WO2012145678A1 - Anti-cancer and anti-inflammatory parthenolide compounds - Google Patents

Anti-cancer and anti-inflammatory parthenolide compounds Download PDF

Info

Publication number
WO2012145678A1
WO2012145678A1 PCT/US2012/034527 US2012034527W WO2012145678A1 WO 2012145678 A1 WO2012145678 A1 WO 2012145678A1 US 2012034527 W US2012034527 W US 2012034527W WO 2012145678 A1 WO2012145678 A1 WO 2012145678A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
heteroaryl
cycloalkyl
compound
Prior art date
Application number
PCT/US2012/034527
Other languages
French (fr)
Inventor
Daniel A. HARKI
Dan Wang
Original Assignee
Regents Of The University Of Minnesota
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 Regents Of The University Of Minnesota filed Critical Regents Of The University Of Minnesota
Publication of WO2012145678A1 publication Critical patent/WO2012145678A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/93Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems condensed with a ring other than six-membered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/92Naphthofurans; Hydrogenated naphthofurans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems

Definitions

  • the natural product parthenolide (PTL, 1):
  • DMAPT dimethylamino-parthenolide
  • DMAPT (also known as LC-1) is a water-soluble analogue and a clinical drug candidate with efficacy against AML stem and progenitor cells in vivo (canines), and has substantially improved bioavailability as compared to PTL.
  • LC-1 water-soluble analogue
  • canines AML stem and progenitor cells in vivo
  • a Phase I trial with DMAPT has been recently initiated in the United Kingdom for the treatment of AML (Guzman, M. L., et al. Blood 2007, 110, 4427-4435; and Neelakantan, S., et al. Bioorg. Med. Chem. Lett. 2009, 19, 4346-4349).
  • PTL has also been shown to affect other cellular processes, such as blocking STAT3 phosphorylation of IL-6, inhibiting c-Jun N-terminal kinase (JNK), depleting cellular glutathione levels, increasing cellular oxidative stress by the generation of reactive oxygen species (ROS), activation of p53 and cellular caspases, and inhibition of Hsp70.
  • JNK c-Jun N-terminal kinase
  • ROS reactive oxygen species
  • the present invention provides novel compounds that possess anti-cancer and antiinflammatory properties and that are useful for treating cancer and cardiovascular diseases. Accordingly there is provided a compound of the invention which is a compound of formula I, formula II, formula III, formula IV, or formula V:
  • X is O or C3 ⁇ 4, is absent, or taken together with the carbons to which it is attached forms a double bond;
  • Z is O or CH 2 or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
  • R 1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C 1 -C 6 )alkyl,
  • any alkyl of R 1 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R 1 is optionally substituted with one or more R b ;
  • R is H, phosphate, trifluoromethyl, (C 1 -C 6 )alkyl, (C!-C6)alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroaryl(CrC6)alkyl; wherein any alkyl of R 2 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(C 1 -C 6 )alkyl or heteroaryl(C 1 -C6)alkyl of R 2 is optionally substituted with one or more R b ;
  • R is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C 6 )alkyl,
  • any alkyl of R 3 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R 3 is optionally substituted with one or more R b ;
  • R 4 is H or OR v ;
  • R 5 is H or OR v ;
  • R 6 is H or OR v ;
  • R 7 is H or OR v ;
  • R 8 is H or OR v ;
  • R 9 is H or OR v ;
  • each R a is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (C 1 -C6)alkyl,
  • each R° and R d is independently selected from H, (d-C ⁇ alkyl, (C 3 -C 6 )cycloalkyl, (C 3 -
  • each R g and R h is independently selected from H, (C 1 -C6)alkyl, (C3-C 6 )cycloalkyl, (C 3 - C 6 )cycloalkyl(C]-C 6 )alkyl, aryl, heteroaryl, ar l(CrC6) alkyl and heteroaryl(d-C 6 ) alkyl; or R 8 andR h together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C 6 )alkyl, (C 3 -C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(d-C 6 )alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(C 1 -C6)alkyl of R 8 and R h is optionally substituted with one or more
  • each R k is independently selected from H, (C 1 -C )alkyl, (C 3 -C 6 )cycloalkyl, (C 3 -
  • each R m and R n is independently selected from H, (d-C 6 )alkyl, (C 3 -C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(CrC 6 )alkyl, aryl, heteroaryl, aryl(d-C 6 ) alkyl and heteroaryl(d-C 6 ) alkyl; or R m andR n together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C )alkyl, (C 3 -C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(Ci-C 6 )alkyl, aryl, heteroaryl, aryl(d-C 6 ) alkyl or heteroaryl(C r C 6 )alkyl of R m and R" is optionally substituted
  • each R l and R u is independently selected from H, (d-C 6 )alkyl, (C 3 -C 6 )cycloalkyl, (C 3 - C 6 )cycloalkyl(d-C 6 )alkyl, aryl, heteroaryl, aryl(C r C 6 ) alkyl and heteroaryl(d-C 6 ) alkyl; or R'andR" together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
  • each R v is H, (d-C 6 )alkyl, phosphate, (d-C 6 )alkoxycarbonyl, aryl, heteroaryl, aryl(C 1 -C6)alkyl, or heteroaryl(C 1 -C 6 )alkyl; wherein any alkyl of R v is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(C 1 -C6)alkyl or heteroaryl(d- C )alkyl of R v is optionally substituted with one or more R b ; or a salt thereof.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
  • the invention also provides a method for treating cancer in a mammal, comprising administering a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, to the mammal.
  • the invention also provides a method for treating a cardiovascular diseases (e.g. atherosclerosis) in a mammal, comprising administering a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, to the mammal.
  • a cardiovascular diseases e.g. atherosclerosis
  • the invention also provides a method for inhibiting the NF- ⁇ signaling pathway in a cell, comprising contacting the cell in vitro or in vivo with an effective amount of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof.
  • the invention provides a therapeutic method for preventing or treating a pathological condition or symptom in a mammal, such as a human, wherein the activity of the NF-KB signaling pathway is implicated and antagonism of its action is desired comprising administering to a mammal in need of such therapy, an effective amount of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof.
  • the invention provides a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of cancer.
  • the invention provides a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of a cardiovascular disease.
  • the invention provides a compound of formula I, formula ⁇ , formula III, formula IV, or formula
  • V or a pharmaceutically acceptable salt thereof, for use in medical therapy (e.g. for use in treating cancer or a cardiovascular diseases such as atherosclerosis).
  • the invention provides the use of a compound of formula I, formula II, formula ⁇ , formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of cancer in a mammal, such as a human.
  • the invention provides the use of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of a cardiovascular disease (e.g. atherosclerosis) in a mammal, such as a human.
  • a cardiovascular disease e.g. atherosclerosis
  • the invention also provides synthetic processes and synthetic intermediates disclosed herein that are useful for preparing compounds formula I, formula II, formula HI, formula IV, or formula V, or salts thereof.
  • halo is fluoro, chloro, bromo, or iodo.
  • Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as propyl embraces only the straight chain radical, a branched chain isomer such as isopropyl being specifically referred to.
  • Aryl denotes a phenyl radical or an ortho-fused bicyclic carbocyclic radical having about nine to ten ring atoms in which at least one ring is aromatic.
  • Heteroaryl encompasses a radical of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non- peroxide oxygen, sulfur, and N(Y) wherein Y is absent or is H, O, (C 1 -C4)alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms comprising one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y).
  • (d-C ⁇ alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3-pentyl, or hexyl;
  • (C 3 -C 6 )cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
  • (C 1 -C 6 )alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec- butoxy, pentoxy, 3-pentoxy, or hexyloxy;
  • (CrC ⁇ alkanoyl can be acetyl, propanoyl or butanoyl;
  • (Q-C ⁇ alkoxycarbonyl can be methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxy
  • a s ecific compound of the invention is a compound of formula la, Ila, or Ilia:
  • a s ecific compound of the invention is a compound of formula lb, Ic, or Id:
  • a s ecific compound of the invention is a compound of formula le, If, or I :
  • a specific compound of the invention is a compound of formula Im, In, or Io:
  • a s ecific compound of the invention is a compound of formula Ip, Iq, or Ir:
  • a specific compound of the invention is a compound of formula lib, lie, or lid:
  • a specific value for R 4 is H.
  • a specific value for R 5 is H.
  • a specific value for R 6 is H.
  • a specific value for R 4 is OH.
  • a specific value for R 5 is OH.
  • a specific value for R 6 is OH.
  • a specific value for R 7 is H.
  • a specific value for R 8 is H.
  • a specific value for R 9 is H.
  • a specific value for R 7 is OH.
  • a specific value for R 8 is OH.
  • a specific value for R 9 is OH.
  • a specific value for R 1 is H.
  • R 1 A specific value for R 1 is -NR c R d .
  • a specific value for R 2 is H.
  • R 2 is (C 1 -C 6 )alkyl, that is optionally substituted with one or more (e.g. 1 , 2, 3, or 4) R a .
  • a specific value for R 3 is H.
  • a specific value for R 3 is -NR c R d .
  • a specific compound of the invention is a compound of formula I, formula II, or formula III:
  • X is O or CH 2 , is absent, or taken together with the carbons to which it is attached forms a cis- double bond;
  • Z is O or C3 ⁇ 4 or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
  • R 1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C ⁇ alkyl, (C 3 -C 6 )cycloalkyl, (CrC 6 )alkoxy, (C 1 -C 6 )alkoxycarbonyl, (C 1 -C 6 )alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
  • any alkyl of R 1 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R 1 is optionally substituted with one or more R b ;
  • R is H, trifluoromethyl, (C 1 -C 6 )alkyl, (Q-C ⁇ alkoxycarbonyl, aryl, heteroaryl, arylid-C ⁇ alkyl, or heteroary ⁇ CrC ⁇ alkyl; wherein any alkyl of R 2 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(Ci-C )alkyl or heteroary ⁇ C C 6 )alkyl of R is optionally substituted with one or more R ; R is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Q-C ⁇ alkyl, (C3-C )cycloalkyl, (d-C 6 )alkoxy, (Ci-C6)alkoxycarbonyl, (Q-Csialkanoyloxy, aryl, heteroaryl, ary
  • any alkyl of R 3 is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R 3 is optionally substituted with one or more R b ;
  • R 4 is H or OR v ;
  • R 5 is H or OR v ;
  • R 6 is H or OR v ;
  • R 7 is H or OR v ;
  • R 8 is H or OR v ;
  • R 9 is H or OR v ;
  • each R a is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (Ci-C 6 )alkyl, (C 3 -C )cycloalkyl, (C 1 -C6)alkoxy, (CrC ⁇ alkoxycarbonyl, (C 1 -C 6 )alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (C 1 -C 6 )alkylthio, -S(0)R k , -S(0) 2 R k , -S(0) 3 R k , -S(0) 2 NR e R f , and -NR R f ; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C!-C6)alkyl,
  • each R b is independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C 1 -C 6 )alkyl, (C 3 -C 6 )cycloalkyl, (C]-C6)alkoxy, (Ci-C 6 )alkoxycarbonyl, (Q- C 6 )alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (C 1 -C 6 )alkylthio, -S(0)R k , -S(0) 2 R k , -S(0) 3 R k , -S(0) 2 NR m R n , and -NR m R n ; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluorometh
  • each R c and R d is independently selected from H, (CrC ⁇ alkyl, (C 3 -C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(C 1 -C6)alkyl, hydroxy, aryl, heteroaryl, ary ⁇ Q-Ce) alkyl and heteroaryliCrCe) alkyl; or R c and R d together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C 1 -C 6 )alkyl, (C 3 -C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(C 1 -C )alkyl, aryl, heteroaryl, aryl(C C 6 ) alkyl or heteroaryl(C 1 -C6)alkyl of R c and R d is optionally substituted
  • each R e and R f is independently selected from H, (C C ⁇ alkyl, (C3-C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(C 1 -C 6 )alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroaryliCi-Ce) alkyl; or R e andR f together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C3-C 6 )cycloalkyl, (C 3 - C )cycloalkyl(C 1 -C 6 )alkyl, aryl, heteroaryl, aryl(CrC 6 ) alkyl or heteroaryliQ-C ⁇ alkyl of R e and R f is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and
  • each R 8 and R h is independently selected from H, (C C ⁇ alkyl, (C3-C6)cycloalkyl, (C 3 - C 6 )cycloalkyl(C 1 -C 6 )alkyl, aryl, heteroaryl, aryl(C r C 6 ) alkyl and heteroary C Ce) alkyl; or R g andR h together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C 3 -C6)cycloalkyl, (C 3 - C f cycloalky CrC f alkyl, aryl, heteroaryl, aryl(Ci-C 6 ) alkyl or heteroaryl(C 1 -C 6 )alkyl of R g and R h is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NR l
  • each R k is independently selected from H, (C 1 -C 6 )alkyl, (C 3 -C 6 )cycloalkyl, (C 3 -
  • each R m and R n is independently selected from H, (C 1 -C 6 )alkyl, (C 3 -C 6 )cycloalkyl, (C 3 - C 6 )cycloalkyl(C 1 -C 6 )alkyl, aryl, heteroaryl, aryl(CrC 6 ) alkyl and heteroarylCCrCe) alkyl; or R m and R n together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C !
  • R m and R n is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NR l R u ;
  • each R l and R u is independently selected from H, (CrC ⁇ alkyl, (C 3 -C 6 )cycloalkyl, (C 3 - C ⁇ cycloalkylCC C ⁇ alkyl, aryl, heteroaryl, aiyl(Ci-C 6 ) alkyl and heteroarylCd-Ce) alkyl; or R l andR u together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
  • each R v is H, (d-C 6 )alkyl, (C 1 -C 6 )alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroary ⁇ d-C ⁇ alkyl; wherein any alkyl of R v is optionally substituted with one or more R a ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryliC C ⁇ alkyl or heteroaryl(C ! - C 6 )alkyl of R v is optionally substituted with one or more R b ;
  • the compound when a compound is shown with a wedged (up) or dashed (back) bond the compound may be enriched by about 60%, 80%, 90%, 95%, 98%, or 99% in the absolute stereoisomer represented.
  • a specific compound of the invention is compound 3, 4, 6, 8, 13, 14, 15, or 16, or a salt thereof.
  • a specific compound of the invention is compound 3, 4, 14, or 15, or a salt thereof.
  • the compound of the invention is not compound 1, 2, 5, 7,
  • a salt of a compound of formula I, formula II, formula III, formula IV, or formula V can be useful as an intermediate for isolating or purifying a compound of formula I.
  • administration of a compound of formula I, formula II, formula III, formula IV, or formula V as a pharmaceutically acceptable acid or base salt may be appropriate.
  • pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, fumarate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and a- glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • the compounds of formula I, formula II, formula III, formula IV, or formula V can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1 % of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as com starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • the active compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water- alcohol/ glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compounds to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No.4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compounds can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
  • the compound is conveniently formulated in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
  • the invention provides a composition comprising a compound of the invention formulated in such a unit dosage form.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • the invention also provides a composition comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, and a pharmaceutically acceptable diluent or carrier.
  • the invention also provides a kit comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, packaging material, and instructions for administering the compound of the invention or the pharmaceutically acceptable salt thereof and the other therapeutic agent or agents to an animal to treat cancer.
  • cytotoxicity of a compound of the invention may be determined using pharmacological models which are well known, or using Test A described below.
  • CCRF-CEM acute lymphoblastic leukemia cells (ATCC, CCL-119) were cultured in RPMI- 1640 media (ATCC, 30-2001) supplemented with 10% fetal bovine serum (FBS, Gibco), penicillin (100 I.U./mL), and streptomycin (100 ⁇ g/mL) (ATCC, 30-2300) at a density of 2 10 5 - 2 10 6 cells/mL.
  • HL-60 acute promyelocytic leukemia cells (ATCC, CCL-240) were cultured in IMDM media (ATCC, 30-2005) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 ⁇ g/mL) at a density of 1 10 5 - 1 10 6 cells/mL.
  • DU- 145 prostate cancer cells (ATCC, HTB-81 ) were cultured in EMEM media (ATCC, 30-2003) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 ⁇ g/mL).
  • U-87 MG glioblastoma cells (ATCC, HTB-14) were cultured in EMEM media (ATCC, 30-2003) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 ⁇ g/mL).
  • GBM6 primary glioblastoma cells (a gift from Professor John Ohlfest, University of Minnesota) were cultured under serum-free conditions utilizing Neural Stem Cell (NSC) complete growth media. This media was prepared by treating a 500 mL bottle of DMEM/F12 (1 : 1) media containing L-glutamine (ThermoSci.
  • SH3027101 with B-27 supplement without Vitamin A (10 mL of a 50X solution; Gibco 12587-010), N-2 supplement (5 mL of a 100X solution; Gibco 17502- 048), normocin (100 ⁇ g/mL; Invivogen ant-nr-1), penicillin (50 I.U./mL; Cellgro 30-001-CI), and streptomycin (50 pg/mL; Cellgro 30-001-CI).
  • cytokines human EGF (20 ng/ ⁇ , in 0.1% BS A/PBS solution; PeptroTech; 100-15) and human FGF-basic (20 ng pL in 0.1% BS A/PBS solution; PeproTech; 100-18B), were added to aliquots of the NSC media prior to splitting and plating the cell cultures. Trypsinization of GBM6 cells was achieved with the use of TrypLE Express solution (Gibco 12604013) in place of trypsin.
  • CCRF-CEM and HL-60 cells were seeded at a density of 10,000 cells/well in cell culture media (50 ⁇ ) in standard 96-well plates (Costar) 24 h prior to treatment.
  • DU- 145, U87-MG, and GBM6 cells were seeded at a density of 5,000 cells/well in cell culture media (50 L) in standard 96-well plates (Costar).
  • Blank (no cells) wells and control (vehicle control treated) wells were prepared with each experiment.
  • Alamar Blue (Invitrogen) cell viability reagent was added to each well (10 ⁇ ). This procedure yields a quantitative measure of cell viability by evaluating the ability of metabolically active cells (which are proportional to the number of living cells) to convert resazurin (non-fluorescent dye) to red-fluorescent resorufin. Fluorescence data were obtained on either a Molecular Devices SpectraMax M2 plate reader or an LJL BioSystems HT Analyst plate reader.
  • Murine Cytotoxicity Assays The anti-leukemic activities of 1-19 were studied in murine cell culture models of drug-resistant acute myeloid leukemia (AML).
  • the cell lines used are B 117P, B 117H, B 140P and B140H.
  • Bl 17P and B140P are murine AML cell lines derived from a BXH-2 strain of mice that develop AML (J Virol 1995, 69, 5095). These cell lines are cytarabine-sensitive models of AML. Continuous low-dose culturing of Bl 17P and B140P cell lines with cytarabine yielded Bl 17H and B140H, respectively, which can tolerate cytarabine concentrations 500-1000 times greater than the parental cell lines (Exp.
  • AML Colony Forming Assay AML proliferation is dependent on the existence of the cancer stem cell population, and drug-mediated elimination of AML CSCs will prohibit colony formation (Nat. Immunol. 2004, 5, 738).
  • a colony-forming assay with 5 was performed.
  • a low concentration of primary murine leukemia cells (harboring MII/AF9 and N/L4S G/2F transgenes) were treated with vehicle control or 38 ⁇ 5 and then plated in methylcellulose semi-solid media containing IL-3, IL-6, GM-CSF, and SCF.
  • individual cells were suspended in the media, and therefore, resulting colonies represent the outgrowth from single cells with replicative capacity (stem cells). Colonies were then counted for both vehicle control and 5.
  • Dosing of the vehicle control yields approximately 26 colonies per 10,000 nucleated cells in this assay. Repeating this experiment with 5 revealed no colony formation, suggesting the
  • the pharmacokinetic properties of a compound of the invention may be determined using pharmacological models which are well known, or using Test B described below.
  • PK Pharmacokinetics
  • the signal was optimized for each compound by ESI positive or negative ionization mode.
  • An MS2 scan or a SIM scan was used to optimize the fragmenter voltage and a product ion analysis was used to identify the best fragment for analysis, and the collision energy was optimized using a product ion or MRM scan.
  • An ionization ranking was assigned indicating the compound's ease of ionization.
  • Plasma and brain samples were thawed on ice and kept at 4 °C during processing. Brain tissues were homogenized in 50 mM potassium phosphate, pH 7.4. An aliquot of plasma or brain homogenate sample or calibration sample were mixed with three volumes of methanol containing internal standard, incubated on ice for 5 min, and centrifuged. The protein-free supernatant was used for analysis.
  • Test Article Formulation Test articles were formulated in 0.5% methylcellulose; the dosing solution was freshly made on the dosing day and dosed the animals immediately: PO at 100 mg/Kg, (10 mg/mL, 10 mL/kg). The following is the study table:
  • mice were provided a single dose by body weight via oral gavage at 100 mg/Kg (10 mg/mL, 10 mL/kg) Sampling. Blood samples were collected into K 2 EDTA microtainer tubes for plasma separation. Blood samples were centrifuged at 4 °C at 6,000 RPM for 5 minutes. Decanted plasma samples were stored at -80 °C until analysis. Brains were perfused and collected for the oral study. The tissue samples were stored at -80 °C until analysis.
  • brain ng/g
  • brain ng/g
  • DMAPT Dimethylamino parthenolide
  • a 0.20 M solution of Zn(CH 2 I) 2 DME complex was made in the following manner: diethyl zinc (1.0 M solution in hexanes, 4.0 mL, 4 mmol) was added to CH 2 C1 2 (20 mL) and DME (0.50 mL) at 0 °C under N 2 . Diiodomethane (0.80 mL, 9.92 mmol) was added and the mixture was stirred for 10 minutes. This material was added dropwise over 10 minutes to a solution of parthenolide (1, 90 mg, 0.36 mmol) in CH 2 C1 2 (2 mL) at 0 °C. The mixture was stirred for 1 hour at 0 °C, and the stirring was continued overnight with gradual warming to room temperature.
  • the crude material was quenched by pouring into NH 4 CI (saturated, aq., 20 mL) and then extracted with CH 2 C1 2 (20 mL, 4x). The combined organic layers were washed with NaHC0 3 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na 2 S0 4 .
  • the crude material was MPLC purified using Si0 2 (10-30% EtOAc in hexanes over 15 minutes) to yield compound 3 (36 mg, 40%) as a colorless oil and recovered 1 (37 mg, 41%).
  • Cyclopropyl parthenolide (3, 35.0 mg, 0.134 mmol) was dissolved in anhydrous MeOH (1 mL). Dimethylamine (2.0 M solution in methanol, 100.0 ⁇ , 0.200 mmol) was added and the mixture was allowed to stir overnight at room temperature. The solvent and excess dimethylamine were removed in vacuo, and the crude material was MPLC purified using Si0 2 (5-30% MeOH in CH 2 C1 2 over 15 minutes) to afford 4 (34.0 mg, 83%).
  • This molecule was synthesized from melampomagnolide B (9) by photochemical isomerization of the CI -CIO olefin. A protocol for this reaction is described for a related molecule in Eur. J. Org. Chem. 2003, 2003, 3969.
  • This molecule was synthesized from melampomagnolide B (9) by incorporation of the dimethylamino group as described in the synthesis of 4.
  • This molecule is the C 1 -C 10 isomer of parthenolide and was prepared according to the known method in Eur. J. Org. Chem. 2003, 2003, 3969. HRMS calcd for (C 17 H 27 N0 4 + H) + 310.2018, found 310.2019
  • This molecule is dimethylamino costunolide. It was synthesized by addition of a dimemylamino group to costunolide (commercial) according to the method described in the synthesis of 4. Example 10. Preparation of Compound 12.
  • This molecule is a glutathione adduct of parthenolide and was prepared by reacting parthenolide
  • Example 17 The following illustrate representative pharmaceutical dosage forms, containing a compound of the invention, or a pharmaceutically acceptable salt thereof ('Compound X'), for therapeutic or prophylactic use in humans.

Abstract

The invention provides compounds of formula I, formula II, formula III, formula IV, and formula V: and salts thereof wherein X, Z, ---, and R1-R9 have any of the values defined in the specification, as well as compositions comprising the compounds and methods for their use in therapy. The compounds are useful for the treatment of cancer, the treatment of anti-inflammatory diseases, and for the treatment of cardiovascular diseases.

Description

Anti-cancer and Anti-inflammatory Parthenolide Compounds
Priority of Invention
This application claims priority to United States Provisional Application Number 61/477,380 that was filed on April 20, 2011. The entire content of this provisional application is hereby incorporated herein by reference.
Background of the Invention
The natural product parthenolide (PTL, 1):
Figure imgf000002_0001
a component of traditional herbal remedy from the feverfew plant Tanacetum parthenium, has demonstrated cytotoxic activity against a spectrum of human cancers, including brain cancer, breast cancer, chronic lymphocytic leukemia, pancreatic cancer, and prostate cancer. See Anderson, K. N., et al. J Pharmacol. Sci. 2008, 106, 318-320; Nakshatri, H., et al. Oncogene 2004, 23, 7330-7344; Sweeney, C. J., et al. Mol. Cancer Ther. 2005, 4, 1004-1012; Steele, A. J., et al. Leukemia 2006, 20, 1073-1079; Ramachandran, P. V., et al. Bioorg. Med. Chem. Lett. 2010, 20, 6620-6623; Sweeney, C, et al. Clinical Cancer Research 2004, 10, 5501-5507; and Shanmugam, R., et al. Prostate 2006, 66, 1498- 1511.
Excitement towards the therapeutic potential of PTL was heightened in 2005 when PTL was shown to induce apoptosis in primary human acute myeloid leukemia (AML) cells and blast crisis chronic myeloid leukemia (CML) cells without targeting normal hematopoietic stem cells (HSCs) (Guzman, M. L., et al. Blood 2005, 105, 4163-4169). PTL was also shown to induce apoptosis in AML CD34+/CD38" cells, which are AML cancer stem cells, and diminish the repopulating ability of AML in immunocompromised mice (Guzman, M. L., et al. Blood 2005, 105, 4163-4169). However, the clinical utility of PTL has been limited due to its poor water solubility and low serum bioavailability (Sweeney, C. J., et al. Mol. Cancer Ther. 2005, 4, 1004-1012). To overcome this limitation, a prodrug of PTL, dimethylamino-parthenolide (DMAPT, 2), has been prepared:
Figure imgf000003_0001
DMAPT (also known as LC-1) is a water-soluble analogue and a clinical drug candidate with efficacy against AML stem and progenitor cells in vivo (canines), and has substantially improved bioavailability as compared to PTL. Notably, a Phase I trial with DMAPT has been recently initiated in the United Kingdom for the treatment of AML (Guzman, M. L., et al. Blood 2007, 110, 4427-4435; and Neelakantan, S., et al. Bioorg. Med. Chem. Lett. 2009, 19, 4346-4349).
Despite the promising cytotoxicity of PTL, the mechanism of action by which PTL targets CSCs is unclear. Previous studies have characterized PTL as an inhibitor of NF-κΒ signaling via the covalent modification of ΙΚΚβ kinase and the alkylation of NF-κΒ p65 protein (Garcia-Pineres, A. J., et al. J. Biol. Chem. 2001, 276, 39713-39720; Hehner, S. P., et al. J. Immunol. 1999, 163, 5617-5623; and Kwok, B. H. B., et al. Chem. Biol. 2001, 8, 1 '59-766). However, others have shown that inhibition of NF-KB signaling in primary AML cells with an ΙκΒ superrepressor allele, which abolishes NF-KB activation via production a degradation-resistant ΙκΒα suppressor protein, fails to promote the loss of leukemia CSC viability (Guzman, M. L., et al. Proc. Natl. Acad. Sci. USA 2002, 99, 16220-16225). PTL has also been shown to affect other cellular processes, such as blocking STAT3 phosphorylation of IL-6, inhibiting c-Jun N-terminal kinase (JNK), depleting cellular glutathione levels, increasing cellular oxidative stress by the generation of reactive oxygen species (ROS), activation of p53 and cellular caspases, and inhibition of Hsp70. See Sobota, R., et al. Biochem. Biophys. Res. Comm. 2000, 267, 329-333; Won, Y. K., et al. Carcinogenesis 2004, 25, 1449-1458; Wen, J., et al. J. Biol. Chem. 2002, 277, 38954-38964; Zhang, S. Y, et al. Cancer Letters 2004, 211, 175-188; and Pei, S. S., et al. In American Society of Hematology Conference Abstract 2734 2009; Vol. 114.
The level to which any or all of these events contribute to PTL's cytotoxicity towards AML CSCs is largely unknown. However, the feature that PTL and DMAPT (LC-1) can eradicate CSC populations and not target normal hematopoietic stem cells make these natural products extremely promising molecules for future development.
Currently, there is a need for novel anti-cancer agents, especially those agents that target differentiated as well as stem cell populations in cancer (Wang, J. C. Y. Cell Stem Cell 2007, 1, 497- 501 ; and Park, C. Y., et al. Mol. Ther. 2009, 17, 219-230). Therapy regimens that fail to address cancer stem cell populations are predicted to fail by the cancer stem cell model (Ward, R. J., et al. Ann. Rev. Pathol. Mech. 2007, 2, 175-189). There is also an additional need for novel parthenolide compounds that can be used to study the mechanism of action of parthenolide and that have improved potency or pharmacological properties. Summary of the Invention
The present invention provides novel compounds that possess anti-cancer and antiinflammatory properties and that are useful for treating cancer and cardiovascular diseases. Accordingly there is provided a compound of the invention which is a compound of formula I, formula II, formula III, formula IV, or formula V:
Figure imgf000004_0001
wherein: X is O or C¾, is absent, or taken together with the carbons to which it is attached forms a double bond;
Z is O or CH2 or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
R1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl,
(C3-C6)cycloalkyl, (Ci-C6)alkoxy, (CrCeialkoxycarbonyl, (C]-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000005_0001
or -NRcRd; wherein any alkyl of R1 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R1 is optionally substituted with one or more Rb;
R is H, phosphate, trifluoromethyl, (C1-C6)alkyl, (C!-C6)alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroaryl(CrC6)alkyl; wherein any alkyl of R2is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(C1-C6)alkyl or heteroaryl(C1-C6)alkyl of R2 is optionally substituted with one or more Rb;
R is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C6)alkyl,
(C3-C6)cycloalkyl, (CrC6)alkoxy, (Ct-C^alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000005_0002
or -NRcRd; wherein any alkyl of R3 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R3 is optionally substituted with one or more Rb;
R4 is H or ORv;
R5 is H or ORv;
R6 is H or ORv;
R7 is H or ORv;
R8 is H or ORv;
R9 is H or ORv;
each Ra is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (C1-C6)alkyl,
(C3-C6)cycloalkyl, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (Q-C^alkylthio, -OP(=0)(OH)2, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NReRf, and -NReRf; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (CrC^alkyl, (C3-C6)cycloalkyl, (CrCe^lkoxy, (Ci-C^alkoxycarbonyl, (C\- C6)alkanoyloxy,
Figure imgf000006_0001
-S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NR8Rh, and -NR8Rh;
each Rb is independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Q-C^alkyl, (C3-C6)cycloalkyl, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, (C\- C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (Q^^lkylthio, -OP(=0)(OH)2, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C6)alkyl, (C3-C6)cycloalkyl, (Cr C6)alkoxy, (CrC6)alkoxycarbonyl, (C1-C6)alkanoyloxy, (Q-C^alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRraRn;
each R° and Rd is independently selected from H, (d-C^alkyl, (C3-C6)cycloalkyl, (C3-
CeicycloalkylCQ-C^alkyl, hydroxy, aryl, heteroaryl, aryl(C!-C6) alkyl and heteroary^CrCe) alkyl; or Rc and Rd together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3-
Figure imgf000006_0002
aryl, heteroaryl, arylCQ-Q) alkyl or heteroaryl(C]-C6)alkyl of Rc and Rd is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu; each Re and Rf is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(Ci-C6) alkyl and heteroaryl(d-C6) alkyl; or R andRf together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C]-C6)alkyl, aryl, heteroaryl, aryl(C!-C6) alkyl or heteroaryl(C1-C6)alkyl of Re and Rf is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rg and Rh is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C]-C6)alkyl, aryl, heteroaryl, ar l(CrC6) alkyl and heteroaryl(d-C6) alkyl; or R8andRh together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(d-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(C1-C6)alkyl of R8 and Rh is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rk is independently selected from H, (C1-C )alkyl, (C3-C6)cycloalkyl, (C3-
C6)cycloalkyl(C!-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl and heteroaryl(d-C6) alkyl;
each Rm and Rn is independently selected from H, (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(CrC6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl and heteroaryl(d-C6) alkyl; or RmandRn together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C )alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(Ci-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(CrC6)alkyl of Rm and R" is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rl and Ru is independently selected from H, (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(d-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroaryl(d-C6) alkyl; or R'andR" together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
each Rv is H, (d-C6)alkyl, phosphate, (d-C6)alkoxycarbonyl, aryl, heteroaryl, aryl(C1-C6)alkyl, or heteroaryl(C1-C6)alkyl; wherein any alkyl of Rv is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(C1-C6)alkyl or heteroaryl(d- C )alkyl of Rv is optionally substituted with one or more Rb; or a salt thereof.
The invention also provides a pharmaceutical composition comprising a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
The invention also provides a method for treating cancer in a mammal, comprising administering a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, to the mammal.
The invention also provides a method for treating a cardiovascular diseases (e.g. atherosclerosis) in a mammal, comprising administering a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, to the mammal.
The invention also provides a method for inhibiting the NF-κΒ signaling pathway in a cell, comprising contacting the cell in vitro or in vivo with an effective amount of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof.
Additionally, the invention provides a therapeutic method for preventing or treating a pathological condition or symptom in a mammal, such as a human, wherein the activity of the NF-KB signaling pathway is implicated and antagonism of its action is desired comprising administering to a mammal in need of such therapy, an effective amount of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof.
The invention provides a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of cancer.
The invention provides a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of a cardiovascular disease.
The invention provides a compound of formula I, formula Π, formula III, formula IV, or formula
V, or a pharmaceutically acceptable salt thereof, for use in medical therapy (e.g. for use in treating cancer or a cardiovascular diseases such as atherosclerosis).
The invention provides the use of a compound of formula I, formula II, formula ΠΙ, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of cancer in a mammal, such as a human. The invention provides the use of a compound of formula I, formula II, formula III, formula IV, or formula V, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of a cardiovascular disease (e.g. atherosclerosis) in a mammal, such as a human.
The invention also provides synthetic processes and synthetic intermediates disclosed herein that are useful for preparing compounds formula I, formula II, formula HI, formula IV, or formula V, or salts thereof.
Detailed Description
The following definitions are used, unless otherwise described: halo is fluoro, chloro, bromo, or iodo. Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as propyl embraces only the straight chain radical, a branched chain isomer such as isopropyl being specifically referred to. Aryl denotes a phenyl radical or an ortho-fused bicyclic carbocyclic radical having about nine to ten ring atoms in which at least one ring is aromatic. Heteroaryl encompasses a radical of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non- peroxide oxygen, sulfur, and N(Y) wherein Y is absent or is H, O, (C1-C4)alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms comprising one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(Y). As used herein (e.g. in the definition for R2 and Rv) "phosphate" means a group -P(=0)(OH)2.
It will be appreciated by those skilled in the art that compounds of the invention having a chiral center may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein, it being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
Specific values listed below for radicals, substituents, and ranges, are for illustration only; they do not exclude other defined values or other values within defined ranges for the radicals and substituents.
Specifically, (d-C^alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3-pentyl, or hexyl; (C3-C6)cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; (C1-C6)alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec- butoxy, pentoxy, 3-pentoxy, or hexyloxy; (CrC^alkanoyl can be acetyl, propanoyl or butanoyl; (Q-C^alkoxycarbonyl can be methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, or hexyloxycarbonyl; (C2-C6)alkanoyloxy can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy; aryl can be phenyl, indenyl, or naphthyl; and heteroaryl can be furyl, imidazolyl, triazolyl, triazinyl, oxazoyl, isoxazoyl, thiazolyl, isothiazoyl, pyrazolyl, pyrrolyl, pyrazinyl, tetrazolyl, pyridyl, (or its N-oxide), thienyl, pyrimidinyl (or its N-oxide), indolyl, isoquinolyl (or its N-oxide) or quinolyl (or its N-oxide).
A s ecific compound of the invention is a compound of formula la, Ila, or Ilia:
Figure imgf000010_0001
or a salt thereof.
A s ecific compound of the invention is a compound of formula lb, Ic, or Id:
Figure imgf000010_0002
or a salt thereof. A s ecific compound of the invention is a compound of formula le, If, or I :
Figure imgf000011_0001
or a salt thereof.
A specific compound of the invention is a compound of formula Im, In, or Io:
Figure imgf000011_0002
or a salt thereof.
A s ecific compound of the invention is a compound of formula Ip, Iq, or Ir:
Figure imgf000011_0003
a salt thereof.
A specific compound of the invention is a compound of formula lib, lie, or lid:
Figure imgf000012_0001
or a salt thereof.
A specific value for R4 is H.
A specific value for R5 is H.
A specific value for R6 is H.
A specific value for R4 is OH.
A specific value for R5 is OH.
A specific value for R6 is OH.
A specific value for R7 is H.
A specific value for R8 is H.
A specific value for R9 is H.
A specific value for R7 is OH.
A specific value for R8 is OH.
A specific value for R9 is OH.
A specific value for R1 is H.
A specific value for R1 is -NRcRd.
A specific value for R2 is H.
A specific value for R2 is (C1-C6)alkyl, that is optionally substituted with one or more (e.g. 1 , 2, 3, or 4) Ra .
A specific value for R2 is -P(=0)(OH)2.
A specific value for R3 is H.
A specific value for R3 is -NRcRd. A specific compound of the invention is a compound of formula I, formula II, or formula III:
Figure imgf000013_0001
wherein:
X is O or CH2, is absent, or taken together with the carbons to which it is attached forms a cis- double bond;
Z is O or C¾ or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
R1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C^alkyl, (C3-C6)cycloalkyl, (CrC6)alkoxy, (C1-C6)alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000013_0002
or -NR°Rd; wherein any alkyl of R1 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R1 is optionally substituted with one or more Rb;
R is H, trifluoromethyl, (C1-C6)alkyl, (Q-C^alkoxycarbonyl, aryl, heteroaryl, arylid-C^alkyl, or heteroary^CrC^alkyl; wherein any alkyl of R2 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(Ci-C )alkyl or heteroary^C C6)alkyl of R is optionally substituted with one or more R ; R is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Q-C^alkyl, (C3-C )cycloalkyl, (d-C6)alkoxy, (Ci-C6)alkoxycarbonyl, (Q-Csialkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000014_0001
or -NRcRd; wherein any alkyl of R3 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R3 is optionally substituted with one or more Rb;
R4 is H or ORv;
R5 is H or ORv;
R6 is H or ORv;
R7 is H or ORv;
R8 is H or ORv;
R9 is H or ORv;
each Ra is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (Ci-C6)alkyl, (C3-C )cycloalkyl, (C1-C6)alkoxy, (CrC^alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (C1-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NReRf, and -NR Rf; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C!-C6)alkyl, (C3-C6)cycloalkyl, (Q-C^alkoxy, (C1-C6)alkoxycarbonyl, (Q- C6)alkanoyloxy, (C1-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NR8Rh, and -NRgRh;
each Rb is independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C]-C6)alkoxy, (Ci-C6)alkoxycarbonyl, (Q- C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (C1-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Cj-C6)alkyl, (C3-C6)cycloalkyl, (d-C6)alkoxy, (Q- C6)alkoxycarbonyl, (C C6)alkanoyloxy,
Figure imgf000015_0001
-S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn;
each Rc and Rd is independently selected from H, (CrC^alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, hydroxy, aryl, heteroaryl, ary^Q-Ce) alkyl and heteroaryliCrCe) alkyl; or Rc and Rd together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C )alkyl, aryl, heteroaryl, aryl(C C6) alkyl or heteroaryl(C1-C6)alkyl of Rc and Rd is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Re and Rf is independently selected from H, (C C^alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroaryliCi-Ce) alkyl; or ReandRf together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any
Figure imgf000015_0002
(C3-C6)cycloalkyl, (C3- C )cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl or heteroaryliQ-C^alkyl of Re and Rf is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each R8 and Rh is independently selected from H, (C C^alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroary C Ce) alkyl; or RgandRh together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any
Figure imgf000015_0003
(C3-C6)cycloalkyl, (C3- Cf cycloalky CrCf alkyl, aryl, heteroaryl, aryl(Ci-C6) alkyl or heteroaryl(C1-C6)alkyl of Rg and Rh is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rk is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3-
C6)cycloalkyl(C]-C6)alkyl, aryl, heteroaryl, aryl(C]-C6) alkyl and heteroaryl(Ci-C6) alkyl;
each Rm and Rn is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroarylCCrCe) alkyl; or Rm and Rn together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C!-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl or heteroaryl(C1-C6)alkyl of Rm and Rn is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rl and Ru is independently selected from H, (CrC^alkyl, (C3-C6)cycloalkyl, (C3- C^cycloalkylCC C^alkyl, aryl, heteroaryl, aiyl(Ci-C6) alkyl and heteroarylCd-Ce) alkyl; or RlandRu together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
each Rv is H, (d-C6)alkyl, (C1-C6)alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroary^d-C^alkyl; wherein any alkyl of Rv is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryliC C^alkyl or heteroaryl(C!- C6)alkyl of Rv is optionally substituted with one or more Rb;
or a salt thereof.
Figure imgf000016_0001
or a salt thereof.
Figure imgf000017_0001
or a salt thereof.
In one embodiment of the invention, when a compound is shown with a wedged (up) or dashed (back) bond the compound may be enriched by about 60%, 80%, 90%, 95%, 98%, or 99% in the absolute stereoisomer represented.
A specific compound of the invention is compound 3, 4, 6, 8, 13, 14, 15, or 16, or a salt thereof.
Figure imgf000018_0001
1 (Parthenolide) 2 (DMAPT; LC-1)
Figure imgf000018_0002
A specific compound of the invention is compound 3, 4, 14, or 15, or a salt thereof.
In one embodiment of the invention, the compound of the invention is not compound 1, 2, 5, 7,
9, 10, 11, 12, 17, 18, or 19, or a salt thereof.
In cases where compounds are sufficiently basic or acidic, a salt of a compound of formula I, formula II, formula III, formula IV, or formula V can be useful as an intermediate for isolating or purifying a compound of formula I. Additionally, administration of a compound of formula I, formula II, formula III, formula IV, or formula V as a pharmaceutically acceptable acid or base salt may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, fumarate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and a- glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
The compounds of formula I, formula II, formula III, formula IV, or formula V can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
Thus, the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1 % of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as com starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.
The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
For topical administration, the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water- alcohol/ glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
Examples of useful dermatological compositions which can be used to deliver the compounds to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No.4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
Useful dosages of the compounds can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
In general, however, a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
The compound is conveniently formulated in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form. In one embodiment, the invention provides a composition comprising a compound of the invention formulated in such a unit dosage form. The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
Compounds of the invention can also be administered in combination with other therapeutic agents, for example, other agents that are useful for the treatment of cancer. Accordingly, in one embodiment the invention also provides a composition comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, and a pharmaceutically acceptable diluent or carrier. The invention also provides a kit comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, at least one other therapeutic agent, packaging material, and instructions for administering the compound of the invention or the pharmaceutically acceptable salt thereof and the other therapeutic agent or agents to an animal to treat cancer.
The cytotoxicity of a compound of the invention may be determined using pharmacological models which are well known, or using Test A described below.
Test A.
Mammalian Cell Culture. Cell lines were maintained in a humidified 5% C02 environment at 37 °C. CCRF-CEM acute lymphoblastic leukemia cells (ATCC, CCL-119) were cultured in RPMI- 1640 media (ATCC, 30-2001) supplemented with 10% fetal bovine serum (FBS, Gibco), penicillin (100 I.U./mL), and streptomycin (100 μg/mL) (ATCC, 30-2300) at a density of 2 105 - 2 10 6 cells/mL. HL-60 acute promyelocytic leukemia cells (ATCC, CCL-240) were cultured in IMDM media (ATCC, 30-2005) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 μg/mL) at a density of 1 105 - 1 10 6 cells/mL. DU- 145 prostate cancer cells (ATCC, HTB-81 ) were cultured in EMEM media (ATCC, 30-2003) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 μg/mL). U-87 MG glioblastoma cells (ATCC, HTB-14) were cultured in EMEM media (ATCC, 30-2003) supplemented with 10% FBS, penicillin (100 I.U./mL), and streptomycin (100 μg/mL). GBM6 primary glioblastoma cells (a gift from Professor John Ohlfest, University of Minnesota) were cultured under serum-free conditions utilizing Neural Stem Cell (NSC) complete growth media. This media was prepared by treating a 500 mL bottle of DMEM/F12 (1 : 1) media containing L-glutamine (ThermoSci. SH3027101) with B-27 supplement without Vitamin A (10 mL of a 50X solution; Gibco 12587-010), N-2 supplement (5 mL of a 100X solution; Gibco 17502- 048), normocin (100 μg/mL; Invivogen ant-nr-1), penicillin (50 I.U./mL; Cellgro 30-001-CI), and streptomycin (50 pg/mL; Cellgro 30-001-CI). In addition, cytokines, human EGF (20 ng/μΐ, in 0.1% BS A/PBS solution; PeptroTech; 100-15) and human FGF-basic (20 ng pL in 0.1% BS A/PBS solution; PeproTech; 100-18B), were added to aliquots of the NSC media prior to splitting and plating the cell cultures. Trypsinization of GBM6 cells was achieved with the use of TrypLE Express solution (Gibco 12604013) in place of trypsin.
Mammalian Cytotoxicity Assays. CCRF-CEM and HL-60 cells were seeded at a density of 10,000 cells/well in cell culture media (50 μί) in standard 96-well plates (Costar) 24 h prior to treatment. DU- 145, U87-MG, and GBM6 cells were seeded at a density of 5,000 cells/well in cell culture media (50 L) in standard 96-well plates (Costar). Blank (no cells) wells and control (vehicle control treated) wells were prepared with each experiment. Compounds were serially diluted in pre-warmed media and dosed to cells (final volume/well = 100 μΐ,; final DMSO concentration = 0.5%). Approximately 2 h before the end of the treatment period (48 h), Alamar Blue (Invitrogen) cell viability reagent was added to each well (10 μΕ). This procedure yields a quantitative measure of cell viability by evaluating the ability of metabolically active cells (which are proportional to the number of living cells) to convert resazurin (non-fluorescent dye) to red-fluorescent resorufin. Fluorescence data were obtained on either a Molecular Devices SpectraMax M2 plate reader or an LJL BioSystems HT Analyst plate reader. Background fluorescence (no cell controls) was subtracted from each well and cellular viability values following compound treatment were normalized to vehicle-only treated wells (control wells only treated with aqueous DMSO, which were arbitrarily assigned 100% viability). Individual IC50 curves were generated by fitting data to the sigmoidal (dose response) function of varied slope in GraphPad Prism (v. 5.0) software. Only curve fits with r2 > 0.95 were deemed sufficient. Each experiment was performed in biological triplicate and mean IC50 values (with standard deviation) were calculated from the individual IC50 values obtained from each replicate.
Murine Cytotoxicity Assays. The anti-leukemic activities of 1-19 were studied in murine cell culture models of drug-resistant acute myeloid leukemia (AML). The cell lines used are B 117P, B 117H, B 140P and B140H. Bl 17P and B140P are murine AML cell lines derived from a BXH-2 strain of mice that develop AML (J Virol 1995, 69, 5095). These cell lines are cytarabine-sensitive models of AML. Continuous low-dose culturing of Bl 17P and B140P cell lines with cytarabine yielded Bl 17H and B140H, respectively, which can tolerate cytarabine concentrations 500-1000 times greater than the parental cell lines (Exp. Hematol. 2006, 34, 631). These cell lines are cytarabine-resistant models of AML and are excellent tools for studying drug resistant disease. Importantly, the Bl 17P cells display phenotypic markers consistent with stem cells or early progenitors, such as CD34 and c-kit, while the B140P cells do not. This diversity between the cell lines, plus the abundance of knowledge already accumulated, makes these lines a valuable tool for studying drug responses in AML. Cell viability against these cell lines was measured by standard MTS colorimetric assay.
Data for representative compounds of the invention are shown in the following tables.
DU-145 Prostate Cancer Cells
Figure imgf000024_0001
Figure imgf000024_0002
CCRF-CEM T-Cell Leukemia Cells
Figure imgf000025_0001
HL-60 Acute Myelogenous Leukemia Cells
Figure imgf000025_0002
U-87 MG Glioblastoma Cells
Figure imgf000026_0001
GBM6 Primary Glioblastoma Cells
Figure imgf000026_0002
Screening in Murine AML Cell lines B117P, B117H, B140P, and B140H
Figure imgf000027_0001
AML Colony Forming Assay. AML proliferation is dependent on the existence of the cancer stem cell population, and drug-mediated elimination of AML CSCs will prohibit colony formation (Nat. Immunol. 2004, 5, 738). A colony-forming assay with 5 was performed. A low concentration of primary murine leukemia cells (harboring MII/AF9 and N/L4SG/2Ftransgenes) were treated with vehicle control or 38 μΜ 5 and then plated in methylcellulose semi-solid media containing IL-3, IL-6, GM-CSF, and SCF. As a result, individual cells were suspended in the media, and therefore, resulting colonies represent the outgrowth from single cells with replicative capacity (stem cells). Colonies were then counted for both vehicle control and 5. Dosing of the vehicle control yields approximately 26 colonies per 10,000 nucleated cells in this assay. Repeating this experiment with 5 revealed no colony formation, suggesting the
Figure imgf000027_0002
The pharmacokinetic properties of a compound of the invention may be determined using pharmacological models which are well known, or using Test B described below.
Test B. Pharmacokinetics (PK) Studies in Mice Pharmacokinetics of 2 and 4. A pharmacokinetics study was performed with compounds 2 and 4 in CD1 male mice. Both compounds 2 and 4 were synthesized as their mono-fumarate salts. Testing was performed at Apredica in Watertown, MA by contract service. Details of the PK experiment are as follows:
LC/MS/MS Method development. The signal was optimized for each compound by ESI positive or negative ionization mode. An MS2 scan or a SIM scan was used to optimize the fragmenter voltage and a product ion analysis was used to identify the best fragment for analysis, and the collision energy was optimized using a product ion or MRM scan. An ionization ranking was assigned indicating the compound's ease of ionization.
Analysis. Samples were analyzed by LC/MS/MS using an Agilent 6410 mass spectrometer coupled with an Agilent 1200 HPLC and a CTC PAL chilled autosampler, all controlled by MassHunter software (Agilent). After separation on a CI 8 reverse phase HPLC column (Agilent, Waters, or equivalent) using an acetonitrile-water gradient system, peaks were analyzed by mass spectrometry (MS) using ESI ionization in MRM mode.
Sample preparation. Plasma and brain samples were thawed on ice and kept at 4 °C during processing. Brain tissues were homogenized in 50 mM potassium phosphate, pH 7.4. An aliquot of plasma or brain homogenate sample or calibration sample were mixed with three volumes of methanol containing internal standard, incubated on ice for 5 min, and centrifuged. The protein-free supernatant was used for analysis.
Calibration samples. A working dilution of test agent in DMSO at 50 times the final concentration was prepared and serially diluted samples were prepared. These samples were diluted 50-fold into Mouse plasma or brain homogenate and processed as above.
Animals. Species and sex: Mouse, male. Strain: CD1. Number: 12. All procedures in this protocol are in compliance with the Animal Welfare Act, the Guide for the Care and Use of Laboratory Animals, and the Office of Laboratory Animal Welfare. In the opinion of the Sponsor and Principal Investigator, this study did not unnecessarily duplicate any previous work.
Study Design. Test Article Formulation: Test articles were formulated in 0.5% methylcellulose; the dosing solution was freshly made on the dosing day and dosed the animals immediately: PO at 100 mg/Kg, (10 mg/mL, 10 mL/kg). The following is the study table:
Molecule Route Type of # of animals Dose Sample Collection
Tested Mouse (mg/Kc ) Collected time points
4 PO CD-1 3 100 Plasma 1, 4 hours
4 PO CD-1 3 100 Brain 1, 4 hours
2 PO CD-1 3 100 Plasma 1, 4 hours
2 PO CD-1 3 100 Brain — 1, 4 hours
Test Article administration. Mice were provided a single dose by body weight via oral gavage at 100 mg/Kg (10 mg/mL, 10 mL/kg) Sampling. Blood samples were collected into K2EDTA microtainer tubes for plasma separation. Blood samples were centrifuged at 4 °C at 6,000 RPM for 5 minutes. Decanted plasma samples were stored at -80 °C until analysis. Brains were perfused and collected for the oral study. The tissue samples were stored at -80 °C until analysis.
Clinical Observations. There were no clinical observations to report as all the mice were normal.
Results. The following tables show the results from the PK study of 2 and 4.
Compound 2 Measured in Mouse Plasma;
Figure imgf000029_0001
Compound 2 Measured in Mouse Brain:
Mouse ID Dosage Time (hr) Compound in Compound 1 in
brain (ng/g) brain (ng/g)
1-1 PO (100 mg/kg) 1 4996 2129
1-2 PO (100 mg/kg) 1 7505 2676
1-4 PO (100 mg/kg) 4 240 74.7
1-4 PO (100 mg/kg) 4 204 66.1
1-4 PO (100 mg/kg) 4 581 197
Compound 4 Measured in Mouse Plasma:
Mouse ID Dosage Time (hr) Compound 4 in Compound 3 in
plasma (ngf mL) plasma (ng/mL)
1-1 PO (100 mg/kg) 1 525 106
1-2 PO (100 mg/kg) 1 368 103
1-4 PO (100 mg/kg) 4 10.0 16.2
1-4 PO (100 mg/kg) 4 5.3 15.8
1-4 PO (100 mg/kg) 4 7.6 1.7
Compound 4 Measured in Mouse Brain:
Figure imgf000029_0002
The above results demonstrate that representative compounds of the invention (compounds 2 and 4) accumulate to micromolar levels in serum and, therefore, reach efficacious levels with respect to their IC50 anticancer activties as determined by cell culture experiments. Therefore, biologically relevant levels of these molecules can be achieved in serum. Additionally, both compounds 2 and 4 are highly brain penetrant and accumulate to higher levels in brain tissue than serum. The ratio of brain/serum levels (about 2:1) surpasses that observed with temozolomide (about 1 :2.5), which is the front-line small molecule used to treat glioblastomas. Since most small molecules against brain tumors fail due to inability to cross the blood-brain barrier (or are rapidly removed by efflux mechanisms), the feature that these molecules accumulate to high levels in brain tissue is significant.
The invention will now be illustrated by the following non-limiting Examples.
EXAMPLES
General. Chemical reagents were typically from Aldrich or Acros and were used without additional purification unless explicitly noted. Solvents were from Fisher Scientific. Parthenolide (1) was purchased from Enzo Life Sciences and repurified by Si02 chromatography before use. Unless otherwise noted, reactions were performed under an atmosphere of dry nitrogen. MPLC chromatography was preformed on a Teledyne-Isco Combiflash Rf-200 instrument utilizing Redisep Rf Gold High Performance silica gel columns or self-packed columns with SiliCycle SiliaFlash 60A silica gel. Preparative HPLC purifications were performed on an Agilent 1200 series instrument equipped with a Zorbax SB-C18 column (21.2 x 250 mm, 7 μπι; Agilent Technologies). Nuclear magnetic resonance (NMR) spectroscopy employed a Bruker Avance 400 MHz (for lH) instrument. Chemical shifts were normalized to internal solvent peaks or tetramethylsilane. X-ray crystal structures were solved by Victor G. Young, Jr. at the X-Ray Crystallographic Laboratory, University of Minnesota, Department of Chemistry.
Dimethylamino parthenolide (DMAPT; LC-1; compound 2) was prepared as previously described (Bioorg. Med. Chem. Lett. 2009, 19, 4346).
Example 1. Preparation of Compound 3
A 0.20 M solution of Zn(CH2I)2 DME complex was made in the following manner: diethyl zinc (1.0 M solution in hexanes, 4.0 mL, 4 mmol) was added to CH2C12 (20 mL) and DME (0.50 mL) at 0 °C under N2. Diiodomethane (0.80 mL, 9.92 mmol) was added and the mixture was stirred for 10 minutes. This material was added dropwise over 10 minutes to a solution of parthenolide (1, 90 mg, 0.36 mmol) in CH2C12 (2 mL) at 0 °C. The mixture was stirred for 1 hour at 0 °C, and the stirring was continued overnight with gradual warming to room temperature. The crude material was quenched by pouring into NH4CI (saturated, aq., 20 mL) and then extracted with CH2C12 (20 mL, 4x). The combined organic layers were washed with NaHC03 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na2S04. The crude material was MPLC purified using Si02 (10-30% EtOAc in hexanes over 15 minutes) to yield compound 3 (36 mg, 40%) as a colorless oil and recovered 1 (37 mg, 41%). 1H NMR (400 MHz, CDC13) δ: 6.28 (d, J= 3.5 Hz, 1H), 5.57 (d, J= 3.3 Hz, 1H), 3.96 (t, J= 3.3 Hz, 1H), 2.98 (d, J= 9.0 Hz, 1H), 2.67 (ddd, J= 9.1, 5.9, 2.8 Hz, 1H), 2.39 (dd, J= 14.7, 7.9 Hz, 1H), 2.19 (dd, J= 8.3, 2.3 Hz, 1H), 2.00 - 1.88 (m, 2H), 1.70 (ddd, J= 15.9, 10.7, 5.8 Hz, 1H), 1.40 (s, 3H), 1.30 -1.24 (m, 2H), 1.09 (s, 3H), -0.08 (dd, J= 5.7, 4.4 Hz, 1H), 0.39 (dd, J= 9.5 4.3 Hz, 1H), 0.64 (td, J= 9.5, 5.9 Hz, 1H), 0.85 (dd, J = 14.6, 11.0 Hz, 1H),. 13C NMR (100 MHz, CDC13) δ: 169.4, 139.86, 120.5,. 82.7, 65.5, 60.6, 48.0, 42.3, 38.4, 25.7, 24.5, 22.3, 20.5, 18.8, 18.5, 17.1. The structure of 3 (shown below) was further confirmed b small molecule X-ray crystallography.
Figure imgf000032_0001
Example 2. Preparation of Compound 4
Cyclopropyl parthenolide (3, 35.0 mg, 0.134 mmol) was dissolved in anhydrous MeOH (1 mL). Dimethylamine (2.0 M solution in methanol, 100.0 μί, 0.200 mmol) was added and the mixture was allowed to stir overnight at room temperature. The solvent and excess dimethylamine were removed in vacuo, and the crude material was MPLC purified using Si02 (5-30% MeOH in CH2C12 over 15 minutes) to afford 4 (34.0 mg, 83%). 1H NMR (400 MHz, CDC13) S: 3.94 (t, J= 9.4 Hz, 1H), 2.93 (d,J= 9.0 Hz, 1H), 2.73 (dd,J= 13.0, 5.3, lH), 2.59 (dd, J= 13.3, 5.3 Hz, 1H), 2.40 (dt,J= 12.3, 5.5 Hz, 1H), 2.33 - 2.21 (m, 7H), 2.20 - 2.07 (m, 2H), 1.97 - 1.95 (m, 1H), 1.83 (dd, J= 15.6, 8.8 Hz, 1H), 1.64 (ddd,J= 16.3, 9.8, 6.0 Hz, 1H), 1.40 (s, 3H), 1.30 - 1.24 (m, 2H), 1.07 (s, 3H), 0.81 (dd,J= 14.8, 10.3 Hz, 1H), 0.64 (td, J= 9.8, 6.0 Hz, 1H), 0.37 (dd, J= 9.4, 4.1 Hz, 1H),-0.10 (dd, J= 5.8, 4.3 Hz, 1H). 13C NMR (100 MHz, CDC13) δ: 176.5, 82.4, 65.38, 60.6, 57.5, 48.3, 47.0, 45.9, 42.2, 38.6, 25.3, 24.4, 22.6, 20.3, 18.8, 18.5, 17.1. The fumarate salt of compound 4 was also prepared using standard procedures. The fumarate salt of compound 4 is also a representative compound of the invention.
Example 3. Preparation of Compound 5
This compound was synthesized by a modification to the procedure reported in J. Nat. Prod 1993, 59, 90. In brief, parthenolide (1, 20 mg, 0.08 mmol) was dissolved in toluene (0.8 mL) and BF3 Et20 (5 uL, 0.048 mmol) was added to the solution at 0 °C. The reaction was stirred at room temperature for 1 hour and quenched by the addition of NaHC03 (saturated, aq., 20 mL). The crude material was diluted with CH2C12 (20 mL) and then extracted with CH2C12 (20 mL, 4x). The combined organic layers were washed with NaHC03 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na2S04. The crude material was MPLC purified on Si02 (10-35% EtOAc in hexanes over 14 min) to yield compound 5 (11 mg, 55%). 1H NMR (400 MHz, CDC13) δ: 6.21 (d, J= 3.3 Hz, 1H), 5.50 (d,J= 3.0 Hz, 1H), 3.81 (t, J= 10.3 Hz, 1H), 2.74 - 2.62 (m, 3H), 2.39 (dd,J= 16.2, 8.4 Hz, 1H), 2.27 - 2.23 (m, 2H), 2.13 - 2.05 (m, 1H), 1.88 - 1.72 (m, 2H), 1.69 (s, 3H), 1.63 (br. s., 1H), 1.42 - 1.29 (m, 4H).
Example 4. Preparation of Compound 6.
This compound was prepared from 5, followed by installation of the dimethylamino group as described in the synthesis of 4. 1H NMR (400 MHz, CDC13) S: 3.83 (t, J- 10.4 Hz, 1H), 2.76 (dd, J= 12.8, 5.2 Hz, 1H), 2.63 (m, 2H), 2.45 - 2.35 (m, 3H), 2.30 (s, 6H), 2.17 - 1.98 (m, 5H), 1.84 - 1.72 (m, 2H), 1.67 (s, 3H), 1.31 - 1.25 (m, 4H).
Example 5. Preparation of Compound 7.
This molecule was synthesized from melampomagnolide B (9) by photochemical isomerization of the CI -CIO olefin. A protocol for this reaction is described for a related molecule in Eur. J. Org. Chem. 2003, 2003, 3969.
Example 6. Preparation of Compound 8.
This molecule was synthesized from melampomagnolide B (9) by incorporation of the dimethylamino group as described in the synthesis of 4.
Example 7. Preparation of Compound 9.
This compound is melampomagnolide B and was prepared from parthenolide (1) according to the method described in Phytochemistry 1984, 23, 2372. 1H NMR (400 MHz, CDC13) δ: 6.32 (d, J= 3.6 Hz, 1H), 5.61 (d, J= 3.6 Hz, 1H), 5.20 (dd, J= 12.1 Hz, 2.4 Hz, 1H), 3.85 (t, J= 8.6 Hz, 1H), 2.79 - 2.76 (m, 2H), 2.43 (dd,J= 13.7, 5.2 Hz, 1H), 2.37 (dd,J= 13.0, 5.2 Hz, 1H), 2.20 - 2.11 (m, 4H), 1.76 - 1.67 (m, 4H), 1.29 - 1.19 (m, 4H). Example 8. Preparation of Compound 10.
This molecule is the C 1 -C 10 isomer of parthenolide and was prepared according to the known method in Eur. J. Org. Chem. 2003, 2003, 3969. HRMS calcd for (C17H27N04 + H)+ 310.2018, found 310.2019
Example 9. Preparation of Compound 11.
This molecule is dimethylamino costunolide. It was synthesized by addition of a dimemylamino group to costunolide (commercial) according to the method described in the synthesis of 4. Example 10. Preparation of Compound 12.
This molecule was synthesized as previously described (Bioorg. Med. Chem. 2006, 14, 83). ^ NMR (400 MHz, CDC13) «5: 5.17 (d, J= 12.0 Hz, 1H), 3.80 (t, J= 9.2 Hz, 1H), 2.69 (d, J= 8.8 Hz, 1H), 2.79 - 2.76 (m, 2H), 2.39 - 2.26 (m, 3H), 2.18 - 2.01 (m, 3H), 1.83 - 1.92 (m, 2H), 1.69 (s, 3H), 1.29 (s, 3H), 1.76 - 1.67 (m, 4H), 1.24 - 1.20 (m, 4H).
Example 11. Preparation of Compound 13.
This molecule was synthesized from a mixture of 14 and 15 by addition of the dimethylamino group as described in the synthesis of 4. Example 12. Preparation of Compounds 14 and 15.
To a solution of compound 2 in THF (5 mL) in a Parr bomb was added Pd/C (10% w/w, 60.0 mg). The bomb was charged with H2 (50 psi) and the reaction was stirred at room temperature for 24 hours. The crude reaction products were then filtered through a pad of Celite and the pad was washed with CH2C12 (15 mL, 4x). Concentration of the eluant in vacuo afforded the hydrogenated products as a mixture of diasteromers. The crude reaction products were redissolved in THF (2 mL), treated with CH3I (0.10 mL, 1.59 mmol), and allowed to stir overnight at room temperature. The material was then concentrated in vacuo to a yellow solid, resuspended in H20 (5.0 mL), and stirred at 50 C for 30 minutes. The heterogeneous solution solubilized within a few minutes. NaHC03 (saturated, aq., 20 mL) was added to the flask, and the crude material was extracted with CH2C12 (20 mL, 4x). The combined extracts were washed with NaHC03 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na2S04. The solvent was removed in vacuo and the crude material was purified by preparative HPLC (Zorbax SB-C18 column; 10-95% MeCN in H20 over 47 min) to give compounds 14 and 15 (11.8 mg, 63%) as a mixture of diastereomers. The mixture was then subjected to further preparative HPLC purification (Zorbax SB-C18 column; isocratic 40:50: 10 H20:MeCN:/-PrOH) to provide separated 14 (tR = 55 min) and 15 (tR = 59 min). Compound 14 (4.40 mg, 23%): 1H NMR (400 MHz, CDC13) δ: 6.25 (dd, J= 11.2, 4.2 Hz, 1H), 5.54 (d, J= 3.2 Hz, 1H), 3.96 (t,J= 8.9 Hz, 1H), 3.10 (d,J= 8.7 Hz, 1H), 2.98 - 2.78 (m, 1H), 2.25 - 2.10 (m, 2H), 1.73 (ddd, J= 13.0, 8.3, 4.2 Hz, 1H), 1.67 - 1.49 (m, 4H), 1.39 - 1.14 (m, 5H), 1.04 - 0.84 (m, 3H); 13C NMR (100 MHz, CDCl3) <S: 169.5, 139.4, 120.5, 81.5, 63.7, 61.9, 49.4, 39.5, 38.4, 36.5, 33.4, 28.8, 22.7, 21.7, 18.6; HRMS calcd for (C15H2203 + Na)+ 273.1467, found 273.1470; HPLC retention time, 15.71 min. Compound 15 (7.40 mg, 39%). 1HNMR (400 MHz, CDCI3) δ: 6.16 (d, J= 3.5 Hz, 1H), 5.45 (d, J= 3.2 Hz, 1H), 3.76 (t, J- 9.6 Hz, 1H), 3.02 (d, J= 9.6 Hz, 1H), 2.89 (ddd, J= 10.0, 6.9, 3.6 Hz, 1H), 2.19 - 1.93 (m, 2H), 1.87 - 1.63 (m, 2H), 1.45 - 1.27 (m, 5H), 1.18 (s, 2H), 1.16 - 0.99 (m, 2H), 0.95 - 0.77 (m, 4H); 13C NMR (100 MHz, CDCl3) cS: 169.8, 139.6, 119.8, 81.1, 66.5, 61.4, 44.0, 36.8, 36.2, 30.2, 28.0, 24.8, 21.4, 20.7, 19.3; HRMS calcd for (Ci5H2203 +Na)+ 273.1467, found 273.1463; HPLC retention time, 15.84 min. The structure of 15 (shown below) was confirmed by small molecule X-ra crystallography.
Figure imgf000035_0001
Example 13. Preparation of Compound 16.
To a stirred solution of compound 8 (40 mg, 0.133 mmol) in dry THF was added Pd/C (100 mg). The reaction was then stirred under hydrogen gas for 2 days at room temperature. Pd/C was removed by filtration through celite. Mel (50 μί, 0.803 mmol) was added to the resulting filtrate and the reaction was allowed to stir overnight. The solvent was removed under reduced pressure and the residue was resuspended in H20 (1 mL) and heated to 50 °C for 30 min. NaHC03 (saturated, aq., 20 mL) was added to the flask, and the crude material was extracted with CH2C12 (20 mL, 4x). The combined extracts were washed with NaHC03 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na2S04. The solvent was removed in vacuo and the crude material was purified by preparative HPLC (Zorbax SB-C18 column; 10-95% MeCN in H20 over 47 min) to give compounds 16 (5 mg, 15%). HRMS calcd for (C17H29N03 + H)+ 296.2226, found 296.2222
Example 14. Preparation of Compound 17.
This molecule is a glutathione adduct of parthenolide and was prepared by reacting parthenolide
(1) with glutathione.
Example 15. Preparation of Compound 18.
This compound was isolated as a minor product (8% yield) from the reaction that yielded compound 5. 1H NMR (400 MHz, CDC13) S: 6.19 (dd, J= 13.6, 3.8 Hz, IH), 5.47 (t, J= 3.5 Hz, IH), 5.10 (d, J= 3.1 Hz, IH), 4.00 (dd, J= 9.8, 3.3 Hz, IH), 3.03 - 2.94 (m, IH), 2.65 - 2.35 (m, 2H), 2.27 - 1.94 (m, 2H) 1.82 - 1.76 (m, 3H), 1.68 - 1.53 (m, 2H), 1.32 - 1.24 (m, 2H), 1.07 (dd, J= 6.9, 2.8 Hz, 3H). Example 16. Preparation of Compound 19.
To a stirred solution of compound 5 (20 mg, 0.081 mmol) in pyridine (1 mL) was added POCl3 (75 μί, 0.81 mmol) dropwise at 0 C. The reaction was warmed to room temperature and stirred for 2 hours. Pyridine was removed under reduced pressure and the residue was dissolved in DCM (2 mL). NaHC03 (saturated, aq., 20 mL) was added to the flask, and the crude material was extracted with CH2C12 (20 mL, 4x). The combined extracts were washed with NaHC03 (saturated, aq., 20 mL), brine (saturated, aq., 20 mL) and then dried over Na2S04. The crude material was MPLC purified on Si02 (10-50% EtOAc in hexanes over 18 min) to yield compound 19 (12 mg, 65%). 1H NMR (400 MHz, CDC13) δ: 6.11 (d, J= 3.2 Hz, IH), 5.52 (m, IH), 5.37 (d, J= 3.0 Hz, IH), 3.63 (t, J= 9.8 Hz, IH), 3.39 (d, J= 10.0 Hz, IH), 2.96 (br s, 2H), 2.81 - 2.74 (m, IH), 2.30 (t, j= 13.3 Hz, IH), 2.16 (ddd, J= 14.8, 5.8, 2.0 Hz, IH), 2.08 (dq, J= 13.1, 2.3 Hz, IH), 1.93 (s, 3H), 1.71 (d, J= 0.8 Hz, 3H). HRMS calcd for (C15H2204 + Na)+289.1416, found 289.1421. Example 17. The following illustrate representative pharmaceutical dosage forms, containing a compound of the invention, or a pharmaceutically acceptable salt thereof ('Compound X'), for therapeutic or prophylactic use in humans.
(i) Tablet 1 mg/tablet
Compound X= 100.0
Lactose 77.5
Povidone 15.0
Croscarmellose sodium 12.0
Microcrystalline cellulose 92.5
Magnesium stearate
300.0
(ii Tablet 2 me/tablet
Compound X= 20.0
Microcrystalline cellulose 410.0
Starch 50.0
Sodium starch glycolate 15.0
Magnesium stearate 5J0
500.0
(iii Capsule mg/capsule
Compound X= 10.0
Colloidal silicon dioxide 1.5
Lactose 465.5
Pregelatinized starch 120.0
Magnesium stearate 3J)
600.0 (iv) Injection 1 (1 mg/ml) mg/ml
Compound X= (free acid form) 1.0
Dibasic sodium phosphate 12.0
Monobasic sodium phosphate 0.7
Sodium chloride 4.5
1.0 N Sodium hydroxide solution
(pH adjustment to 7.0-7.5) q.s.
Water for injection q.s. ad 1 mL
(v) Injection 2 (10 mg/ml) mg/ml
Compound X= (free acid form) 10.0
Monobasic sodium phosphate 0.3
Dibasic sodium phosphate 1.1
Polyethylene glycol 400 200.0
01 N Sodium hydroxide solution
(pH adjustment to 7.0-7.5) q.s.
Water for injection q.s. ad 1 mL (vi) Aerosol mg/can
Compound X= 20.0
Oleic acid 10.0
Trichloromonofluoromethane 5,000.0
Dichlorodifluoromethane 10,000.0
Dichlorotetrafluoroethane 5,000.0
The above formulations may be obtained by conventional procedures well known in the pharmaceutical art.
All publications, patents, and patent documents are incorporated by reference herein, as though individually incorporated by reference. The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention.

Claims

What is claimed is
1. A compound of formula I, formula II, formula III, formula IV, or formula V:
Figure imgf000039_0001
wherein:
X is O or CH2, is absent, or taken together with the carbons to which it is attached forms a double bond;
Z is O or CH2 or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
R1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C!-C6)alkoxy, (C1-C6)alkoxycarbonyl, (CrC6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000040_0001
or -NRcRd; wherein any alkyl of R1 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R1 is optionally substituted with one or more Rb;
R is H, phosphate, trifluoromethyl, (Ci-C^alkyl, (Q-C^alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroaryliCrC^alkyl; wherein any alkyl of R is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(C1-C6)alkyl or heteroaryl(Ci-C6)alkyl of R is optionally substituted with one or more R ;
R3 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (d-C6)alkoxy, (C1-C6)alkoxycarbonyl, (CrC^alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000040_0002
or -NRcRd; wherein any alkyl of R3 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R3 is optionally substituted with one or more Rb;
R4 is H or ORv;
R5 is H or ORv;
R6 is H or ORv;
R7 is H or ORv: R is H or ORv;
R9 is H or ORv;
each Ra is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (d-C )alkyl, (C3-C6)cycloalkyl, (Ci-C6)alkoxy, (C1-C6)alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (d-C6)alkylthio, -OP(=0)(OH)2, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -SCO^NR^, and -NReRf; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (d-C6)alkoxy, (C1-C6)alkoxycarbonyl, (d- C6)alkanoyloxy, (CrC6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRgRh, and -NR8Rh;
each Rb is independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, (d- C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (Ci-C6)alkylthio, -OP(=0)(OH)2, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (d-C6)alkyl, (C3-C6)cycloalkyl, (d- C6)alkoxy, (d-C6)alkoxycarbonyl, (d-C6)alkanoyloxy, (d-C6)allcylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn;
each Rc and Rd is independently selected from H, (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C )alkyl, hydroxy, aryl, heteroaryl, aryl(d-C6) alkyl and heteroaryl(d-C ) alkyl; or R° and Rd together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(d-C6)alkyl of Rc and Rd is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Re and Rf is independently selected from H, (d-C6)alkyl, (C3-C )cycloalkyl, (C3-
C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl and heteroaryl(d-C6) alkyl; or ReandRf together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C6)alkyl, (C3-C )cycloalkyl, (C3- C6)cycloalkyl(d-C6)alkyl, aryl, heteroaryl, aryl(d-C ) alkyl or heteroaryl(d-C6)alkyl of Re and Rf is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each R8 and Rh is independently selected from H, (d-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, ary C Ce) alkyl andheteroaryKC Ce) alkyl; orRgandRh together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (Ci-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C!-C6)alkyl, aryl, heteroaryl, aryl(Ci-C6) alkyl or heteroaryl(C1-C6)alkyl of Rg and Rh is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rk is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3-
Figure imgf000042_0001
alkyl and heteroaryl(Ci-C6) alkyl;
each Rm and R" is independently selected from H, (d-C^alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(Ci-C6)alkyl, aryl, heteroaryl, aryl(C1-C6) alkyl and heteroaryl(Ci-C6) alkyl; or RmandRn together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any
Figure imgf000042_0002
(C3-C )cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(C1-C6) alkyl or heteroaryl(C1-C6)alkyl of Rm and Rn is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rl and Ru is independently selected from H, (Ci-C6)alkyl, (C3-C6)cycloalkyl, (C3- C^cycloalky^Q-C^alkyl, aryl, heteroaryl, ary C Ce) alkyl and heteroaryl(Ci-C6) alkyl; or R^dR" together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
each Rv is H, (d-C6)alkyl, phosphate, (CrC^alkoxycarbonyl, aryl, heteroaryl, ary Q-C^alkyl, or heteroaryl(C1-C6)alkyl; wherein any alkyl of Rv is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any arylid-C^alkyl or heteroary^C C6)alkyl of Rv is optionally substituted with one or more Rb;
or a salt thereof.
compound of claim 1 which is a compound of formula I, formula Π, or formula III:
Figure imgf000043_0001
wherein:
X is O or C¾, is absent, or taken together with the carbons to which it is attached forms a cis- double bond;
Z is O or CH2 or taken together with the carbons to which it is attached forms a double bond; the bond represented by— is a single or a double bond;
R1 is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl,
Figure imgf000043_0002
aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000043_0003
or -NRcRd; wherein any alkyl of R1 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R1 is optionally substituted with one or more Rb;
R2 is H, trifluoromethyl, (C1-C6)alkyl, (Q-C^alkoxycarbonyl, aryl, heteroaryl, aryliQ-C^alkyl, or heteroaryl(Ci-C6)alkyl; wherein any alkyl of R2 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryltd-C^alkyl or heteroary^Ci- C6)alkyl of R is optionally substituted with one or more R ; R is H, halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, (Q-C^alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, a group of formula:
Figure imgf000044_0001
or -NR°Rd; wherein any alkyl of R3 is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryloxy, or heteroaryloxy of R3 is optionally substituted with one or more Rb;
R4 is H or OR1
R5 is H or ORv;
R6 is H or ORv
R7 is H or ORv
R8 is H or ORv
R9 is H or ORv
each Ra is independently selected from halo, cyano, nitro, hydroxy, carboxy, oxo, (Q-C^alkyl, (C3-C6)cycloalkyl, (C Ceialkoxy, (C1-C6)alkoxycarbonyl, (C1-C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy, (Ci-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NReRf, and -NReRf; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (Ci-C )ak , (C3-C )cycloalkyl, (C1-C6)alkoxy, (Q-C^alkoxycarbonyl, (C\- C6)alkanoyloxy, (C1-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRgRh, and -NRgRh;
each Rb is independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C]-C )alkyl, (C3-C6)cycloalkyl, (C1-C6)alkoxy, (C1-C6)alkoxycarbonyl, (Cr C6)alkanoyloxy, aryl, heteroaryl, aryloxy, heteroaryloxy,
Figure imgf000044_0002
-S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn; wherein each aryl, heteroaryl, aryloxy, and heteroaryloxy is optionally substituted with one or more groups independently selected from halo, cyano, nitro, hydroxy, carboxy, trifluoromethyl, trifluoromethoxy, (C1-C6)alkyl, (C3-C6)cycloalkyl, (CrC6)alkoxy, (d- C6)alkoxycarbonyl, (d-C6)alkanoyloxy, (C1-C6)alkylthio, -S(0)Rk, -S(0)2Rk, -S(0)3Rk, -S(0)2NRmRn, and -NRmRn;
each Rc and Rd is independently selected from H, (Ci-C6)alkyl, (C3-C )cycloalkyl, (C3- C6)cycloalkyl(CrC6)alkyl, hydroxy, aryl, heteroaryl, aryl(d-C ) alkyl and heteroaryl(d-C6) alkyl; or Rc and Rd together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(CrC )alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(d-C )alkyl of Rc and Rd is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Re and Rf is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl and heteroarylid-Ce) alkyl; or ReandRf together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(Ci-C6) alkyl or heteroaryl(d-C6)alkyl of Re and Rf is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each R8 and Rh is independently selected from H, (C1-C6)alkyl, (C3-C )cycloalkyl, (C3- C6)cycloalkyl(C]-C )alkyl, aryl, heteroaryl, aryl(CrC6) alkyl andheteroaryl(d-C6) alkyl; or RgandRh together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(d-C6)alkyl of R8 and Rh is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rk is independently selected from H, (Ci-C6)alkyl, (C3-C6)cycloalkyl, (C3-
C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(CrC6) alkyl and heteroaryl(d-C6) alkyl;
each Rm and R" is independently selected from H, (C1-C6)alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(Ci-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl and heteroaryl(Ci-C6) alkyl; or RmandRn together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; wherein any (d-C )alkyl, (C3-C )cycloalkyl, (C3- C6)cycloalkyl(C1-C6)alkyl, aryl, heteroaryl, aryl(d-C6) alkyl or heteroaryl(Ci-C6)alkyl of Rm and Rn is optionally substituted with one or more groups independently selected from hydroxy, carboxy, and NRlRu;
each Rl and Ru is independently selected from H, (CrC^alkyl, (C3-C6)cycloalkyl, (C3- C6)cycloalkyl(Ci-C )alkyl, aryl, heteroaryl, aryl(Ci-C6) alkyl and heteroary^Q-Ce) alkyl; or RlandRu together with the nitrogen to which they are attached form a aziridino, azetidino, morpholino, piperazino, pyrrolidino or piperidino; and
each Rv is H, (C1-C6)alkyl, (C1-C6)alkoxycarbonyl, aryl, heteroaryl, aryl(Ci-C6)alkyl, or heteroarylid-C^alkyl; wherein any alkyl of Rv is optionally substituted with one or more Ra ; and wherein any aryl, heteroaryl, or any aryl or heteroaryl portion of any aryl(Ci-C )alkyl or heteroaryl(Ci- C6)alkyl of Rv is optionally substituted with one or more Rb;
or a salt thereof.
Figure imgf000046_0001
or a salt thereof.
4. The com ound of claim 1 which is a compound of formula lb, Ic, or Id:
Figure imgf000046_0002
or a salt thereof.
5. The compound of claim 1 which is a compound of formula le, If, or I :
Figure imgf000047_0001
a salt thereof.
6. The compound of claim 1 which is a compound of formula Im, In, or Io:
Figure imgf000047_0002
a salt thereof.
7. The compound of claim 1 which is a compound of formula Ip, Iq, or Ir:
Figure imgf000047_0003
a salt thereof.
8. The compound of claim 1 which is a compound of formula lib, lie, or lid:
Figure imgf000048_0001
or a salt thereof.
9. The compound of claim 1 which is a com ound formula IVc, or Va:
Figure imgf000048_0002
or a salt thereof.
10. The compound of claim 1 which is a compound of formula IVd, or Vb:
Figure imgf000048_0003
or a salt thereof.
11. The compound of claim 1 which is compound 3, 4, 6, 8, 13, 14, 15, or 16, or a salt thereof.
12. The compound of claim 1 which is not compound 1, 2, 5, 7, 9, 10, 11, 12, 17, 18, or 19, or a salt thereof.
13. The compound of any one of claims 1-7 wherein R1 is H.
14. The compound of any one of claims 1-7 wherein R1 is -NRcRd. 15. The compound of claim 1 , 2, 3, 8, or 9 wherein R2 is H.
16. The compound of claim 1 , 2, 3, 8, or 9 wherein R2 is (C1-C6)alkyl, that is optionally substituted with one or more Ra . 17. The compound of claim 1, 2, 3, 8, or 9 wherein R is H.
18. The compound of claim 1 , 2, 3, 8, or 9 wherein R3 is -NRcRd.
19. A pharmaceutical composition comprising a compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable diluent or carrier.
20. A method for treating cancer in a mammal, comprising administering a compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof, to the mammal. 21. A method for treating a cardiovascular disease in a mammal, comprising administering a compound as described in any one of claims 1 - 18, or a pharmaceutically acceptable salt thereof, to the mammal.
22. A method for inhibiting the NF-κΒ signaling pathway in a cell, comprising contacting the cell in vitro or in vivo with an effective amount of a compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof.
23. A method for preventing or treating a pathological condition or symptom in a mammal, such as a human, wherein the activity of the NF-κΒ signaling pathway is implicated and antagonism of its action is desired comprising administering to a mammal in need of such therapy, an effective amount of a compound as described in any one of claims 1-14, or a pharmaceutically acceptable salt thereof.
24. The method of claim 20 wherein the cancer is a blood cancer, or a cancerous solid tumor.
25. The method of claim 20 wherein the cancer is leukemia, breast, pancreatic, or prostate cancer. 26. The method of claim 20 wherein the cancer is a cancer that has an established cancer stem cell population.
27. A compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof, for use in medical therapy.
28. A compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of cancer.
29. A compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof, for use in the prophylactic or therapeutic treatment of a cardiovascular disease.
30. The use of a compound as described in any one of claims 1 - 18, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of cancer in a mammal. 31. The invention provides the use of compound as described in any one of claims 1-18, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament useful for the treatment of a cardiovascular disease in a mammal.
PCT/US2012/034527 2011-04-20 2012-04-20 Anti-cancer and anti-inflammatory parthenolide compounds WO2012145678A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161477380P 2011-04-20 2011-04-20
US61/477,380 2011-04-20

Publications (1)

Publication Number Publication Date
WO2012145678A1 true WO2012145678A1 (en) 2012-10-26

Family

ID=46018127

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/034527 WO2012145678A1 (en) 2011-04-20 2012-04-20 Anti-cancer and anti-inflammatory parthenolide compounds

Country Status (1)

Country Link
WO (1) WO2012145678A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014067379A1 (en) * 2012-10-30 2014-05-08 天津尚德药缘科技有限公司 Deuterated dimethyl amine parthenolide, preparation method and use thereof in drug preparation
CN104211669A (en) * 2013-06-05 2014-12-17 天津尚德药缘科技有限公司 Micheliolide derivative and preparation method and use thereof
CN104876899A (en) * 2014-02-28 2015-09-02 天津尚德药缘科技股份有限公司 Dimethylamino sphaelactone fumarate and use thereof
WO2016090166A1 (en) * 2014-12-03 2016-06-09 Board Of Trustees Of The University Of Arkansas Melampomagnolide b dimers
US9469650B2 (en) 2013-11-08 2016-10-18 Board Of Trustees Of The University Of Arkansas Melampomagnolide B derivatives
US9487536B2 (en) 2013-11-08 2016-11-08 Board Of Trustees Of The University Of Arkansas Melampomagnolide B derivatives
EP3019165A4 (en) * 2013-07-11 2017-01-25 The Board of Trustees of the University of Arkansas Dehydroleucodine derivatives and uses thereof
CN106366088A (en) * 2015-07-24 2017-02-01 天津尚德药缘科技股份有限公司 Parthenolide derivative, and medicinal composition, preparation method and use thereof
EP3003317A4 (en) * 2013-06-06 2017-04-19 University Of Rochester Parthenolide derivatives, methods for their preparation and their use as anticancer agents
DE102015117882A1 (en) * 2015-10-21 2017-04-27 Mehrdad Ghashghaeinia Pharmaceutical composition
CN108484550A (en) * 2018-05-07 2018-09-04 中国人民解放军第四军医大学 One kind having a smile on one's face lactone derivatives and its preparation method and application
CN109897022A (en) * 2017-12-07 2019-06-18 中国医学科学院药物研究所 Lactone derivatives, its pharmaceutical composition and its preparation method and application with a smile
US10428082B2 (en) 2016-01-29 2019-10-01 Bioventures, Llc Triazole derivatives of melampomagnolide B and methods of use thereof
CN111303099A (en) * 2018-12-12 2020-06-19 天津尚德药缘科技股份有限公司 Sphaelactone dimethylamine fumarate crystal form F and preparation method thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor

Non-Patent Citations (39)

* Cited by examiner, † Cited by third party
Title
ANDERSON, K. N. ET AL., J PHARMACOL. SCI., vol. 106, 2008, pages 318 - 320
BIOORG. MED CHEM. LETT., vol. 19, 2009, pages 4346
BIOORG. MED. CHEM., vol. 14, 2006, pages 83
ESSAM ABDEL SATTAR ET AL: "Antitumor Germacranolides from Anvillea garcinii", JOURNAL OF NATURAL PRODUCTS, vol. 59, no. 4, 1 January 1996 (1996-01-01), pages 403 - 405, XP055029130, ISSN: 0163-3864, DOI: 10.1021/np960064g *
EUR. J. ORG. CHEM., vol. 2003, 2003, pages 3969
EXP. HEMATOL., vol. 34, 2006, pages 631
GARCIA-PINERES, A. J. ET AL., J BIOL. CHEM., vol. 276, 2001, pages 39713 - 39720
GUZMAN, M. L. ET AL., BLOOD, vol. 105, 2005, pages 4163 - 4169
GUZMAN, M. L. ET AL., BLOOD, vol. 110, 2007, pages 4427 - 4435
GUZMAN, M. L. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 99, 2002, pages 16220 - 16225
HEHNER, S. P. ET AL., J. IMMUNOL., vol. 163, 1999, pages 5617 - 5623
J NAT. PROD, vol. 59, 1993, pages 90
J. ALBERTO MARCO ET AL: "New Germacranolides and Eudesmanolides from North African Artemisia herba-alba", JOURNAL OF NATURAL PRODUCTS, vol. 57, no. 7, 1 July 1994 (1994-07-01), pages 939 - 946, XP055029105, ISSN: 0163-3864, DOI: 10.1021/np50109a010 *
J. VIROL, vol. 69, 1995, pages 5095
JOSÉ CASTAÑEDA-ACOSTA ET AL: "The molecular structures of three germacrolides related to parthenolide", JOURNAL OF CHEMICAL CRYSTALLOGRAPHY, vol. 27, no. 11, 1 November 1997 (1997-11-01), pages 635 - 639, XP055029099, ISSN: 1074-1542, DOI: 10.1007/BF02576404 *
KWOK, B. H. B. ET AL., CHEM. BIOL., vol. 8, 2001, pages 759 - 766
MOHAMED MOUMOU ET AL: "10[alpha]-Hydroxy-4,9-dimethyl-13-(pipyridin-1-ylmethyl)-3,8,15-trioxatetracyclo[10.3.0.0 2,4 .0 7,9 ]tetradecan-14-one", ACTA CRYSTALLOGRAPHICA SECTION E STRUCTURE REPORTS ONLINE, vol. 67, no. 11, 22 October 2011 (2011-10-22), pages O3003 - O3004, XP055029148, DOI: 10.1107/S1600536811042644 *
MOHAMED MOUMOU ET AL: "10[alpha]-Hydroxy-4,9-dimethyl-13-[(4-phenylpiperazin-1-yl)methyl]-3,8,15-trioxatetracyclo[10.3.0.0 2,4 .0 7,9 ]tetradecan-14-one", ACTA CRYSTALLOGRAPHICA SECTION E STRUCTURE REPORTS ONLINE, vol. 67, no. 11, 22 October 2011 (2011-10-22), pages O2981 - O2982, XP055029145, DOI: 10.1107/S1600536811042012 *
MOHAMED MOUMOU ET AL: "10[alpha]-Hydroxy-4,9-dimethyl-13-[(pyrrolidin-1-yl)methyl]-3,8,15-trioxatetracyclo[10.3.0.0 2,4 .0 7,9 ]pentadecan-14-one", ACTA CRYSTALLOGRAPHICA SECTION E STRUCTURE REPORTS ONLINE, vol. 67, no. 7, 30 June 2011 (2011-06-30), pages O1842 - O1843, XP055029149, DOI: 10.1107/S1600536811024688 *
NAKSHATRI, H. ET AL., ONCOGENE, vol. 23, 2004, pages 7330 - 7344
NAT. IMMUNOL., vol. 5, 2004, pages 738
NEELAKANTAN, S. ET AL., BIOORG. MED. CHEM. LETT., vol. 19, 2009, pages 4346 - 4349
PARK, C. Y. ET AL., MOL. THER., vol. 17, 2009, pages 219 - 230
PEI, S. S. ET AL., AMERICAN'SOCIETY OFHEMATOLOGY CONFERENCE ABSTRACT, vol. 114, 2009, pages 2734
PHYTOCHEMISTRY, vol. 23, 1984, pages 2372
RAMACHANDRAN, P. V. ET AL., BIOORG. MED CHEM. LETT., vol. 20, 2010, pages 6620 - 6623
SHANMUGAM, R ET AL., PROSTATE, vol. 66, 2006, pages 1498 - 1511
SOBOTA, R. ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 267, 2000, pages 329 - 333
STEELE, A. J. ET AL., LEUKEMIA, vol. 20, 2006, pages 1073 - 1079
STEFFEN WAGNER ET AL: "Neural Networks as Valuable Tools To Differentiate between Sesquiterpene Lactones' Inhibitory Activity on Serotonin Release and on NF-[kappa]B", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, no. 5, 1 March 2008 (2008-03-01), pages 1324 - 1332, XP055028926, ISSN: 0022-2623, DOI: 10.1021/jm701318x *
SWEENEY, C. J. ET AL., MOL. CANCER THER., vol. 4, 2005, pages 1004 - 1012
SWEENEY, C., AL. CLINICAL CANCER RESEARCH, vol. 10, 2004, pages 5501 - 5507
WAGNER S ET AL: "Development of a Structural Model for NF-.kappa.B Inhibition of Sesquiterpene Lactones Using Self-Organizing Neural Networks", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 49, no. 7, 1 January 2006 (2006-01-01), pages 2241 - 2252, XP002536718, ISSN: 0022-2623, DOI: 10.1021/JM051125N *
WANG, J. C. Y., CELL STEM CELL, vol. 1, 2007, pages 497 - 501
WARD, R. J. ET AL., ANN. REV. PATHOL. MECH., vol. 2, 2007, pages 175 - 189
WEDGE D E ET AL: "Fungicidal activity of natural and synthetic sesquiterpene lactone analogs", PHYTOCHEMISTRY, PERGAMON PRESS, GB, vol. 53, no. 7, 1 April 2000 (2000-04-01), pages 747 - 757, XP004291370, ISSN: 0031-9422, DOI: 10.1016/S0031-9422(00)00008-X *
WEN, J. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 38954 - 38964
WON, Y. K. ET AL., CARCINOGENESIS, vol. 25, 2004, pages 1449 - 1458
ZHANG, S. Y ET AL., CANCER LETTERS, vol. 211, 2004, pages 175 - 188

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103788103A (en) * 2012-10-30 2014-05-14 天津尚德药缘科技有限公司 Deuterated dimethylamino parthenolide, preparation method thereof and applications thereof in medicine preparation
CN103788103B (en) * 2012-10-30 2016-08-03 天津尚德药缘科技股份有限公司 Deuterated dimethyl amine parthenolide, its preparation method and the purposes in preparing medicine
WO2014067379A1 (en) * 2012-10-30 2014-05-08 天津尚德药缘科技有限公司 Deuterated dimethyl amine parthenolide, preparation method and use thereof in drug preparation
CN104211669A (en) * 2013-06-05 2014-12-17 天津尚德药缘科技有限公司 Micheliolide derivative and preparation method and use thereof
CN104211669B (en) * 2013-06-05 2018-01-19 天津尚德药缘科技股份有限公司 Lactone derivatives with a smile, and its production and use
EP3003317A4 (en) * 2013-06-06 2017-04-19 University Of Rochester Parthenolide derivatives, methods for their preparation and their use as anticancer agents
EP3019165A4 (en) * 2013-07-11 2017-01-25 The Board of Trustees of the University of Arkansas Dehydroleucodine derivatives and uses thereof
US11059800B2 (en) 2013-07-11 2021-07-13 Universidad Tecnica Particular De Loja Dehydroleucodine derivatives and uses thereof
US9469650B2 (en) 2013-11-08 2016-10-18 Board Of Trustees Of The University Of Arkansas Melampomagnolide B derivatives
US9908892B2 (en) 2013-11-08 2018-03-06 Bioventures, Llc Melampomagnolide B derivatives
US9487536B2 (en) 2013-11-08 2016-11-08 Board Of Trustees Of The University Of Arkansas Melampomagnolide B derivatives
US9920063B2 (en) 2013-11-08 2018-03-20 Bioventures, Llc Melampomagnolide B derivatives
CN104876899A (en) * 2014-02-28 2015-09-02 天津尚德药缘科技股份有限公司 Dimethylamino sphaelactone fumarate and use thereof
US9981990B2 (en) 2014-12-03 2018-05-29 Bioventures, Llc Melampomagnolide B dimers
WO2016090166A1 (en) * 2014-12-03 2016-06-09 Board Of Trustees Of The University Of Arkansas Melampomagnolide b dimers
US10118935B2 (en) 2014-12-03 2018-11-06 Bioventures, Llc Melampomagnolide B dimers
CN106366088A (en) * 2015-07-24 2017-02-01 天津尚德药缘科技股份有限公司 Parthenolide derivative, and medicinal composition, preparation method and use thereof
US10420746B2 (en) 2015-10-21 2019-09-24 Mehrdad Ghashghaeinia Pharmaceutical composition containing Bay 11-7082, parthenolide or a combination thereof for the treatment of obesity or cardiovascular diseases
DE102015117882A1 (en) * 2015-10-21 2017-04-27 Mehrdad Ghashghaeinia Pharmaceutical composition
US10428082B2 (en) 2016-01-29 2019-10-01 Bioventures, Llc Triazole derivatives of melampomagnolide B and methods of use thereof
CN109897022A (en) * 2017-12-07 2019-06-18 中国医学科学院药物研究所 Lactone derivatives, its pharmaceutical composition and its preparation method and application with a smile
CN108484550A (en) * 2018-05-07 2018-09-04 中国人民解放军第四军医大学 One kind having a smile on one's face lactone derivatives and its preparation method and application
CN111303099A (en) * 2018-12-12 2020-06-19 天津尚德药缘科技股份有限公司 Sphaelactone dimethylamine fumarate crystal form F and preparation method thereof
CN111303099B (en) * 2018-12-12 2022-12-02 天津尚德药缘科技股份有限公司 Sphaelactone dimethylamine fumarate crystal form F and preparation method thereof

Similar Documents

Publication Publication Date Title
WO2012145678A1 (en) Anti-cancer and anti-inflammatory parthenolide compounds
US8563596B2 (en) Enantiomerically pure compounds for the treatment of proliferative disorders
US7884127B2 (en) Inhibitors of cyclin dependent kinases and their use
US20100197686A1 (en) Therapeutic compounds
EP2190426B1 (en) Griseofulvin analogues for the treatment of cancer by inhibition of centrosomal clustering
JP2008500366A (en) Migrastatin analogues in the treatment of cancer
WO2011159926A1 (en) Antimicrobial agents
JP2011105766A (en) Migrastatin analog composition and use thereof
WO2013109972A2 (en) Therapeutic compounds
HUT74681A (en) N-(2-hydroxy-1-indenyl)-2-phenylmethyl-4-hydroxy-5-(2-carboxamido-piperazinyl)-pentaneamide derivatives of hiv protease inhibitor activity and phrmaceutical compositions contining them
US9108941B2 (en) Griseofulvin analogues for the treatment of cancer by inhibition of centrosomal clustering
MXPA04004607A (en) Solubilized topoisomerase poison agents.
WO2014007412A1 (en) Novel compound or pharmaceutically acceptable salt thereof, and pharmaceutical composition containing same as active ingredient
Kiełczewska et al. The synthesis of solasodine F-homo-analogues
JP4418627B2 (en) Micanolide derivatives, their production and their medical use
CN101230015B (en) Substituted cinnamic acid derivatives containing amine substituent group and tumor cytotoxicity thereof
JP2017503026A (en) Water-soluble 4-azapodophyllotoxin analog
US6673937B2 (en) Syntheses and methods of use of new antimitotic agents
US11407761B2 (en) Derivative of β-apopicropodophyllin and method of preparing thereof
KR100742060B1 (en) NEW BENZO[b]PYRANO[3,2-h]ACRIDIN-7-ONE CINNAMATE COMPOUNDS, A PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
RU2418786C2 (en) Enantiomerically pure flavone derivatives for treatment of proliferative disorders and methods of their obtaining
EP1399455A2 (en) Novel tricyclic dihydro-quinoline derivatives, method for preparing same and pharmaceutical compositions containing same

Legal Events

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

Ref document number: 12717565

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12717565

Country of ref document: EP

Kind code of ref document: A1