WO2008028057A2 - Shikimic acid-derived compounds for inhibition and detection of aurora a-associated tumors - Google Patents

Shikimic acid-derived compounds for inhibition and detection of aurora a-associated tumors Download PDF

Info

Publication number
WO2008028057A2
WO2008028057A2 PCT/US2007/077266 US2007077266W WO2008028057A2 WO 2008028057 A2 WO2008028057 A2 WO 2008028057A2 US 2007077266 W US2007077266 W US 2007077266W WO 2008028057 A2 WO2008028057 A2 WO 2008028057A2
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
branched
unbranched
moiety
Prior art date
Application number
PCT/US2007/077266
Other languages
French (fr)
Other versions
WO2008028057A3 (en
Inventor
Karl Munger
Hiroyuki Hayakawa
Peter M. Howley
Angela N. Koehler
Timothy A. Lewis
Hua Miao
Stuart L. Schreiber
John Tallarico
Original Assignee
President And Fellows Of Harvard College
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 President And Fellows Of Harvard College filed Critical President And Fellows Of Harvard College
Publication of WO2008028057A2 publication Critical patent/WO2008028057A2/en
Publication of WO2008028057A3 publication Critical patent/WO2008028057A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/14Radicals substituted by nitrogen atoms, not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/46Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C229/48Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups and carboxyl groups bound to carbon atoms of the same non-condensed ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/40Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/10Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/14Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/10Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/16Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/12Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted in position 3 and unsubstituted in position 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/16Systems containing only non-condensed rings with a six-membered ring the ring being unsaturated

Definitions

  • Aurora kinase family Members of the Aurora kinase family are serine/threoinin kinases which are involved in mitosis (Keen and Taylor., Nat. Rev. Cancer (2004) 4:927). These kinases have been found to regulate multiple steps in mitosis including centrosome duplication, formation of bipolar mitotic spindles, and chromosome alignment on the mitotic spindle. Centrosomes act as mitotic spindle pole bodies during mitosis. Abnormal centrosome numbers are frequently detected in human solid tumors and are a hallmark of genomically unstable cells. Three Aurora kinases, A, B, and C, are known.
  • Aurora A kinase localizes to the duplicated centrosomes and to the spindle poles during mitosis and assists with centrosome maturation and separation.
  • Aurora B kinase is a chromosomal passenger protein which is localized to the centromeric regions of the chromosomes in the early stages of mitosis and accumulates in the spindle midzone and midbody.
  • the role of Aurora C kinase is unknown currently.
  • the Aurora kinase family is involved in regulating multiple steps in mitosis, including centrosome duplication, formation of bipolar mitotic spindles, and chromosome alignment on the mitotic spindle (Keen and Taylor, Nat. Rev. Cancer (2004) 4:921).
  • Centrosomes act as mitotic spindle pole bodies during mitosis. Abnormal centrosome numbers are frequently detected in human solid tumors and are a hallmark of genomically unstable cells. Small molecules that associate with, but do not inhibit, centrosomal components can serve as markers for cells with centrosome abnormalities in tumor cells, and thus be useful as diagnostic tools in the detection of cancer cells. [0005] Amplification and overexpression of Aurora A kinase has been observed in several types of human cancers, including cancers of the colon, breast, pancreas, ovaries, and stomach, among others.
  • the present invention provides a method of treating a proliferative disease (e.g., cancer) comprising the step of adminiserting a therapeutically effective amount of a compound of formulae (I) or (II), or a pharmaceutically acceptable form thereof, to a subject in need thereof.
  • a proliferative disease e.g., cancer
  • the present invention provides a method of detecting proliferative cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
  • the present invention provides a method of detecting pre-cancerous cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
  • the present invention provides a method of identifying compounds which bind to Aurora-A kinase, comprising the steps of: (i) contacting a cancer cell with a test compound of formulae (I) or (II) conjugated to an imaging agent and (ii) screening for binding to Aurora-A kinase.
  • the present invention contemplates all such compounds, including cis- and ⁇ r ⁇ ws-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • an isomer/enantiomer may, in some embodiments, be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched.”
  • “Optically enriched,” as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer.
  • the compound of the present invention is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer.
  • Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses.
  • HPLC high pressure liquid chromatography
  • Jacques, et al. Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen, S. H., et al., Tetrahedron 33:2725 (1977); Eliel, E.L. Stereochemistry of Carbon Compounds (McGraw- Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972).
  • the compounds of the present invention may be substituted with any number of substituents or functional moieties.
  • substituted whether preeceded by the term “optionally” or not, and substituents contained in formulas of this invention, refer to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent.
  • the substituent may be either the same or different at every position.
  • substituted is contemplated to include substitution with all permissible substituents of organic compounds, any of the substituents described herein (for example, aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, etc.), and any combination thereof (for example, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, arylthio; heteroaryloxy,
  • acyl refers to a group having the general formula -
  • acyl groups include aldehydes (-CHO), carboxylic acids (-CO 2 H), ketones, acyl halides, esters, amides, imines, carbonates, carbamates, and ureas.
  • Acyl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl
  • aliphatic includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, and cyclic (i.e., carbocyclic) hydrocarbons, which are optionally substituted with one or more functional groups.
  • aliphatic is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties.
  • alkyl includes straight, branched and cyclic alkyl groups (for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl, cyclodecenyl,and the like, and which may bear one or more substituents).
  • aliphatic is used to indicate those aliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms (i.e., Ci_ 2 o aliphatic) [in situations wherein the number of carbons is specified, such as a Ci 5 aliphatic, is meant a carbon chain 15 carbons in length].
  • the aliphatic group employed by the invention is an optionally substituted C 1 -C 15 aliphatic group. In certain embodiments, the aliphatic group employed by the invention is an optionally substituted C 1 -C 10 aliphatic group. In certain embodiments, the aliphatic group employed by the invention is an optionally substituted C 1 -C 5 aliphatic group.
  • Aliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, iso
  • alkyloxy refers to an aliphatic group attached to the parent molecule via an oxygen atom.
  • alkylthio refers to an aliphatic group attached to the parent molecule via a sulfur atom.
  • alkyl refers to a saturated, straight- or branched- chain hydrocarbon radical derived from a hydrocarbon moiety which is optionally substituted with one or more functional groups.
  • the alkyl group employed in the invention contains 1-20 carbon atoms (i.e., Ci_ 2 o alkyl) [in situations wherein the number of carbons is specified, such as a C 15 alkyl, is meant a fully saturated carbon chain 15 carbons in length].
  • the alkyl group employed by the invention is an optionally substituted C 1 -C 15 alkyl group.
  • the alkyl group employed by the invention is an optionally substituted Ci-Ci 0 alkyl group.
  • the alkyl group employed by the invention is an optionally substituted C 1 -C 5 alkyl group.
  • alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, sec-pentyl, iso-pentyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, sec- hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, and the like, which may bear one or
  • Alkyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, iso
  • alkenyl denotes a monovalent group derived from a straight- or branched-chain hydrocarbon moiety having at least one carbon-carbon double bond, and which is optionally substituted with one or more functional groups.
  • the alkenyl group employed in the invention contains 2-20 carbon atoms (i.e., C 2 - 2 0 alkenyl) [in situations wherein the number of carbons is specified, such as a C 15 alkenyl, is meant a carbon chain 15 carbons containing at least one sp 2 (double bonded) carbon atom].
  • the alkenyl group employed by the invention is an optionally substituted C 2 -C 1 5 alkenyl group.
  • the alkenyl group employed by the invention is an optionally substituted C 2 -C 1 0 alkenyl group. In certain embodiments, the alkenyl group employed by the invention is an optionally substituted C2-C5 alkenyl group.
  • Alkenyl groups include, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, and the like, which may bear one or more substituents.
  • Alkenyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, is
  • alkynyl refers to a monovalent group derived from a straight- or branched-chain hydrocarbon having at least one carbon-carbon triple bond, and which is optionally substituted with one or more functional groups.
  • the alkynyl group employed in the invention contains 2-20 carbon atoms (i.e., C 2 - 2 0 alkenyl) [in situations wherein the number of carbons is specified, such as a Ci 5 alkynyl, is meant a carbon chain 15 carbons containing at least one sp (triple bonded) carbon atom].
  • the alkynyl group employed by the invention is an optionally substituted C 2 -C 15 alkynyl group. In certain embodiments, the alkynyl group employed by the invention is an optionally substituted C 2 -C 1 0 alkynyl group. In certain embodiments, the alkynyl group employed by the invention is an optionally substituted C 2 -C5 alkynyl group.
  • Representative alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl, and the like, which may bear one or more substituents.
  • Alkynyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano,
  • amino refers to a group of the formula (-NH 2 ).
  • substituted amino refers to a mono-substituted amino group of the formula (-NHR ⁇ ) or a di-substituted amino group of the formula (-NR ⁇ 2 ).
  • Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., an amino protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, and the like, each of which may or may not be further substituted).
  • NR ⁇ 2 form a 5- to 6- membered hetereocyclic ring.
  • aryl refers to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which all the ring atoms are carbon, and which may be substituted or unsubstituted.
  • aryl refers to a mono, bi, or tricyclic C 4 -C 2 0 aromatic ring system having one, two, or three aromatic rings which include, but not limited to, phenyl, biphenyl, naphthyl, and the like, which may bear one or more substituents.
  • Aryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino
  • aryloxy refers to an aryl group attached to the parent molecule via an oxygen atom.
  • arylthio refers to an aryl group attached to the parent molecule via a sulfur atom.
  • “optionally substituted azido” refers to a group of the formula (-N3R ⁇ ), wherein R ⁇ can be any substitutent (other than hydrogen).
  • Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • cyano refers to a group of the formula (-CN).
  • halo and halogen as used herein refer to an atom selected from fluorine (fluoro, -F), chlorine (chloro, -Cl), bromine (bromo, -Br), and iodine (iodo, -I).
  • heteroaliphatic refers to an aliphatic moiety, as defined herein, which includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, cyclic (i.e., heterocyclic), or poly cyclic hydrocarbons, which are optionally substituted with one or more functional groups, and that contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • heteroaliphatic moieties are substituted by independent replacement of one or more of the hydrogen atoms thereon with one or more substituents.
  • heteroaliphatic is intended herein to include, but is not limited to, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl moieties.
  • heteroalkyl includes straight, branched and cyclic alkyl groups that contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms.
  • An analogous convention applies to other generic terms such as “heteroalkenyl", “heteroalkynyl", and the like.
  • heteroalkyl encompass both substituted and unsubstituted groups.
  • heteroaliphatic is used to indicate those heteroaliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms (i.e., Ci_ 2 o heteroaliphatic) [in situations wherein the number of carbons is specified, such as a C 15 heteroaliphatic, is meant a carbon chain 15 carbons in length].
  • the heteroaliphatic group employed by the invention is an optionally substituted C 1 -C 15 heteroaliphatic group. In certain embodiments, the heteroaliphatic group employed by the invention is an optionally substituted C 1 -C 10 heteroaliphatic group. In certain embodiments, the heteroaliphatic group employed by the invention is an optionally substituted C 1 -C 5 heteroaliphatic group.
  • Heteroaliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano
  • heteroaryloxy refers to a heteroaryl group attached to the parent molecule through an oxygen atom.
  • heteroarylthio refers to a heteroaryl group attached to the parent molecule through a sulfur atom.
  • heterocyclic refers to a cyclic heteroaliphatic.
  • a heterocyclic group refers to a non-aromatic, partially unsaturated or fully saturated, 3- to 10-membered ring system, which includes single rings of 3 to 8 atoms in size, and bi- and tri-cyclic ring systems which may include aromatic five- or six-membered aryl or heteroaryl groups fused to a non-aromatic ring.
  • heterocyclic rings include those having from one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, in which the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • heterocylic refers to a non-aromatic 5-, 6-, or 7-membered ring or polycyclic group wherein at least one ring atom is a heteroatom selected from O, S, and N (wherein the nitrogen and sulfur heteroatoms may be optionally oxidized), and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms.
  • Heterocycyl groups include, but are not limited to, a bi- or tri-cyclic group, comprising fused five, six, or seven-membered rings having between one and three heteroatoms independently selected from the oxygen, sulfur, and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds, and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl ring.
  • heterocycles include azacyclopropanyl, azacyclobutanyl, 1,3-diazatidinyl, piperidinyl, piperazinyl, azocanyl, thiaranyl, thietanyl, tetrahydrothiophenyl, dithiolanyl, thiacyclohexanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropuranyl, dioxanyl, oxathiolanyl, morpholinyl, thioxanyl, tetrahydronaphthyl, and the like, which may bear one or more substituents.
  • Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyan
  • heteroaryl refers to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which one ring atom is selected from S, O, and N; zero, one, or two ring atoms are additional heteroatoms independently selected from S, O, and N; and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms.
  • heteroaryls include, but are not limited to pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, pyyrolizinyl, indolyl, quinolinyl, isoquinolinyl, benzoimidazolyl, indazolyl, quinolinyl, isoquinolinyl, quinolizinyl, cinnolinyl, quinazolynyl, phthalazinyl, naphthridinyl, quinoxalinyl, thiophenyl, thianaphthenyl, furanyl, benzofuranyl, benzothiazolyl, thiazolynyl, isothiazolyl, thiadiazolynyl, oxazolyl, isoxazolyl, oxadiazi
  • Heteroaryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a stable moiety e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, is
  • hydroxy refers to a group of the formula (-OH).
  • An “optionally substituted hydroxyl” refers to a group of the formula (- OR ⁇ ), wherein R ⁇ can be hydrogen, or any substitutent which results in a stable moiety (e.g., a suitable hydroxyl protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
  • a suitable amino protecting group for example, aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, and the like, each of which may or may not be further substituted.
  • isocyano refers to a group of the formula (-NC).
  • nitro refers to a group of the formula (-NO 2 ).
  • An "optionally substituted thiol” refers to a group of the formula (-SR ⁇ ), wherein R ⁇ can be hydrogen, or any substitutent.
  • Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., a suitable thiol protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, cyano, nitro, alkylaryl, arylalkyl, and the like, each of which may or may not be further substituted).
  • stable moiety preferably refers to a moiety which possess stability sufficient to allow manufacture, and which maintains its integrity for a sufficient period of time to be useful for the purposes detailed herein.
  • resin refers to a resin useful for solid phase synthesis.
  • a resin may be functionalized with one or more linker groups prior to attachment of the compound to the solid phase, and such functionalization with linker groups is well within the scope of this invention.
  • Solid phase synthesis is a well-known synthetic technique; see generally, Atherton, E., Sheppard, R.C. Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, Oxford, England, 1989, and Stewart J.M., Young, J.D. Solid Phase Peptide Synthesis, 2nd edition, Pierce Chemical Company, Rockford, 1984, the entire contents of each of which are hereby incorporated herein by reference.
  • Exemplary resins which may be employed by the present invention include, but are not limited to:
  • alkenyl resins e.g., REM resin, vinyl sulfone polymer-bound resin, vinyl- polystyrene resin
  • alkenyl resins e.g., REM resin, vinyl sulfone polymer-bound resin, vinyl- polystyrene resin
  • amine functionalized resins e.g., amidine resin, N-(4- Benzyloxybenzyl)hydroxylamine polymer bound, (aminomethyl)polystyrene, polymer bound (7?)-(+)-a-methylbenzylamine, 2-Chlorotrityl Knorr resin, 2-N-Fmoc-Amino- dibenzocyclohepta-l,4-diene, polymer-bound resin, 4-[4-(l-Fmoc-aminoethyl)-2- methoxy-5-nitrophenoxy]butyramidomethyl-polystyrene resin, 4-Benzyloxybenzylamine, polymer-bound, 4-Carboxybenzenesulfonamide, polymer-bound, Bis(tert- butoxycarbonyl)thiopseudourea, polymer-bound, Dimethylaminomethyl-polystyrene, Fmoc- 3-amino-3-(2-nitrophenyl
  • benzhydrylamine (BHA) resins e.g., 2-Chlorobenzhydryl chloride, polymer- bound, HMPB-benzhydrylamine polymer bound, 4-Methylbenzhydrol, polymer-bound, Benzhydryl chloride, polymer-bound, Benzhydrylamine polymer-bound;
  • Br-functionalized resins e.g., 4-(Benzyloxy)benzyl bromide polymer bound, A- Bromopolystyrene, Brominated PPOA resin, Brominated Wang resin, Bromoacetal, polymer-bound, Bromopolystyrene, HypoGel ® 200 Br, Polystyrene A-Br for peptide synthesis, Selenium bromide, polymer-bound, TentaGel HL-Br, TentaGel MB-Br, TentaGel S-Br, TentaGel S-Br); (5) Chloromethyl resins (e.g., 5-[4-(Chloromethyl)phenyl]pentyl]styrene, polymer- bound, 4-(Benzyloxy)benzyl chloride polymer bound, 4-Methoxybenzhydryl chloride, polymer-bound);
  • Chloromethyl resins e.g., 5-[
  • CHO-functionalized resins e.g., (4-Formyl-3- methoxyphenoxymethyl)polystyrene, (4-Formyl-3-methoxyphenoxymethyl)polystyrene, 3- Benzyloxybenzaldehyde, polymer-bound, 4-Benzyloxy-2,6- dimethoxybenzaldehyde,polymer-bound, Formylpolystyrene, HypoGel ® 200 CHO, Indole resin, Polystyrene A-CH(OEt) 2 , TentaGel HL-CH(OEt) 2 );
  • Cl-functionalized resins e.g., Benzoyl chloride polymer bound, (Chloromethyl)polystyrene, Merrifield's resin
  • CO 2 H functionalized resins e.g., Carboxyethylpolystryrene, HypoGel ® 200 COOH, Polystyrene AM-COOH, TentaGel HL-COOH, TentaGel MB-COOH, TentaGel S- COOH;
  • Hypo-Gel resins e.g., HypoGel ® 200 FMP, HypoGel ® 200 PHB , HypoGel ® 200 Trt-OH , HypoGel ® 200 HMB );
  • I-functionalized resins e.g., 4-Iodophenol, polymer-bound, Iodopolystyrene
  • Janda-JelsTM JandaJef- Rink amide, JandaJel-NH 2 , JandaJel-Cl, JandaJeM- Mercaptophenol, JandaJel-OH, JandaJel-l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide, JandaJel- 1, 3,4,6,7, 8-hexahydro-2H-pyrimido-[l,2- ⁇ ] pyrimidine, JandaJel-morpholine, JandaJel-polypyridine, JandaJel-Triphenylphosphine, JandaJel-Wang);
  • MBHA resins (3 [4'-( ⁇ ydroxymethyl)phenoxy] propionic acid ⁇ - methylbenzhydrylamine resin, 4-(Hydroxymethyl)phenoxyacetic acid polymer-bound to MBHA resin, HMBA ⁇ -methylbenzhydrylamine polymer bound, 4-Methylbenzhydrylamine hydrochloride polymer bound Capacity (amine));
  • NH 2 functionalized resins ((Aminomethyl)polystyrene, (Aminomethyl)polystyrene, HypoGel ® 200 NH2, Polystyrene AM-NH 2 , Polystyrene Microspheres 2-aminoethylated, Polystyrol Microspheres 2-bromoethylated, Polystyrol Microspheres 2-hydroxyethylated, TentaGel HL-NH 2 , Tentagel M Br, Tentagel M NH 2 , Tentagel M OH, TentaGel MB-NH 2 , TentaGel S-NH 2 , TentaGel S-NH 2 );
  • OH-functionalized resins e.g., 4-Hydroxymethylbenzoic acid, polymer-bound, Hydroxymethyl Resins, OH-functionalized Wang Resins
  • 4-Hydroxymethylbenzoic acid e.g., 4-Hydroxymethylbenzoic acid, polymer-bound, Hydroxymethyl Resins, OH-functionalized Wang Resins
  • oxime resins e.g., 4-Chlorobenzophenone oxime polymer bound, Benzophenone oxime polymer bound, 4-Methoxybenzophenone oxime polymer bound;
  • PEG resins e.g., ethylene glycol polymer bound
  • Boc-/Blz peptide synthesis resins e.g., Boc-Lys(Boc)-Lys[Boc-Lys(Boc)]- Cys(Acm)-b-Ala-O-PAM resin, Boc-Lys(Fmoc)-Lys[Boc-Lys(Fmoc)]-b-Ala-O-Pam resin, Boc-Lys ⁇ o ⁇ -LysfBoc-Lys ⁇ oc ⁇ -Lys (BOC-LyS(BoC)-LyS[BoC-LyS(BoC)]! ⁇ - AIa-O-PAM resin, Boc-Lys(Fmoc)-Lys[Boc-Lys(Fmoc)]-Lys ⁇ Boc-Lys(Fmoc)-Lys[
  • Fmoc-/t-Bu peptide synthesis resins e.g., Fmoc-Lys(Fmoc)-Lys[Fmoc- Lys(Fmoc)]-b-Ala-O-Wang resin, Fmoc-Lys(Fmoc)-Lys[Fmoc-Lys(Fmoc)]-Lys ⁇ Fmoc- Lys(Fmoc)-Lys[Fmoc-Lys(Fmoc)] ⁇ -b-Ala-O-Wang resin, Preloaded TentaGel® S Trityl Resins, Preloaded TentaGel® Resins, Preloaded Trityl Resins, Preloaded Wang Resins, Trityl Resins Preloaded with Amino Alcohols);
  • thiol-functionalized resins e.g., HypoGel ® 200 S-Trt, Polystyrene AM-S- Trityl, TentaGel HL-S-Trityl, TentaGel MB-S-Trityl, TentaGel S-S-Trityl;
  • silicon-functionalized resin e.g., 4-methoxyphenyl)diisopropylsilylpropyl polystyrene.
  • linker refers to a chemical moiety utilized to covalently attach a resin, a fluoroescent or luminescent moiety to a compound of formulae (I) or (II).
  • a "fluorescent label” or "fluorophore” or “fluroescent moiety” as used herein, is a component of a molecule which causes a molecule to be fluorescent. It is a functional group in a molecule which will absorb energy of a specific wavelength and re-emit energy at a different (but equally specific) wavelength. The amount and wavelength of the emitted energy depend on both the fluorophore and the chemical environment of the fluorophore. This technology has particular importance in the field of biochemistry and protein studies, eg. in immunofluorescence and immunohistochemistry.
  • Exemplary fluorescent labels are provided in Figure 7, and include reactive and conjugated probes, Alexa Fluor dyes (Molecular Probes) Cy Dyes (AP Biotech) Nucleic acid probes, Cell function probes, Fluorescent Proteins, and are also described in U.S. Patent Publication 2004/0067503; Valeur, B., "Molecular Fluorescence: Principles and Applications,” John Wiley and Sons, 2002; and Handbook of Fluorescent Probes and Research Products, Molecular Probes, 9 th edition, 2002.
  • a "luminescent label” or “luminescent moiety” as used herein, is a component of a molecule which causes a molecule to be chemiluminescent. Chemiluminescence is a common technique for a variety of detection assays in biology. A horseradish peroxidase enzyme (HRP) is tethered to the molecule of interest (usually through labeling an immunoglobulin that specifically recognizes the molecule). This enzyme complex, then catalyzes the conversion of the luminescent moiety into a sensitized moiety, which on further oxidation by hydrogen peroxide, produces a triplet (excited) carbonyl which emits light when it decays to the singlet carbonyl. Enhanced chemiluminescence allows detection of minute quantities of a molecule. Proteins can be detected down to femtomole quantities well below the detection limit for most assay systems.
  • HRP horseradish peroxidase enzyme
  • a “protecting group” refers to an amino, hydroxyl, thiol, or carboxylic acid protecting group.
  • amino-protecting group is well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3 rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference.
  • Suitable amino-protecting groups include methyl carbamate, ethyl carbamante, 9-fluorenylmethyl carbamate (Fmoc), 9-(2- sulfo)fluorenylmethyl carbamate, 9-(2,7-dibromo)fluoroenylmethyl carbamate, 2,7— di— f— butyl-[9-( 10,10-dioxo- 10, 10, 10, 10-tetrahydrothioxanthy I)] methyl carbamate (DBD-Tmoc), 4-methoxyphenacyl carbamate (Phenoc), 2,2,2-trichloroethyl carbamate (Troc), 2- trimethylsilylethyl carbamate (Teoc), 2-phenylethyl carbamate (hZ), 1 -(I -adamanty I)-I- methylethyl carbamate (Adpoc), l,l-dimethyl-2-haloethyl carbamate
  • a "hydroxyl protecting group” as used herein, is well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3 rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference. Suitable hydroxyl protecting groups include methyl, methoxylmethyl (MOM), methylthiomethyl (MTM), ⁇ -butylthiomethyl,
  • the protecting groups include methylene acetal, ethylidene acetal, l-?-butylethylidene ketal, 1-phenylethylidene ketal, (4- methoxyphenyl)ethylidene acetal, 2,2,2-trichloroethylidene acetal, acetonide, cyclopentylidene ketal, cyclohexylidene ketal, cycloheptylidene ketal, benzylidene acetal, p— methoxybenzylidene acetal, 2,4-dimethoxybenzylidene ketal, 3,4-dimethoxybenzylidene acetal, 2-nitrobenzylidene acetal, methoxymethylene acetal, ethoxymethylene acetal, dimethoxy methylene ortho ester, 1-methoxy ethylidene ortho
  • a "thiol protecting group,” as used herein, are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3 rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference.
  • Examples of suitably protected thiol groups further include, but are not limited to, thioesters, carbonates, sulfonates allyl thioethers, thioethers, silyl thioethers, alkyl thioethers, arylalkyl thioethers, and alkyloxyalkyl thioethers.
  • ester groups include formates, acetates, proprionates, pentanoates, crotonates, and benzoates.
  • Specific examples of suitable ester groups include formate, benzoyl formate, chloroacetate, trifluoroacetate, methoxyacetate, triphenylmethoxyacetate, p- chlorophenoxyacetate, 3-phenylpropionate, 4-oxopentanoate, 4,4-
  • Suitable carbonates include 9-fluorenylmethyl, ethyl, 2,2,2-trichloroethyl, 2-(trimethylsilyl)ethyl, 2- (phenylsulfonyl)ethyl, vinyl, allyl, and p-nitrobenzyl carbonate.
  • silyl groups include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl ether, and other trialkylsilyl ethers.
  • suitable alkyl groups include methyl, benzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, trityl, t-butyl, and allyl ether, or derivatives thereof.
  • Suitable arylalkyl groups include benzyl, p- methoxybenzyl (MPM), 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl, 2- and 4-picolyl ethers.
  • a "carboxylic acid protecting group,” as used herein, are well known in the art and include those described in detail in Greene (1999).
  • Examples of suitably protected carboxylic acids further include, but are not limited to, silyl-, alkyl-, alkenyl-, aryl-, and arylalkyl-protected carboxylic acids.
  • Examples of suitable silyl groups include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl, and the like.
  • suitable alkyl groups include methyl, benzyl, p-methoxybenzyl, 3,4- dimethoxybenzyl, trityl, t-butyl, tetrahydropyran-2-yl.
  • suitable alkenyl groups include allyl.
  • suitable aryl groups include optionally substituted phenyl, biphenyl, or naphthyl.
  • Suitable arylalkyl groups include optionally substituted benzyl (e.g., p-methoxybenzyl (MPM), 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl), and 2- and 4-picolyl.
  • MPM p-methoxybenzyl
  • 3-4-dimethoxybenzyl 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl
  • 2- and 4-picolyl 2- and 4-picolyl.
  • a "pharmaceutically acceptable form thereof includes any pharmaceutically acceptable salts, prodrugs, tautomers, isomers, and/or polymorphs of a compound of the present invention, as defined below and herein.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1911 , 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C i_ 4 alky I) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • prodrug refers to a derivative of a parent compound that requires transformation within the body in order to release the parent compound.
  • a prodrug has improved physical and/or delivery properties over the parent compound.
  • Prodrugs are typically designed to enhance pharmaceutically and/or pharmacokinetically based properties associated with the parent compound.
  • the advantage of a prodrug can lie in its physical properties, such as enhanced water solubility for parenteral administration at physiological pH compared to the parent compound, or it enhances absorption from the digestive tract, or it may enhance drug stability for long-term storage.
  • bioreversible derivatives have been exploited for utilization in designing prodrugs.
  • esters as a prodrug type for compounds containing a carboxyl or hydroxyl functionality is known in the art as described, for example, in "The Organic Chemistry of Drug Design and Drug Interaction” Richard Silverman, published by Academic Press (1992).
  • the term "tautomer” includes two or more interconvertable compounds resulting from at least one formal migration of a hydrogen atom and at least one change in valency (e.g., a single bond to a double bond, a triple bond to a single bond, or vice versa). The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH.
  • Tautomerizations may catalyzed by acid or base.
  • exemplary tautomerizations include keto-to-enol; amide-to-imide; lactam-to-lactim; enamine-to-imine; and enamine-to-(a different) enamine tautomerizations.
  • isomers includes any and all geometric isomers and stereoisomers.
  • isomers include cis— and trans-isomsrs, E- and Z- isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • an isomer/enantiomer may, in some embodiments, be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched.”
  • “Optically- enriched,” as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer.
  • the compound of the present invention is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer.
  • Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses.
  • HPLC high pressure liquid chromatography
  • Jacques, et al. Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen, S.H., et al., Tetrahedron 33:2725 (1977); Eliel, EX. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S. H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972).
  • polymorph refers to a crystalline inventive compound existing in more than one crystalline form/structure. When polymorphism exists as a result of difference in crystal packing it is called packing polymorphism. Polymorphism can also result from the existence of different conformers of the same molecule in conformational polymorphism. In pseudopolymorphism the different crystal types are the result of hydration or solvation. Brief Description of the Figures
  • Aurora A kinase protein Aurora A kinase protein
  • FIGs 2A-2C Figures 2A-2C.
  • Figure 2A Compounds (1-1) to (1-7) at 5 ⁇ M were tested for binding to Aurora A kinase by surface plasmon resonance.
  • DOS Diversity-oriented synthesis
  • Figure 4. Sensograms for compounds (1-1), (1-9) to (1-10), (1-12) to (1-20), (I-
  • Figure 6 Table of exemplary fluorochromes useful for flow cytometry or fluorescence microscopy provided, within groups, roughly in order of excitation wavelength. MW: molecular weight; Ex: Peak excitation wavelength (nm); Em: Peak emission wavelength (nm).
  • FIGs 7A-7C HeLa (cervical cancer), compound (1-33) (FITC-labeled compound; 4 ⁇ M). Similar images have been obtained with U2OS and MCF7 cells.
  • Figure 7 A image of fluorescent tagged compound (1-33) bound to a centrosome (red color);
  • Figure IB image of cell sample;
  • Figure 1C a merge of the two images. The centrosome (i.e., two centrioles) within the cell are easily visualized (red color).
  • Figure 9 Numerical centrosome aberrations in normal cervical epithelium and various cervical lesions (Skyldberg et al., Modern Pathology (2001) 14:279-284). Expression of HPV E7 protein uncouples centrosome and chromosome duplication. Supernumerary centrosomes are a biomarker for HPV E7 oncogene expression. [0069] Figure 10. Images of cells undergoing normal and abnormal mitosis. Detailed Description of Certain Embodiments of the Invention
  • the present invention provides inventive compounds, pharmaceutical compositions thereof, and use of such compounds.
  • Compounds of the present invention are useful as anti-proliferative agents and diagnostic agents for the detection of proliferative (e.g., cancerous) and pre-cancerous cells.
  • proliferative e.g., cancerous
  • pre-cancerous cells e.g., cancerous cells
  • the present invention provides compounds of the formulae:
  • Ri is hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -OR A ; -CN; -SCN; -SR A ; or -N(R A ) 2 ; wherein each occurrence of R A is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety;
  • R 2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
  • R 4 is hydrogen; a hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
  • Ri is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -OR A ; -CN; -SCN; -SR A ; - N(R A ) 2 ; or -C(R A )3.
  • Ri is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; -OR A ; -SR A ; -N(R A ) 2 ; or -C(R A )3. In certain embodiments, Ri is -ORA;-SRA; or -N(RA) 2 .
  • Ri is -N(R A ) 2 , wherein each occurrence of R A is independently a hydrogen, an amino protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
  • R A is a substituted or unsubstituted aliphatic moiety.
  • R A is a substituted or unsubstituted heteroaliphatic moiety.
  • Ri is - NHR A .
  • Ri is -NH 2 .
  • Ri is -N(R A ) 2 , wherein each occurrence of R A is independently a hydrogen or a substituted or unsubstituted heteroaliphatic moiety. In certain embodiments, Ri corresponds to any one of the following groups:
  • R G is hydrogen, a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety; a fluorescent label; or a resin.
  • Ri corresponds to group:
  • Ri corresponds to any of the following groups:
  • is a resin
  • Ri is -N(R A ) 2 , wherein each occurrence of R A is independently a hydrogen or a substituted or unsubstituted aliphatic moiety. In certain embodiments, Ri corresponds to any one of the following groups:
  • R 0 is a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety; a fluorescent label; or a resin; and each instance of q is independently 0, 1, 2, or 3.
  • Ri corresponds to the groups: t ⁇ N-(CH 2 ) q -CH 3
  • Ri is -OR A , wherein each occurrence of R A is independently a hydrogen, a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
  • Ri is -OH.
  • Ri is -OR A , wherein R A is a substituted or unsubstituted aliphatic moiety or a substituted or unsubstituted heteroaliphatic moiety.
  • Ri is -OR A , wherein R A is a substituted or unsubstituted aliphatic moiety. In certain embodiments, R A is a substituted or unsubstitued methyl, ethyl, n-propyl, isopropyl, t-butyl, sec-butyl, or iso-butyl group. In certain embodiments, Ri is -OCH3.
  • R 2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl.
  • R 2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl.
  • R 2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic or cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic. In certain embodiments, R 2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic.
  • R 2 corresponds to a -(CH 2 ) m R H group, wherein R H is hydrogen, or a substituted or unsubstituted aryl moeity, a substituted or unsubstituted heteroaryl moeity, a substituted or unsubstituted cycloalkyl moeity, a substituted or unsubstituted cycloheteroalkyl moeity, a substituted or unsubstituted alkyloxy moeity, or - CF 3 ; and m is 0, 1, 2, 3, 4, 5 or 6.
  • R H corresponds to a substituted or unsubstituted aryl moeity or a substituted or unsubstituted heteroaryl moeity.
  • m is 1 or 2.
  • R 2 corresponds to any one of the following groups:
  • R3 is substituted or unsubstituted, branched or unbranched acyl. In certain embodiments, R3 is hydrogen.
  • R3 corresponds to any one of the following groups:
  • R 4 is hydrogen; hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl.
  • R 4 is hydrogen, a hydroxyl protecting group, cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic, or substituted or unsubstituted, branched or unbranched acyl. In certain embodiments, R 4 is hydrogen, a hydroxyl protecting group or C 1 -S alkyl. In certain embodiments, R4 is hydrogen.
  • each occurrence of Re is independently hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; -
  • Re is not halogen.
  • PGi is a silyl protecting group.
  • Exemplary silyl protecting groups include trimethylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), and tertbutyldimethylsilyl (TBDMS).
  • n is 0, 1 or 2. In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2.
  • n 0.
  • n is 1, and Re is substituted ortho to the phenolic oxygen. In certain embodiments, n is 1, and Re is substituted para to the phenolic oxygen.
  • n is 2, and Re is substituted ortho and para to the phenolic oxygen.
  • Rs is hydrogen; substituted or unsubstituted aryl; or substituted or unsubstituted heteroaryl. In certain embodiments, Rs is substituted or unsubstituted aryl. In certain embodiments, Rs corresponds to the groups:
  • n and Re are as defined above and herein, and X is halogen or -S(O) 2 Ri, wherein Ri is a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
  • Ri is a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
  • X is -I, -Cl, or -F.
  • X is -I.
  • Rs corresponds to any of the following groups: wherein R 6 and X are as defined above and herein.
  • R5 corresponds to the groups:
  • R 6 and X are as defined above and herein.
  • R5 corresponds to any one of the following groups:
  • compounds of formula (I), or any pharmaceutically acceptable forms thereof correspond to any one of the following stereoisomers of formula
  • compounds of formula (I), or any pharmaceutically acceptable forms thereof correspond to any one of the following stereoisomers of formula
  • compounds of formula (I), or pharmaceutically acceptable forms thereof are any one of the following compounds:
  • compounds of formula (II), or pharmaceutically acceptable forms thereof correspond any one of the following stereoisomers of formula (II):
  • compounds of formula (II), or pharmaceutically acceptable forms thereof correspond any one of the following stereoisomers of formula (II):
  • compounds of formula (II), or pharmaceutically acceptable forms thereof are any one of the following compounds:
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound, or pharmaceutically acceptable form thereof, of the formulae (I) or (II), or any subsets thereof, and a pharmaceutically acceptable excipient.
  • active ingredient generally refers to an inventive compound, as described above and herein.
  • the formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with a carrier and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition of the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
  • compositions of the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • exemplary pharmaceutically acceptable excipients include any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form.
  • the pharmaceutically acceptable excipient is at least
  • the excipient is approved for use in humans and for veterinary use. In some embodiments, the excipient is approved by United States Food and Drug Administration. In some embodiments, the excipient is pharmaceutical grade. In some embodiments, the excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
  • USP United States Pharmacopoeia
  • EP European Pharmacopoeia
  • British Pharmacopoeia the British Pharmacopoeia
  • International Pharmacopoeia International Pharmacopoeia
  • compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in the inventive formulations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and perfuming agents can be present in the composition, according to the judgment of the formulator.
  • Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and combinations thereof
  • Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation- exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked polyvinylpyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and combinations thereof.
  • crospovidone cross-linked polyvinylpyrrolidone
  • sodium carboxymethyl starch sodium starch glyco
  • Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • natural emulsifiers e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin
  • colloidal clays e.g. bentonite [aluminum silicate]
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • Cremophor polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij 30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic F 68, Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.
  • polyoxyethylene ethers e.g. polyoxyethylene lauryl ether [Brij 30]
  • poly(vinyl-pyrrolidone) diethylene glycol monolaurate
  • triethanolamine oleate sodium oleate
  • potassium oleate ethyl oleate
  • oleic acid ethyl laurate
  • Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.); natural and synthetic gums (e.g.
  • acacia sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.
  • Exemplary preservatives may include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and other preservatives.
  • Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and sodium sulfite.
  • Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and trisodium edetate.
  • EDTA ethylenediaminetetraacetic acid
  • citric acid monohydrate disodium edetate
  • dipotassium edetate dipotassium edetate
  • edetic acid fumaric acid, malic acid
  • phosphoric acid sodium edetate
  • tartaric acid tartaric acid
  • trisodium edetate trisodium edetate.
  • antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and thimerosal.
  • Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and sorbic acid.
  • Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and phenylethyl alcohol.
  • Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and phytic acid.
  • preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus, Phenonip, methylparaben, Germall 115, Germaben II, Neolone, Kathon, and Euxyl.
  • the preservative is an anti-oxidant.
  • the preservative is a chelating agent.
  • Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, is
  • Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.
  • Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana,
  • Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and combinations thereof.
  • Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzy
  • the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • the conjugates of the invention are mixed with solubilizing agents such as Cremophor, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and combinations thereof.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U. S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing the conjugates of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and g
  • Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
  • Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active ingredients can be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active ingredient may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may comprise buffering agents. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • Dosage forms for topical and/or transdermal administration of a compound of this invention may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches.
  • the active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and/or any needed preservatives and/or buffers as may be required.
  • the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of an active ingredient to the body.
  • Such dosage forms may be prepared, for example, by dissolving and/or dispensing the active ingredient in the proper medium.
  • the rate may be controlled by either providing a rate controlling membrane and/or by dispersing the active ingredient in a polymer matrix and/or gel.
  • Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Patents 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662.
  • Intradermal compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof.
  • Jet injection devices which deliver liquid vaccines to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S.
  • Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable.
  • Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.
  • Topically-administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of the active ingredient may be as high as the solubility limit of the active ingredient in the solvent.
  • Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity.
  • a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 to about 7 nanometers or from about 1 to about 6 nanometers.
  • Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container.
  • Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers.
  • Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
  • Low boiling propellants generally include liquid propellants having a boiling point of below 65 0 F at atmospheric pressure. Generally the propellant may constitute 50 to 99.9% (w/w) of the composition, and the active ingredient may constitute 0.1 to 20% (w/w) of the composition. The propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient). [00145] Pharmaceutical compositions of the invention formulated for pulmonary delivery may provide the active ingredient in the form of droplets of a solution and/or suspension.
  • Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization and/or atomization device.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate.
  • a flavoring agent such as saccharin sodium
  • a volatile oil such as a volatile oil
  • a buffering agent such as a a surface active agent
  • a preservative such as methylhydroxybenzoate.
  • the droplets provided by this route of administration may have an average diameter in the range from about 0.1 to about 200 nanometers.
  • formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition of the invention.
  • Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nares.
  • Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient, and may comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for buccal administration.
  • a formulation suitable for buccal administration may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, 0.1 to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein.
  • formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising the active ingredient.
  • Such powdered, aerosolized, and/or aerosolized formulations when dispersed, may have an average particle and/or droplet size in the range from about 0.1 to about 200 nanometers, and may further comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration.
  • Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid carrier.
  • Such drops may further comprise buffering agents, salts, and/or one or more other of the additional ingredients described herein.
  • opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this invention. [00149] General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21 st ed., Lippincott Williams & Wilkins, 2005.
  • compositions suitable for administration to humans are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.
  • kits comprising one or more inventive compounds (or pharmaceutically acceptable forms thereof), and/or an inventive pharmaceutical composition.
  • Kits are typically provided in a suitable container (e.g., for example, a foil, plastic, or cardboard package).
  • an inventive kit may include one or more pharmaceutical excipients, pharmaceutical additives, therapeutically active agents, and the like, as described herein.
  • an inventive kit may include means for proper administration, such as, for example, graduated cups, syringes, needles, cleaning aids, and the like.
  • an inventive kit may include instructions for proper administration and/or preparation for proper administration.
  • the present invention provides a method of treating a proliferative disease, disorder or condition comprising the step of adminiserting a therapeutically effective amount of a compound of formulae I or II, or a pharmaceutically acceptable form thereof, to a subject in need thereof.
  • Subjects to which administration is contemplated include, but are not limited to, humans (e.g., male, female, infant, child, adolescant, adult, elderly, etc.) and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, and/or turkeys.
  • Treating refers to partially or completely inhibiting or reducing the proliferative disease, disorder or condition from which the subject is suffering.
  • “Therapeutically effective amount” refers to the minimal amount or concentration of an inventive compound or inventive pharmaceutical composition that, when administered, is sufficient in treating the subject.
  • a proliferative disease, condition, or disorder includes, but is not limited to, cancer, hematopoietic neoplastic disorders, proliferative breast disease, proliferative disorders of the lung, proliferative disorders of the colon, proliferative disorders of the liver, and proliferative disorders of the ovary.
  • cancers treatable by the above method include carcinoma, sarcoma, or metastatic disorders, breast cancer, ovarian cancer, colon cancer, lung cancer, gastric cancer, esophageal cancer, rectal cancer, pancreatic cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, blood cancer, bone cancer, stomach cancer, liver cancer, kidney cancer, skin cancer, brain cancer, fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinom
  • hematopoietic neoplastic disorders treatable by the above method includes diseases involving hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • the diseases arise from poorly differentiated acute leukemias, e.g., erythroblastic leukemia and acute megakaryoblastic leukemia.
  • Additional exemplary myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev.
  • lymphoid malignancies include, but are not limited to acute lymphoblastic leukemia (ALL) which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM).
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • HLL hairy cell leukemia
  • W Waldenstrom's macroglobulinemia
  • malignant lymphomas include, but are not limited to non-Hodgkin lymphoma and variants thereof, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Stemberg disease.
  • Examples of proliferative breast disease treatable by the above method includes epithelial hyperplasia, sclerosing adenosis, and small duct papillomas; tumors, e.g., stromal tumors such as fibroadenoma, phyllodes tumor, and sarcomas, and epithelial tumors such as large duct papilloma; carcinoma of the breast including in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ, and invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, medullary carcinoma, colloid (mucinous) carcinoma, tubular carcinoma, and invasive papillary carcinoma, and miscellaneous malignant neoplasms.
  • Disorders in the male breast include, but are not limited to, gynecomastia and carcinoma.
  • proliferative disorders of the lung treatable by the above method include, but are not limited to, bronchogenic carcinoma, including paraneoplastic syndromes, bronchioloalveolar carcinoma, neuroendocrine tumors, such as bronchial carcinoid, miscellaneous tumors, and metastatic tumors; pathologies of the pleura, including inflammatory pleural effusions, noninflammatory pleural effusions, pneumothorax, and pleural tumors, including solitary fibrous tumors (pleural fibroma) and malignant mesothelioma.
  • bronchogenic carcinoma including paraneoplastic syndromes, bronchioloalveolar carcinoma, neuroendocrine tumors, such as bronchial carcinoid, miscellaneous tumors, and metastatic tumors
  • pathologies of the pleura including inflammatory pleural effusions, noninflammatory pleural effusions, pneumothorax, and pleural tumors, including solitary fibrous tumors (pleural fibroma) and
  • proliferative disorders of the colon treatable by the above method include, but are not limited to, non-neoplastic polyps, adenomas, familial syndromes, colorectal carcinogenesis, colorectal carcinoma, and carcinoid tumors.
  • proliferative disorders of the liver treatable by the above method include, but are not limited to, nodular hyperplasias, adenomas, and malignant tumors, including primary carcinoma of the liver and metastatic tumors.
  • Examples of proliferative disorders of the ovary treatable by the above method include, but are not limited to, ovarian tumors such as, tumors of coelomic epithelium, serous tumors, mucinous tumors, endometeriod tumors, clear cell adenocarcinoma, cystadenofibroma, brenner tumor, surface epithelial tumors; germ cell tumors such as mature (benign) teratomas, monodermal teratomas, immature malignant teratomas, dysgerminoma, endodermal sinus tumor, choriocarcinoma; sex cord-stomal tumors such as, granulosa-theca cell tumors, thecomafibromas, androblastomas, hill cell tumors, and gonadoblastoma; and metastatic tumors such as Krukenberg tumors.
  • ovarian tumors such as, tumors of coelomic epithelium, serous tumors, mucinous tumors
  • inventive compounds may be administered using any amount and any route of administration effective for treatment.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular composition, its mode of administration, its mode of activity, and the like.
  • the compounds of the present invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts.
  • inventive compounds and compositions of the present invention may be administered by any route.
  • the inventive compounds and compositions are administered via a variety of routes, including oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, bucal, enteral, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol.
  • routes including oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal,
  • Specifically contemplated routes are systemic intravenous injection, regional administration via blood and/or lymph supply, and/or direct administration to an affected site.
  • the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), the condition of the subject (e.g., whether the subject is able to tolerate oral administration), etc.
  • the oral and/or nasal spray and/or aerosol route is most commonly used to deliver therapeutic agents directly to the lungs and/or respiratory system.
  • the invention encompasses the delivery of the inventive pharmaceutical composition by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
  • the exact amount of a compound required to achieve a therapeutically effective amount will vary from subject to subject, depending on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound(s), mode of administration, and the like.
  • the desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks.
  • the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • a therapeutically effective amount of an inventive compound for administration one or more times a day to a 70 kg adult human may comprise about 0.0001 mg to about 5000 mg of an inventive compound per unit dosage form. It will be appreciated that dose ranges as described herein provide guidance for the administration of inventive pharmaceutical compositions to an adult.
  • the amount to be administered to, for example, a child or an adolescent can be determined by a medical practitioner or person skilled in the art and can be lower or the same as that administered to an adult.
  • the inventive pharmaceutical composition comprises about 0.0001 mg to about 1000 mg of an inventive compound per unit dosage form.
  • the composition comprises from about 0.0001 mg to about 1000 mg, from about 0.001 mg to about 1000 mg, from about 0.01 mg to about 1000 mg, from about 0.01 mg to about 1000 mg, from about 0.1 mg to about 1000 mg, from about 1 mg to about 1000 mg, from about 2 mg to about 1000 mg, from about 4 mg to about 1000 mg, from about 6 mg to about 1000 mg, from about 8 mg to about 1000 mg, from about 10 mg to about 1000 mg, from about 20 mg to about 1000 mg, from about 30 mg to about 1000 mg, from about 40 mg to about 1000 mg, from about 60 mg to about 1000 mg, from about 80 mg to about 1000 mg, from about 100 mg to about 1000 mg, from about 200 mg to about 1000 mg, from about 300 mg to about 1000 mg, from about 400 mg to about 1000 mg, from about 500 mg to about 1000 mg, from about 600 mg to about 1000 mg, from about 700 mg to about 1000 mg, from
  • the composition comprises at least about 0.0001 mg, at least about 0.001 mg, at least about 0.01 mg, at least about 0.1 mg, at least about 1 mg, at least about 2 mg, at least about 4 mg, at least about 6 mg, at least about 8 mg, at least about 10 mg, at least about 20 mg, at least about 30 mg, at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 120 mg, at least about 140 mg, at least about 160 mg, at least about 180 mg, at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at least about 460 mg, at least about 480 mg, at least
  • an inventive compound or composition as described above and herein, can be employed in combination with one or more additional medical procedures, such as surgery or radiation therapy.
  • compositions can be administered concurrently with, prior to, or subsequent to, one or more other additional therapeutically active agents.
  • a “therapeutically active agent” or “active agent” refers to any substance that is useful for therapy, including prophylactic and therapeutic treatment.
  • An active agent includes a compound that increases the effect or effectiveness of another compound, for example, by enhancing potency or reducing adverse effects of the other compound (i.e., an adjuvant).
  • each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the invention encompasses the delivery of the inventive pharmaceutical compositions in combination with agents that may improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
  • the additional therapeutically active agent utilized in this combination may be administered together in a single composition or administered separately in different compositions.
  • the therapy employed may achieve a desired effect for the same disorder (for example, an inventive compound may be administered in combination with another anti-proliferative agent), and/or they may achieve different effects (e.g., control of any adverse side-effects).
  • an active agent is an anti-cancer agent, antibiotic, anti-viral agent, anti-HTV agent, anti-parasite agent, anti-protozoal agent, anesthetic, anticoagulant, inhibitor of an enzyme, steroidal agent, steroidal or non-steroidal antiinflammatory agent, antihistamine, immunosuppressant agent, anti-neoplastic agent, antigen, vaccine, antibody, decongestant, sedative, opioid, analgesic, anti-pyretic, birth control agent, hormone, prostaglandin, progestational agent, anti-glaucoma agent, ophthalmic agent, anticholinergic, analgesic, anti-depressant, anti-psychotic, neurotoxin, hypnotic, tranquilizer, anti-convulsant, muscle relaxant, anti-Parkinson agent, anti-spasmodic, muscle contractant, channel blocker, miotic agent, anti-secretory agent, anti-thrombotic agent, anticoagulant, anti-cholinergic, ⁇
  • Exemplary active agents include, but are not limited to, small organic molecules such as drug compounds, peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins, and cells.
  • the active agent is a cell.
  • Exemplary cells include immune system cells (e.g., mast, lymphocyte, plasma cell, macrophage, dendritic cell, neutrophils, eosinophils), connective tissue cells (e.g., blood cells, erythrocytes, leucocytes, megakarocytes, fibroblasts, osteoclasts), stem cells (e.g., embryonic stem cells, adult stem cells), bone cells, glial cells, pancreatic cells, kidney cells, nerve cells, skin cells, liver cells, muscle cells, adipocytes, Schwann cells, Langerhans cells, as well as (micro)-tissues such as the Islets of Langerhans.
  • immune system cells e.g., mast, lymphocyte, plasma cell, macrophage, dendritic cell, neutrophils, eosinophils
  • connective tissue cells e.g., blood cells, erythrocytes, leucocytes, megakarocytes, fibroblasts, osteoclasts
  • stem cells
  • the active agent is a small organic molecule.
  • a small organic molecule is non-peptidic.
  • a small organic molecule is non-oligomeric.
  • a small organic molecule is a natural product or a natural product-like compound having a partial structure (e.g., a substructure) based on the full structure of a natural product.
  • Exemplary natural products include steroids, penicillins, prostaglandins, venoms, toxins, morphine, paclitaxel (Taxol), morphine, cocaine, digitalis, quinine, tubocurarine, nicotine, muscarine, artemisinin, cephalosporins, tetracyclines, aminoglycosides, rifamycins, chloramphenicol, asperlicin, lovastatin, ciclosporin, curacin A, eleutherobin, discodermolide, bryostatins, dolostatins, cephalostatins, antibiotic peptides, epibatidine, ⁇ -bungarotoxin, tetrodotoxin, teprotide, and neurotoxins from Clostridium botulinum.
  • a small organic molecule is a drug approved by the
  • centrosome-associated protein Aurora A is amplified in cancer cells, indicating that it is important for cancer formation and/or progression (Keen and Taylor, Nature Reviews Cancer (2004) 4:927-936).
  • a diagnostic agent which binds to centrosome-associated proteins, such as Aurora-A, Centrin and ⁇ -tubulin, can allow for detection of centrosome abnormalities in a cell of a biological sample, and thus detection of a cancerous or precancerous condition.
  • the present invention provides a method of detecting proliferative cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
  • the present invention provides a method of detecting precancerous cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
  • screening is meant visually viewing the sample, and
  • screening may be further employ visual screening aids, such as, for example, an automated device, instrument (e.g., microscope), light source (e.g., high or low UV light, Infrared light, etc.), and/or stain etc.
  • instrument e.g., microscope
  • light source e.g., high or low UV light, Infrared light, etc.
  • imaging agents include, but are not limited to, fluorescent and luminescent moieties.
  • the imaging agent is a fluorescent moiety.
  • the fluorescent moiety is selected from Hydroxycoumarin; Aminocoumarin;
  • the fluoresent moiety is Fluorescein.
  • an abnormal number of centrioles or centrosomes means more than one centrosome per cell not undergoing mitosis, or three or more centrioles per cell undergoing mitosis.
  • biological samples include tissue samples, organ biopsies and biological fluids such as saliva, blood, plasma, urine, semen, seminal fluid, amniotic fluid, cerebrospinal fluid, and the like.
  • the present invention also provides a method of identifying compounds which bind to Aurora-A kinase, comprising the steps of: (i) contacting a cancer cell with a test compound of formulae (I) or (II) conjugated to an imaging agent and (ii) screening for binding to Aurora-A kinase.
  • Verification of binding will be visually apparent by a localization of the visual marker (e.g., a fluorescence or luminescence) in the area of the cell where Aurora-A kinase is over-expressed (e.g., the centrosomes).
  • the visual marker e.g., a fluorescence or luminescence
  • the compound of formulae (I) or (II) binds toAurora
  • a kinase protein but does not modulate (e.g., activate, inhibit or partially inhibit) Aurora A kinase protein activity.
  • (II) binds to Aurora A kinase protein, and activates Aurora A kinase protein activity.
  • the compound of formulae (I) or (II) binds to Aurora A kinase protein, and inhibits or partially inhibits Aurora A kinase protein activity.
  • the proliferative cells are cancer cells.
  • the cancer cells are selected from a group consisting of HeLa cells, U2OS cells, MCF7 cells, SW620 human colon adenocarcinoma cells, LS174T human colorectal adenocarcinoma cells, KM 12 human colon carcinoma cells, HCTl 16 human colon carcinoma cells, SR human leukemia cells, RL human non-Hodgkin's lymphoma B-lymphoblasts, PA682 Burkitt's human lymphoma cells, Leukemia (acute myeloid) human cells, HL60 human acute promyelocytic leukemia cells, HCTl 16 human colon carcinoma cells, LS174T human colorectal adenocarcinoma cells, MDAMB231 human breast carcinoma cells, HeLa human cervix adenocarcinoma cells, A375 human malignant melanoma cells, PC3 human prostate cancinoma cells (androgen
  • Shikimic acid is a naturally occurring small molecule, and has been used to synthesize many biologically active natural products (see, for example, Floss Nat. Prod. Rep. (1997) 433) and several potent non-natural compounds such as neuraminidase inhibitors, e.g. Oseltamivir (TamifluTM) (Karpf and Trussardi, J. Org. Chem. (2001) 66: 2044). Shikimic acid-based libraries have been synthesized and tested as glycomimetics (Schuster et al, Org. Lett. (2003) 5:1407).
  • (-)-Shikimic acid (1) was converted to sjw-hydroxyepoxide 2 using a previously reported procedure (McGowan and Berchtold, J. Org. Chem. (1985) 50: 1557) (steps a-c) (Scheme 2).
  • (-)-Shikimic acid (1) was first esterified (Tan et al., J. Am. Chem. Soc. 1998, 120, 8565. Tan et al., J. Am. Chem. Soc. 1999, 121, 9073) then the trans diol was converted to an epoxide and the ester was hydrolyzed to give carboxylic acid 2. Both enantiomeric trans stereoisomers of 2 are also accessible from 1 (Wood and Ganem, J. Am. Chem. Soc. (1990) 112:8907). Scheme 2.
  • Opening of the solid-phase epoxide with amines was tested with varying amounts of an amine (e.g., benzylamine, N-methylbenzylamine) in a number of solvents (THF, CH3C ⁇ , DMF, dioxane, CH 2 CI 2 ) and Lewis acids (MgBr 2 -Et 2 O, LiClO 4 , Yb(OTf) 3 , LiOTf) at various temperatures.
  • This survey revealed that 20-40 equivalents of amine with 1-1.1 equiv of a Lewis acid (e.g., MgBr 2 -Et 2 O) in a solvent (e.g., CH 3 CN) is optimal.
  • SMMs small-molecule microarrays
  • the activity of 1-1 was assessed in a cell-free Aurora A kinase enzymatic activity assay (Aurora-A kinase Assay/Inhibitor Screening Kit, CycLex Co., Ltd., cat. no. CY-1165). Although the binding assay demonstrated that 1-1 possessed good binding affinity, this Aurora A-binding compound did not inhibit kinase activity.
  • Table 2 The activity of 1-1 was assessed in a cell-free Aurora A kinase enzymatic activity assay (Aurora-A kinase Assay/Inhibitor Screening Kit, CycLex Co., Ltd., cat. no. CY-1165). Although the binding assay demonstrated that 1-1 possessed good binding affinity, this Aurora A-binding compound did not inhibit
  • the probed arrays were washed three times in TBST (5 min for each wash), followed by doubly distilled water for 3 min on an orbital platform shaker. Arrays were dried by centrifugation and scanned for fluorescence at 635 nm using a Genepix 4000B microarray scanner.
  • Biacore S51 instrument using Biacore CM5 sensor chips Ethanolamine, EDC, NHS, and P- 20 surfactant were all obtained from Biacore, Inc. Aurora A was directly immobilized through primary amines using standard EDC/NHS chemistry. The sensor surface was conditioned using alternating injection of 10 mM glycine pH 2.2 and 50 mM NaOH. The surface was then activated with 1: 1 4 M EDC: 1 M NHS for 10 minutes. Aurora A diluted to 15 ⁇ g/mL in 10 mM acetate pH 5.5 was exposed to the activated sensor. The surface was quenched by a seven minute injection of 1 M ethanolamine. Protein activity was assessed by observing the binding of staurosporine.
  • Aurora A (2.4 mg/mL) was thawed before the first use and divided into 5.5 ⁇ L aliquots that were stored at -20 0 C. Freshly thawed Aurora A was used for each day's assay.
  • the running buffer used in the preparations was PBS-P pH 7.4 and the temperature of the assay was 25°C.
  • Aurora A was diluted to 150 ug/ml in 1OmM Acetate Buffer pH 5.5 and immobilized on the sensor surface on Spot 1 (day 1) and Spot 2 (day 2) at 5 ⁇ L/min for 10 min using standard EDC/NHS chemistry. Between 10,990 and 11,118 Response Units (RU) of protein were immobilized in each assay.
  • An anti-RGSHis antibody (Qiagen) immobilized at a level of -3,000 RU was used as a reference surface.
  • Compounds 1-1 to 1-7 were diluted from 5 mM stocks in DMF to the appropriate concentration in buffer with the same solvent concentration as the running buffer (2%). Compounds were injected at a flow rate of 30 ⁇ L/min into the flow cell for 60 seconds followed by 90 seconds of buffer with no compound. Compound 1-1 was further characterized by measuring the binding at concentrations from 391 nM to 25 ⁇ M in half dilution. A flow rate of 90 ⁇ L/minute was used in the characterization of compound 1-1. [00196] Compounds of Table 3 were diluted from 10 mM stocks in DMSO to the appropriate concentration in buffer to give the same solvent concentration as the running buffer (PBS-P, 2% DMSO).
  • U2OS osteosarcoma
  • HeLa cervix cancer
  • MCF7 breast cancer cells were used for immunofluorescence analyses.
  • Cells were grown on glass coverslips, pre- extracted at room temperature with CSK buffer (100 mM NaCl, 300 mM sucrose, 10 mM PIPES-pH6.8, and 3 mM MgCl 2 ) with 0.1% Tritin-XIOO and rinsed with CSK buffer. Cells were then fixed and permeabilized in cold methanol for 10 min at -20 C, followed by rehydrated with PBS for 5 min.
  • CSK buffer 100 mM NaCl, 300 mM sucrose, 10 mM PIPES-pH6.8, and 3 mM MgCl 2
  • Normal goat serum (Jackson ImmunoResearch) was used for blocking samples at the concentrations of 10% in SNBP buffer (BSA (5g/L), saponin (0.1 g/L), sodium azide (0.25 g/L) in PBS) for 1 h at room temperature.
  • FITC-labeled compound 1-33 (provided by method of Scheme 4) was incubated at the concentrations of 2.2 ⁇ M in the presence of polyclonal antibody against Aurora-A (Abeam, cat. #abl3408) at a dilution of 1 : 1000 for 30 min at 37 C, followed by additional 30 min at room temperature. Samples were washed with SNBP buffer and incubated with Alexa Fluor 647 goat anti-rabbit IgG antibody (Molecular Probes, cat.
  • Enzyme inhibition assays were run with a commercially available Aurora-A kinase Assay/Inhibitor Screening Kit, CycLex Co., Ltd., (cat. no. CY-1165) as instructed. Briefly, compounds at various concentrations were incubated with Aurora-A positive control (CycLex Co., Ltd., cat. no. CY-El 165) and ATP in kinase reaction buffer in microtiter wells at 30 C for 45 minutes. Wells were washed with wash buffer (5x200 ⁇ l). 100 ⁇ l of anti- phospho-Lats2-S83 antibody solution (supplied with the kit) was added to each well and incubated at room temperature for 30 min.
  • Resin samples were washed as indicated and solvent was removed under argon flow and/or in vacuum on a Vac-Man® laboratory vacuum manifold (Promega, Madison, WI; A7231) fitted with nylon 3-way stopcocks (Biorad 732-8107). Resin sample in glass vials were filtered in 10 mL Amersham column and washed on the Vac-Man vacuum manifold.
  • the beads were allowed to swell in anhydrous DMF for 30 min before adding a solution of 20% piperidine in DMF (10 mL) and the reaction tube was agitated for 5 h before the beads were drained, and subjected to rinsing method A. Loading level was determined spectrophotometrically as per Tallarico, et al, J. Comb. Chem. 2001, 3, 312. The resin (3) was air-dried for 2 h and then placed under vacuum for 12 h to remove solvent. [00207] Solid Phase Amide Coupling (4).
  • Resin 3 (15 mg, 1.17 mmol Si/g, 18 ⁇ mol) was allowed to swell for 30 min in 5 mL of anhydrous ⁇ MP before the addition of PyBOP (14 mg, 26 ⁇ mol), DIPEA (9.2 ⁇ L, 53 ⁇ mol) and organic acid (4.1 mg, 26 ⁇ mol) and the reaction mixture was gently agitated for 24 h. The beads were drained and rinsed using method A. The resin (4) was then air-dried for 2 h and then placed under vacuum for 12 h.
  • the beads were drained and subjected to rinsing with method B after washing with NaCN solution (7/3 THF/ 1 M NaCN (aq ), 10 mL x 3); cleavage from the resin resulted in 6.2 mg of the expected fused-ring product (75% yield from resin 7).
  • o-iodophenol DEAD, Ph 3 P/THF, -78 0 C to -20 0 C, 90%
  • c) TBDMSOCH 2 CH 2 NH 2 , EDAC, HOBt, PyBOP, DIPEA/CH 2 C1 2 , 80%, d) 2-methoxyethylamme, MgBr 2 OEt 2 / CH 3 CN, 60 0 C, 88%, e) pentanoyl chloride, DIPEA/CH 2 C1 2 , 90%, f) HF py ⁇ dine/py ⁇ dme/THF 5 10 85, 3 h then TMSOEt, 0 5 h, 86%

Abstract

The present invention provides inventive compounds of formulae (I) and (II), pharmaceutically acceptable forms thereof, pharmaceutical compositions thereof, and methods for treatment and their use.

Description

Shikimic Acid-Derived Compounds for Inhibition and Detection of Aurora A-Associated Tumors
Government Support
[0001] This invention was made with Government support under grant 20XS139A awarded by the Initiative for Chemical Genetics, grant GM38627 awarded by the National Institute of General Medical Sciences, and grants 1U01HL80731 and 1K08HL077186 awarded by the National Institutes of Health. The United States government has certain rights in the invention.
Priority Information
[0002] This application claims priority under 35 U.S. C. § 119(e) to United States provisional patent application, U.S. S.N. 60/841,035, filed August 30, 2006, which is incorportated herein by reference.
Background to the Invention
[0003] Members of the Aurora kinase family are serine/threoinin kinases which are involved in mitosis (Keen and Taylor., Nat. Rev. Cancer (2004) 4:927). These kinases have been found to regulate multiple steps in mitosis including centrosome duplication, formation of bipolar mitotic spindles, and chromosome alignment on the mitotic spindle. Centrosomes act as mitotic spindle pole bodies during mitosis. Abnormal centrosome numbers are frequently detected in human solid tumors and are a hallmark of genomically unstable cells. Three Aurora kinases, A, B, and C, are known. Aurora A kinase localizes to the duplicated centrosomes and to the spindle poles during mitosis and assists with centrosome maturation and separation. Aurora B kinase is a chromosomal passenger protein which is localized to the centromeric regions of the chromosomes in the early stages of mitosis and accumulates in the spindle midzone and midbody. The role of Aurora C kinase is unknown currently. [0004] The Aurora kinase family is involved in regulating multiple steps in mitosis, including centrosome duplication, formation of bipolar mitotic spindles, and chromosome alignment on the mitotic spindle (Keen and Taylor, Nat. Rev. Cancer (2004) 4:921). Centrosomes act as mitotic spindle pole bodies during mitosis. Abnormal centrosome numbers are frequently detected in human solid tumors and are a hallmark of genomically unstable cells. Small molecules that associate with, but do not inhibit, centrosomal components can serve as markers for cells with centrosome abnormalities in tumor cells, and thus be useful as diagnostic tools in the detection of cancer cells. [0005] Amplification and overexpression of Aurora A kinase has been observed in several types of human cancers, including cancers of the colon, breast, pancreas, ovaries, and stomach, among others. High level Aurora A expression in human head and neck squamous cell carcinoma is a negative predictor for various clinical parameters including disease stage, presence of local and distant metastases as well as overall patient survival (Reiter et al., Clin. Cancer Res. (2006) 72:5136). Several small-molecule inhibitors of Aurora A kinase are in clinical trials and have been show to inhibit cell-cycle progression and induce apoptosis in multiple human tumor types in vitro and in animal models (Harrington et al., Nat. Med. (2004) 70:262; Gadea and Ruderman, MoL Biol. Cell. (2005) 76: 1305; Hauf et al., J. Cell Biol. (2003) 767:281)
Summary of the Invention [0006] The present invention provides inventive compounds of formulae (I) and (II),
Figure imgf000004_0001
or pharmaceutically acceptable forms thereof; wherein Ri, R2, R3, R4, R5, Re and n are described herein; and pharmaceutical compositions thereof. Compounds of the present invention bind to Aurora kinase, and are useful as anti-proliferative agents and diagnostic agents for the detection of proliferating and pre-cancerous cells.
[0007] For example, in one aspect, the present invention provides a method of treating a proliferative disease (e.g., cancer) comprising the step of adminiserting a therapeutically effective amount of a compound of formulae (I) or (II), or a pharmaceutically acceptable form thereof, to a subject in need thereof.
[0008] In another aspect, the present invention provides a method of detecting proliferative cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes. [0009] In yet another aspect, the present invention provides a method of detecting pre-cancerous cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes. [0010] In still yet another aspect, the present invention provides a method of identifying compounds which bind to Aurora-A kinase, comprising the steps of: (i) contacting a cancer cell with a test compound of formulae (I) or (II) conjugated to an imaging agent and (ii) screening for binding to Aurora-A kinase.
[0011] This application refers to various issued patents, published patent applications, journal articles, and other publications, all of which are incorporated herein by reference. [0012] The details of one or more embodiments of the invention are set forth in the accompanying Figures and the Detailed Description of Certain Embodiments, as described below. Other features, objects, and advantages of the invention will be apparent from the description, the figures, and from the claims.
Definitions
[0013] Definitions of specific functional groups and chemical terms are described in more detail below. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed., inside cover, and specific functional groups are generally defined as described therein. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Organic Chemistry, Thomas Sorrell, University Science Books, Sausalito, 1999; Smith and March March 's Advanced Organic Chemistry, 5l Edition, John Wiley & Sons, Inc., New York, 2001; Larock, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; Carruthers, Some Modern Methods of Organic Synthesis, 3rd Edition, Cambridge University Press, Cambridge, 1987; the entire contents of each of which are incorporated herein by reference. [0014] The compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis- and ϊrαws-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
[0015] Where an isomer/enantiomer is preferred, it may, in some embodiments, be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched." "Optically enriched," as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer. In certain embodiments the compound of the present invention is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer. Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses. See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen, S. H., et al., Tetrahedron 33:2725 (1977); Eliel, E.L. Stereochemistry of Carbon Compounds (McGraw- Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972).
[0016] It will be appreciated that the compounds of the present invention, as described herein, may be substituted with any number of substituents or functional moieties. In general, the term "substituted" whether preeceded by the term "optionally" or not, and substituents contained in formulas of this invention, refer to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent. When more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. [0017] As used herein, the term "substituted" is contemplated to include substitution with all permissible substituents of organic compounds, any of the substituents described herein (for example, aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, etc.), and any combination thereof (for example, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, arylthio; heteroaryloxy, heteroarylthio, and the like) that results in the formation of a stable moiety. The present invention contemplates any and all such combinations in order to arrive at a stable substituent/moiety. For purposes of this invention, heteroatoms such as nitrogen may have hydrogen substituents and/or any suitable substituent as described herein which satisfy the valencies of the heteroatoms and results in the formation of a stable moiety. [0018] The term "acyl," as used herein, refers to a group having the general formula -
C(O)R , -C(O)OR , -C(O)-O-C(O)R , -C(O)SR , -C(O)N(R)2, -C(=S)R , - C(=S)N(R')2, and -C(=S)S(R ), -C(=NR')R', -C(=NR')OR', -C(=NR')SR', and - C(=NR )N(R )2, wherein R is hydrogen; halogen; optionally substituted hydroxyl; optionally substituted thiol; optionally substituted amino; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched alkyl; cyclic or acyclic, substituted or unsubstituted, branched or unbranched alkenyl; substituted or unsubstituted alkynyl; optionally substituted aryl, optionally substituted heteroaryl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy, mono- or di- aliphaticamino, mono- or di- heteroaliphaticamino, mono- or di- alkylamino, mono- or di- heteroalkylamino, mono- or di- arylamino, or mono- or di- heteroarylamino; or two R groups taken together form a 5- to 6- membered heterocyclic ring. Exemplary acyl groups include aldehydes (-CHO), carboxylic acids (-CO2H), ketones, acyl halides, esters, amides, imines, carbonates, carbamates, and ureas. Acyl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, aliphaticamino, heteroaliphaticamino, alkylamino, heteroalkylamino, arylamino, heteroarylamino, alkylaryl, arylalkyl, aliphaticoxy, heteroaliphaticoxy, alkyloxy, heteroalkyloxy, aryloxy, heteroaryloxy, aliphaticthioxy, heteroaliphaticthioxy, alkylthioxy, heteroalkylthioxy, arylthioxy, heteroarylthioxy, and the like, each of which may or may not be further substituted).
[0019] The term "aliphatic," as used herein, includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, and cyclic (i.e., carbocyclic) hydrocarbons, which are optionally substituted with one or more functional groups. As will be appreciated by one of ordinary skill in the art, "aliphatic" is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties. Thus, as used herein, the term "alkyl" includes straight, branched and cyclic alkyl groups (for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl, cyclodecenyl,and the like, and which may bear one or more substituents). An analogous convention applies to other generic terms such as "alkenyl", "alkynyl", and the like. Furthermore, as used herein, the terms "alkyl", "alkenyl", "alkynyl", and the like encompass both substituted and unsubstituted groups. In certain embodiments, as used herein, "aliphatic" is used to indicate those aliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms (i.e., Ci_2o aliphatic) [in situations wherein the number of carbons is specified, such as a Ci 5 aliphatic, is meant a carbon chain 15 carbons in length]. In certain embodiments, the aliphatic group employed by the invention is an optionally substituted C1-C15 aliphatic group. In certain embodiments, the aliphatic group employed by the invention is an optionally substituted C1-C10 aliphatic group. In certain embodiments, the aliphatic group employed by the invention is an optionally substituted C1-C5 aliphatic group. Aliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0020] The term "alkyloxy" or "alkoxy" as used herein, refers to an aliphatic group attached to the parent molecule via an oxygen atom.
[0021] The term "alkylthio" as used herein, refers to an aliphatic group attached to the parent molecule via a sulfur atom.
[0022] The term "alkyl," as used herein, refers to a saturated, straight- or branched- chain hydrocarbon radical derived from a hydrocarbon moiety which is optionally substituted with one or more functional groups. In some embodiments, the alkyl group employed in the invention contains 1-20 carbon atoms (i.e., Ci_2o alkyl) [in situations wherein the number of carbons is specified, such as a C15 alkyl, is meant a fully saturated carbon chain 15 carbons in length]. In certain embodiments, the alkyl group employed by the invention is an optionally substituted C1-C15 alkyl group. In certain embodiments, the alkyl group employed by the invention is an optionally substituted Ci-Ci0 alkyl group. In certain embodiments, the alkyl group employed by the invention is an optionally substituted C1-C5 alkyl group. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, sec-pentyl, iso-pentyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, sec- hexyl, n-heptyl, n-octyl, n-decyl, n-undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, and the like, which may bear one or more sustitutents. Alkyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0023] The term "alkenyl," as used herein, denotes a monovalent group derived from a straight- or branched-chain hydrocarbon moiety having at least one carbon-carbon double bond, and which is optionally substituted with one or more functional groups. In certain embodiments, the alkenyl group employed in the invention contains 2-20 carbon atoms (i.e., C2-20 alkenyl) [in situations wherein the number of carbons is specified, such as a C15 alkenyl, is meant a carbon chain 15 carbons containing at least one sp2 (double bonded) carbon atom]. In certain embodiments, the alkenyl group employed by the invention is an optionally substituted C2-C15 alkenyl group. In certain embodiments, the alkenyl group employed by the invention is an optionally substituted C2-C10 alkenyl group. In certain embodiments, the alkenyl group employed by the invention is an optionally substituted C2-C5 alkenyl group. Alkenyl groups include, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, and the like, which may bear one or more substituents. Alkenyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted). [0024] The term "alkynyl," as used herein, refers to a monovalent group derived from a straight- or branched-chain hydrocarbon having at least one carbon-carbon triple bond, and which is optionally substituted with one or more functional groups. In certain embodiments, the alkynyl group employed in the invention contains 2-20 carbon atoms (i.e., C2-20 alkenyl) [in situations wherein the number of carbons is specified, such as a Ci 5 alkynyl, is meant a carbon chain 15 carbons containing at least one sp (triple bonded) carbon atom]. In certain embodiments, the alkynyl group employed by the invention is an optionally substituted C2-C15 alkynyl group. In certain embodiments, the alkynyl group employed by the invention is an optionally substituted C2-C10 alkynyl group. In certain embodiments, the alkynyl group employed by the invention is an optionally substituted C2-C5 alkynyl group. Representative alkynyl groups include, but are not limited to, ethynyl, 2-propynyl (propargyl), 1-propynyl, and the like, which may bear one or more substituents. Alkynyl group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0025] The term "amino," as used herein, refers to a group of the formula (-NH2).
An "substituted amino" refers to a mono-substituted amino group of the formula (-NHRΨ) or a di-substituted amino group of the formula (-NRΨ 2). Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., an amino protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, and the like, each of which may or may not be further substituted). In certain embodiments, the Rψ substituents of the di-substituted amino group(-
NRΨ 2) form a 5- to 6- membered hetereocyclic ring.
[0026] The term "aryl," as used herein, refer to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which all the ring atoms are carbon, and which may be substituted or unsubstituted. In certain embodiments of the present invention, "aryl" refers to a mono, bi, or tricyclic C4-C20 aromatic ring system having one, two, or three aromatic rings which include, but not limited to, phenyl, biphenyl, naphthyl, and the like, which may bear one or more substituents. Aryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted, and the like, each of which may or may not be further substituted).
[0027] The term "aryloxy" refers to an aryl group attached to the parent molecule via an oxygen atom.
[0028] The term "arylthio" refers to an aryl group attached to the parent molecule via a sulfur atom.
[0029] The term "azido," as used herein, refers to a group of the formula (-N3). An
"optionally substituted azido" refers to a group of the formula (-N3RΦ), wherein Rφ can be any substitutent (other than hydrogen). Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0030] The term "cyano," as used herein, refers to a group of the formula (-CN). [0031] The terms "halo" and "halogen" as used herein refer to an atom selected from fluorine (fluoro, -F), chlorine (chloro, -Cl), bromine (bromo, -Br), and iodine (iodo, -I). [0032] The term "heteroaliphatic," as used herein, refers to an aliphatic moiety, as defined herein, which includes both saturated and unsaturated, nonaromatic, straight chain (i.e., unbranched), branched, acyclic, cyclic (i.e., heterocyclic), or poly cyclic hydrocarbons, which are optionally substituted with one or more functional groups, and that contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms. In certain embodiments, heteroaliphatic moieties are substituted by independent replacement of one or more of the hydrogen atoms thereon with one or more substituents. As will be appreciated by one of ordinary skill in the art, "heteroaliphatic" is intended herein to include, but is not limited to, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl moieties. Thus, as used herein, the term "heteroalkyl" includes straight, branched and cyclic alkyl groups that contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms, e.g., in place of carbon atoms. An analogous convention applies to other generic terms such as "heteroalkenyl", "heteroalkynyl", and the like. Furthermore, as used herein, the terms "heteroalkyl", "heteroalkenyl", "heteroalkynyl", and the like encompass both substituted and unsubstituted groups. In certain embodiments, as used herein, "heteroaliphatic" is used to indicate those heteroaliphatic groups (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms (i.e., Ci_2o heteroaliphatic) [in situations wherein the number of carbons is specified, such as a C15 heteroaliphatic, is meant a carbon chain 15 carbons in length]. In certain embodiments, the heteroaliphatic group employed by the invention is an optionally substituted C1-C15 heteroaliphatic group. In certain embodiments, the heteroaliphatic group employed by the invention is an optionally substituted C1-C10 heteroaliphatic group. In certain embodiments, the heteroaliphatic group employed by the invention is an optionally substituted C1-C5 heteroaliphatic group. Heteroaliphatic group substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0033] The term "heteroaryloxy" refers to a heteroaryl group attached to the parent molecule through an oxygen atom. [0034] The term "heteroarylthio" refers to a heteroaryl group attached to the parent molecule through a sulfur atom.
[0035] The term "heterocyclic," "heterocycles," or "heterocyclyl," as used herein, refers to a cyclic heteroaliphatic. A heterocyclic group refers to a non-aromatic, partially unsaturated or fully saturated, 3- to 10-membered ring system, which includes single rings of 3 to 8 atoms in size, and bi- and tri-cyclic ring systems which may include aromatic five- or six-membered aryl or heteroaryl groups fused to a non-aromatic ring. These heterocyclic rings include those having from one to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, in which the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. In certain embodiments, the term heterocylic refers to a non-aromatic 5-, 6-, or 7-membered ring or polycyclic group wherein at least one ring atom is a heteroatom selected from O, S, and N (wherein the nitrogen and sulfur heteroatoms may be optionally oxidized), and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms. Heterocycyl groups include, but are not limited to, a bi- or tri-cyclic group, comprising fused five, six, or seven-membered rings having between one and three heteroatoms independently selected from the oxygen, sulfur, and nitrogen, wherein (i) each 5-membered ring has 0 to 2 double bonds, each 6-membered ring has 0 to 2 double bonds, and each 7-membered ring has 0 to 3 double bonds, (ii) the nitrogen and sulfur heteroatoms may be optionally oxidized, (iii) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above heterocyclic rings may be fused to an aryl or heteroaryl ring. Exemplary heterocycles include azacyclopropanyl, azacyclobutanyl, 1,3-diazatidinyl, piperidinyl, piperazinyl, azocanyl, thiaranyl, thietanyl, tetrahydrothiophenyl, dithiolanyl, thiacyclohexanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropuranyl, dioxanyl, oxathiolanyl, morpholinyl, thioxanyl, tetrahydronaphthyl, and the like, which may bear one or more substituents. Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0036] The term "heteroaryl," as used herein, refer to stable aromatic mono- or polycyclic ring system having 3-20 ring atoms, of which one ring atom is selected from S, O, and N; zero, one, or two ring atoms are additional heteroatoms independently selected from S, O, and N; and the remaining ring atoms are carbon, the radical being joined to the rest of the molecule via any of the ring atoms. Exemplary heteroaryls include, but are not limited to pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, pyyrolizinyl, indolyl, quinolinyl, isoquinolinyl, benzoimidazolyl, indazolyl, quinolinyl, isoquinolinyl, quinolizinyl, cinnolinyl, quinazolynyl, phthalazinyl, naphthridinyl, quinoxalinyl, thiophenyl, thianaphthenyl, furanyl, benzofuranyl, benzothiazolyl, thiazolynyl, isothiazolyl, thiadiazolynyl, oxazolyl, isoxazolyl, oxadiaziolyl, oxadiaziolyl, and the like, which may bear one or more substituents. Heteroaryl substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted). [0037] The term "hydroxy," or "hydroxyl," as used herein, refers to a group of the formula (-OH). An "optionally substituted hydroxyl" refers to a group of the formula (- OR§), wherein R§ can be hydrogen, or any substitutent which results in a stable moiety (e.g., a suitable hydroxyl protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, oxo, imino, thiooxo, cyano, isocyano, amino, azido, nitro, hydroxyl, thiol, halo, and the like, each of which may or may not be further substituted).
[0038] The term "imino," as used herein, refers to a group of the formula (=NR¥), wherein R¥ corresponds to hydrogen or any substitutent as described herein, that results in the formation of a stable moiety (for example, a suitable amino protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, and the like, each of which may or may not be further substituted).
[0039] The term "isocyano," as used herein, refers to a group of the formula (-NC).
[0040] The term "nitro," as used herein, refers to a group of the formula (-NO2).
[0041] The term "oxo," as used herein, refers to a group of the formula (=0).
[0042] The term "thiooxo," as used herein, refers to a group of the formula (=S).
[0043] The term "thio," or "thiol," as used herein, refers to a group of the formula (-
SH). An "optionally substituted thiol" refers to a group of the formula (-SRΩ), wherein RΩ can be hydrogen, or any substitutent. Substituents include, but are not limited to, any of the substituents described herein, that result in the formation of a stable moiety (e.g., a suitable thiol protecting group; aliphatic, alkyl, alkenyl, alkynyl, heteroaliphatic, heterocyclic, aryl, heteroaryl, acyl, sulfinyl, sulfonyl, cyano, nitro, alkylaryl, arylalkyl, and the like, each of which may or may not be further substituted). [0044] The term "stable moiety," as used herein, preferably refers to a moiety which possess stability sufficient to allow manufacture, and which maintains its integrity for a sufficient period of time to be useful for the purposes detailed herein.
[0045] As used herein, the term "resin" refers to a resin useful for solid phase synthesis. A resin may be functionalized with one or more linker groups prior to attachment of the compound to the solid phase, and such functionalization with linker groups is well within the scope of this invention. Solid phase synthesis is a well-known synthetic technique; see generally, Atherton, E., Sheppard, R.C. Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, Oxford, England, 1989, and Stewart J.M., Young, J.D. Solid Phase Peptide Synthesis, 2nd edition, Pierce Chemical Company, Rockford, 1984, the entire contents of each of which are hereby incorporated herein by reference. Exemplary resins which may be employed by the present invention include, but are not limited to:
(1) alkenyl resins (e.g., REM resin, vinyl sulfone polymer-bound resin, vinyl- polystyrene resin);
(2) amine functionalized resins (e.g., amidine resin, N-(4- Benzyloxybenzyl)hydroxylamine polymer bound, (aminomethyl)polystyrene, polymer bound (7?)-(+)-a-methylbenzylamine, 2-Chlorotrityl Knorr resin, 2-N-Fmoc-Amino- dibenzocyclohepta-l,4-diene, polymer-bound resin, 4-[4-(l-Fmoc-aminoethyl)-2- methoxy-5-nitrophenoxy]butyramidomethyl-polystyrene resin, 4-Benzyloxybenzylamine, polymer-bound, 4-Carboxybenzenesulfonamide, polymer-bound, Bis(tert- butoxycarbonyl)thiopseudourea, polymer-bound, Dimethylaminomethyl-polystyrene, Fmoc- 3-amino-3-(2-nitrophenyl)propionic acid, polymer-bound, Ν-Methyl aminomethylated polystyrene, PAL resin, Sieber amide resin, tert-BvXyl N-(2-mercaptoethyl)carbamate, polymer-bound, Triphenylchloromethane-4-carboxamide polymer bound);
(3) benzhydrylamine (BHA) resins (e.g., 2-Chlorobenzhydryl chloride, polymer- bound, HMPB-benzhydrylamine polymer bound, 4-Methylbenzhydrol, polymer-bound, Benzhydryl chloride, polymer-bound, Benzhydrylamine polymer-bound);
(4) Br-functionalized resins (e.g., 4-(Benzyloxy)benzyl bromide polymer bound, A- Bromopolystyrene, Brominated PPOA resin, Brominated Wang resin, Bromoacetal, polymer-bound, Bromopolystyrene, HypoGel® 200 Br, Polystyrene A-Br for peptide synthesis, Selenium bromide, polymer-bound, TentaGel HL-Br, TentaGel MB-Br, TentaGel S-Br, TentaGel S-Br); (5) Chloromethyl resins (e.g., 5-[4-(Chloromethyl)phenyl]pentyl]styrene, polymer- bound, 4-(Benzyloxy)benzyl chloride polymer bound, 4-Methoxybenzhydryl chloride, polymer-bound);
(6) CHO-functionalized resins (e.g., (4-Formyl-3- methoxyphenoxymethyl)polystyrene, (4-Formyl-3-methoxyphenoxymethyl)polystyrene, 3- Benzyloxybenzaldehyde, polymer-bound, 4-Benzyloxy-2,6- dimethoxybenzaldehyde,polymer-bound, Formylpolystyrene, HypoGel® 200 CHO, Indole resin, Polystyrene A-CH(OEt)2, TentaGel HL-CH(OEt)2);
(7) Cl-functionalized resins (e.g., Benzoyl chloride polymer bound, (Chloromethyl)polystyrene, Merrifield's resin);
(8) CO2H functionalized resins (e.g., Carboxyethylpolystryrene, HypoGel® 200 COOH, Polystyrene AM-COOH, TentaGel HL-COOH, TentaGel MB-COOH, TentaGel S- COOH);
(9) Hypo-Gel resins ( e.g., HypoGel® 200 FMP, HypoGel® 200 PHB , HypoGel® 200 Trt-OH , HypoGel® 200 HMB );
(10) I-functionalized resins (e.g., 4-Iodophenol, polymer-bound, Iodopolystyrene); Janda-Jels™ (JandaJef- Rink amide, JandaJel-NH2, JandaJel-Cl, JandaJeM- Mercaptophenol, JandaJel-OH, JandaJel-l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide, JandaJel- 1, 3,4,6,7, 8-hexahydro-2H-pyrimido-[l,2-α] pyrimidine, JandaJel-morpholine, JandaJel-polypyridine, JandaJel-Triphenylphosphine, JandaJel-Wang);
(11) MBHA resins (3 [4'-(Ηydroxymethyl)phenoxy] propionic acid^- methylbenzhydrylamine resin, 4-(Hydroxymethyl)phenoxyacetic acid polymer-bound to MBHA resin, HMBA^-methylbenzhydrylamine polymer bound, 4-Methylbenzhydrylamine hydrochloride polymer bound Capacity (amine));
(12) NH2 functionalized resins ((Aminomethyl)polystyrene, (Aminomethyl)polystyrene, HypoGel® 200 NH2, Polystyrene AM-NH2, Polystyrene Microspheres 2-aminoethylated, Polystyrol Microspheres 2-bromoethylated, Polystyrol Microspheres 2-hydroxyethylated, TentaGel HL-NH2, Tentagel M Br, Tentagel M NH2, Tentagel M OH, TentaGel MB-NH2, TentaGel S-NH2, TentaGel S-NH2);
(13) OH-functionalized resins (e.g., 4-Hydroxymethylbenzoic acid, polymer-bound, Hydroxymethyl Resins, OH-functionalized Wang Resins);
(14) oxime resins (e.g., 4-Chlorobenzophenone oxime polymer bound, Benzophenone oxime polymer bound, 4-Methoxybenzophenone oxime polymer bound);
(15) PEG resins (e.g., ethylene glycol polymer bound); (16) Boc-/Blz peptide synthesis resins (e.g., Boc-Lys(Boc)-Lys[Boc-Lys(Boc)]- Cys(Acm)-b-Ala-O-PAM resin, Boc-Lys(Fmoc)-Lys[Boc-Lys(Fmoc)]-b-Ala-O-Pam resin, Boc-Lys^o^-LysfBoc-Lys^oc^-Lys (BOC-LyS(BoC)-LyS[BoC-LyS(BoC)]!^- AIa-O-PAM resin, Boc-Lys(Fmoc)-Lys[Boc-Lys(Fmoc)]-Lys {Boc-Lys(Fmoc)-Lys[Boc- Lys(Fmoc)]}-b-Ala-O-PAM resin, Boc-Lys(Boc)-Lys[Boc-Lys(Boc)]-Lys{Boc- Lys(Boc)-Lys[Boc-Lys(Boc)]}-Cys(Acm)-b-Ala-O-PAM resin, Preloaded PAM resins);
(17) Fmoc-/t-Bu peptide synthesis resins (e.g., Fmoc-Lys(Fmoc)-Lys[Fmoc- Lys(Fmoc)]-b-Ala-O-Wang resin, Fmoc-Lys(Fmoc)-Lys[Fmoc-Lys(Fmoc)]-Lys {Fmoc- Lys(Fmoc)-Lys[Fmoc-Lys(Fmoc)]}-b-Ala-O-Wang resin, Preloaded TentaGel® S Trityl Resins, Preloaded TentaGel® Resins, Preloaded Trityl Resins, Preloaded Wang Resins, Trityl Resins Preloaded with Amino Alcohols);
(19) thiol-functionalized resins (e.g., HypoGel® 200 S-Trt, Polystyrene AM-S- Trityl, TentaGel HL-S-Trityl, TentaGel MB-S-Trityl, TentaGel S-S-Trityl);
(20) Wang resins (e.g., Fmoc-Ala-Wang resin, Fmoc-Arg(Pbf)-Wang resin, Fmoc- Arg(Pmc)-Wang resin, Fmoc-Asn(Trt)-Wang resin, Fmoc-Asp(OtBu)-Wang resin, Fmoc- Cys(Acm)-Wang resin, Fmoc-Cys(StBu)-Wang resin, , Fmoc-Cys(Trt) Wang resin, Fmoc- Gln(Trt)-Wang resin, Fmoc-Glu(OtBu)-Wang resin, Fmoc-Gly-Wang resin, Fmoc- His(Trt)-Wang resin, Fmoc-Ile-Wang resin, Fmoc-Leu-Wang resin, Fmoc-Lys(Boc)- Wang resin, Fmoc-Met-Wang resin, Fmoc-D-Met-Wang resin, Fmoc-Phe-Wang resin, Fmoc-Pro-Wang resin, Fmoc-Ser(tBu)-Wang resin, Fmoc-Ser(Trt)-Wang resin, Fmoc- Thr(tBu)-Wang resin, Fmoc-Trp(Boc) Wang resin, Fmoc-Trp-Wang resin, Fmoc-Tyr(tBu)- Wang resin, Fmoc-Val-Wang resin); and
(21) silicon-functionalized resin (e.g., 4-methoxyphenyl)diisopropylsilylpropyl polystyrene).
[0046] The term "linker," as used herein, refers to a chemical moiety utilized to covalently attach a resin, a fluoroescent or luminescent moiety to a compound of formulae (I) or (II).
[0047] A "fluorescent label" or "fluorophore" or "fluroescent moiety" as used herein, is a component of a molecule which causes a molecule to be fluorescent. It is a functional group in a molecule which will absorb energy of a specific wavelength and re-emit energy at a different (but equally specific) wavelength. The amount and wavelength of the emitted energy depend on both the fluorophore and the chemical environment of the fluorophore. This technology has particular importance in the field of biochemistry and protein studies, eg. in immunofluorescence and immunohistochemistry. Exemplary fluorescent labels are provided in Figure 7, and include reactive and conjugated probes, Alexa Fluor dyes (Molecular Probes) Cy Dyes (AP Biotech) Nucleic acid probes, Cell function probes, Fluorescent Proteins, and are also described in U.S. Patent Publication 2004/0067503; Valeur, B., "Molecular Fluorescence: Principles and Applications," John Wiley and Sons, 2002; and Handbook of Fluorescent Probes and Research Products, Molecular Probes, 9th edition, 2002.
[0048] A "luminescent label" or "luminescent moiety" as used herein, is a component of a molecule which causes a molecule to be chemiluminescent. Chemiluminescence is a common technique for a variety of detection assays in biology. A horseradish peroxidase enzyme (HRP) is tethered to the molecule of interest (usually through labeling an immunoglobulin that specifically recognizes the molecule). This enzyme complex, then catalyzes the conversion of the luminescent moiety into a sensitized moiety, which on further oxidation by hydrogen peroxide, produces a triplet (excited) carbonyl which emits light when it decays to the singlet carbonyl. Enhanced chemiluminescence allows detection of minute quantities of a molecule. Proteins can be detected down to femtomole quantities well below the detection limit for most assay systems.
[0049] A "protecting group" refers to an amino, hydroxyl, thiol, or carboxylic acid protecting group.
[0050] An "amino-protecting group," as used herein, is well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference. Suitable amino-protecting groups include methyl carbamate, ethyl carbamante, 9-fluorenylmethyl carbamate (Fmoc), 9-(2- sulfo)fluorenylmethyl carbamate, 9-(2,7-dibromo)fluoroenylmethyl carbamate, 2,7— di— f— butyl-[9-( 10,10-dioxo- 10, 10, 10, 10-tetrahydrothioxanthy I)] methyl carbamate (DBD-Tmoc), 4-methoxyphenacyl carbamate (Phenoc), 2,2,2-trichloroethyl carbamate (Troc), 2- trimethylsilylethyl carbamate (Teoc), 2-phenylethyl carbamate (hZ), 1 -(I -adamanty I)-I- methylethyl carbamate (Adpoc), l,l-dimethyl-2-haloethyl carbamate, l,l-dimethyl-2,2- dibromoethyl carbamate (DB-ϊ-BOC), l,l-dimethyl-2,2,2-trichloroethyl carbamate (TCBOC), l-methyl-l-(4-biphenylyl)ethyl carbamate (Bpoc), l-(3,5-di-?-butylphenyl)-l- methylethyl carbamate (ϊ-Bumeoc), 2-(2'- and 4'-pyridyl)ethyl carbamate (Pyoc), 2-(N,N- dicyclohexylcarboxamido)ethyl carbamate, ?-butyl carbamate (BOC), 1-adamantyl carbamate (A doc), vinyl carbamate (Voc), allyl carbamate (Alloc), 1-isopropylallyl carbamate (Ipaoc), cinnamyl carbamate (Coc), 4-nitrocinnamyl carbamate (Noc), 8-quinolyl carbamate, N-hydroxypiperidinyl carbamate, alkyldithio carbamate, benzyl carbamate (Cbz), p-methoxybenzyl carbamate (Moz), p-nitobenzyl carbamate, p-bromobenzyl carbamate, p- chlorobenzyl carbamate, 2,4-dichlorobenzyl carbamate, 4-methylsulfinylbenzyl carbamate (Msz), 9-anthrylmethyl carbamate, diphenylmethyl carbamate, 2-methylthioethyl carbamate, 2-methylsulfonylethyl carbamate, 2-(p-toluenesulfonyl)ethyl carbamate, [2— (1,3— dithianyl)]methyl carbamate (Dmoc), 4-methylthiophenyl carbamate (Mtpc), 2,4- dimethylthiophenyl carbamate (Bmpc), 2-phosphonioethyl carbamate (Peoc), 2- triphenylphosphonioisopropyl carbamate (Ppoc), l,l-dimethyl-2-cyanoethyl carbamate, m— chloro-p-acyloxybenzyl carbamate, />-(dihydroxyboryl)benzyl carbamate, 5- benzisoxazolylmethyl carbamate, 2-(trifluoromethyl)-6-chromonylmethyl carbamate (Tcroc), m-nitrophenyl carbamate, 3,5-dimethoxybenzyl carbamate, o-nitrobenzyl carbamate, 3,4-dimethoxy-6-nitrobenzyl carbamate, phenyl(ø-nitrophenyl)methyl carbamate, phenothiazinyl-(10)-carbonyl derivative, N'-p-toluenesulfonylaminocarbonyl derivative, N'-phenylaminothiocarbonyl derivative, ?-amyl carbamate, S-benzyl thiocarbamate, />-cyanobenzyl carbamate, cyclobutyl carbamate, cyclohexyl carbamate, cyclopentyl carbamate, cyclopropylmethyl carbamate, />-decyloxybenzyl carbamate, 2,2- dimethoxycarbonylvinyl carbamate, o-(N,N-dimethylcarboxamido)benzyl carbamate, 1,1- dimethyl-3-(N,N-dimethylcarboxamido)propyl carbamate, 1,1-dimethylpropynyl carbamate, di(2-pyridyl)methyl carbamate, 2-furanylmethyl carbamate, 2-iodoethyl carbamate, isoborynl carbamate, isobutyl carbamate, isonicotinyl carbamate, p-(p '~ methoxyphenylazo)benzyl carbamate, 1-methylcyclobutyl carbamate, 1-methylcyclohexyl carbamate, 1-methyl-l-cyclopropylmethyl carbamate, 1 -methyl- 1 -(3,5- dimethoxyphenyl)ethyl carbamate, l-methyl-l-(p-phenylazophenyl)ethyl carbamate, 1- methyl-1-phenylethyl carbamate, l-methyl-l-(4-pyridyl)ethyl carbamate, phenyl carbamate, ^-(phenylazo)benzyl carbamate, 2,4,6-tri-?-butylphenyl carbamate, A- (trimethylammonium)benzyl carbamate, 2,4,6-trimethylbenzyl carbamate, formamide, acetamide, chloroacetamide, trichloroacetamide, trifluoroacetamide, phenylacetamide, 3- phenylpropanamide, picolinamide, 3-pyridylcarboxamide, N-benzoylphenylalanyl derivative, benzamide, p-phenylbenzamide, o-nitophenylacetamide, o- nitrophenoxyacetamide, acetoacetamide, (N'-dithiobenzyloxycarbonylamino)acetamide, 3- (p-hydroxyphenyl)propanamide, 3-(o-nitrophenyl)propanamide, 2-methyl-2-(o- nitrophenoxy)propanamide, 2-methyl-2-(o-phenylazophenoxy)propanamide, A- chlorobutanamide, 3-methyl-3-nitrobutanamide, o-nitrocinnamide, N-acetylmethionine derivative, o-nitrobenzamide, o-(benzoyloxymethyl)benzamide, 4,5-diphenyl-3-oxazolin- 2-one, N-phthalimide, N-dithiasuccinimide (Dts), N-2,3-diphenylmaleimide, N-2,5- dimethylpyrrole, N-l,l,4,4-tetramethyldisilylazacyclopentane adduct (STABASE), 5- substituted l^-dimethyl-l^S-triazacyclohexan^-one, 5-substituted l,3-dibenzyl-l,3,5- triazacyclohexan-2-one, 1-substituted 3,5-dinitro-4-pyridone, N-methylamine, N- allylamine, N-[2-(trimethylsilyl)ethoxy]methylamine (SEM), N-3-acetoxypropylamine, N- (l-isopropyl^-nitro-2-oxo-3-pyroolin-3-yl)amine, quaternary ammonium salts, N- benzylamine, N-di(4-methoxyphenyl)methylamine, N-5-dibenzosuberylamine, N- triphenylmethylamine (Tr), N-[(4-methoxyphenyl)diphenylmethyl] amine (MMTr), N-9- phenylfluorenylamine (PhF), N-2,7-dichloro-9-fluorenylmethyleneamine, N- ferrocenylmethylamino (Fcm), N-2-picolylamino N'-oxide, N-1, 1- dimethylthiomethyleneamine, N-benzylideneamine, N-p-methoxybenzylideneamine, N- diphenylmethyleneamine, N-[(2-pyridyl)mesityl]methyleneamine, N-(N ',N'- dimethylaminomethylene)amine, NN'-isopropylidenediamine, N-p-nitrobenzylideneamine, N-salicylideneamine, N-5-chlorosalicylideneamine, N-(5-chloro-2- hydroxyphenyl)phenylmethyleneamine, N-cyclohexylideneamine, N-(5,5-dimethyl-3-oxo- l-cyclohexenyl)amine, N-borane derivative, N-diphenylborinic acid derivative, N- [phenyl(pentacarbonylchromium- or tungsten)carbonyl] amine, N-copper chelate, N-zinc chelate, N-nitroamine, N-nitrosoamine, amine N-oxide, diphenylphosphinamide (Dpp), dimethylthiophosphinamide (Mpt), diphenylthiophosphinamide (Ppt), dialkyl phosphoramidates, dibenzyl phosphoramidate, diphenyl phosphoramidate, benzenesulfenamide, o-nitrobenzenesulfenamide (Νps), 2,4-dinitrobenzenesulfenamide, pentachlorobenzenesulfenamide, 2-nitro^-methoxybenzenesulfenamide, triphenylmethylsulfenamide, 3-nitropyridinesulfenamide (Νpys), p-toluenesulfonamide (Ts), benzenesulfonamide, 2,3,6,-trimethyl^-methoxybenzenesulfonamide (Mtr), 2,4,6- trimethoxybenzenesulfonamide (Mtb), 2,6-dimethyl^-methoxybenzenesulfonamide (Pme), 2,3 ,5,6-tetramethyl^-methoxybenzenesulfonamide (Mte), 4-methoxybenzenesulfonamide (Mbs), 2,4,6-trimethylbenzenesulfonamide (Mts), 2,6-dimethoxy-4- methylbenzenesulfonamide (iMds), 2,2,5, 7,8-pentamethylchroman-6-sulfonamide (Pmc), methanesulfonamide (Ms), β-trimethylsilylethanesulfonamide (SES), 9- anthracenesulfonamide, 4-(4',8'-dimethoxynaphthylmethyl)benzenesulfonamide (DΝMBS), benzylsulfonamide, trifluoromethylsulfonamide, and phenacylsulfonamide. [0051] A "hydroxyl protecting group" as used herein, is well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference. Suitable hydroxyl protecting groups include methyl, methoxylmethyl (MOM), methylthiomethyl (MTM), ϊ-butylthiomethyl,
(phenyldimethylsilyl)methoxymethyl (SMOM), benzyloxymethyl (BOM), p— methoxybenzyloxymethyl (PMBM), (4-methoxyphenoxy)methyl (p-AOM), guaiacolmethyl (GUM), ϊ-butoxymethyl, 4-pentenyloxymethyl (POM), siloxymethyl, 2- methoxyethoxymethyl (MEM), 2,2,2-trichloroethoxymethyl, bis(2-chloroethoxy)methyl, 2- (trimethylsilyl)ethoxymethyl (SEMOR), tetrahydropyranyl (THP), 3- bromotetrahydropyranyl, tetrahydrothiopyranyl, 1-methoxycyclohexyl, 4- methoxytetrahydropyranyl (MTHP), 4-methoxytetrahydrothiopyranyl, 4- methoxytetrahydrothiopyranyl S,S-dioxide, l-[(2-chloro-4-methyl)phenyl]-4- methoxypiperidin-4-yl (CTMP), 1 ,4-dioxan-2-yl, tetrahydrofuranyl, tetrahydrothiofuranyl, 2,3,3a,4,5,6,7,7a-octahydro-7,8,8-trimethyl-4,7-methanobenzofuran-2-yl, 1-ethoxy ethyl, l-(2-chloroethoxy)ethyl, 1 -methyl- 1-methoxy ethyl, 1-methyl-l-benzyloxyethyl, 1- methyl-l-benzyloxy-2-fluoroethyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 2- (phenylselenyl)ethyl, ?-butyl, allyl, />-chlorophenyl, />-methoxyphenyl, 2,4-dinitrophenyl, benzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, o-nitrobenzyl, p-nitrobenzyl, p- halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl, p-phenylbenzyl, 2-picolyl, 4-picolyl, 3- methyl-2-picolyl N-oxido, diphenylmethyl, p,p -dinitrobenzhydryl, 5-dibenzosuberyl, triphenylmethyl, α-naphthyldiphenylmethyl, /»-methoxyphenyldiphenylmethyl, di(p- methoxyphenyl)phenylmethyl, tri(p-methoxyphenyl)methyl, 4-(4'- bromophenacyloxyphenyl)diphenylmethyl, 4,4' ,4' '-tris(4,5- dichlorophthalimidophenyl)methyl, 4,4',4"-tris(levulinoyloxyphenyl)methyl, 4,4',4"- tris(benzoyloxyphenyl)methyl, 3-(imidazol-l-yl)bis(4',4"-dimethoxyphenyl)methyl, 1,1- bis(4-methoxyphenyl)-l'-pyrenylmethyl, 9-anthryl, 9-(9-phenyl)xanthenyl, 9-(9-phenyl- 10-oxo)anthryl, l,3-benzodithiolan-2-yl, benzisothiazolyl S,S-dioxido, trimethylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), dimethylisopropylsilyl (IPDMS), diethylisopropylsilyl (DEIPS), dimethylthexylsilyl, ϊ-butyldimethylsilyl (TBDMS), t- butyldiphenylsilyl (TBDPS), tribenzylsilyl, tri— /?— xy Iy Is ily 1, triphenylsilyl, diphenylmethylsilyl (DPMS), ϊ-butylmethoxyphenylsilyl (TBMPS), formate, benzoylformate, acetate, chloroacetate, dichloroacetate, trichloroacetate, trifluoroacetate, methoxyacetate, triphenylmethoxyacetate, phenoxyacetate, />-chlorophenoxyacetate, 3- phenylpropionate, 4-oxopentanoate (levulinate), 4,4-(ethylenedithio)pentanoate (levulinoyldithioacetal), pivaloate, adamantoate, crotonate, 4-methoxycrotonate, benzoate, p— phenylbenzoate, 2,4,6-trimethylbenzoate (mesitoate), alkyl methyl carbonate, 9- fluorenylmethyl carbonate (Fmoc), alkyl ethyl carbonate, alkyl 2,2,2-trichloroethyl carbonate (Troc), 2-(trimethylsilyl)ethyl carbonate (TMSEC), 2-(phenylsulfonyl) ethyl carbonate (Psec), 2-(triphenylphosphonio) ethyl carbonate (Peoc), alkyl isobutyl carbonate, alkyl vinyl carbonate alkyl allyl carbonate, alkyl p-nitrophenyl carbonate, alkyl benzyl carbonate, alkyl />-methoxybenzyl carbonate, alkyl 3,4-dimethoxybenzyl carbonate, alkyl o-nitrobenzyl carbonate, alkyl p-nitrobenzyl carbonate, alkyl S-benzyl thiocarbonate, 4-ethoxy-l- napththyl carbonate, methyl dithiocarbonate, 2-iodobenzoate, 4-azidobutyrate, 4-nitro^- methylpentanoate, o-(dibromomethyl)benzoate, 2-formylbenzenesulfonate, 2- (methylthiomethoxy)ethyl, 4-(methylthiomethoxy)butyrate, 2-
(methylthiomethoxymethyl)benzoate, 2,6-dichloro-4-methylphenoxyacetate, 2,6-dichloro- 4-( 1 , 1 ,3 ,3-tetramethylbutyl)phenoxyacetate, 2,4-bis( 1 , l-dimethylpropyl)phenoxyacetate, chlorodiphenylacetate, isobutyrate, monosuccinoate, (is)-2-methyl-2-butenoate, o— (methoxycarbonyl)benzoate, α-naphthoate, nitrate, alkyl N,N,N',N'- tetramethylphosphorodiamidate, alkyl N-phenylcarbamate, borate, dimethylphosphinothioyl, alkyl 2,4-dinitrophenylsulfenate, sulfate, methanesulfonate (mesylate), benzylsulfonate, and tosylate (Ts). For protecting 1,2- or 1,3— diols, the protecting groups include methylene acetal, ethylidene acetal, l-?-butylethylidene ketal, 1-phenylethylidene ketal, (4- methoxyphenyl)ethylidene acetal, 2,2,2-trichloroethylidene acetal, acetonide, cyclopentylidene ketal, cyclohexylidene ketal, cycloheptylidene ketal, benzylidene acetal, p— methoxybenzylidene acetal, 2,4-dimethoxybenzylidene ketal, 3,4-dimethoxybenzylidene acetal, 2-nitrobenzylidene acetal, methoxymethylene acetal, ethoxymethylene acetal, dimethoxy methylene ortho ester, 1-methoxy ethylidene ortho ester, 1-ethoxyethylidine ortho ester, 1,2-dimethoxy ethylidene ortho ester, α-methoxybenzylidene ortho ester, 1-(N, N- dimethylamino)ethylidene derivative, α-(N,N'-dimethylamino)benzylidene derivative, 2- oxacyclopentylidene ortho ester, di-ϊ-butylsilylene group (DTBS), 1,3— (1, 1,3,3— tetraisopropyldisiloxanylidene) derivative (TIPDS), tetra-?-butoxydisiloxane-l,3-diylidene derivative (TBDS), cyclic carbonates, cyclic boronates, ethyl boronate, and phenyl boronate. [0052] A "thiol protecting group," as used herein, are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, the entirety of which is incorporated herein by reference. Examples of suitably protected thiol groups further include, but are not limited to, thioesters, carbonates, sulfonates allyl thioethers, thioethers, silyl thioethers, alkyl thioethers, arylalkyl thioethers, and alkyloxyalkyl thioethers. Examples of suitable ester groups include formates, acetates, proprionates, pentanoates, crotonates, and benzoates. Specific examples of suitable ester groups include formate, benzoyl formate, chloroacetate, trifluoroacetate, methoxyacetate, triphenylmethoxyacetate, p- chlorophenoxyacetate, 3-phenylpropionate, 4-oxopentanoate, 4,4-
(ethylenedithio)pentanoate, pivaloate (trimethylacetate), crotonate, 4-methoxy-crotonate, benzoate, p-benylbenzoate, 2,4,6-trimethylbenzoate. Examples of suitable carbonates include 9-fluorenylmethyl, ethyl, 2,2,2-trichloroethyl, 2-(trimethylsilyl)ethyl, 2- (phenylsulfonyl)ethyl, vinyl, allyl, and p-nitrobenzyl carbonate. Examples of suitable silyl groups include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl ether, and other trialkylsilyl ethers. Examples of suitable alkyl groups include methyl, benzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, trityl, t-butyl, and allyl ether, or derivatives thereof. Examples of suitable arylalkyl groups include benzyl, p- methoxybenzyl (MPM), 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl, 2- and 4-picolyl ethers.
[0053] A "carboxylic acid protecting group," as used herein, are well known in the art and include those described in detail in Greene (1999). Examples of suitably protected carboxylic acids further include, but are not limited to, silyl-, alkyl-, alkenyl-, aryl-, and arylalkyl-protected carboxylic acids. Examples of suitable silyl groups include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl, and the like. Examples of suitable alkyl groups include methyl, benzyl, p-methoxybenzyl, 3,4- dimethoxybenzyl, trityl, t-butyl, tetrahydropyran-2-yl. Examples of suitable alkenyl groups include allyl. Examples of suitable aryl groups include optionally substituted phenyl, biphenyl, or naphthyl. Examples of suitable arylalkyl groups include optionally substituted benzyl (e.g., p-methoxybenzyl (MPM), 3,4-dimethoxybenzyl, O-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-cyanobenzyl), and 2- and 4-picolyl. [0054] As used herein, a "pharmaceutically acceptable form thereof includes any pharmaceutically acceptable salts, prodrugs, tautomers, isomers, and/or polymorphs of a compound of the present invention, as defined below and herein.
[0055] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1911 , 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C i_4alky I)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0056] As used herein, the term "prodrug" refers to a derivative of a parent compound that requires transformation within the body in order to release the parent compound. In certain cases, a prodrug has improved physical and/or delivery properties over the parent compound. Prodrugs are typically designed to enhance pharmaceutically and/or pharmacokinetically based properties associated with the parent compound. The advantage of a prodrug can lie in its physical properties, such as enhanced water solubility for parenteral administration at physiological pH compared to the parent compound, or it enhances absorption from the digestive tract, or it may enhance drug stability for long-term storage. In recent years several types of bioreversible derivatives have been exploited for utilization in designing prodrugs. Using esters as a prodrug type for compounds containing a carboxyl or hydroxyl functionality is known in the art as described, for example, in "The Organic Chemistry of Drug Design and Drug Interaction" Richard Silverman, published by Academic Press (1992). [0057] As used herein, the term "tautomer" includes two or more interconvertable compounds resulting from at least one formal migration of a hydrogen atom and at least one change in valency (e.g., a single bond to a double bond, a triple bond to a single bond, or vice versa). The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Tautomerizations (i.e., the reaction providing a tautomeric pair) may catalyzed by acid or base. Exemplary tautomerizations include keto-to-enol; amide-to-imide; lactam-to-lactim; enamine-to-imine; and enamine-to-(a different) enamine tautomerizations.
[0058] As used herein, the term "isomers" includes any and all geometric isomers and stereoisomers. For example, "isomers" include cis— and trans-isomsrs, E- and Z- isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. For instance, an isomer/enantiomer may, in some embodiments, be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched." "Optically- enriched," as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer. In certain embodiments the compound of the present invention is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer. Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses. See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen, S.H., et al., Tetrahedron 33:2725 (1977); Eliel, EX. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S. H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972).
[0059] As used herein, "polymorph" refers to a crystalline inventive compound existing in more than one crystalline form/structure. When polymorphism exists as a result of difference in crystal packing it is called packing polymorphism. Polymorphism can also result from the existence of different conformers of the same molecule in conformational polymorphism. In pseudopolymorphism the different crystal types are the result of hydration or solvation. Brief Description of the Figures
[0060] Figure 1. Sensorgram depicting binding of Compound (1-1) to immobilized
Aurora A kinase protein.
[0061] Figures 2A-2C. Figure 2A. Compounds (1-1) to (1-7) at 5 μM were tested for binding to Aurora A kinase by surface plasmon resonance. Compound (1-1) was further characterized (n = 4) by measuring binding in a dilution series (391 nM to 25 μM) and calculating affinity by (Figure 2B) kinetic and (Figure 2C) equilibrium analyses. Kinetic kon = 1.2 x 1(T4 M 1SeC"1, koff = 6.8 x 1(T2 sec"1, KD = 5.3 ± 1 μM; Equilibrium KD = 6.0 ± 1 μM. [0062] Figure 3. Diversity-oriented synthesis (DOS): chemical diversity and complexity in a limited number of steps (Kumagai, N. et al. Angew. Chem. Int. Ed. (2006) 45:3635).
[0063] Figure 4. Sensograms for compounds (1-1), (1-9) to (1-10), (1-12) to (1-20), (I-
24) and (1-34).
[0064] Figure 5. Sensograms for compounds (1-1), (1-8), (1-11), (1-21), (1-25), (I-
27), (1-29) to (1-35), (II-l) and (11-35).
[0065] Figure 6. Table of exemplary fluorochromes useful for flow cytometry or fluorescence microscopy provided, within groups, roughly in order of excitation wavelength. MW: molecular weight; Ex: Peak excitation wavelength (nm); Em: Peak emission wavelength (nm).
[0066] Figures 7A-7C. HeLa (cervical cancer), compound (1-33) (FITC-labeled compound; 4 μM). Similar images have been obtained with U2OS and MCF7 cells. Figure 7 A: image of fluorescent tagged compound (1-33) bound to a centrosome (red color); Figure IB: image of cell sample; Figure 1C: a merge of the two images. The centrosome (i.e., two centrioles) within the cell are easily visualized (red color). [0067] Figure 8. Early detection of cervical cancer by Pap-smear.
[0068] Figure 9. Numerical centrosome aberrations in normal cervical epithelium and various cervical lesions (Skyldberg et al., Modern Pathology (2001) 14:279-284). Expression of HPV E7 protein uncouples centrosome and chromosome duplication. Supernumerary centrosomes are a biomarker for HPV E7 oncogene expression. [0069] Figure 10. Images of cells undergoing normal and abnormal mitosis. Detailed Description of Certain Embodiments of the Invention
[0070] The present invention provides inventive compounds, pharmaceutical compositions thereof, and use of such compounds. Compounds of the present invention are useful as anti-proliferative agents and diagnostic agents for the detection of proliferative (e.g., cancerous) and pre-cancerous cells. [0071] In one aspect, the present invention provides compounds of the formulae:
Figure imgf000026_0001
or pharmaceutically acceptable forms thereof; wherein:
Ri is hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORA; -CN; -SCN; -SRA; or -N(RA)2; wherein each occurrence of RA is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety;
R2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
R3 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)RB; -CC^RB; -SORB; or -SO2RB; wherein each occurrence of RB is independently a halogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety;
R4 is hydrogen; a hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
R5 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)Rc; -CO2Rc; -SORc; or -SO2Rc; wherein each occurrence of Rc is independently a hydrogen, a halogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety; n is 0, 1, 2, 3 or 4; and each occurrence of Re is independently hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORD; -C(=O)RD; -CO2RD; -CN; -SCN; -SRD; -NO2; -N(RD)2; or -NHC(O)R0; wherein each occurrence of RF is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety. [0072] In certain embodiments, Ri is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORA; -CN; -SCN; -SRA; - N(RA)2; or -C(RA)3. In certain embodiments, Ri is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; -ORA; -SRA; -N(RA)2; or -C(RA)3. In certain embodiments, Ri is -ORA;-SRA; or -N(RA)2.
[0073] In certain embodiments, Ri is -N(RA)2, wherein each occurrence of RA is independently a hydrogen, an amino protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety. In certain embodiments, RA is a substituted or unsubstituted aliphatic moiety. In certain embodiments, RA is a substituted or unsubstituted heteroaliphatic moiety. In certain embodiments, Ri is - NHRA. In certain embodiments, Ri is -NH2.
[0074] In certain embodiments, Ri is -N(RA)2, wherein each occurrence of RA is independently a hydrogen or a substituted or unsubstituted heteroaliphatic moiety. In certain embodiments, Ri corresponds to any one of the following groups:
Figure imgf000028_0001
wherein RG is hydrogen, a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety; a fluorescent label; or a resin. [0075] In certain embodiments, Ri corresponds to group:
Figure imgf000028_0002
[0076] In certain embodiments, Ri corresponds to any of the following groups:
Figure imgf000029_0001
wherein ^^ is a resin.
[0077] In certain embodiments, Ri is -N(RA)2, wherein each occurrence of RA is independently a hydrogen or a substituted or unsubstituted aliphatic moiety. In certain embodiments, Ri corresponds to any one of the following groups:
A ^N-(CH2)q-CH3 >^NV
N-(CH2)q-CH3 -O-R H (CH2)q-CH3
Figure imgf000029_0002
wherein R0 is a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety; a fluorescent label; or a resin; and each instance of q is independently 0, 1, 2, or 3. [0078] In certain embodiments, Ri corresponds to the groups: t ^N-(CH2)q-CH3
^N-(CH2)q-CH3 I _
H H or <CH2)q CH3 wherein each instance of q is independently 0, 1, 2, or 3
[0079] In certain embodiments, Ri is -ORA, wherein each occurrence of RA is independently a hydrogen, a hydroxyl protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety. In certain embodiments, Ri is -OH. In certain embodiments, Ri is -ORA, wherein RA is a substituted or unsubstituted aliphatic moiety or a substituted or unsubstituted heteroaliphatic moiety.
[0080] In certain embodiments, Ri is -ORA, wherein RA is a substituted or unsubstituted aliphatic moiety. In certain embodiments, RA is a substituted or unsubstitued methyl, ethyl, n-propyl, isopropyl, t-butyl, sec-butyl, or iso-butyl group. In certain embodiments, Ri is -OCH3.
[0081] In certain embodiments, R2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl. In certain embodiments, R2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl. In certain embodiments, R2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic or cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic. In certain embodiments, R2 is cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic. [0082] In certain embodiments, R2 corresponds to a -(CH2)mRH group, wherein RH is hydrogen, or a substituted or unsubstituted aryl moeity, a substituted or unsubstituted heteroaryl moeity, a substituted or unsubstituted cycloalkyl moeity, a substituted or unsubstituted cycloheteroalkyl moeity, a substituted or unsubstituted alkyloxy moeity, or - CF3; and m is 0, 1, 2, 3, 4, 5 or 6. In certain embodiments, RH corresponds to a substituted or unsubstituted aryl moeity or a substituted or unsubstituted heteroaryl moeity. [0083] In certain embodiments, m is 1 or 2.
[0084] In certain embodiments, R2 corresponds to any one of the following groups:
Figure imgf000030_0001
[0085] In certain embodiments, R3 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)RB; -CO2RB; -SORB; or -SO2RB.
[0086] In certain embodiments, R3 is substituted or unsubstituted, branched or unbranched acyl. In certain embodiments, R3 is hydrogen.
[0087] In certain embodiments, R3 is -C(=O)RB; -CO2RB; -SORB; or -SO2RB- In certain embodiments, R3 is -C(=O)RB or -CO2RB- In certain embodiments, R3 is -C(=O)RB-
[0088] In certain embodiments, R3 is -C(=O)RB, wherein RB is independently a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
[0089] In certain embodiments, R3 corresponds to any one of the following groups:
Figure imgf000031_0001
[0090] In certain embodiments, R4 is hydrogen; hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl. In certain embodiments, R4 is hydrogen, a hydroxyl protecting group, cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic, or substituted or unsubstituted, branched or unbranched acyl. In certain embodiments, R4 is hydrogen, a hydroxyl protecting group or C1-S alkyl. In certain embodiments, R4 is hydrogen. [0091] In certain embodiments, each occurrence of Re is independently hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; -
C(=O)RD; -CO2RD; -ORD; -SRD; -N(RD)2; or -NHC(O)R0. However, in certain embodiments, Re is not halogen.
[0092] In certain embodiments, each occurrence of Re is independently hydrogen; substituted or unsubstituted aliphatic; substituted or unsubstituted heteroaliphatic; -C(=O)RD;
-CO2RD or -ORD. In certain embodiments, each occurrence of Re is independently hydrogen; alkyloxy, hydroxyalkyl, or -C(=O)RD. In certain embodiments, each occurrence
Of R6 is independently hydrogen, -C(=O)H, -(CH2)POH or -(CH2)pOPGi wherein p is 0, 1, 2 or 3, and PGi is a hydroxyl protecting group. In certain embodiments, PGi is a silyl protecting group. Exemplary silyl protecting groups include trimethylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), and tertbutyldimethylsilyl (TBDMS).
[0093] In certain embodiments, n is 0, 1 or 2. In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2.
[0094] In certain embodiments, n is 0.
[0095] In certain embodiments, n is 1, and Re is substituted ortho to the phenolic oxygen. In certain embodiments, n is 1, and Re is substituted para to the phenolic oxygen.
[0096] In certain embodiments, n is 2, and Re is substituted ortho and para to the phenolic oxygen.
[0097] In certain embodiments, Rs is hydrogen; substituted or unsubstituted aryl; or substituted or unsubstituted heteroaryl. In certain embodiments, Rs is substituted or unsubstituted aryl. In certain embodiments, Rs corresponds to the groups:
Figure imgf000032_0001
wherein n and Re are as defined above and herein, and X is halogen or -S(O)2Ri, wherein Ri is a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety. In certain embodiments, X is -I, -Cl, or -F. In certain embodiments, X is -I. [0098] In certain embodiments, Rs corresponds to any of the following groups:
Figure imgf000033_0001
wherein R6 and X are as defined above and herein. [0099] In certain embodiments, R5 corresponds to the groups:
Figure imgf000033_0002
wherein R6 and X are as defined above and herein. [00100] In certain embodiments, R5 corresponds to any one of the following groups:
Figure imgf000033_0003
wherein PGi is as defined above and herein.
Compounds of Formula (I)
[00101] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to any one of the following stereoisomers of formula
(I):
Figure imgf000033_0004
Figure imgf000034_0001
(I-e) (I-f) wherein R1, R2, R3, R4 and R5 are as defined above and herein.
[00102] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to any one of the following stereoisomers of formula
(I):
Figure imgf000034_0002
(I-a) (I-b) wherein R1, R2, R3, R4 and R5 are as defined above and herein.
[00103] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to the formula (I-a):
Figure imgf000034_0003
(I-a) wherein R1, R2, R3, R4 and R5 are as defined above and herein.
[00104] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to the formulae:
Figure imgf000035_0001
(I-g) (I-h) wherein Ri, R2, R3, R4, R5, Re, X and n are as defined above and herein. [00105] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to any one of the following stereoisomers of formulae (I-g) or (I-h):
Figure imgf000035_0002
(I-k) (I-m)
Figure imgf000035_0003
(I-n) (I-o)
Figure imgf000035_0004
(I-P) (I-q)
Figure imgf000036_0001
(I-t) (I-u) wherein Ri, R2, R3, R4, R5, Re, X and n are as defined above and herein. [00106] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to any one of the following stereoisomers of formulae (I-g) or (I-h):
Figure imgf000036_0002
(I-P) (I-q) wherein Ri, R2, R3, R4, R5, Re, X and n are as defined above and herein. [00107] In certain embodiments, compounds of formula (I), or any pharmaceutically acceptable forms thereof, correspond to the formula (I-i):
Figure imgf000036_0003
(I-i) wherein Ri, R2, R3, R4, R5, Re, X and n are as defined above and herein. [00108] In certain embodiments, the present invention provides compounds of formula
(I), or any subsets thereof, wherein at least one of R2 and R3 is hydrogen. In other embodiments, the present invention provides compounds of formula (I), or any subsets thereof, wherein neither of R2 and R3 is hydrogen.
[00109] In certain embodiments, compounds of formula (I), or pharmaceutically acceptable forms thereof, correspond to the formula (I-v):
Figure imgf000037_0001
(I-V) wherein R2, R3, Re, X and n are defined above and herein.
[00110] In certain embodiments, compounds of formula (I), or pharmaceutically acceptable forms thereof, are any one of the following compounds:
Figure imgf000037_0002
(1-4) (1-5) (1-6)
Figure imgf000038_0001
(1-7) (1-8) (1-9)
Figure imgf000038_0002
(1-10) (1-11) (1-12)
Figure imgf000038_0003
(1-13) (1-14) (1-15)
Figure imgf000038_0004
(1-16) (1-17) (1-18)
Figure imgf000039_0001
(1-19) (1-20) (1-21)
Figure imgf000039_0002
(1-22) (1-23) (1-24)
Figure imgf000039_0003
(1-25) (1-26) (1-27)
Figure imgf000039_0004
(1-28) (1-29) (1-30)
Figure imgf000039_0005
(1-31) (1-32)
Figure imgf000040_0001
(1-33)
Figure imgf000040_0002
(1-36) (1-37)
Figure imgf000040_0003
(1-38)
Figure imgf000041_0001
(1-40)
Figure imgf000041_0002
(1-41)
Figure imgf000041_0003
(1-42)
39
Figure imgf000042_0001
(1-43)
Figure imgf000042_0002
(1-44)
Figure imgf000042_0003
(1-45)
Compounds of Formula (II)
[00111] In certain embodiments, compounds of formula (II), or pharmaceutically acceptable forms thereof, correspond any one of the following stereoisomers of formula (II):
Figure imgf000043_0001
(II-e) (Il-f) wherein Ri, R2, R3, R4, Re and n are as defined above and herein.
[00112] In certain embodiments, compounds of formula (II), or pharmaceutically acceptable forms thereof, correspond any one of the following stereoisomers of formula (II):
Figure imgf000043_0002
(II-a) (II-b) wherein Ri, R2, R3, R4, Re and n are as defined above and herein. [00113] In certain embodiments, compounds of formula (II), or pharmaceutically acceptable forms thereof, correspond to the formula (II-a):
Figure imgf000044_0001
(II-a) wherein Ri, R2, R3, R4, Re and n are as defined above and herein.
[00114] In certain embodiments, compounds of formula (II), or pharmaceutically acceptable forms thereof, correspond to the formula (II-g):
Figure imgf000044_0002
(II-g) wherein R2, R3, Re and n are as defined above and herein.
[00115] In certain embodiments, compounds of formula (II), or pharmaceutically acceptable forms thereof, are any one of the following compounds:
Figure imgf000044_0003
(ii-i) (II-2) (II-3)
Figure imgf000045_0001
(II-4) (II-5) (II-6)
Figure imgf000045_0002
(II-7) (II-8) (II-9)
Figure imgf000045_0003
(11-10) (11-11) (11-12)
Figure imgf000045_0004
Figure imgf000046_0001
(11-22) (11-24) (11-25)
Figure imgf000046_0002
(11-32) (11-35) (11-37)
Figure imgf000047_0001
(11-33)
Figure imgf000047_0002
(11-38)
Figure imgf000047_0003
(11-39)
Figure imgf000048_0001
(11-40)
Figure imgf000048_0002
(11-41)
Figure imgf000048_0003
(11-42)
Figure imgf000048_0004
(11-43)
Figure imgf000049_0001
(11-44)
Figure imgf000049_0002
(11-45)
Pharmaceutical Compositions and Formulations
[00116] In another apsect, the present invention also provides a pharmaceutical composition comprising a compound, or pharmaceutically acceptable form thereof, of the formulae (I) or (II), or any subsets thereof, and a pharmaceutically acceptable excipient. [00117] For the purposes of the present invention, the phrase "active ingredient" generally refers to an inventive compound, as described above and herein. [00118] The formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with a carrier and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
[00119] A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
[00120] The relative amounts of the active ingredient, the pharmaceutically acceptable carrier, and/or any additional ingredients in a pharmaceutical composition of the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient. [00121] Exemplary pharmaceutically acceptable excipients include any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form. Remington's The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, (Lippincott, Williams & Wilkins, Baltimore, MD, 2006) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.
[00122] In some embodiments, the pharmaceutically acceptable excipient is at least
95%, 96%, 97%, 98%, 99%, or 100% pure. In some embodiments, the excipient is approved for use in humans and for veterinary use. In some embodiments, the excipient is approved by United States Food and Drug Administration. In some embodiments, the excipient is pharmaceutical grade. In some embodiments, the excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
[00123] Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in the inventive formulations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and perfuming agents can be present in the composition, according to the judgment of the formulator. [00124] Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and combinations thereof
[00125] Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation- exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked polyvinylpyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and combinations thereof. [00126] Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate [Tween 20], polyoxyethylene sorbitan [Tween 60], polyoxyethylene sorbitan monooleate [Tween 80], sorbitan monopalmitate [Span 40], sorbitan monostearate [Span 60], sorbitan tristearate [Span 65], glyceryl monooleate, sorbitan monooleate [Span 80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [Myrj 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. Cremophor), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij 30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic F 68, Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.
[00127] Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof. [00128] Exemplary preservatives may include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and other preservatives. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and sorbic acid. Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus, Phenonip, methylparaben, Germall 115, Germaben II, Neolone, Kathon, and Euxyl. In certain embodiments, the preservative is an anti-oxidant. In other embodiments, the preservative is a chelating agent.
[00129] Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl alcohol, etc., and combinations thereof.
[00130] Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof. [00131] Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and combinations thereof. [00132] Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredients, the liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. In certain embodiments for parenteral administration, the conjugates of the invention are mixed with solubilizing agents such as Cremophor, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and combinations thereof.
[00133] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U. S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[00134] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00135] In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. [00136] Compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing the conjugates of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
[00137] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.
[00138] Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[00139] The active ingredients can be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active ingredient may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may comprise buffering agents. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
[00140] Dosage forms for topical and/or transdermal administration of a compound of this invention may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches. Generally, the active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and/or any needed preservatives and/or buffers as may be required. Additionally, the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of an active ingredient to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the active ingredient in the proper medium. Alternatively or additionally, the rate may be controlled by either providing a rate controlling membrane and/or by dispersing the active ingredient in a polymer matrix and/or gel. [00141] Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Patents 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662. Intradermal compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof. Jet injection devices which deliver liquid vaccines to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Patents 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189; 5,704,911; 5,383,851; 5,893,397; 5,466,220; 5,339,163; 5,312,335; 5,503,627; 5,064,413; 5,520,639; 4,596,556; 4,790,824; 4,941,880; 4,940,460; and PCT publications WO 97/37705 and WO 97/13537. Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable. Alternatively or additionally, conventional syringes may be used in the classical mantoux method of intradermal administration. [00142] Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions. Topically-administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of the active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
[00143] A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 to about 7 nanometers or from about 1 to about 6 nanometers. Such compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
[00144] Low boiling propellants generally include liquid propellants having a boiling point of below 65 0F at atmospheric pressure. Generally the propellant may constitute 50 to 99.9% (w/w) of the composition, and the active ingredient may constitute 0.1 to 20% (w/w) of the composition. The propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient). [00145] Pharmaceutical compositions of the invention formulated for pulmonary delivery may provide the active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate. The droplets provided by this route of administration may have an average diameter in the range from about 0.1 to about 200 nanometers.
[00146] The formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition of the invention. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nares. [00147] Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, 0.1 to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising the active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 to about 200 nanometers, and may further comprise one or more of the additional ingredients described herein. [00148] A pharmaceutical composition of the invention may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid carrier. Such drops may further comprise buffering agents, salts, and/or one or more other of the additional ingredients described herein. Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this invention. [00149] General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005.
[0001] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.
Kits
[00150] Still further encompassed by the invention are kits comprising one or more inventive compounds (or pharmaceutically acceptable forms thereof), and/or an inventive pharmaceutical composition. Kits are typically provided in a suitable container (e.g., for example, a foil, plastic, or cardboard package). In certain embodiments, an inventive kit may include one or more pharmaceutical excipients, pharmaceutical additives, therapeutically active agents, and the like, as described herein. In certain embodiments, an inventive kit may include means for proper administration, such as, for example, graduated cups, syringes, needles, cleaning aids, and the like. In certain embodiments, an inventive kit may include instructions for proper administration and/or preparation for proper administration.
Methods and Uses
[00151] In another apsect, the present invention provides a method of treating a proliferative disease, disorder or condition comprising the step of adminiserting a therapeutically effective amount of a compound of formulae I or II, or a pharmaceutically acceptable form thereof, to a subject in need thereof.
[00152] Subjects to which administration is contemplated include, but are not limited to, humans (e.g., male, female, infant, child, adolescant, adult, elderly, etc.) and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, and/or turkeys. "Treating," as used herein, refers to partially or completely inhibiting or reducing the proliferative disease, disorder or condition from which the subject is suffering. "Therapeutically effective amount," as used herein, refers to the minimal amount or concentration of an inventive compound or inventive pharmaceutical composition that, when administered, is sufficient in treating the subject. [00153] As used herein a proliferative disease, condition, or disorder includes, but is not limited to, cancer, hematopoietic neoplastic disorders, proliferative breast disease, proliferative disorders of the lung, proliferative disorders of the colon, proliferative disorders of the liver, and proliferative disorders of the ovary.
[00154] Examples of cancers treatable by the above method include carcinoma, sarcoma, or metastatic disorders, breast cancer, ovarian cancer, colon cancer, lung cancer, gastric cancer, esophageal cancer, rectal cancer, pancreatic cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, blood cancer, bone cancer, stomach cancer, liver cancer, kidney cancer, skin cancer, brain cancer, fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or Kaposi sarcoma.
[00155] Examples of hematopoietic neoplastic disorders treatable by the above method includes diseases involving hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof. In certain embodiments, the diseases arise from poorly differentiated acute leukemias, e.g., erythroblastic leukemia and acute megakaryoblastic leukemia. Additional exemplary myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11:267-97); lymphoid malignancies include, but are not limited to acute lymphoblastic leukemia (ALL) which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM). Additional forms of malignant lymphomas include, but are not limited to non-Hodgkin lymphoma and variants thereof, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Stemberg disease.
[00156] Examples of proliferative breast disease treatable by the above method includes epithelial hyperplasia, sclerosing adenosis, and small duct papillomas; tumors, e.g., stromal tumors such as fibroadenoma, phyllodes tumor, and sarcomas, and epithelial tumors such as large duct papilloma; carcinoma of the breast including in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ, and invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, medullary carcinoma, colloid (mucinous) carcinoma, tubular carcinoma, and invasive papillary carcinoma, and miscellaneous malignant neoplasms. Disorders in the male breast include, but are not limited to, gynecomastia and carcinoma.
[00157] Examples of proliferative disorders of the lung treatable by the above method include, but are not limited to, bronchogenic carcinoma, including paraneoplastic syndromes, bronchioloalveolar carcinoma, neuroendocrine tumors, such as bronchial carcinoid, miscellaneous tumors, and metastatic tumors; pathologies of the pleura, including inflammatory pleural effusions, noninflammatory pleural effusions, pneumothorax, and pleural tumors, including solitary fibrous tumors (pleural fibroma) and malignant mesothelioma.
[00158] Examples of proliferative disorders of the colon treatable by the above method include, but are not limited to, non-neoplastic polyps, adenomas, familial syndromes, colorectal carcinogenesis, colorectal carcinoma, and carcinoid tumors.
[00159] Examples of proliferative disorders of the liver treatable by the above method include, but are not limited to, nodular hyperplasias, adenomas, and malignant tumors, including primary carcinoma of the liver and metastatic tumors.
[00160] Examples of proliferative disorders of the ovary treatable by the above method include, but are not limited to, ovarian tumors such as, tumors of coelomic epithelium, serous tumors, mucinous tumors, endometeriod tumors, clear cell adenocarcinoma, cystadenofibroma, brenner tumor, surface epithelial tumors; germ cell tumors such as mature (benign) teratomas, monodermal teratomas, immature malignant teratomas, dysgerminoma, endodermal sinus tumor, choriocarcinoma; sex cord-stomal tumors such as, granulosa-theca cell tumors, thecomafibromas, androblastomas, hill cell tumors, and gonadoblastoma; and metastatic tumors such as Krukenberg tumors.
Administration
[0002] The inventive compounds may be administered using any amount and any route of administration effective for treatment. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular composition, its mode of administration, its mode of activity, and the like. The compounds of the present invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts.
[0003] The inventive compounds and compositions of the present invention may be administered by any route. In some embodiments, the inventive compounds and compositions are administered via a variety of routes, including oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, bucal, enteral, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. Specifically contemplated routes are systemic intravenous injection, regional administration via blood and/or lymph supply, and/or direct administration to an affected site. In general the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), the condition of the subject (e.g., whether the subject is able to tolerate oral administration), etc. At present the oral and/or nasal spray and/or aerosol route is most commonly used to deliver therapeutic agents directly to the lungs and/or respiratory system. However, the invention encompasses the delivery of the inventive pharmaceutical composition by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
[0004] The exact amount of a compound required to achieve a therapeutically effective amount will vary from subject to subject, depending on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound(s), mode of administration, and the like. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). [0005] In certain embodiments of the present invention, a therapeutically effective amount of an inventive compound for administration one or more times a day to a 70 kg adult human may comprise about 0.0001 mg to about 5000 mg of an inventive compound per unit dosage form. It will be appreciated that dose ranges as described herein provide guidance for the administration of inventive pharmaceutical compositions to an adult. The amount to be administered to, for example, a child or an adolescent can be determined by a medical practitioner or person skilled in the art and can be lower or the same as that administered to an adult.
[0006] In certain embodiments, the inventive pharmaceutical composition comprises about 0.0001 mg to about 1000 mg of an inventive compound per unit dosage form. In certain embodiments, the composition comprises from about 0.0001 mg to about 1000 mg, from about 0.001 mg to about 1000 mg, from about 0.01 mg to about 1000 mg, from about 0.01 mg to about 1000 mg, from about 0.1 mg to about 1000 mg, from about 1 mg to about 1000 mg, from about 2 mg to about 1000 mg, from about 4 mg to about 1000 mg, from about 6 mg to about 1000 mg, from about 8 mg to about 1000 mg, from about 10 mg to about 1000 mg, from about 20 mg to about 1000 mg, from about 30 mg to about 1000 mg, from about 40 mg to about 1000 mg, from about 60 mg to about 1000 mg, from about 80 mg to about 1000 mg, from about 100 mg to about 1000 mg, from about 200 mg to about 1000 mg, from about 300 mg to about 1000 mg, from about 400 mg to about 1000 mg, from about 500 mg to about 1000 mg, from about 600 mg to about 1000 mg, from about 700 mg to about 1000 mg, from about 800 mg to about 1000 mg, from about 900 mg to about 1000 mg; from about 0.0001 mg to about 1000 mg, from about 0.0001 mg to about 900 mg, from about 0.0001 mg to about 800 mg, from about 0.0001 mg to about 700 mg, from about 0.0001 mg to about 600 mg, from about 0.0001 mg to about 500 mg, from about 0.0001 mg to about 400 mg, from about 0.0001 mg to about 300 mg, from about 0.0001 mg to about 200 mg, from about 0.0001 mg to about 100 mg, from about 0.0001 mg to about 90 mg, from about 0.0001 mg to about 80 mg, from about 0.0001 mg to about 70 mg, from about 0.0001 mg to about 60 mg, from about 0.0001 mg to about 50 mg, from about 0.0001 mg to about 40 mg, from about 0.0001 mg to about 30 mg, from about 0.0001 mg to about 20 mg, from about 0.0001 mg to about 10 mg, from about 0.0001 mg to about 8 mg, from about 0.0001 mg to about 6 mg, from about 0.0001 mg to about 4 mg, from about 0.0001 mg to about 2 mg; from about 0.0001 mg to about 1 mg; from about 0.0001 mg to about 0.1 mg; from about 0.0001 mg to about 0.01 mg; from about 0.0001 mg to about 0.001 mg of an inventive compound per unit dosage form. In certain embodiments, the composition comprises at least about 0.0001 mg, at least about 0.001 mg, at least about 0.01 mg, at least about 0.1 mg, at least about 1 mg, at least about 2 mg, at least about 4 mg, at least about 6 mg, at least about 8 mg, at least about 10 mg, at least about 20 mg, at least about 30 mg, at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 120 mg, at least about 140 mg, at least about 160 mg, at least about 180 mg, at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at least about 460 mg, at least about 480 mg, at least about 500 mg, at least about 520 mg, at least about 540 mg, at least about 560 mg, at least about 580 mg, at least about 600 mg, at least about 620 mg, at least about 640 mg, at least about 660 mg, at least about 680 mg, at least about 700 mg, at least about 720 mg, at least about 740 mg, at least about 760 mg, at least about 780 mg, at least about 800 mg, at least about 820 mg, at least about 840 mg, at least about 860 mg, at least about 880 mg, at least about 900 mg, at least about 920 mg, at least about 940 mg, at least about 960 mg, at least about 980 mg, or at least about 1000 mg of an inventive compound per unit dosage form.
Combination therapy
[0007] It will be appreciated that an inventive compound or composition, as described above and herein, can be employed in combination with one or more additional medical procedures, such as surgery or radiation therapy.
[0008] It will be also appreciated that an inventive compound or composition, as described above and herein, can be employed in combination with one or more additional therapeutically active agents. [0009] By "in combination with," it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the invention. The compositions can be administered concurrently with, prior to, or subsequent to, one or more other additional therapeutically active agents.
[0010] By a "therapeutically active agent" or "active agent" refers to any substance that is useful for therapy, including prophylactic and therapeutic treatment. [0011] An active agent includes a compound that increases the effect or effectiveness of another compound, for example, by enhancing potency or reducing adverse effects of the other compound (i.e., an adjuvant).
[0012] In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.
[0013] Additionally, the invention encompasses the delivery of the inventive pharmaceutical compositions in combination with agents that may improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
[0014] In will further be appreciated that the additional therapeutically active agent utilized in this combination may be administered together in a single composition or administered separately in different compositions.
[0015] In general, it is expected that additional therapeutically active agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
[0016] The particular combination to employ in a regimen will take into account compatibility of the inventive compound with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved.
[0017] It will also be appreciated that the therapy employed may achieve a desired effect for the same disorder (for example, an inventive compound may be administered in combination with another anti-proliferative agent), and/or they may achieve different effects (e.g., control of any adverse side-effects).
[0018] In certain embodiments, an active agent is an anti-cancer agent, antibiotic, anti-viral agent, anti-HTV agent, anti-parasite agent, anti-protozoal agent, anesthetic, anticoagulant, inhibitor of an enzyme, steroidal agent, steroidal or non-steroidal antiinflammatory agent, antihistamine, immunosuppressant agent, anti-neoplastic agent, antigen, vaccine, antibody, decongestant, sedative, opioid, analgesic, anti-pyretic, birth control agent, hormone, prostaglandin, progestational agent, anti-glaucoma agent, ophthalmic agent, anticholinergic, analgesic, anti-depressant, anti-psychotic, neurotoxin, hypnotic, tranquilizer, anti-convulsant, muscle relaxant, anti-Parkinson agent, anti-spasmodic, muscle contractant, channel blocker, miotic agent, anti-secretory agent, anti-thrombotic agent, anticoagulant, anti-cholinergic, β-adrenergic blocking agent, diuretic, cardiovascular active agent, vasoactive agent, vasodilating agent, anti-hypertensive agent, angiogenic agent, modulators of cell-extracellular matrix interactions (e.g. cell growth inhibitors and anti-adhesion molecules), or inhibitors/intercalators of DNA, RNA, protein-protein interactions, protein- receptor interactions, etc.
[0019] Exemplary active agents include, but are not limited to, small organic molecules such as drug compounds, peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins, and cells. In certain embodiments, the active agent is a cell. Exemplary cells include immune system cells (e.g., mast, lymphocyte, plasma cell, macrophage, dendritic cell, neutrophils, eosinophils), connective tissue cells (e.g., blood cells, erythrocytes, leucocytes, megakarocytes, fibroblasts, osteoclasts), stem cells (e.g., embryonic stem cells, adult stem cells), bone cells, glial cells, pancreatic cells, kidney cells, nerve cells, skin cells, liver cells, muscle cells, adipocytes, Schwann cells, Langerhans cells, as well as (micro)-tissues such as the Islets of Langerhans.
[0020] In certain embodiments, the active agent is a small organic molecule. In certain embodiments, a small organic molecule is non-peptidic. In certain embodiments, a small organic molecule is non-oligomeric. In certain embodiments, a small organic molecule is a natural product or a natural product-like compound having a partial structure (e.g., a substructure) based on the full structure of a natural product. Exemplary natural products include steroids, penicillins, prostaglandins, venoms, toxins, morphine, paclitaxel (Taxol), morphine, cocaine, digitalis, quinine, tubocurarine, nicotine, muscarine, artemisinin, cephalosporins, tetracyclines, aminoglycosides, rifamycins, chloramphenicol, asperlicin, lovastatin, ciclosporin, curacin A, eleutherobin, discodermolide, bryostatins, dolostatins, cephalostatins, antibiotic peptides, epibatidine, α-bungarotoxin, tetrodotoxin, teprotide, and neurotoxins from Clostridium botulinum. [0021] In certain embodiments, a small organic molecule is a drug approved by the
Food and Drugs Administration as provided in the Code of Federal Regulations (CFR).
Screening and Assays
[00161] Centrosomes, or mitotic spindle pole bodies, are the main microtubule organizing centers in mammalian cells. Centrosomes are duplicated in synchrony with chromosomes during S-phase, with duplication complete by G2, to assure bipolar mitosis and equal chromosome segregation during mitosis. If a centrosome replicates too often or fails to replicate, the daughter cells (each inheriting one centrosome = two centrioles) can get abnormal chromosomal compositions, which can predispose the cells to cancer. Researchers have found that pre-cancerous as well as cancerous cells have abnormal numbers of centrioles (e.g., more than two) or centrosomes (e.g., more than one) (Pihan et ah, Cancer Research (1998) 58:3974-3985; Skyldberg et ah, Modern Pathology (2001) 279-284). In breast, prostate and cervical cancer, such abnormalities arise in to form early premalignant lesions. Multi-polar mitoses are diagnostic hallmarks of high-risk HPV-associated lesions. [00162] Many mitotic reglulators are aberrantly expressed in proliferating cells. For example, centrosome-associated protein Aurora A is amplified in cancer cells, indicating that it is important for cancer formation and/or progression (Keen and Taylor, Nature Reviews Cancer (2004) 4:927-936). A diagnostic agent which binds to centrosome-associated proteins, such as Aurora-A, Centrin and γ-tubulin, can allow for detection of centrosome abnormalities in a cell of a biological sample, and thus detection of a cancerous or precancerous condition.
[00163] Thus, in one aspect, the present invention provides a method of detecting proliferative cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes. [00164] In another aspect, the present invention provides a method of detecting precancerous cells in a biological sample, comprising the steps of: (i) contacting the biological sample with a compound of formulae (I) or (II) conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes. [00165] As used herein, "screening" is meant visually viewing the sample, and
"screening" may be further employ visual screening aids, such as, for example, an automated device, instrument (e.g., microscope), light source (e.g., high or low UV light, Infrared light, etc.), and/or stain etc.
[00166] As used herein, when two entities are "conjugated" to one another they are linked by a direct or indirect covalent interaction. An indirect covalent interaction is when two entities are covalently connected through a linker group. Exemplary imaging agents include, but are not limited to, fluorescent and luminescent moieties.
[00167] In certain embodiments, the imaging agent is a fluorescent moiety. In certain embodiments, the fluorescent moiety is selected from Hydroxycoumarin; Aminocoumarin;
Methoxycoumarin; Cascade Blue; Lucifer yellow; NBD; R-Phycoerythrin (PE); PE-Cy5; PE-
Cy7; APC-Cy7; Red 613; Fluorescein (FITC); FluorX; BODIPY-FL; TRITC; X-Rhodamine;
Lissamine Rhodamine B; PerCP; Texas Red; Allophycocyanin (APC); TruRed; Alexa Fluor dyes; and Cy dyes. In certain embodiments, the fluoresent moiety is Fluorescein.
[00168] As used herein, an abnormal number of centrioles or centrosomes means more than one centrosome per cell not undergoing mitosis, or three or more centrioles per cell undergoing mitosis.
[00169] Examples of biological samples include tissue samples, organ biopsies and biological fluids such as saliva, blood, plasma, urine, semen, seminal fluid, amniotic fluid, cerebrospinal fluid, and the like.
[00170] The present invention also provides a method of identifying compounds which bind to Aurora-A kinase, comprising the steps of: (i) contacting a cancer cell with a test compound of formulae (I) or (II) conjugated to an imaging agent and (ii) screening for binding to Aurora-A kinase.
[00171] Verification of binding will be visually apparent by a localization of the visual marker (e.g., a fluorescence or luminescence) in the area of the cell where Aurora-A kinase is over-expressed (e.g., the centrosomes).
[00172] In certain embodiments, the compound of formulae (I) or (II) binds toAurora
A kinase protein, but does not modulate (e.g., activate, inhibit or partially inhibit) Aurora A kinase protein activity. However, in certain embodiments, the compound of formulae (I) or
(II) binds to Aurora A kinase protein, and activates Aurora A kinase protein activity. In certain embodiments, the compound of formulae (I) or (II) binds to Aurora A kinase protein, and inhibits or partially inhibits Aurora A kinase protein activity.
[00173] In certain embodiments, the proliferative cells are cancer cells. In certain embodiments, the cancer cells are selected from a group consisting of HeLa cells, U2OS cells, MCF7 cells, SW620 human colon adenocarcinoma cells, LS174T human colorectal adenocarcinoma cells, KM 12 human colon carcinoma cells, HCTl 16 human colon carcinoma cells, SR human leukemia cells, RL human non-Hodgkin's lymphoma B-lymphoblasts, PA682 Burkitt's human lymphoma cells, Leukemia (acute myeloid) human cells, HL60 human acute promyelocytic leukemia cells, HCTl 16 human colon carcinoma cells, LS174T human colorectal adenocarcinoma cells, MDAMB231 human breast carcinoma cells, HeLa human cervix adenocarcinoma cells, A375 human malignant melanoma cells, PC3 human prostate cancinoma cells (androgen-independent), MiaPaCa2 human pancreas adenocarcinoma cells, and MDAMB231 human breast carcinoma cells. [00174] In certain embodiments, the compound or test compound is a member of a diversity oriented synthesis (DOS) library of compounds, a purchased library of compounds and/or a historical collection of compounds.
[00175] In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner.
Examples
[00176] Modern methods for small-molecule synthesis are enabling the examination of a far greater array of chemical diversity in biological processes than previously possible (Schreiber, Chem. Eng. News (2003) 81:51). When high yielding and selective chemical transformations are performed on functionalized solid supports, thousands of compounds can be synthesized in only 3 to 4 chemical steps. High-throughput cell-based assays and small- molecule microarray-based assays provide a broad assessment of the properties of these compounds in biological settings (for high-throughput cell-based assays see, for example, Strausberg and Schreiber, Science (2003) 300:294; for small-molecule microarray-based assays see, for example, Bradner et al, Chem. Biology (2006) 13:493). [00177] Shikimic acid is a naturally occurring small molecule, and has been used to synthesize many biologically active natural products (see, for example, Floss Nat. Prod. Rep. (1997) 433) and several potent non-natural compounds such as neuraminidase inhibitors, e.g. Oseltamivir (Tamiflu™) (Karpf and Trussardi, J. Org. Chem. (2001) 66: 2044). Shikimic acid-based libraries have been synthesized and tested as glycomimetics (Schuster et al, Org. Lett. (2003) 5:1407). Previously, a large collection of compounds was prepared using trans 4,5-epoxy 3-hydroxy-2-cyclohexenoic acid, both enantiomers of which are derived from (- )-shikimic acid (Tan et al., J. Am. Chem. Soc. (1998) 120:8565; Tan et al., J. Am. Chem. Soc. (1999) 121 :9073). [00178] In this example, a solid phase resin is used that permits epoxide opening by amines and subsequent acylation of the resulting amine (Scheme 1). A novel intramolecular Heck reaction yields tricyclic dihydrobenzofurans, a structure found in a number of biologically active natural products. This strategy was recently used in the total synthesis of galanthamine (Trost et ah, J. Am. Chem. Soc. (2005) 127: 14785), a reversible inhibitor of acetylcholinesterase used for the treatment of Alzheimer's disease (Guillou et al, Bioorg. Med. Chem. Lett. (2000) 10:637). Scheme 1.
Figure imgf000070_0001
*" °Penln9
(-) Shikimic acid (1)
Figure imgf000070_0002
[00179] The Schemes 2 and 3 demonstrate how the compounds were synthesized on the solid phase and in solution.
[00180] (-)-Shikimic acid (1) was converted to sjw-hydroxyepoxide 2 using a previously reported procedure (McGowan and Berchtold, J. Org. Chem. (1985) 50: 1557) (steps a-c) (Scheme 2). (-)-Shikimic acid (1) was first esterified (Tan et al., J. Am. Chem. Soc. 1998, 120, 8565. Tan et al., J. Am. Chem. Soc. 1999, 121, 9073) then the trans diol was converted to an epoxide and the ester was hydrolyzed to give carboxylic acid 2. Both enantiomeric trans stereoisomers of 2 are also accessible from 1 (Wood and Ganem, J. Am. Chem. Soc. (1990) 112:8907). Scheme 2.
Figure imgf000070_0003
a) MeOH, Amberlite-IR120; b) PPh3, DEAD/THF; c) LiOH, THF/H2O
[00181] Polystyrene resin (500-600 μm bead size) functionalized with a diisopropyl
4-methoxyphenyl silyl group was activated under the recommended conditions (Tallarico et al., J. Comb. Chem. (2001) 3:312) and coupled to N-Fmoc ethanolamine (step a) (Scheme 3). Deprotection with piperidine/DMF gave the amino modified resin (step b) to which 2 was coupled under standard conditions (step c). [00182] A Mitsunobu reaction on the solid phase was first tested with 2-iodophenol using standard conditions (Mitsunobu, Synthesis (1981) 1). The reaction mixture was agitated overnight, rinsed and treated with HF in order to cleave the silyl linker. Using HPLC/MS, the product was detected as a single entity, with no starting alcohol. Previous studies with shikimic acid-based systems showed that MgBr2-catalyzed epoxide opening by amines is highly regioselective (Schuster et al, Org. Lett. (2003) 5: 1407). Opening of the solid-phase epoxide with amines was tested with varying amounts of an amine (e.g., benzylamine, N-methylbenzylamine) in a number of solvents (THF, CH3CΝ, DMF, dioxane, CH2CI2) and Lewis acids (MgBr2-Et2O, LiClO4, Yb(OTf)3, LiOTf) at various temperatures. This survey revealed that 20-40 equivalents of amine with 1-1.1 equiv of a Lewis acid (e.g., MgBr2-Et2O) in a solvent (e.g., CH3CN) is optimal. HPLC and NMR (500 MHz) analyses following cleavage indicated the presence of only one amine product. Acylation was tested using an acylating agent (e.g., an acyl chloride or a carboxylic acid) with various coupling agents (e.g. DCC, EDAC, PyBOP, PyBrOP, and the like). Finally, Heck cyclization was tested using various Pd sources (e.g., Pd(PPh3)4, PdCl2(PPh3)2, Pd(OAc)2), ligands (e.g., dppp, dcpe, dcpp) and bases (e.g., DIPEA, K2CO3) in different solvents (e.g., DMF, DMA, dioxane). l,2-(Dicyclohexylphosphino)-ethane (dcpe), which has been used for similar cyclizations (Trost and Toste, J. Am. Chem. 5bc.(2000) 122: 11262) and palladium acetate at 80 0C for 24 hr proved optimal, yielding single tricyclic products. Later it was discovered that the chemical tags used as a means of encoding the reaction history were cleaved from the resin at this temperature (Ohlmeyer et al., Proc. Natl. Acad. ScL U.S.A. (1993) 10922; Nestler et al., J. Org. Chem. (1994) 59:4723; Blackwell et al., Chem. Biol. (2001) 8: 1167). The addition of tetrabutylammonium acetate led to equally clean reaction mixtures in less time (12 h) and at lower temperature (45-50 C) such that the chemical tags were retained. [00183] A library synthesis (Scheme 3) was achieved using the one-bead/one-stock solution procedure (Tallarico et al., J. Comb. Chem.(200l) 3:312) with chemical tagging of the resin being performed after each step using a variation of Still's procedure (Ohlmeyer et al., Proc. Natl. Acad. ScL U.S.A. (1993) 10922; Nestler et al., J. Org. Chem. (1994) 59:4723; Blackwell et al., Chem. Biol. (2001) 8: 1167).
Figure imgf000072_0001
12 - 90 compounds 13 - 2700 compounds 14 - 2700 compounds a\ trifluoromethanesulfonic acid, then 2,6-lutidene, CH2Cl2, then 7V-Fmoc-ethanolamine; b) piperidine/DMF; c) 2, PyBOP, DIPEA/NMP/CH2C12; (d) optionally substituted iodophenol, PPh3, DIAD/THF; (e) amine, Lewis acid, solvent; (f) acylating agent, coupling agent, solvent, base; (g) Heck cyclization; (h) cleavage from resin Qp = polystyrene macrobeads
[00184] The reactions were all performed with shaking overnight followed by extensive washings. Cleavage of the products from the resin was performed using HF-pyridine for 3 h followed by treatment with TMSOEt. The Mitsunobu reactions were achieved with 2-iodophenol, 4-(ter?-butyldimethylsilyloxy)methyl-6-methoxy-2- iodophenol, and 4-formyl-6-methoxy-2-iodophenol (Doria, Eur. J. Med. Chem. (1978) 13:33), the last two phenols having handles for chemical transformation after library realization (Strausberg and Schreiber Science (2003) 300:294). HPLC and NMR analyses after cleavage from a single bead indicated that the desired product was formed in each case, generally as a predominantly single entity. [00185] Each of the three products was then chemically tagged and the products were combined, mixed thoroughly and divided into 30 even portions. An additional tagging was then performed to track 30 structurally diverse amines which were used in the library synthesis to open the resin-bound epoxide (Table 1). Thirty beads (out of 90) were cleaved, decoded, and analyzed by HPLC/MS; no starting epoxides were detected and the product amines were generally >90% pure. After mixing and dividing, the resins were further functionalized with 30 carboxylic acids (Table 1) followed by tagging. Thirty bead portions were analyzed; no starting amines were detected and the products were typically >90% pure. The collection of beads was divided in half at this point, one half was subjected to the Heck reaction conditions. The Heck cyclization gave less pure products; analysis of a set of 20 products indicated that purities ranged mostly between 70-90%. The structures of the side products could not be determined. One expected side product, the reduced, non-cyclized des- iodo compound, was not detected in the crude reaction mixture; authentic material was synthesized by running the entire reaction sequence replacing 2-iodo phenol with phenol. [00186] The overall process should theoretically produce a total of 5,493 (3 + 90 +
2,700 + 2,700) distinct compounds. Enough resin (3.3 g, ca. 5 beads/mg, ca. 16,500 beads) was used to make three copies of each product, some of which are used for quality control in the library synthesis. Prior to the Heck reaction, little bead breakage was observed. The Heck reaction did result in bead breakage such that only about 70% of the beads remained intact after this reaction. A total of 6,336 wells were filled with compound for multi-purpose screening.
Table 1.
Figure imgf000073_0001
Table 1. (continued)
Amines
Figure imgf000074_0001
s-11 s-12 s-13 s-14 s-15
Figure imgf000074_0002
s-21 s-22 s-23 s-24 s-25
Figure imgf000074_0003
s-31 s-32
Table 1 (continued)
Carboxylic Acids
Figure imgf000075_0001
[00187] Stock solutions corresponding to 1408 members of the resulting collection of small-molecule products were printed onto isocyanate-functionalized glass slides to prepare small-molecule microarrays (SMMs) (Bradner et al. Chem. Bio. (2006)13:493). The SMMs were screened for binding to a number of enzymes, hits being detected in various assays for amine, amide, and Heck products. Among the screens producing screening positives was the anti-cancer target Aurora A kinase.
[00188] Seven positives from the SMM assay (1-1 to 1-7, Table 2) were resynthesized either in solution (with a TBDMS group as a surrogate for the resin, Scheme 3) or on solid phase; 20 mg of functionalized resin typically produced 5-10 mg of purified product. Compounds 1-1 to 1-7 were analyzed for binding to Aurora A kinase in a secondary binding assay using surface plasmon resonance (SPR, Figures 1 and 2A) (Barnes-Seeman et al., Angew. Chem. Int. Ed. (2003) 42:2376). Compounds 1-2 to 1-7 each bound to Aurora A weakly with affinities greater than 40 μM. Compound 1-1 had a KD value of 5.3 ± 1 μM (n = 4) by kinetic analysis and a KD value of 6.0 ± 1 μM by equilibrium analysis (n = 4) (Figure 2). The activity of 1-1 was assessed in a cell-free Aurora A kinase enzymatic activity assay (Aurora-A kinase Assay/Inhibitor Screening Kit, CycLex Co., Ltd., cat. no. CY-1165). Although the binding assay demonstrated that 1-1 possessed good binding affinity, this Aurora A-binding compound did not inhibit kinase activity. Table 2.
Figure imgf000076_0001
(1-4) (1-5) (1-6)
Figure imgf000076_0002
(1-7)
[00189] Compound 1-1 was used as a starting point for the synthesis of derivatives
(compounds 1-8 to 1-34, synthesized in amounts varying from 0.76 mg to 2.82 mg and purities ranging from 81-92%) that exhibited better binding affinity for Aurora A without inhibiting the protein's enzymatic activity (Table 3). Such molecules may be useful for diagnostic purposes, to detect cells in clinical specimens that overexpress Aurora A. [00190] Compounds were ranked according to the percent theoretical maximum response they gave at 40 μM as the majority of compounds gave little response at 400 nM. Of those that did give a response at 400 nM, only one, compound, HUM287, did not give a higher response than compound 1-1 at 40 μM. Nonetheless, it should also be considered for further evaluation.
[00191] To rank the compounds at 40 μM, the percent theoretical maximum response was normalized for the molecular weight of the compounds and for the response relative to the response seen by compound 1-1 for that assay. Overall, two compounds (1-33) and (1-30) gave a substantial increase in response over 1-1 with a total of eleven compounds showing an increase in response [i.e., compounds (1-30), (11-35), (1-33), and (1-34) bind better than compound 1-1 by more than 1.5-fold; compounds (1-27), (TI-I), (1-25), (1-24), (1-12), (I- 10), and (1-8) bind better than compound 1-1 by less than 1.5-fold]. In both assays, the estimated equilibrium KD for compound 1-1 was 4.9 μM, validating the assay conditions. Table 3.
Figure imgf000077_0001
(1-8) (1-9) (i-io) (1 11)
Figure imgf000077_0002
(1-12) (1-13) (1-14) (1-15)
Figure imgf000077_0003
(1-16) (1-17) (1-18) (1-19)
Figure imgf000078_0001
(1-20) α-2i) α-22) (1-23)
Figure imgf000078_0002
(1-24) (1-25) (II I) (1-26)
Figure imgf000078_0003
(1-27) (1-28) (1-29) (1-30)
Figure imgf000078_0004
(1-33) (1-34)
Figure imgf000079_0001
(1-36) (1-37)
Small-Molecule Microarray Assays
[00192] Recombinant Aurora A was expressed in E. coli Rosetta2 (DE3) cells
(Novagen) as a C-terminal 6xHis-tagged protein and was purified using Ni-NTA agarose affinity chromatography according to the standard protocols. Small-molecule microarrays were incubated in triplicate with 400 μL of a 10 μg/mL solution of purified Aurora A-6xHis in TBST buffer for 30 min at room temperature. The arrays were washed three times in TBST (1 min for each wash) on an orbital platform shaker. Arrays were then incubated with 300 μL of a 0.2 μg/mL solution of Cy5-labeled anti-5xHis antibody (Qiagen) in TBST for 30 min at room temperature. The probed arrays were washed three times in TBST (5 min for each wash), followed by doubly distilled water for 3 min on an orbital platform shaker. Arrays were dried by centrifugation and scanned for fluorescence at 635 nm using a Genepix 4000B microarray scanner.
Surface Plasmon Resonance Experiments
[00193] The Aurora A kinase surface plasmon resonance assay was conducted on a
Biacore S51 instrument using Biacore CM5 sensor chips. Ethanolamine, EDC, NHS, and P- 20 surfactant were all obtained from Biacore, Inc. Aurora A was directly immobilized through primary amines using standard EDC/NHS chemistry. The sensor surface was conditioned using alternating injection of 10 mM glycine pH 2.2 and 50 mM NaOH. The surface was then activated with 1: 1 4 M EDC: 1 M NHS for 10 minutes. Aurora A diluted to 15 μg/mL in 10 mM acetate pH 5.5 was exposed to the activated sensor. The surface was quenched by a seven minute injection of 1 M ethanolamine. Protein activity was assessed by observing the binding of staurosporine. Small molecule binding assays were conducted in 23 mM Tris buffer pH 7.4 with 137 mM NaCl, 3 mM KCl, 0.005% P-20 surfactant and DMSO or DMF co-solvent. Co-solvent was varied from 2-5% and 5 mM MgCl2 was used in some assays, however neither had an effect on compound 1-1 affinity. Sensor data was analyzed using the Scrubber 2 software (BioLogic Software Pty Ltd). Data was double reference subtracted and corrected for variation in solvent concentration. Binding affinity was calculated using kinetic and equilibrium analyses. A least-squares fit of a Langmuir 1 : 1 binding model was implemented for kinetic analysis.
Sensor Chip Preparation
[00194] Aurora A was directly immobilized using standard EDC/NHS chemistry.
Aurora A (2.4 mg/mL) was thawed before the first use and divided into 5.5 μL aliquots that were stored at -200C. Freshly thawed Aurora A was used for each day's assay. The running buffer used in the preparations was PBS-P pH 7.4 and the temperature of the assay was 25°C. Aurora A was diluted to 150 ug/ml in 1OmM Acetate Buffer pH 5.5 and immobilized on the sensor surface on Spot 1 (day 1) and Spot 2 (day 2) at 5 μL/min for 10 min using standard EDC/NHS chemistry. Between 10,990 and 11,118 Response Units (RU) of protein were immobilized in each assay. An anti-RGSHis antibody (Qiagen) immobilized at a level of -3,000 RU was used as a reference surface.
Assay Parameters
[00195] Compounds 1-1 to 1-7 (Table 2) were diluted from 5 mM stocks in DMF to the appropriate concentration in buffer with the same solvent concentration as the running buffer (2%). Compounds were injected at a flow rate of 30 μL/min into the flow cell for 60 seconds followed by 90 seconds of buffer with no compound. Compound 1-1 was further characterized by measuring the binding at concentrations from 391 nM to 25 μM in half dilution. A flow rate of 90 μL/minute was used in the characterization of compound 1-1. [00196] Compounds of Table 3 were diluted from 10 mM stocks in DMSO to the appropriate concentration in buffer to give the same solvent concentration as the running buffer (PBS-P, 2% DMSO). Compounds were injected into the flow cell at 90 μL/min for 120 seconds followed by 120 seconds of buffer with no compound. An extra buffer injection was performed after each injection to account for compounds that were slow to dissociate. Each compound was tested with duplicate injections at 400 nM and 40 μM and compared to the established affinity for compound 1-1.
Fluorescence Imaging Experiments
[00197] U2OS (osteosarcoma), HeLa (cervix cancer), and MCF7 (breast cancer) cells were used for immunofluorescence analyses. Cells were grown on glass coverslips, pre- extracted at room temperature with CSK buffer (100 mM NaCl, 300 mM sucrose, 10 mM PIPES-pH6.8, and 3 mM MgCl2) with 0.1% Tritin-XIOO and rinsed with CSK buffer. Cells were then fixed and permeabilized in cold methanol for 10 min at -20 C, followed by rehydrated with PBS for 5 min. Normal goat serum (Jackson ImmunoResearch) was used for blocking samples at the concentrations of 10% in SNBP buffer (BSA (5g/L), saponin (0.1 g/L), sodium azide (0.25 g/L) in PBS) for 1 h at room temperature. FITC-labeled compound 1-33 (provided by method of Scheme 4) was incubated at the concentrations of 2.2 μM in the presence of polyclonal antibody against Aurora-A (Abeam, cat. #abl3408) at a dilution of 1 : 1000 for 30 min at 37 C, followed by additional 30 min at room temperature. Samples were washed with SNBP buffer and incubated with Alexa Fluor 647 goat anti-rabbit IgG antibody (Molecular Probes, cat. #A21244) at a dilution of 1: 1000 for 1 h at room temperature. Nuclei were stained with Hoechst 33258 DNA dye. Confocal images were acquired by using a PCM2000 (Nikon) coupled to a Zeiss microscope using SIMPLE 32 software (Nikon) (Figure 11). Green and far-red channels were used for the imaging. Scheme 4.
Figure imgf000081_0001
(a) 5-fluoresceine 5-carbonyl azide diacetate (b) hydroxylamine
Enzyme Inhibition Assays
[00198] Enzyme inhibition assays were run with a commercially available Aurora-A kinase Assay/Inhibitor Screening Kit, CycLex Co., Ltd., (cat. no. CY-1165) as instructed. Briefly, compounds at various concentrations were incubated with Aurora-A positive control (CycLex Co., Ltd., cat. no. CY-El 165) and ATP in kinase reaction buffer in microtiter wells at 30 C for 45 minutes. Wells were washed with wash buffer (5x200 μl). 100 μl of anti- phospho-Lats2-S83 antibody solution (supplied with the kit) was added to each well and incubated at room temperature for 30 min. Wells were washed with wash buffer (5x200 μl - supplied with the kit). 100 μl of HRP-conjugated anti-mouse IgG solution was added per well and incubated at room temperature for 30 min. Wells were washed with wash buffer (5x200 μl), and 100 μl of substrate reagent (supplied with the kit) was added and incubated at room temperature for 10 min. Reactions were stopped by adding 100 μl of stop solution (supplied with the kit) and absorbance was measured in each well using a spectrophotomeric plate reader (Molecular Devices, Spectra MAX 190) at dual wavelengths of 450 and 540 nm.
Synthetic Methods
[00199] General Information. Starting materials and regents were purchased from commercial suppliers and used without further purification. All reactions were run under inert atmosphere. CH2CI2, THF, and Et2θ were passed through two activated alumina columns to remove impurities prior to use (as described in Timmers et ah, Organometallics (1996) 15: 1518-1520). 2,6-Lutidine was distilled from CaEt under nitrogen. Brominated polystyrene macrobeads were purchased from Polymer Labs. The functionalized 500-600μm macrobeads used were prepared according to published procedure (Tallarico et ah, J. Comb. Chem. (2001) 3:312). The loading level was determined according to published procedure, and the amount of eluted compound was quantified by 1H NMR using (PhMe2Si)2θ as internal standard.
[00200] Purification of reaction products was carried out by flash chromatography using E. Merck silica gel 60 (230-400 mesh). Analytical thin layer chromatography was performed on E. Merck 0.25 mm silica gel 60-F plates. Visualization was accomplished with UV light and aqueous cerium ammonium molybdenate (CAM) solution followed by heating. Analytic LC/MS chromatography was performed on Waters Alliance 2690 HPLC system with a Waters Symmetry C18 column (3.5 um, 4.6X 100mm column) gradient of 20-80% CH3CN in water, constant 0.1% formic acid, with UV detection at 214 and 280 nm and a Micromass LCZ (ESI) spectrometer.
[00201] 1H NMR spectra were recorded on a Varian 500 MHz spectrometer and are reported in ppm and referenced to residual protons in the NMR solvent. Data are reported as shift, splitting (s = singlet, d = doublet, t = triplet, m = multiplet; br = broad), coupling constant in Hz; intergration. 13C NMR spectra were recorded at 125 MHz on Varian spectrometers, 13C shifts are reported in ppm and referenced to carbon resonances in the NMR solvent. Solid Phase Experimental Procedures
[00202] Small-scale solid phase reactions (5-10 mg resin) were performed in 1 mL fritted polyacrylamide Bio-Spin® chromatography columns (Bio-Rad Laboratories, Hercules, CA; 732-6008) or Wheaton glass vials, fitted with Teflon-coated caps with gentle mixing provided by Thermoline Vari-Mix shaker or a Vortex Genie-2 vortexer (VWR 58815-178, setting V1-V2). Large-scale solid phase reactions (>500 mg resin) were performed in 10-50 mL Amersham columns or silanized 50 or 100 mL fritted glass tubes equipped for vacuum filtration and N2 bubbling. The tubes were silanized by treatment with 20% dichlorodimethylsilane/CH2Cl2 for 15 min, MeOH for 15 min, followed by oven heating at 120 0C for at least 2 h.
[00203] Resin samples were washed as indicated and solvent was removed under argon flow and/or in vacuum on a Vac-Man® laboratory vacuum manifold (Promega, Madison, WI; A7231) fitted with nylon 3-way stopcocks (Biorad 732-8107). Resin sample in glass vials were filtered in 10 mL Amersham column and washed on the Vac-Man vacuum manifold.
[00204] The following standard wash procedures were used for rinsing: Method A - 3 x THF, 3 x DMF, 3 x iPrOH, 3 x DMF, 3x THF; Method B - 3 x THF, 3x THF/H2O (9: 1), 3 x DMF, 3 x iPrOH, 3 x DMF, 3x THF.
[00205] All compounds were cleaved from macrobeads using the following standard procedure: To resin in a polypropylene eppendorf tube was added freshly prepared solution of 85% THF, 10% pyridine, and 5% HF pyridine. The resulting mixture was then agitated at room temperature for 3 h before quenching with ethoxytrimethylsilane (10/1 v/v relative to THF). The resulting mixture was agitated at room temperature for 30 min, then transferred to an eppendorf vial and evaporated on a Speedvac. Resin was washed twice with THF and the wash solutions were combined, concentrated and analyzed by HPLC and NMR. [00206] Loading of iV-Fmoc-ethanolamine onto silicon-functionalized polystyrene macrobeads and deprotection (3). Dry 3-[diisopropyl(p- methoxyphenyl)silyl]propyl functionalized macrobeads (400 mg, 1.41 mmol Si/g, 0.56 mmol) in a 20 mL polypropylene tube were allowed to swell in anhydrous CH2CI2 (15 mL) for 30 min under argon atmosphere. To the mixture was added 4% (v/v) solution of CF3SO3H in anhydrous CH2CI2 (0.25 mL, 3.4 mmol) by syringe and the reaction tube was shaken periodically. The beads turned orange upon acid treatment and were allowed to stand for 20 min. After filtration under positive argon pressure, the orange-colored beads were washed with anhydrous CH2CI2 (2 x 15 mL) and then resuspended in CH2CI2 (1 mL). Freshly distilled 2,6-lutidine (0.26 mL, 2.2 mmol) was added resulting in bead discoloration, followed by addition of a CH2CI2/DMF (2: 1) solution of N-Fmoc-ethanolamine (476 mg, 1.68 mmol). The reaction mixture was gently agitated for 12 h. The beads were drained, and subjected to rinsing (method A). The resin was air-dried for 2 h and then placed under vacuum for 12 h. The beads were allowed to swell in anhydrous DMF for 30 min before adding a solution of 20% piperidine in DMF (10 mL) and the reaction tube was agitated for 5 h before the beads were drained, and subjected to rinsing method A. Loading level was determined spectrophotometrically as per Tallarico, et al, J. Comb. Chem. 2001, 3, 312. The resin (3) was air-dried for 2 h and then placed under vacuum for 12 h to remove solvent. [00207] Solid Phase Amide Coupling (4). Resin 3 (15 mg, 1.17 mmol Si/g, 18 μmol) was allowed to swell for 30 min in 5 mL of anhydrous ΝMP before the addition of PyBOP (14 mg, 26 μmol), DIPEA (9.2 μL, 53 μmol) and organic acid (4.1 mg, 26 μmol) and the reaction mixture was gently agitated for 24 h. The beads were drained and rinsed using method A. The resin (4) was then air-dried for 2 h and then placed under vacuum for 12 h. Cleavage produced the 3.2 mg of (15r,5R,6i?)-5-hydroxy-N-(2-hydroxyethyl)-7-oxa- bicyclo[4.1.0]hept-3-ene-3-carboxamide as white foam (91%): 1H ΝMR (500 MHz, CD3OD) 8 6.28-6.27 (m, IH), 4.58^.57 (m, IH), 3.61 (t, J = 6.0 Hz, 2H), 3.52-3.49 (m, IH), 3.48-3.46 (m, IH), 3.37 (t, J = 6.0 Hz, 2H), 2.91 (d, J = 19.0 Hz, IH), 2.50 (dd, J = 19.0, 2.5 Hz, IH); ESI-MS (+) m/z 200 (M+H)+.
[00208] Solid Phase Mitsunobu Ether Formation (5). Resin 4 (15 mg, 18 μmol), 2- iodophenol (39 mg, 180 μmol), PPh3 (46 mg, 180 μmol) and THF (0.67 mL) were briefly vortexed prior to cooling to -78 C and addition of DEAD (27 μL, 180 μmol). The reaction mixture was maintained for 0.5 h at -78 C and then at -20 C for 12 h with occasional agitation. Filtering, washing (method A) and cleavage produced 6.2 mg of (lS,5S,6S)-N-(2- hydroxyethyl)-5-(2-iodophenoxy)-7-oxa-bicyclo[4.1.0]hept-3-ene-3-carboxamide (94% yield): 1H NMR (SOO MHz, CD3OD): δ 7.81 (dd, J= 7.5, 1.5 Hz, IH), 7.39 (td, J= 7.5, 1.5 Hz, IH), 7.22 (dd, J= 7.5, 1.5 Hz, IH), 6.81 (td, J= 7.5, 1.5 Hz, IH), 6.53 (m, IH), 5.29 (m, IH ), 3.63 (t, J= 5.5 Hz, 2H ), 3.56 (m, IH), 3.48 (m, IH), 3.37 (t, J= 5.5 Hz, 2H), 2.92 (dd, J = 20.0, 1.5 Hz, IH), 2.85 (dd, J = 20.0, 2.0 Hz, IH); 13C NMR (125 MHz, CD3OD): δ 169.11, 156.93, 139.82, 132.76, 129.83, 124.64, 123.80, 114.90, 87.41, 70.82, 60.31, 51.01, 50.44, 42.11, 24.44; ESI-MS (+) m/z 402 (M+H)+. [00209] Solid Phase Epoxide Opening (6). Resin 5 (20 mg, 23 μmol) was suspended in anhydrous CH3CN (800 μL). After 15 min, MgBr2-OEt2 (7.2 mg, 28 μmol) and N- benzylamine (51 μL, 470 μmol) were added and the reaction mixture was heated at 55-60 C for 24 h. The beads were drained and subjected to rinsing with method B. Cleavage of a single bead produced 5-benzylamino^-hydroxy-3-(2-iodophenoxy)-cyclohex-l- enecarboxylic acid (2-hydroxyethyl)amide: 1H ΝMR (500 MHz, CDCl3): δ 7.79 (dd, J = 8.0, 1.5 Hz, IH), 7.24-7.38 (m, 6H), 7.00 (dd, J = 8.0, 1.0 Hz, IH), 6.77 (td, J= 8.0, 1.0 Hz, IH), 6.38-6.40 (br m, IH), 6.28 (br m, IH), 4.82^.84 (br m, IH), 4.03 (d, J= 12.5 Hz, IH), 3.94 (dd, J= 10.5, 7.5 Hz, IH), 3.74-3.82 (m, 4H), 3.51-3.48 (m, 2H), 3.05 (dd, J= 16.5, 5.0 Hz, IH), 2.93-2.98 (m, IH), 2.20-2.22 (m, IH); ESI-MS (+) m/z 509 (M+H)+. [00210] Solid Phase Amide Formation (7). Resin 6 (20 mg, ca. 23 μmol) was suspended in anhydrous ΝMP (800 μL) for 15 min. Acetic acid (1.3 μL, 230 μmol), and PyBrOP (109 mg, 230 μmol) were added, followed by the addition of DIPEA (82 μL, 47 μmol). The reaction mixture was shaken for 24 h. The beads were drained and subjected to washing method B. Cleavage of a single bead gave 5-(N-acetyl-N-benzylamino)^- hydroxy-3-(2-iodophenoxy)-cyclohex-l-enecarboxylic acid (2-hydroxyethyl)amide. The LC/MS was consistent with clean desired product. The ΝMR spectrum was complicated due to the presence of rotamers: 1H ΝMR (500 MHz, DMSO-^6 + D2O) δ 7.92-7.95 (m, IH), 7.78 (m, IH), 7.13-7.39 (m, 7H), 6.73-6.78 (m, IH), 6.25 (br s, IH), 5.15 (br s, 0.5H), 4.95 (br s, 0.5H), 4.87 (d, J = 16.5 Hz, 0.5H), 4.60^.80 (br s, 0.5 H), 4.43^.60 (br, 0.5H), 4.06- 4.15 (m, IH), 3.92-3.99 (m, IH), 3.31-3.36 (m, 2H), 3.04-3.10 (m, 2H), 2.49 (s, 1.5 H), 2.10-2.40 (m, 3H), 2.18 (s, 1.5 H); ESI-MS (+) m/z 573 (M+Νa)+.
[00211] Solid Phase Heck Cyclization (8b). Resin 7 (20 mg, ca. 23 μmol) was suspended in degassed dioxane (800 μL) for 15 min before adding Pd(OAc)2 (5.3 mg, 23 μmol), l,2-bis(dicyclohexylphosphino)ethane (dcpe, 9.9 mg, 23 μmol), Bu4NOAc (7.1 mg, 23 μmol) and DIPEA (82 μL, 470 μmol). The reaction mixture was heated at 45-50 °C for 12 h. The beads were drained and subjected to rinsing with method B after washing with NaCN solution (7/3 THF/ 1 M NaCN(aq), 10 mL x 3); cleavage from the resin resulted in 6.2 mg of the expected fused-ring product (75% yield from resin 7). The LC/MS showed the desired product with purity of 85%, the NMR spectrum was complicated due to the presence of rotamers: 1H NMR (500 MHz, CD3OD) 7.26-7.60 (m, 4H), 7.04-7.18 (m, 4H), 6.68-6.78 (m, 2H), 6.24 (s, 0.5H), 6.18 (s, 0.5H), 4.66-4.82 (m, 3H), 4.42^.58 (m, 2H), 4.04^.10 (m, 0.5H), 3.60-3.70 (m, 2.5H), 3.38-3.44 (m, 2H), 2.15 (s, 1.5H), 2.12 (s, 1.5H); ESI-MS (+) m/z 445 (M+Na)+.
[00212] Cleavage from resin (8c). The beads were subjected to washing method B
(Method B - 3 x THF, 3x THF/H2O (9: 1), 3 x DMF, 3 x iPrOH, 3 x DMF, 3x THF). To the resin in a polypropylene eppendorf tube was added freshly prepared solution of 85% THF, 10% pyridine, and 5% HF pyridine. The resulting mixture was then agitated at room temperature for 3 h before quenching with ethoxytrimethylsilane (10/1 v/v relative to THF). The resulting mixture was agitated at room temperature for 30 min, then transferred to an eppendorf vial and evaporated on a Speedvac. Resin was washed twice with THF and the wash solutions were combined, concentrated and analyzed by HPLC and NMR. The NMR spectrum of this compound (5-(N-acetyl-N-benzylamino)-4-hydroxy-3-(2-iodophenoxy)- cyclohex-1-enecarboxylic acid (2-hydroxyethyl)-amide, 8c) gives two sets of proton signals which result from hindered rotation about the carbon-nitrogen bond; mass spec was consistent with product. Scheme 7.
Figure imgf000086_0001
a) trifluoromethanesulfonic acid, then 2,6-lutidene, CH2Cl2, then 7V-Fmoc-ethanolamine; b) piperidine/DMF; c) 2, PyBOP, DIPEA/ΝMP/CH2C12; d) iodophenol, PPh3, DIAD/THF; e) 7V-benzylamine, MgBr2 OEt2/CH3CN; f) CH3COOH, PyBrOP, DIPEA/NMP; g) HF pyridine/pyridine/THF then TMSOEt; (h) Heck cyclization
LJ? = polystyrene macrobeads [00213] Solid phase quality check after the acylation step (pre-Heck library)
Figure imgf000087_0001
Y = expected molecular ion detected; N/A = unable to determine; N = no possible expected molecular ion peaks observed
[00214] Solid phase quality check after the Heck reaction (post-Heck library)
Figure imgf000088_0001
Y = expected molecular ion detected; N/A = unable to determine; N = no possible expected molecular ion peaks observed
Solid Phase Synthesis and characterization of compounds 1-2 to 1-7
[00215] Mitsunobu reaction on resin 4 (loading level 0.74 mmol/g) with 4-(tert- butyldimethylsilyloxy)methyl-6-methoxy-2-iodophenol was run as described above.
Cleavage of 30 mg of resin product (rinsing method A) produced 9.4 mg of (lS,5S,6S)-N-(2- hydroxyethyl)-5-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)-7-oxa- bicyclo[4.1.0]hept-3-ene-3-carboxamide (92%): 1H NMR (SOO MHZ1 CDCI3): δ 7.29 (S,
IH), 6.94 (s, IH), 6.32 (bs, IH), 6.22-6.24 (m, IH), 5.06 (bs, IH), 4.65 (s, 2H), 3.87 (s, 3H),
3.70 (t, J= 5.5 Hz, 2H), 3.64 (m, IH), 3.53 (m, IH), 3.41 (t, J= 5.5 Hz, 2H), 3.02 (d, J= 19.5 Hz, IH), 2.82 (dd, J= 19.5 Hz, 2.0 Hz, IH); ESI-MS (+) m/z 462 (M+H)+. [00216] The resin was divided into 20 mg units and solid-phase synthesis and cleavage after the final step was performed as described above with the respective amines and acylating agents to make compounds 1-2 to 1-7, each of which was purified by column chromatography on silica gel and analyzed by LC/MS. NMR spectra of the products was complicated due to the presence of rotamers.
[00217] (35',45',5Λ)-5-(iV-(2-(lH-indol-2-yl)ethyl)butyramido)-4-hydroxy-iV-
(2-hydroxyethyl)-3-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)cyclohex-l- enecarboxamide (1-2): yellow foam, 7.5 mg (88%); 1H NMR (500 MHz, CD3OD) δ 7.76 (d, J = 8.0 Hz, 0.5H), 7.60 (d, J = 8.0 Hz, 0.5H), 7.34-7.37 (m, 2H), 7.01-7.17 (m, 4H), 6.47- 6.49 (m, IH), 5.05-5.10 (m, IH), 4.61 (s, IH), 4.54 (s, IH), 4.06^.19 (m, IH), 3.90 (s, 1.5H), 3.89 (s, 1.5H), 3.60-3.64 (m, 4H), 3.13-3.18 (m, 2H), 2.46-2.53 (m, 3H), 1.96-1.98 (m, 2H), 1.85-1.87 (m, 2H), 1.06 (t, J= 7.5 Hz, 1.5H), 1.02 (t, J= 7.5 Hz, 1.5H); LC/MS: tR = 3.00 min, >95%, ESI-MS (+) m/z 692 (M+H)+.
[00218] 7V-(Cyclohexylmethyl)-iV-((lJR,55',65)-6-hydroxy-3-(2- hydroxyethylcarbamoyl)-5-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)cyclohex- 3-enyl)-4-(trifluoromethyl)benzamide (1-3): yellow foam, 8.7 mg (93%); 1H NMR (500 MHz, CD3OD) δ 8.01 (d, J = 8.0 Hz, 0.5 H), 7.78 (d, J = 8.0 Hz, 2 H), 7.75 (d, J = 8.0 Hz, 0.5 H), 7.63 (d, J= 8.0 Hz, 1 H), 7.40 (s, 0.5 H), 7.36 (s, 0.5 H), 7.07 (s, 0.5 H), 7.01 (s, 0.5 H), 6.49 (s, 0.5 H), 6.27 (s, 0.5 H), 5.10 (br m, 0.5 H), 4.70 (br m, 0.5 H), 4.55 (s, IH), 4.52 (s, IH), 4.15^.19 (m, IH), 3.92 (s, 1.5 H), 3.82 (s, 1.5H), 3.52-3.73 (m, 4H), 3.20 (m, IH), 3.01-2.97 (m, IH), 2.61-2.82 (m, 2 H), 1.59-2.03 (m, 6 H), 1.02-1.41 (m, 5 H), 0.61-0.65 (m, 1 H); LC/MS: tR = 3.67 min, 90%, ESI-MS (+) m/z 747 (M+H)+. [00219] 7V-(Cyclohexylmethyl)-iV-((lJR,55',65)-6-hydroxy-3-(2- hydroxyethylcarbamoyl)-5-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)cyclohex- 3-enyl)-2-oxo-2H-chromene-3-carboxamide (1-4): yellow foam, 7.9 mg (81%), 1H NMR (500 MHz, CD3OD) δ 8.13 (s, 0.5H), 8.09 (s, 0.5H), 7.65-7.75 (m, 2H), 7.35-7.46 (m, 3H), 7.07 (s, 0.5H), 6.99 (s, 0.5H), 6.53 (s, 0.5H), 6.32 (s, 0.5H), 5.15- 5.20 (m, IH), 4.61 (s, IH), 4.51 (s, IH), 4.16-4.19 (m, IH), 3.92 (s, 1.5H), 3.76 (s, 1.5H), 3.60-3.64 (m, 4H), 2.96- 3.04 (m, IH), 2.67-2.70 (m, 2H), 1.59-2.06 (m, 8H), 1.21-1.36 (m, 4H), 1.05-1.14 (m, IH); LC/MS: tR = 3.32 min, 92%, ESI-MS (+) m/z IM (M+H)+. [00220] 7V-Cycloheptyl-iV-((lJR,5S,6S)-6-hydroxy-3-(2- hydroxyethylcarbamoyl)-5-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)cyclohex- 3-enyl)-2-oxo-2H-chromene-3-carboxamide (1-5): yellow foam, 7.1 mg (74%); 1H NMR (500 MHz, CD3OD) δ 7.77-7.65 (m, 3H), 7.35-7.50 (m, 3H), 7.07 (s, 0.5H), 7.00 (s, 0.5H), 6.55 (s, 0.5H), 6.35 (s, 0.5H), 5.21-5.23 (m, IH), 4.55 (s, IH), 4.52 (s, IH), 4.17-4.21 (m, 0.5H), 3.92 (s, 1.5H), 3.85-3.93 (m, 0.5H), 3.89 (s, 1.5H), 3.77-3.79 (m, 2H), 3.71-3.76 (m, 2H), 3.55-3.67 (m, IH), 2.60-2.81 (m, 3H), 1.72-1.91 (m, 4H), 1.56-1.70 (m, 4H), 1.39- 1.52 (m, 2H), 1.24-1.38 (m, 2H); LC/MS: tR = 3.38 min, 85%, ESI-MS (+) m/z IM (M+H)+. [00221] (35',45',5JR)-4-Hydroxy-iV-(2-hydroxyethyl)-3-(4-(hydroxymethyl)-2- iodo-6-methoxyphenoxy)-5-((£)-iV-(((/?)-tetrahydrofuran-2-yl)methyl)pent-2- enamido)cyclohex-l-enecarboxamide (1-6): yellow foam, 6.6 mg (75%); 1H NMR (500 MHz, CD3OD) δ 7.78 (s, IH), 7.05 (s, IH), 6.81-6.84 (m, IH), 6.49 (s, 2H), 6.43 (d, J= 7.0 Hz, IH), 5.06-5.07 (m, 0.5H), 5.00-5.01 (m, 0.5H), 4.54 (s, 2H), 4.17^.24 (m, 2H), 3.89 (s, 3H), 3.77-3.81 (m, IH), 3.60-3.63 (m, 2H), 3.35-3.38 (m, 2H), 2.85-2.93 (m, IH), 2.55- 2.63 (m, 2H), 2.22-2.30 (m, 2H), 2.08-2.10 (m, 2H), 1.88-2.00 (m, 2H), 1.55-1.67 (m, 2H), 1.10 (t, J= 7.5 Hz, 1.5H), 1.07 (t, J= 7.5 Hz, 1.5 H); LC/MS: tR = 2.91 min, 92%, ESI-MS (+) m/z 645 (M+H)+.
[00222] 7V-(4-Fluorobenzyl)-iV-((lJR,5S,6S)-6-hydroxy-3-(2- hydroxyethylcarbamoyl)-5-(4-(hydroxymethyl)-2-iodo-6-methoxyphenoxy)cyclohex- 3-enyl)-tetrahydrofuran-2-carboxamide (1-7): yellow foam, 8.6 mg (91%); 1H NMR (500 MHz, CD3OD) δ 7.35-7.45 (m, 3H), 7.29-7.34 (m, 3H), 6.38-6.55 (m, IH), 5.06-5.10 (m, IH), 4.54 (s, 2H), 4.18-4.36 (m, 2H), 3.96-4.04 (m, 2H), 3.84-3.94 (m, 2H), 3.90 (s, 1.5H), 3.89 (s, 1.5H), 3.55-3.61 (m, 2H), 2.15-2.55 (m, 3H), 1.85-2.12 (m, 6H); LC/MS: tR = 2.84 min, 85%, ESI-MS (+) m/z 685 (M+H)+. Compounds used for both solid and solution phase chemistry
[00223] (15',5/?,6/?)-Methyl 5-hydroxy-7-oxa-bicyclo[4.1.0]hept-3-ene-3- carboxylate (9). Compound 9 was prepared from (-)-shikimic acid (1) according to the literature procedure: McGowan, D. A.; Berchtold, G. A. J. Org. Chem. 1981, 46, 2381-
2383.
[00224] (15',5/?,6/?)-5-hydroxy-7-oxa-bicyclo[4.1.0]hept-3-ene-3-carboxylic acid
(2). To a flask was added 10 mL of THF and 9 (0.50 g, 2.9 mmol) and the solution was cooled to -20 °C before the slow addition Of LiOH H2O (0.14 g, 3.5 mmol) in 0.2 mL Of H2O. Once addition was complete, the reaction mixture was stirred at 0 C for 7 h. The pH of the reaction mixture was adjusted to ca. 5 by adding acidic resin Amberlite IRC-86 before filtration and evaporation to provide white solid product 2 (0.26 g, 57%): 1H NMR (CD3OD) δ 6.54 - 6.56 (m, IH), 4.53 - 4.54 (m, IH), 3.43 - 3.45 (m, IH), 3.40- 3.42 (m, IH), 2.89 (d, J = 19.5 Hz, IH), 2.41 (dd, J = 19.5, 2.5 Hz, IH); 13C NMR (CD3OD) δ ; 175.23, 133.78, 131.79, 66.98, 56.22, 53.12, 26.79; ESI-MS (+) m/z 157 (M+H)+
Solution phase Synthesis and Characterization of Compound 1-1
[00225] (15,5S,6S)-Methyl 5-(2-iodophenoxy)-7-oxa-bicyclo [4.1.0] hept-3-ene-
3-carboxylate (10). To an oven-dried flask was added 9 (8.40 g, 2.13 mmol), o-iodophenol (5.63 g, 25.6 mmol), Ph3P (8.73 g, 33.3 mol) and anhydrous THF (200 mL). The reaction mixture was cooled to -78 C and DEAD (4.04 mL, 25.6 mmol) was added slowly via syringe. The reaction mixture was stirred 1 hr at -78 C and then kept at -20 C overnight. The reaction mixture was concentrated and purified by column chromatography (hexane/Et2O from 5:3 to 3: 1) to afford 7.15 g of 10 as semi-solid (90%): 1H NMR (CD3Cl) δ 7.82 (dd, J=
8.0 , 1.5 Hz, IH), 7.33 (dt, J= 8.0, 1.5 Hz, IH), 7.03 (d, J= 8.0 Hz, IH), 6.92 (m, IH), 6.79 (dt, J = 8.0 , 1.5 Hz, IH), 5.19 (s, IH), 3.78 (s, 3H), 3.55 (d, J = 3.0 Hz, IH), 3.54 (s, IH),
3.01 (dd, J = 20.0, 1.5 Hz, IH), 2.83 ( dd, J = 20 .0, 3.0 Hz); 13C NMR (CD3Cl) δ 166.7, 156.5, 140.1, 130.5, 129.9, 128.9, 124.3, 114.9, 88.4, 70.9, 52.4, 51.1, 50.9, 24.6; ESI-MS (+) m/z 370 (M-H)+ .
[00226] (15',55',65')-5-(2-Iodophenoxy)-7-oxa-bicyclo[4.1.0]hept-3-ene-3- carboxylic acid (11). To an oven-dried flask was added 50 mL of THF and 10 (6.11 g, 16.4 mmol) and the solution was cooled to 0 C before the slow addition Of LiOH-H2O (0.83 g, 20 mmol) in 10 mL H2O. The reaction mixture warmed to room temperature once addition was complete and the reaction was stirred 5 h. The pH of the reaction mixture was adjusted to ca. 5 by adding acidic resin Amberlite IRC-86 before filtration and evaporation to provide 4.98 g of 11 as white solid (85%): 1H NMR (CD3OD) δ 7.79 (dd, J= 8.0, 1.5 Hz, IH), 7.37 (dt, J = 8.5, 1.0 Hz, IH), 7.19 (d, J= 8.5 Hz, IH), 6.89-6.87 (d, J= 1.5 Hz, IH), 6.80 (dt, J= 8.0, 1.0 Hz, IH), 5.28-5.27 (d, J = 1.5 Hz, IH), 3.54 (s, IH), 3.47 (d, J = 1.0 Hz, IH), 2.93 (dd, J = 20.0, 1.5 Hz, IH), 2.76 (ddd, J = 20.0, 1.5, 4.0 Hz, IH); 13C NMR (CD3OD) δ 169.54, 158.08, 141.09, 131.58, 131.04, 130.58, 125.17, 116.38, 88.87, 72.10, 52.21, 51.83, 25.59; ESI-MS (+) m/z 359 (M+H)+.
[00227] (15',55',65')-iV-(2-(terr-Butyldimethylsilyloxy)ethyl)-5-(2-iodophenoxy)-
7-oxa-bicyclo[4.1.0]hept-3-ene-3-carboxamide (12). To an oven-dried flask was added 12 (5.87 g, 16.4 mmol), EDAC (3.45 g, 18.0 mmol), HOBt (2.43 g, 18.0 mmol) and CH2Cl2 (150 mL) and the mixture was cooled to 0 C. 2-(tert-Butyldimethylsilyloxy)ethylamine (4.31 g, 24.6 mmol, prepared per Parsons and Pettifer, J Chem. Soc, Perkin Trans. (1998) 1 :651) was added and the mixture was stirred overnight. The reaction was filtrated and the filtrate was washed with brine twice, dried (Na2SO4), filtered and concentrated. The residue was purified by column chromatography (EtOAc/hexane= 3/7) to afford 12 (6.72 g, 80%). 1H NMR (CDCl3) δ 7.80 (d, J= 8.0 Hz, IH), 7.31 (dt, J= 8.0, 1.5 Hz, IH), 7.01 (d, J= 8.0 Hz, IH), 6.79 (dt, J= 8.0, 1.5 Hz, 1 H), 6.35 (br, IH), 6.20 (br, IH), 5.15 (br, IH), 3.70 (t, J = 4.5 Hz, 2H), 3.54 (s, IH), 3.52 (s, IH), 3.41-3.44 (m, 2H), 2.98 (d, J = 19.5 Hz, IH), 2.85 (dd, J = 19.5, 2.0 Hz, IH); 0.88 (s, 9H), 0.05 (s, 6H); 13C NMR (CDCl3) δ 167.13, 156.27, 139.75, 132.98, 129.62, 123.96, 123.53, 114.84, 88.25, 70.79, 61.42, 50.91, 50.55, 41.76, 25.83, 24.53, 18.12, -5.36; ESI-MS (+) m/z 516 (M+H)+.
[00228] (35',45',5Λ)-iV-(2-(terr-Butyldimethylsilyloxy)ethyl)^-hydroxy-3-(2- iodophenoxy)-5-(2-methoxyethylamino)cyclohex-l-enecarboxamide (1-27). To an oven-dried flask was added 12 (0.12 g, 0.23 mmol), MgBr2-OEt2 (0.66 g, 0.25 mmol), and CH3CN (2 mL). To the above suspension was added 2-methoxyethylamine (40 μL, 0.47 mmol) and the reaction mixture was heated at 60 C for 8 h until the reaction was completed. The solvent was evaporated and the residue was treated with CH2Cl2 and aqueous NH4Cl. The organic layer was washed with brine, dried over Na2SO4. After evaporation of the solvent, the residue was purified by column chromatography (DCM and DCM/MeOH = 60: 1) to afford (1-27) (0.12 g, Yield 88%) as white solid. 1H NMR (CDCl3) δ 7.73 (dd, J= 8.0, 1.5 Hz, IH), 7.25 (dt, J = 8.0, 1.5 Hz, IH), 6.96 (d, J = 8.0 Hz, IH), 6.71 (dt, J = 8.0, 1.5 Hz, IH), 6.30 (br, IH), 6.22 (t, J = 5.0 Hz, IH), 4.80^.82 (m, IH), 3.85 (dd, J = 10.5, 8.0 Hz, IH), 3.65 (t, J= 5.0 Hz, 2H), 3.46-3.49 (m, 2H), 3.35-3.38 (m, 2H), 3.31 (s, 3H), 2.80-2.94 (m, 3H), 2.70-2.74 (m, IH), 0.82 (s, 9H), 0.01 (s, 6H); 13C NMR (CDCl3) δ 167.00, 156.93, 139.72, 135.03, 129.70, 127.83, 123.60, 115.00, 88.48, 81.16, 73.68, 72.14, 61.68, 58.91, 56.86, 46.21, 41.93, 31.04, 26.00, 18.33, -5.19, -5.20; ESI-MS (+) m/z 591 (M+H)+. [00229] (35',45',5/?)-iV-(2-(terr-Butyldimethylsilyloxy)ethyl)^-hydroxy-3-(2- iodophenoxy)-5-(7V-(2-methoxyethyl)pentanamido)cyclohex-l-enecarboxamide (1-32). To an oven-dried flask was added (1-27) (35 mg, 59 μmol), CH2Cl2 (2 mL), DIPEA (16 μL, 89 μmol) followed by the addition of pentanoyl chloride (86 μL, 71 μmol). The reaction mixture was stirred overnight. The solvent was evaporated and the residue was treated with CH2Cl2 and aqueous NH4Cl. The organic layer was washed with brine, dried (Na2SO4), concentrated and purified by column chromatography (CH2Cl2 and CH2Cl2 /MeOH = 100:0.5) to afford (1-32) (36 mg, 90%): 1H NMR (CD3Cl) δ 7.78 (dd, J = 8.0, 1.5 Hz, IH), 7.31 (dt, J = 8.0, 1.5 Hz, IH), 7.14 (dd, J = 8.0, 1.0 Hz, IH), s), 6.77 (dt, J = 8.0, 1.0 Hz, 1 H), 6.33 (br, IH), 6.20 (t, J= 5.5 Hz, IH), 5.23 (d, J= 3.0 Hz, IH, OH), 4.94 - 4.92 (m, IH), 4.11- 4.04 (m, 2H), 3.98 - 3.93 (m, IH), 3.77 (d, J = 14.0 Hz, IH), 3.71 - 3.67 (m, 2H), 3.47- 3.40 (m, 3H), 3.39 (s, 3H), 3.14 - 3.09 (m, IH), 2.61 (dd, J= 17.5, 5.5 Hz, IH), 2.52 - 2.43 (m, 2H) , 2.39 - 2.30 (m, IH), 1.67 - 1.61 (m, 2H), 1.39 - 1.34 (m, 2H), 0.94 (t, J= 6.5 Hz, 3H), 0.86 (s, 9H), 0.05 (s, 6H); 13C NMR (CD3Cl) δ 174.95, 166.60, 157.71, 139.67, 134.66, 129.81, 128.43, 124.08, 116.38, 89.14, 81.62, 70.83, 70.48, 61.72, 59.10, 58.21, 42.37, 42.02, 33.82, 29.03, 27.32, 26.07, 22.65, 18.42, 14.15, - 5.11, -5.13; ESI-MS (+) m/z 675 (M+H)+.
[00230] (35',45',5Λ)-4-Hydroxy-iV-(2-hydroxyethyl)-3-(2-iodophenoxy)-5-(iV-
(2-methoxyethyl)pentanamido)cyclohex-l-enecarboxamide (1-1). Compound (1-32) (15 mg) was dissolved in THF (1.25 mL) and pyridine (0.150 mL) was added. HF-pyridine (75 μL) was added, and the mixture was shaken. After 3 h, TMSOEt (0.75 mL) was added and the mixture was shaken 0.5 h. The reaction mixture was put in the SpeedVac and concentrated. The residue was purified by column chromatograph (CH2Cl2 then CH2Cl2 /MeOH = 95:5) to afford compound (1-1) (11 mg, 86%): 1H NMR (CDCl3) δ 7.79 (dd, J = 8.0, 1.5 Hz, IH), ), 7.31 (d, J = 8.0 Hz, IH), 7.14 (d, J = 8.0 Hz, IH), 7.78 (dt, J = 8.0, 1.5 Hz, IH), 6.40 - 6. 37 (br, IH), 6.26 (t, J= 5.5 Hz, IH), 5.20 (d, J= 3.0 Hz, IH, OH), 4.94 - 4.90 (b, IH), 4.11 - 4.04 (m, 2H), 3.98 - 3.93 (m, IH), 3.80 - 3.74 (m, 3H), 3.52 -3.42 (m, 4H), 3.39 (s, 3H), 3.14 - 3.09 (m, IH), 2.62 (dd, J= 17.5, 5.5 Hz, IH), 2.52 - 2.46 9m, 2H), 2.38 - 2. 30 (m, IH), 1.68 - 1.58 (m, 2H), 1.40 - 1.34 (m, 2H), 0.93 (t, J= 7.5 Hz, 3H); ESI- MS (+) m/z 561 (M+H)+. Scheme 8.
Figure imgf000094_0001
a) o-iodophenol, DEAD, Ph3P/THF, -78 0C to -20 0C, 90%, b) LiOH H2O, H20/THF, 85%, c) TBDMSOCH2CH2NH2, EDAC, HOBt, PyBOP, DIPEA/CH2C12, 80%, d) 2-methoxyethylamme, MgBr2 OEt2/ CH3CN, 60 0C, 88%, e) pentanoyl chloride, DIPEA/CH2C12, 90%, f) HF pyπdine/pyπdme/THF= 5 10 85, 3 h then TMSOEt, 0 5 h, 86%
Solution phase Synthesis and Characterization of Compound 1-35
[00231] (3S,4S,5R)-N-(2-(ter*-butyldimethylsilyloxy)ethyl)-4-hydroxy-3-(2- iodophenoxy)-5-(3-methoxyphenethylamino)cyclohex-l-enecarboxamide (1-36). To an oven-dried flask was added 12 (0.91 g, 1.8 mmol), MgBr2OEt2 (0.50 g, 1.9 mmol), and CH3CN (7 mL). To the suspension was added 2-(3-methoxyphenyl)ethylamine (0.51 mL, 3.5 mmol) and the reaction mixture was heated at 6O0C for 6 h. The solvent was evaporated and the residue was partitioned between CH2Cl2 and aqueous NH4Cl. The organic layer was washed with brine, dried (Na2SO4), and filtered. After evaporation of the solvent, the residue was purified by column chromatography (CH2CI2 and CH2CI2:Me0H = 60: 1) to afford (1-36) as white solid (1.05 g, 90%): 1H NMR (CDCI3) δ 7.79 (d, J= 8.0, IH), 7.30 (d, J= 8.0, IH), 7.23 (t, J = 8.0 Hz, IH), 6.99 (d, J = 7.5 Hz, IH), 6.83 (d, J = 7.5 Hz, IH), 6.79-6.75 (m, 3H), 6.34 (bs, IH), 6.18 (t, J= 5 Hz, IH), 4.86-4.85 (m, IH), 3.96 (m, IH), 3.82 (s, 3H), 3.70 (t, J= 5.0 Hz, 2H), 3.42 (q, J= 5.0 Hz, 2H), 3.16 (m, IH), 3.03 (d, J= 6.0 Hz, IH), 3.00 (t, J = 5.0 Hz, IH), 2.97-2.89 (m, 4H), 2.28-2.18 (br, IH), 0.86 (s, 9H), 0.05 (s, 6H); 13C NMR (CD3Cl) δ 166.81, 159.70, 156.67, 141.16, 139.63, 134.77, 129.61, 129.51, 127.72, 123.44, 121.04, 114.45, 111.63, 88.33, 80.75, 73.27, 61.53, 56.63, 55.16, 47.89, 41.81, 36.49, 30.79, 25.90, 18.21,-5.27; ESI-MS (+) m/z 667 (M+H)+. [00232] N-((l/?,5S;6S)-3-2(2-(tert-butyldimethylsilyloxy)ethylcarbamoyl)-6- hydroxy-5-(2-iodophenoxy)cyclohex-3-enyl)-iV-(3-methoxyphenethyl)-2-oxo-2H- chromene-3-carboxamide (1-37). To an oven-dried flask was added (1-36) (0.14 g, 0.21 mmol), PyBOP (0.1 Ig, 0.23 mmol), coumarin-3-carboxylic acid (0.44 g, 0.23 mmol) and CH2CZ2 (5 mL) followed by the addition of DIPEA (86 μL, 0.63 mmol). The reaction mixture was stirred overnight at room temperature. The solvent was evaporated and the residue was partitioned between CH2CZ2 and aqueous NH4CZ. The organic layer was washed with brine, dried (Na2SO4), and filtered. After concentration, the residue was purified by column chromatography (CH2CZ2 and CH2CZ2MeOH = 100:0.5) to afford (1-37) as white solid (0.15 g, 82%): 1H NMR (CDCZ3) two rotamers) δ 8.04 (s), 7.78 (dd, J = 8.0, 1.5 Hz), 7.63-7.54 (m), 7.40-7.22 (m), 7.17 (t, J= 8.0 Hz), 7.12 (d, J= 8.0 Hz), 6.95 (d, J= 7.5 Hz), 6.93 (s), 6.82 (dd, J= 8.5, 2.5 Hz), 6.81-6.72 (m), 6.69 (d, J= 7.0 Hz), 6.56 (s), 6.36 (s), 6.28 (t, J = 5.5 Hz), 4.87^.89 (br), 4.64^.68 (br), 4.06^.12 (m), 3.9-3.5 (m), 3.46 (q, J = 5.5 Hz), 3.42-3.34 (m), 3.24-3.10 (m), 3.06-2.98 (br), 2.90-2.82 (m), 2.60 (dd, J = 5.5, 17.5 Hz), 2.40-2.26 (br), 2.10-1.90 (m), 1.89-1.79 (br), 1.35-1.15 (m), 0.88 (s), 0.86 (s), 0.07 (s), 0.06 (s), 0.05 (s), 0.04 (s); ESI-MS (+) m/z 839 (M+H)+.
[00233] Synthesis of Compound (11-37). To an oven-dried flask was added (1-37)
(45 mg, 54 mmol), Pd(OAc)2, l,2-bis(dicyclohexylphosphino)ethane (dcpe, 11 mg, 27 mmol), Bu4NOAc (8 mg, 27 mmol) and dioxane (1 mL). After degassing for 10 min with Ar, DIPEA (19 μL, 0.11 mmol) was added and the reaction was heated at 5O0C for 2 h. The reaction mixture was concentrated and purified by column chromatography (CH2Cl2 and CH2CL2MeOH =100:0.5) to afford (11-37) as yellow solid (35 mg, 91%):^ (CDCl3) (two rotamers): 8.05 (s), 7.98 (s), 7.62-7.54 (m), 7.40- 7.32 (m), 7.21 (t, J= 7.0 Hz), 7.17-7.09 (m), 6.90-6.70 (m), 6.63 (d, J= 7.5 Hz), 6.54 (s), 638 (s), 5.95 (br), 5.84 (s), 4.94 (t, J= 9.0 Hz), 4.89 (s), 4.87 (s), 4.72 (d, J= 9.0 Hz), 4.63 (t, J= 9.0 Hz), 4.444.36 (br), 4.20 (d, J= 9.0 Hz), 3.82-3.78 (m), 3.77 (s), 3.68-3.60 (m), 3.59 (s), 3.59-3.55 (m), 3.45-3.22 (m), 3.20- 3.18 (m), 2.90-2.80 (m), 2.01-1.95 (m), 1,62-1.60 (m),1.42-1.40(m), 1.35-1.26 (br), 1.02- 1.00 (m), 0.89 (s), 0.88 (s), 0.10 (s), 0.09 (s); ESI-MS (+) m/z 111 (M+H)+. [00234] Synthesis of Compound (11-35). Compound (11-37) (25 mg) was dissolved in THF (1.3 mL) in a Teflon vial. Pyridine (0.15 mL) and HF pyridine (75 μL) were added to the solution, the vial was capped shaken for 3 h. After the starting material was disappeared (3 h), TMSOEt (0.75 mL) was added and shaken for 0.5 h. The reaction mixture was put in the SpeedVac and the solvent was evaporated. The residue was purified by column chromatograph (CH2Cl2 and then CH2Ci /MeOH = 95:5) to afford compound (11-35) (18 mg, 86%): 1H NMR (CD3OD) two rotamers: 8.42-8.38 (m), 8.16 (bs), 8.14 (bs), 7.78-7.67 (m), 7.62-7.59 (m), 7.43-738 (m), 7.1.7-7.06 (m), 6.87-6.83 (m), 6.74-6.69 (m), 6.59 (bs), 6.43 (bs), 6.23 (bs), 4,94^.87 (m), 4.82 (d., J= 8.5 Hz), 4.75 (t, J= 9.0 Hz), 4.68 (d,.J= 9.0 Hz), 4.49 (d, J= 8.5 Hz), 3.69 (s), 3.56 (s), 3.84-3.40 (m), 335-3.20 (in), 2.92-2.72 (m), 2.02- 1.90 (m), 1.70-1.60 (m), 1.46-1.20 (m), 1.06-1.02 (m), 0.94-0.89 (an); ESI-MS (+) m/z 597 (M+H)+.
Scheme 9.
Figure imgf000096_0001
(1-37) (11-37) (11-35)
(a) amine addition, (b) acylation, (c) Heck coupling, (d) deprotection
Other Embodiments
[00235] The foregoing has been a description of certain non-limiting preferred embodiments of the invention. Those of ordinary skill in the art will appreciate that various changes and modifications to this description may be made without departing from the spirit or scope of the present invention, as defined in the following claims.

Claims

ClaimsWhat is claimed is:
1. A compound of the formulae (I):
Figure imgf000098_0001
(I) or a pharmaceutically acceptable form thereof; wherein:
Ri is hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORA; -CN; -SCN; -SRA; or -N(RA)2; wherein each occurrence of RA is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety;
R2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
R3 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)RB; -CO2RB; -SORB; or -SO2RB; wherein each occurrence of RB is independently a halogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety;
R4 is hydrogen; a hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl; and
R5 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)Rc; -CChRc; -SORc; or -SChRc; wherein each occurrence of Rc is independently a hydrogen, a halogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety.
2. The compound of claim 1, or a pharmaceutically acceptable form thereof, having any one of the formulae:
Figure imgf000099_0001
wherein X is halogen; n is O, 1, 2, 3 or 4; and each occurrence Of R6 is independently hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORD; -C(=0)RD; -CO2RD; -CN; -SCN; -SRD; -NO2; -N(RD)2; or -NHC(O)R0; wherein each occurrence of RF is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety.
3. The compound according to claim 2, or a pharmaceutically acceptable form thereof, having any one of the formulae:
Figure imgf000100_0001
Figure imgf000100_0002
Figure imgf000100_0003
Figure imgf000101_0001
4. The compound of claim 1, wherein Ri is -NHRA.
5. The compound of claim 1, wherein R2 is substituted or unsubstituted, branched or unbranched aliphatic or substituted or unsubstituted, branched or unbranched heteroaliphatic.
6. The compound of claim 4, wherein R2 corresponds to any of the following groups:
Figure imgf000101_0002
7. The compound of claim 1, wherein R3 is -C(=O)Rβ.
8. The compound of claim 6, wherein R3 corresponds to any of the following groups:
Figure imgf000102_0001
9. The compound of claim 1, wherein R4 is hydrogen or an hydroxyl protecting group.
10. The compound of claim 1, wherein R5 is a substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
11. The compound of claim 1, wherein R5 corresponds to the formulae:
Figure imgf000102_0002
wherein X is halogen or -S(O)2Ri, wherein Ri is a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety.
12. The compound of claim 10, wherein n is 0 or 2.
13. The compound of claim 1, wherein said compound, or a pharmaceutically acceptable form thereof, is selected from the group consisting of:
Figure imgf000103_0001
Figure imgf000103_0002
Figure imgf000104_0001
Figure imgf000104_0002
Figure imgf000104_0003
Figure imgf000105_0001
Figure imgf000105_0002
Figure imgf000106_0001
Figure imgf000106_0002
Figure imgf000106_0003
Figure imgf000106_0004
Figure imgf000106_0005
Figure imgf000107_0001
Figure imgf000107_0002
14. A compound of the formula (II):
Figure imgf000107_0003
(H) or a pharmaceutically acceptable form thereof; wherein:
Ri is hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORA; -CN; -SCN; -SRA; or -N(RA)2; wherein each occurrence of RA is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; or a substituted or unsubstituted heteroaryl moiety;
R2 is hydrogen; an amino protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl;
R3 is hydrogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -C(=O)RB; -CO2RB; -SORB; or -SO2RB; wherein each occurrence of RB is independently a halogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety;
R4 is hydrogen; a hydroxyl protecting group; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; or substituted or unsubstituted, branched or unbranched heteroaryl; n is 0, 1, 2, 3 or 4; and each occurrence of Re is independently hydrogen; halogen; cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic; cyclic or acyclic, substituted or unsubstituted, branched or unbranched heteroaliphatic; substituted or unsubstituted, branched or unbranched acyl; substituted or unsubstituted, branched or unbranched aryl; substituted or unsubstituted, branched or unbranched heteroaryl; -ORD; -C(=O)RD; -CO2RD; -CN; -SCN; -SRD; -NO2; -N(RD)2; or -NHC(O)R0; wherein each occurrence of RF is independently a hydrogen, a protecting group, a substituted or unsubstituted aliphatic moiety, a substituted or unsubstituted heteroaliphatic moiety, an acyl moiety; a substituted or unsubstituted aryl moiety; a substituted or unsubstituted heteroaryl moiety; substituted or unsubstituted alkoxy; substituted or unsubstituted aryloxy; substituted or unsubstituted alkylthio; substituted or unsubstituted arylthio; substituted or unsubstituted amino, substituted or unsubstituted alkylamino, substituted or unsubstituted dialkylamino, substituted or unsubstituted heteroaryloxy; or substituted or unsubstituted heteroarylthio moiety.
15. The compound of claim 14, or a pharmaceutically acceptable form thereof, having any one of the formulae:
Figure imgf000109_0001
Figure imgf000109_0002
16. The compound of claim 14, wherein Ri is -NHRA.
17. The compound of claim 14, wherein R2 is substituted or unsubstituted, branched or unbranched aliphatic or substituted or unsubstituted, branched or unbranched heteroaliphatic.
18. The compound of claim 17, wherein R2 corresponds to any of the following groups:
Figure imgf000110_0001
19. The compound of claim 14, wherein R3 is -C(=O)Rβ.
20. The compound of claim 19, wherein R3 corresponds to any of the following groups:
Figure imgf000110_0002
Figure imgf000111_0001
21. The compound of claim 14, wherein R4 is hydrogen or an hydroxyl protecting group.
22. The compound of claim 14, wherein n is 0 or 2.
23. The compound of claim 1, wherein said compound, or a pharmaceutically acceptable form thereof, is selected from the group consisting of:
Figure imgf000111_0002
Figure imgf000111_0003
Figure imgf000112_0001
Figure imgf000112_0002
Figure imgf000112_0003
Figure imgf000112_0004
Figure imgf000113_0001
Figure imgf000113_0002
Figure imgf000113_0003
Figure imgf000113_0004
Figure imgf000114_0001
Figure imgf000114_0002
Figure imgf000114_0003
Figure imgf000114_0004
Figure imgf000115_0001
Figure imgf000115_0002
Figure imgf000115_0003
Figure imgf000116_0001
24. A pharmaceutical composition comprising a compound of any one of claims 1 to 13 or any one of claims 14 to 23, or a pharmaceutically acceptable form thereof, and a pharmaceutically acceptable excipient.
25. A method of treating a proliferative disease, said method comprising the steps of administering a therapeutically effective amount of a compound according to any one of claims 1 to 13 or any one of claims 14 to 23, or a pharmaceutically acceptable form thereof, to a subject in need thereof.
26. A method of detecting proliferating cells in a biological sample, comprising the steps of: (i) contacting a biological sample with a compound according to any one of claims 1 to 13 or any one of claims 14 to 23 conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
27. A method of detecting pre-cancerous cells in a biological sample, comprising the steps of: (i) contacting a biological sample with a compound according to any one of claims 1 to 13 or any one of claims 14 to 23 conjugated to an imaging agent, and (ii) screening the sample for abnormal numbers of centrioles or centrosomes.
28. A method of identifying compounds which bind to Aurora kinase, comprising the steps of: (i) contacting a cell comprising Aurora A kinase activity with a test compound according to any one of claims 1 to 13 or any one of claims 14 to 23 conjugated to an imaging agent and (ii) screening for binding to Aurora-A kinase.
29. The method according to any one of claims 26 to 28, wherein the imaging agent is a fluroescent moiety.
PCT/US2007/077266 2006-08-30 2007-08-30 Shikimic acid-derived compounds for inhibition and detection of aurora a-associated tumors WO2008028057A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84103506P 2006-08-30 2006-08-30
US60/841,035 2006-08-30

Publications (2)

Publication Number Publication Date
WO2008028057A2 true WO2008028057A2 (en) 2008-03-06
WO2008028057A3 WO2008028057A3 (en) 2008-12-18

Family

ID=39136896

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/077266 WO2008028057A2 (en) 2006-08-30 2007-08-30 Shikimic acid-derived compounds for inhibition and detection of aurora a-associated tumors

Country Status (1)

Country Link
WO (1) WO2008028057A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6939986B2 (en) * 1999-06-11 2005-09-06 Hoffmann-La Roche Inc. Process for preparing 1,2-diamino compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6939986B2 (en) * 1999-06-11 2005-09-06 Hoffmann-La Roche Inc. Process for preparing 1,2-diamino compounds

Also Published As

Publication number Publication date
WO2008028057A3 (en) 2008-12-18

Similar Documents

Publication Publication Date Title
JP7448966B2 (en) Proline-locked stapled peptide and its uses
US10017520B2 (en) Myc modulators and uses thereof
US9567301B2 (en) Pyrrol-1-yl benzoic acid derivatives useful as myc inhibitors
US9102697B2 (en) Trioxacarcins and uses thereof
JP2019218363A (en) Stapled and stitched polypeptides and uses thereof
US9023883B2 (en) PRMT1 inhibitors and uses thereof
US20190276426A1 (en) Inhibitors of interleukin-1 receptor-associated kinases and uses thereof
US20090221568A1 (en) Synthesis of Inhibitors of FtsZ
US10844073B2 (en) Palladium-mediated ketolization
US20060287520A1 (en) Synthesis of salinosporamide A and analogues thereof
EP3024327A2 (en) Inhibitors of transcription factors and uses thereof
US11903910B2 (en) Use of metformin and analogs thereof to reduce RAN protein levels in the treatment of neurological disorders
US20110263569A1 (en) Ryanodine channel binders and uses thereof
US20190144466A1 (en) Max binders as myc modulators and uses thereof
WO2008144507A2 (en) Spirooxindole inhibitors of aurora kinase
US20230357320A1 (en) Helical stapled peptides and uses thereof
WO2008028057A2 (en) Shikimic acid-derived compounds for inhibition and detection of aurora a-associated tumors
US20220152036A1 (en) COMPOUNDS FOR USES IN PHARMACOLOGICAL INDUCTION OF HBF FOR TREATMENT OF SICKLE CELL DISEASE AND ß-THALASSEMIA
US20230010562A1 (en) 4,9-dioxo-4,9-dihydronaphtho[2,3-b]furan-3-carboxamide derivatives and uses thereof for treating proliferative diseases and infectious diseases
WO2023196605A1 (en) Inhibiting histone deacetylase 6 (hdac6)
US20230382865A1 (en) Histone demethylase 5 inhibitors and uses thereof

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: 07841631

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07841631

Country of ref document: EP

Kind code of ref document: A2