRP20031081A2 (en) Cdk inhibiting pyrimidines, production thereof and their use as medicaments
CN102850324A (en) Indole compounds
JP2006508997A (en) Chk-, Pdk- and Akt-inhibiting pyrimidines, their preparation and use as pharmaceuticals
JP6152921B2 (en) Anti-angiogenic compounds, intermediates and uses thereof
CN102712601A (en) A compound, a process for its preparation, a pharmaceutical composition, use of a compound, a method for modulating or regulating serine/threonine kinases and a serine/threonine kinases modulating agent
JP2008539277A (en) Protein kinase inhibitor
JPWO2006104161A1 (en) Thienopyridine derivative, quinoline derivative, and quinazoline derivative having c-Met autophosphorylation inhibitory action
KR20160102821A (en) Thienodiazepine derivatives or pharmaceutically acceptable salt thereof, and pharmaceutical composition comprising the same as an active ingredient
JP5603775B2 (en) Antimalarial compounds with flexible side chains
KR102402470B1 (en) Quinazoline Derivative
US10851054B2 (en) Derivatives of indole for the treatment of cancer, viral infections and lung diseases
JP2004525139A (en) FLT-1 ligands and their use in treating diseases regulatable by angiogenesis
CN102268000A (en) Novel spiroheterocyclic compound and application of same serving as therapeutic agent
CN106535894A (en) MCL-1 modulating compounds for cancer treatment
KR102507358B1 (en) Benzofuran-based n-acylhydrazone derivatives and pharmaceutical composition comprising the same

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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