WO2008021211A2 - Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins - Google Patents

Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins Download PDF

Info

Publication number
WO2008021211A2
WO2008021211A2 PCT/US2007/017752 US2007017752W WO2008021211A2 WO 2008021211 A2 WO2008021211 A2 WO 2008021211A2 US 2007017752 W US2007017752 W US 2007017752W WO 2008021211 A2 WO2008021211 A2 WO 2008021211A2
Authority
WO
WIPO (PCT)
Prior art keywords
bcl
compounds
apoptosis
family member
alkyl
Prior art date
Application number
PCT/US2007/017752
Other languages
French (fr)
Other versions
WO2008021211A3 (en
Inventor
Pamela S. Schwartz
Michael K. Manion
John S. Fry
David M. Hockenbery
Original Assignee
Fred Hutchinson Cancer Research Center
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 Fred Hutchinson Cancer Research Center filed Critical Fred Hutchinson Cancer Research Center
Publication of WO2008021211A2 publication Critical patent/WO2008021211A2/en
Publication of WO2008021211A3 publication Critical patent/WO2008021211A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine

Definitions

  • the present invention relates to compounds for modulating apoptosis in cells over expressing Bcl-2 Family member proteins.
  • the present invention also relates to pharmaceutical compositions containing these compounds and methods of using the compounds.
  • neoplasias may result, at least in part, from an apoptosis- resistant state in which cell proliferation signals inappropriately exceed cell death signals.
  • DNA viruses such as Epstein-Barr virus, African swine fever virus and adenovirus, parasitize the host cellular machinery to drive their own replication and at the same time modulate apoptosis to repress cell death and allow the target cell to reproduce the virus.
  • certain diseases such as lymphoproliferative conditions, cancer (including drug resistant cancer), arthritis, inflammation, autoimmune diseases, and the like, may result from a down regulation of cell death signals. In such diseases, it would be desirable to promote apoptotic mechanisms.
  • Bcl-2 and BCI-X L confers resistance to multiple chemotherapeutic agents, including alkylating agents, antimetabolites, topoisomerase inhibitors, microtubule inhibitors and anti-tumor antibiotics, and may constitute a mechanism of clinical chemoresi stance in certain tumors (Minn et al., Blood 86:1903-1910, 1995; Decaudin et al., Cancer Res. 57:62-67, 1997).
  • Bcl-2/ Bcl-XL-directed therapies using either anti-sense oligonucleotides or novel protein-targeted drugs, can increase cellular sensitivity to standard agents in vitro or, in some cases, kill cells as single agents (Jansen et al., Nat. Med. 4:232- 234, 1998).
  • antimycins Two antimycins, antimycin Ai and A3, have been discovered to inhibit the activity of the anti-apoptotic Bcl-2 family member proteins, Bcl-2 or Bcl-x L . These naturally obtained antimycins are toxic, however, because as discussed above, they also inhibit mitochondrial respiration. Therefore, antimycin derivatives that are effective in inducing apoptosis in cells where apoptosis is inappropriately regulated while exhibiting reduced inhibition of mitochondrial respiration have been identified. It would be desirable to identify other compounds that may be effective in inducing apoptosis in cells where apoptosis is inappropriately regulated while minimally inhibiting mitochondrial respiration. Furthermore, it would be desirable to use the identified compounds as to treat diseases related to the down regulation of apoptosis.
  • the present invention provides methods of treating an apoptosis-associated disease.
  • Embodiments of the present invention provide methods for treating an apoptosis-associated disease (e.g., cancer) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an active compound of Formula I:
  • X and X 1 are each independently O or S;
  • Ri, R 2 , R3, R4, ' Rs, Re, R7, Rs, R9, Rio, Rn, R12, and R13 are each independently selected from the group s consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci- 10 -alkyl, Ci-io-alkenyl, Ci-io-alkoxy.
  • halogen e.g., chloro, fluoro, bromo, iodo
  • Embodiments of the present invention provide methods for treating an apoptosis-associated disease (e.g., cancer) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an active compound of Formula II:
  • Xi is O or S
  • X 2 is C or N
  • Ri, R 2 , R3, R4, R5, Re, R7, Rs, R9, Rio, Rn, R12, and R J3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci- 10 -alkyl, d- 10 -alkenyl, Ci- 1 0-alkoxy, Ci-10-alkylsulfanyl, C ⁇ o haloalkyl, Ca-io-cycloalkyl, Cs-io-cycloalkyl-Ci-io-alkyl, Ca-io-heterocycloalkyl, C 3 .10- heterocycloalkyl-Ci- 10 -alkyI, C 3 - 14 aryl, C ⁇ ;-i 4 -arylalkoxy, C ⁇ - ⁇ -aryloxyalkoxy, Ci-10- carbonyloxy, Ca-io
  • the active compound has the strcutre of:
  • the method for treating an apoptosis-associated disease further comprising concurrently adminsterting to said subject another inducer of apopotosis.
  • Embodiments of the present invention further provide pharmaceutical compositions comprising, consisting of or consisting essentially of
  • X and X 1 are each independently O or S;
  • Embodiments of the present invention further provides pharmaceutical compositions comprising, consisting of, or essentially consisting of an active compound of Formula II:
  • Xi is O or S
  • X 2 is C or N
  • Ri, R 2 , R3, R»J R-5 * ROJ R7, Rs 5 Rs>5 Rio, Ri 1, Ri2, and Rn are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Q.io-alkyl, Cj.io-alkenyl, Ci-io-alkoxy, Ci-1 0 -alkylsulfanyl, Ci- 1 0 -haloalkyl, Cs-io-cycloalkyl, Cs-io-cycloalkyl-Ci-io-alkyl, Ca-io-heterocycloalkyl, C 3 -io- heterocycloalkyl-Ci-io-alkyl, C 3 - 14 aryl, C ⁇ -u-arylalkoxy, C ⁇ - H -aryloxyalkoxy, Ci-io- carbony
  • X and X' are each independently O or S;
  • R12, and Rj 3 are each independently selected from the group s consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci- 1 0-alkyl, Ci-10-alkenyl, Ci.io-alkoxy, Ci-10-alkylsulfanyl, Ci-io-haloalkyl, C3.io-cycloalkyl, Cs-io-cycloalkyl-Ci.io-alkyl, Cs-io-heterocycloalkyl, C3-10- heterocycloalkyl-Ci-io-alkyl, C 3 - 14 aryl, C 6 -i4-arylalkoxy, C ⁇ -u-aryloxyalkoxy, Ci-1 0 - !
  • Xi is O or S
  • X 2 is C or N
  • Ri, R 2 , R 3 , R4, R5, Re, R7, Rs, R9, Rio, Rn, Ri 2, and Ri 3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-io-alkyl, Ci-io-alkenyl, Ci-io-alkoxy, Ci_io-alkylsulfanyl, Cj.
  • halogen e.g., chloro, fluoro, bromo, iodo
  • io-haloalkyl Cs-io-cycloalkyl, C 3 _io-cycloalkyl-Ci-io-aIkyl, Cs-w-heterocycloalkyl, C 3 .
  • Figure 1 depicts a scheme for gain-of- function activity with Bcl-xL inhibitor Sl.
  • A fluorescence emission spectra of ANS, Bcl-xL, and the ANS-Bcl-xL complex, with Sl inhibition of ANS binding.
  • B cell viability after treatment with Sl for 24 h in TAMH-neo and TAMH-Bcl-xL cells. Points, mean of at least three separate experiments with quadruplicate assays; bars, SD.
  • C cell viability for TAMH-Bcl-xL cells treated with 10 mmol/L 2-deoxyglucose for 2 h followed by indicated concentrations of 2-MeAA for. an additional 24 h. Representative of three experiments.
  • the compounds, as described herein, may be substituted with any number of substituents or functional moieties.
  • substituted 1 '.. ; whether preceded 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>- i-i • radical of a specified substituent.
  • substituents When more than one position in any given structure may ibe substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • substituted is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms.
  • Alkyl as used herein alone or as part of another group, refers to a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms. In some embodiments, the alkyl employed in the invention contains 1 to 6 carbon atoms.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2- dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl, n-decyl, and the like.
  • Lower alkyl as used herein, is a subset of alkyl, in some embodiments preferred, and refers to a straight or branched chain hydrocarbon group containing from 1 to 4 carbon atoms.
  • Representative examples of lower alkyl include, but are not limited to, methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, and the like.
  • akyl or “loweralkyl” is intended to include both substituted and unsubstituted alkyl or loweralkyl unless otherwise indicated and these groups may be substituted with groups selected from halo ⁇ e.g., haloalkyl), alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocyclo, heterocycloalkyl, hydroxyl, alkoxy (thereby creating a polyalkoxy such as polyethylene glycol), alkenyloxy, alkynyloxy, haloalkoxy, cycloalkoxy, cycloalkylalkyloxy, aryloxy, arylalkyloxy, heterocyclooxy, heterocyclolalkyloxy, mercapto, alkyl-S(O) m , haloalkyl-S(O) m , alkenyl-S(
  • alkenyl refers to a' straight or branched chain hydrocarbon containing from 1 to.lOfcarbon atoms (or in loweralkenyM to 4 • carbon atoms) which include I 1 to 4 double bonds in the normal chain. In some embodiments,- the alkenyl employed in the invention contain 1 to 6 carbonatoms. Representative examples of alkenyl include, but are not limited to, vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2,4-heptadiene, and the like.
  • alkenyl or “loweralkenyl” is intended to include both substituted and unsubstituted alkenyl or loweralkenyl unless otherwise indicated and these groups may be substituted with groups as described in connection with alkyl and loweralkyl above.
  • Alkynyl as used herein alone or as part of another group, refers to a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms (or in loweralkynyl 1 to 4 carbon atoms) which include 1 to 4 triple bond in the normal chain. In some embodiments, the alkynyl employed in the invention contain 1 to 6 carbonatoms.
  • alkynyl include, but are not limited to, 2-propynyl, 3-butynyl, 2- butynyl, 4-pentynyl, 3- pentynyl, and the like.
  • alkynyl or loweralkynyl is intended to include both substituted and unsubstituted alkynyl or loweralknynyl unless otherwise indicated and these groups may be substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
  • Cycloalkyl as used herein alone or as part of another group, refers to groups having 3 to 10 carbon atoms.
  • the cycloalkyl employed in the invention have 3 to 8 carbon atoms.
  • Suitable cycloalkyls include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like, which, as in the case of other aliphatic, heteroaliphatic or hetercyclic moieties, may optionally be substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
  • Heterocycloalkyl or “heterocycle”, as used herein alone or as part of another group, refers to a non-aromatic 3-, 4-, 5-, 6-, 7-, or 8- membered ring or a polycyclic group, including, but not limited to a bi- or tri-cyclic group comprising fused six-membered rings having between one and four heteroatoms independently selected from oxygen, sulfur and nitrogen, wherein (i) the nitrogen and sulfur heteroatoms may be optionally oxidized, (ii) the nitrogen heteroatom may optionally be quaternized, and (iv) may form a spiro ring or be fused with an cycloalkyl, aryl, heterocyclic ring, benzene or a heteroaromatic ring.
  • the heterocycle employed in the invention have 3 to 10 carbon atoms.
  • Representative heterocycles include, but are not limited to, l,4-dioxa-8-azaspiro[4.5]decane, morpholine, azetidine, azepine, aziridine, diazepine',- 1 ,3-dioxolane, dioxane, dithiane, furan, * imidazole, imidazoline, imidazolidine, isothiazole, isothiazoline, isothiazolidine, isoxazole, isoxazoline, isoxazolidine, morpholine, oxadiazole, oxadiazoline, oxadiazolidine, oxazole, oxazoline, oxazolidine, piperazine, piperidine, pyran, pyrazine, pyrazole, pyrazoline, pyrazolidine, pyridine, pyrimidine,
  • benzodioxine 1 ,3-benzodioxole, cinnoline, indazole, indole, indoline, indolizine, naphthyridine, isobenzofuran, isobenzothiophene, isoindole, isoindoline, isoquinoline, phthalazine, purine, pyranopyridine, quinoline, quinolizine, quinoxaline, quinazoline, tetrahydroisoquinoline, tetrahydroquinoline, thiopyranopyridine, and the like.
  • These rings include quaternized derivatives thereof and may be optionally substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
  • Aryl refers to a monocyclic carbocyclic ring system or a bicyclic carbocyclic fused ring system having one or more aromatic rings.
  • the aryl employed in the invention have 3 to 14 carbon atoms.
  • Representative examples of aryl include, azulenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like.
  • the term "aryl” is intended to include both substituted and unsubstituted aryl unless otherwise indicated and these groups may be optionally substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
  • Arylalkyl refers to an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2- phenylethyl, 3-phenylpropyl, 2-naphth-2-ylethyl, and the like.
  • Heteroaryl as used herein alone or as part of another group, refers to a cyclic, aromatic hydrocarbon in which one or more carbon atoms have been replaced with heteroatoms such as O, N, and S. If the heteroaryl group contains more than one heteroatom, the heteroatoms may be the same or different.
  • the heteroaryl employed in the invention have 3 to 14 carbon atoms.
  • heteroaryl groups include pyridyl, pyrimidinyl, imidazolyl, thienyl, furyl, pyrazinyl, pyrrolyl, pyranyl, isobenzofufanyl, chromenyl, xanthenyl, 'iridolyl.; isoindolyU i ⁇ dolizi ⁇ yl, triazplyl, pyridazinyl, indazolyl, ..
  • heteroaryl groups are five and six membered rings and contain from one to three heteroatoms independently selected from O, N, and S.
  • the heteroaryl group, including each heteroatom can be unsubstituted or substituted with from 1 to 4 substituents, as chemically feasible.
  • Alkoxy refers to an alkyl or loweralkyl group, as defined herein (and thus including substituted versions such as polyalkoxy), appended to the parent molecular moiety through an oxy group, -O-, through a sulfur atom.
  • the alkoxy or thioalkyl group contains 1-10 carbon atoms.
  • the alkyl, alkenyl, and alkynyl groups employed in the invention contain 1-8 carbon atoms.
  • the alkyl group contains 1-6 carbon atoms.
  • the alkyl group contains 1-4 carbon atoms.
  • alkoxy include but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, tert-butoxy, neopentoxy and n-hexoxy and the like.
  • thioalkyl include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
  • Aryloxyalkoxy refers to an alkyl or alkoxy radical substituted with an aryl or aryloxy group and includes benzyl, phenethyl, benzyloxy, 2-phenoxyethyl and the like. In some embodiments, the aryloxyalkoxy group contains 6-14 carbon atoms.
  • Arylalkoxy refers to an alkoxy group as defined above that is further substituted with an aryl, as defined above. In some embodiments, the arylalkoxy group contains 6-14 carbon atoms.
  • Carbonyloxy refers to the group -
  • R is hydrogen, loweralkyl, cycloalkyl, heteroclycloalkyl, amino, aryl, heteroaryl or loweraralkyl.
  • the Carbonyloxy group contains 1-10 carbon atoms.
  • Halo as used herein alone or as part of another group, refers to any suitable halogen, including -F, -Cl, -Br, and —I.
  • Aminoalkyl group as used herein alone or as part of another group, refers to the radical -NHR 3 , where R 3 is an alkyl group.
  • Haloalkyl refers to an alkyl group, as defined above, having one, two, or three halogen atoms attached thereto and is exemplified by such groups as chloromethyl, bromoethyl, trifluoromethyl, and the like.
  • apoptosis refers to a regulated network of biochemical events which lead to a selective form of cell suicide, and is characterized by readily observable morphological and biochemical phenomena, such as the fragmentation of the deoxyribo-nucleic acid (DNA), condensation of the chromatin, which may or may not be associated with endonuclease activity, chromosome migration, margination in cell nuclei, the formation of apoptotic bodies, mitochondrial swelling, widening of the mitochondrial cristae, opening of the mitochondrial permeability transition pores and/or dissipation of the mitochondrial proton gradient and the like.
  • DNA deoxyribo-nucleic acid
  • condensation of the chromatin which may or may not be associated with endonuclease activity
  • chromosome migration chromosome migration
  • margination in cell nuclei the formation of apoptotic bodies
  • mitochondrial swelling widening of the mitochondrial cristae
  • apoptosis refers to at least a 5-fold greater stimulation of apoptosis, at a given concentration an agent, including a 2-methoxy antimycin derivative, in cells that over-express a Bcl-2 family member protein as compared with cells that do not over-express the Bcl-2 family member protein (e.g., a 5-fold lower LD 5 0 or ICso).
  • substantially non- toxic refers to an agent including a 2- methoxyantimycin that induces apoptosis in at least about 50 percent of cells that over- express a Bcl-2 family member protein, but does not induce apoptosis in more than about 5%, more preferably less than 1%, of cells that do not over-express the Bcl-2 family member protein.
  • Bcl-2 family member protein(s) refers to an evolutionarily conserved family of proteins characterized by having one or more amino acid homology domains, BHl, BH2, BH3, and/or BH4.
  • the Bcl-2 family member proteins include Bcl-2, Bcl-x L , Bcl-w, Al, McI-I, Bax, Bak, Bad, Bcl-xs, Bid and the like.
  • the "Bcl-2 family member proteins” further include those proteins, or their biologically active fragments, that have at least 70%, preferably at least 80%, and more preferably at least 90% amino acid sequence identity with a Bcl-2*. family member protein.. ' . • • • • • ⁇
  • anti-apoptotic Bcl-2 family member protein refers to Bcl-2, BCI-XL, BCI- w, Al, McI-I, and other proteins characterized by having one or more amino acid homology domains, BHl, BH2, BH3, and/or BH4, and that promote cell survival by attenuating or inhibiting apoptosis.
  • the "anti-apoptotic Bcl-2 family member proteins” further include those proteins, or their biologically active fragments, that have at least 70%, preferably at least 80%, and more preferably at least 90% amino acid sequence identity with an anti- apoptotic Bcl-2 family member protein.
  • identity refers to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using either a PILEUP or BLAST sequence comparison algorithm (see, e.g., J. MoI. Evol. 35:351-360, 1987; Higgins and Sharp, CABIOS 5:151-153, 1989; Altschul et ai. t J. MoI. Biol. 215:403-410, 1990; Zhang et al. t Nucleic Acid Res. 26:3986-3990, 1998; Altschul et al, Nucleic Acid Res. 25:3389-
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman, Adv. Appl. Math. 2:482, 1981, by the homology alignment algorithm of Needleman and Wunsch, J. MoI. Biol. 48:443, 1970, by the search for similarity method of Pearson and Lipman, Proc. Nat. Acad. ScL USA 85:2444, 1988, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see, generally, Ausubel et al., supra).
  • correspondence of one polypeptide sequence to another sequence is based on the convention of numbering according to nucleotide or amino acid position number, and then aligning the sequences in a manner that maximizes the number of nucleotides or amino acids that match at each position, as determined by visual inspection or by using a sequence comparison algorithm such as, for example, PELEUP (see, e.g., supra; Higgins & Sharp, supra) or BLAST (see, e.g., Altschul et al, supra; Zhang et at, supra; Altschul et al., supra).
  • a mutant Bcl-2 family member amino acid sequence having one or more amino acid substitutions ⁇ additions, or deletions as compared to the wild-type protein may correspond to a second Bcl-2 family member amino acid sequence (e.g., the wild-type sequence or a functionally equivalent variant thereof) according to the convention for numbering the second Bcl-2 family member sequence, whereby the mutant sequence is aligned with the second Bcl-2 family member sequence such that at least 50%, typically at least 60%, more typically at least 70%, preferably at least 80%, more preferably at least 90%, and even more preferably at least 95% of the amino acids in a given sequence of at least 20 consecutive amino acids are identical. Because not all positions with a given "corresponding region" need be identical, non-matching positions within a corresponding region are herein regarded as "corresponding positions.”
  • a single amino acid substitution in one ("first") mutant Bcl-2 family member protein "corresponds" to a single amino acid substitution in a second mutant Bcl-2 family member protein (e.g., BCI-X L ) where the corresponding substituted amino acid positions of the first and second mutant proteins are identical.
  • the phrase "no substantial effect on tertiary protein structure relative to the corresponding wild-type Bcl-2 family member protein” or “no substantial alteration of tertiary protein structure relative to the corresponding wild-type Bcl-2 family member protein” means that, when a Ca trace providing a position for each Ca carbon of the mutant protein is superimposed onto a Ca trace of the corresponding wild-type protein and an ⁇ carbon root mean square (RMS) difference root mean square deviation (RMSD) is calculated; i.e., the deviation of the mutant structure from that of the wild-type structure), the RMSD value is no more than about 1.0 A when calculated using the same structural modeling method, typically no more than about 0.75A, even more typically no more than about 0.5 A, preferably no more than about 0.35 A, and even more preferably no more than about 0.25 A.
  • RMS root mean square
  • RMSD difference root mean square deviation
  • biologically active refers to the ability of a molecule to modulate apoptosis, such as by binding to a Bcl-2 family member protein.
  • a biologically active molecule can modulate apoptosis by causing a change in the mitochondrial protonmotive force gradient ⁇ see, e.g., Example 2); by causing a change in mitochondrial swelling or the morphological characteristics of mitochondria (see, e.g., Example 2); by affecting the release of a reporter molecule, such as, for example, rhodamine 123 or calcein, from mitochondria or vesicles (see, e.g., Examples 4 and 8) comprising a pore-forming anti-apoptotic Bcl-2 family member protein (see, e.g., Example 8); or by causing any other morphological change associated with apoptosis.
  • a reporter molecule such as, for example, rhodamine 123 or calcein
  • an amount or “effective” is intended to designate a dose that causes a relief of symptoms of a disease or disorder as noted through clinical testing and evaluation, patient observation, and/or the like. "Effective amount” or “effective” further can further designate a dose that causes a detectable change in biological or chemical activity. The detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process. Moreover, “effective amount” or “effective” can designate an amount that maintains a desired physiological state, i.e.. reduces or prevents significant decline and/or promotes improvement in the condition of interest.
  • an amount of an agent that effectively modulates the apoptotic state of an individual cell such that apoptosis is induced and/or the inappropriately regulated cell death cycle in the cell returns to a normal state.
  • the dosage will vary depending on the administration routes, symptoms and body weight of the patient but also depending upon the compound being administered.
  • terapéuticaally useful and “therapeutically effective” refer to an amount of an agent that effectively modulates the apoptotic state of an individual cell such that apoptosis is induced and/or the inappropriately regulated cell death cycle in the cell returns to a normal state.
  • diagnosis refers to an agent (e.g., an antimycin derivative) for detecting the induction or inhibition of apoptosis in a subject. These terms further include molecules useful for detecting diseases associated with apoptosis, or the susceptibility to such diseases, and for detecting over-expression or under- expression of a Bcl-2 family member protein.
  • an agent e.g., an antimycin derivative
  • over-expression and “under-expression” refer to an increase or decrease, respectively, in the levels of a Bcl-2 family member protein in a cell relative to the level of such a protein found in the same cell or a closely related non-malignant cell under normal physiological conditions.
  • apoptosis-associated disease includes diseases, disorders, and conditions that are linked to an increased or decreased state of apoptosis in at least some of the cells of a subject.
  • diseases include neoplastic disease (e.g., cancer and other proliferative diseases), tumor formation, arthritis, inflammation, autoimmune disease, human immunodeficiency virus (HIV) immunodeficiency syndrome, neurodegenerative diseases, myelodysplastic syndromes (such as aplastic anemia), ischaemic syndromes (such as myocardial infarction), liver diseases which are induced by toxins (such as alcohol), alopecia, damage to the skin due to UV light, lichen planus, atrophy of the skin, cataract, and graft rejections and other premalignant and noneoplastic hyperproliferative disorders.
  • Apoptosis- "associated diseases further include drug resistance associated with increased or decreased levels of an anti-apoptotic Bcl-2 family member protein as well as multiple chemotherapeutic drug resistance.
  • Concurrently administer means that the two compounds or agents are administered closely enough in time to produce a combined effect (that is, concurrently may be simultaneously, or it may be two or more events occurring within a short time period before or after each other, e.g., sequentially). Simultaneous administration may be carried out by mixing the compounds prior to administration, or by administering the compounds at the same point in time but at different anatomic sites and/or by using different routes of administration.
  • a “combinatorial library” is a collection of compounds in which the compounds comprising the collection are composed of one or more types of subunits.
  • the subunits can be selected from natural or unnatural moieties, including dienes, benzene compounds, cycloalkanes, lactones, dilactones, amino acids, alkanes, and the like.
  • the compounds of the combinatorial library differ in one or more ways with respect to the number, order, type or types of modifications made to one or more of the subunits comprising the compounds.
  • a combinatorial library may refer to a collection of "core molecules" which vary as to the number, type or position of R groups they contain and/or the identity of molecules composing the core molecule. The collection of compounds is generated in a systematic way.
  • a combinatorial library can be synthesized on a solid support from one or more solid phase-bound resin starting materials.
  • the library can contain five (5) or more, preferably ten (10) or more, organic molecules, which are different from each other (i.e., five (5) different molecules and not five (5) copies of the same molecule).
  • Each of the different molecules will be present in an amount such that its presence can be determined by some means (e.g., can be isolated, analyzed, detected with a binding partner or suitable probe).
  • each different molecule needed so that its presence can be determined can vary due to the actual procedures used and can change as the technologies for isolation, detection and analysis advance.
  • an amount of about 100 picomoles or more can be detected.
  • Preferred libraries comprise substantially equal molar vamounts of each desired reaction product and do not include relatively large or small amounts of any given molecules so that the presence of such molecules dominates or is completely . suppressed in any assay.
  • Combinatorial libraries are generally prepared by derivatizing a starting compound onto a solid-phase support (such as a bead).
  • the solid support has a commercially available resin attached, such as a Rink or Merrifield Resin, and the like.
  • substituents are attached to the starting compound.
  • the starting compound can comprise the dilactone moiety, or a precursor thereof.
  • Substituents are added to the starting compound, and can be varied by providing a mixture of reactants comprising the substituents. Examples of suitable substituents include, but are not limited to, the following:
  • hydrocarbon substituents that is, aliphatic (e.g., alkyl or alkenyl), alicyclic (e.g., cycloalkyl, cycloalkenyl) substituents, aromatic, aliphatic and alicyclic-substituted aromatic nuclei, and the like, as well as cyclic substituents;
  • substituted hydrocarbon substituents that is, those substituents containing nonhydrocarbon radicals which do not alter the predominantly hydrocarbon substituent; those skilled in the art will be aware of such radicals (e.g., halo (especially chloro and fluoro), alkoxy, mercapto, alkylmercapto, nitro, nitroso, sulfoxy, and the like);
  • hetero substituents that is, substituents which will, while having predominantly hydrocarbyl character, contain other than carbon atoms.
  • heteroatoms will be apparent to those of ordinary skill in the art and include, for example, sulfur, oxygen, nitrogen, and such substituents as pyridyl, furanyl, thiophenyl, imidazolyl, and the like. Heteroatoms, and typically no more than one, will be present for each carbon atom in the hydrocarbon-based substituents. Alternatively, there may be no such radicals or heteroatoms in the hydrocarbon-based substituent and it will, therefore, by purely hydrocarbon.
  • active compounds are provided.
  • the active compounds may be used to modulate apoptosis in cells that over- express a Bcl-2 family member protein, which may be used to treat apoptosis-associated diseases.
  • active compounds include compounds of Formula I and Formula II as set forth below, and including the pharmaceutically acceptable salts and prodrugs>thereof.
  • X and X' are each independently O or S;
  • Ri, R 2 , R3, R4, R5, Re, R7, Rs, R9, Rio, Riu Ri2» and R13 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci- 10 alkyl, d.io alkenyl, Ci- 10 alkoxy, C M 0 alkylsulfanyl, Ci-10 haloalkyl, C3-10 cycloalkyl, C 3 .]o cycloalkyl-Ci-io alkyl, C3.10 heterocycloalkyl, C3-10 heterocycloalkyl-Ci-io alkyl, C 3 -U aryl, C 6 -i 4 -arylalkoxy, C 6 -i4-aryloxyalkoxy, Ci-I 0 - carbonyloxy, C 3 -io-heterocycle-Ci
  • X is O.
  • X 1 is O.
  • Ri, R 4 , Rg, R 9 , and Rio are each independently preferably Ci-10- alkyl, Ci-1 0 -alkenyl, or Ci.io-alkoxy.
  • R 2 , R 3 , R 5 , R 7 , Rs, R 12 and Rj 3 are each independently preferably H or halo.
  • Rn is preferably H, Ci- 10 -alkyl, or Ci-10-alkenyl.
  • X and X' are O;
  • Ri 1 Rj, Re, R 9 , and R] 0 are each independently selected from the group consisting of Ci- 4 -alkyl, and Ci-4-alkenyl;
  • R 2 , R 3 , R 5 , R 7 , R 8 , Ri 2 and Ri 3 are each independently selected from the group consisting of H and halo;
  • Ri 1 is H or Ci- 4 -alkyl; or a pharmaceutically acceptable salt thereof.
  • Ri, R 2 , R3, Rj, Rs, Re, R7, Rs, R9, Rio, Rn, R12, and Ri 3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-io-alkyl, Ci_io alkenyl, Ci- 10 alkoxy, Cj-io alkylsulfanyl, Ci- 10 haloalkyl, C3.
  • halogen e.g., chloro, fluoro, bromo, iodo
  • Xi is O.
  • X 2 is N.
  • R 2 is selected from the group consisting of nitro, halogen, OH, cyano, amine, Ci-4-alkyl and Ci- 4 -alkenyl.
  • Ri, R3, R5, Re, R 7 , Rs, R9, Rio, Ri2 » and Rt 3 are each independently H or halo.
  • R4 and Rn are are each independently H, Ci-10-alkyl, or Ci-1 0 alkenyl.
  • X 2 is N
  • R 2 is selected from the group consisting of Nitro, CN, halogen and Ci-4-alkyl; Ri, R 3 , R5, Re, R 7 , Rs, R9, Rio, Ri2, and R ]3 are each indepently H or halo;
  • R 4 and Rn are each independently H or Ci- 4 -alkyl; or a pharmaceutically acceptable salt thereof.
  • An example of the foregoing is S2 (2-Nitro-6-(N'-quinolin-2-yl- hydrazinomethylene)-cyclohexa-2,4-dienone), having the structure:
  • the compounds used in the methods of the invention may be synthesized by any suitable method known to one of skill in the art. See the examples, infra, for commercial sources for some of the compounds useful in embodiments of the invention.
  • Compounds that modulate apoptosis and are substantially non-toxic to cells that do not over-express a Bcl-2 family member may be identified by a method generally comprises the steps of contacting a candidate compound with a cell that over-expresses a Bcl-2 family member protein; contacting the compound with another cell that does not over-express the Bcl-2 family member protein; and determining whether the compound modulates the activity of the Bcl-2 family member protein to produce a physiological change in the cell that over- expresses the Bcl-2 family member protein, but does not produce a substantial physiological change in the cell which does not over-express that protein.
  • Physiological changes that are indicative of binding of the candidate compound to the Bcl-2 family member protein include an affect on cell death, cell shrinkage, chromosome condensation and migration, mitochondria swelling, and/or disruption of mitochondrial transmembrane potential (i.e., the mitochondrial proton gradient), and/or cell death (e.g., as measured by trypan dye exclusion).
  • Biologically active compounds can also be identified by evaluating the ability of the agents to modulate glucose uptake and/or lactate production in cells expressing an anti-apoptotic Bcl-2 family member protein.
  • Apoptosis-modulating compounds increase cellular glucose uptake or lactate production in proportion to the level of expression of a Bcl-2 family member target protein. Methods for assaying glucose production or lactate production are well-known in the art.
  • Combinatorial libraries of test compounds can be screened for biological activity using any of the assay methods described above.
  • Combinatorial libraries and processes are described, for example, in PCT/USOO/22891.
  • Computer-based methods may also be used to identify biologically active compounds by using a "molecular docking" algorithm to score test compounds for binding to each of a* Bcl-2 family member protein and a corresponding mutant protein as described ⁇ supra.
  • Those test compounds that demonstrate a lower score for binding to the mutant protein relative to the corresponding Bcl-2 family member protein e.g., a mutant BcI- X L protein (having a E92L, F97W, L130A, A142L, F146L, or Y195G mutation) and the wild- type BcI- X L protein, respectively) can be further evaluated in biological assays as described supra to verify biological activity.
  • the orientation of the ligand is evaluated with a shape scoring function (an empirical function resembling the van der Waals attractive energy) and/or a function approximating the ligand-receptor binding energy (which is taken to be approximately the sum of the van der Waals and electrostatic interaction energies).
  • a grid-based rigid body minimization is carried out for the ligand to locate the nearest local energy minimum within the receptor binding site.
  • the position and conformation of each docked molecule can be optimized, for example, using the single anchor search and torsion minimization method of DOCK4.0. ⁇ See, e.g., Ewing and Kuntz, supra; Kuntz, supra.)
  • heuristic docking and consensus scoring strategies can be used in the computer-based identification of biologically active compounds (i.e., different docking and scoring methods can be applied to evaluate the screening results). For example, following a primary screening using, e.g. , DOCK4.0 (supra), top-scoring compounds can be re-scored using other docking algorithms such as, for example, GOLD, FlexX, PM (see Muegge and Martin, J. Med. Chem. 42:791, 1999, and/or AutoDock3.0 (see Morris et al, J. Comput. Chem. 19:1639-1662, 1998).
  • a consensus score can be determined by combining results from any of the individual docking programs used to score the candidate compounds (see Clark et al, J. MoI. Graph Model 20:281-295, 2002).
  • a subset, for example, of the top-scoring molecules from the primary screen can be selected for further analyses (e.g., a tertiary virtual screen or, alternatively or additionally, biological screening assays such as, for example, any of the assays described herein or otherwise known in the art).
  • 2D database similarity searching may be performed using BITMACCS within MOE, and 2Dph, an in-house 2D-pharmacophore-based program, to search for compounds similar to 2-MeAAl, Sl and troglitazone (setting the threshold for the percentage of similarity at >65%) from our screening database.
  • the final "hits" of commercially available compounds may then be saved into the separate databases.
  • Anther approach may focus on partial and whole 3D pharmaophore mapping/ searching with potential bioisosteric replacements of sub- group(s) of our identified initial hits based on a topological pharmacophore description of the fragments.
  • the 5-benzylthiazolidine-2,4-dione moiety of troglitazone can be substituted by various 2-oxy-3-arylcarboxylic acids, and the 9-membered ring of 2-MeAAl could be replaced by two 5- or 6-merbered rings.
  • the Bioster database (39) (Accelrys, San Diego, CA) can be used to search for the most promising replacements, and focused virtual libraries can be built. This method has been used in the pharmaceutical industry to improve the ADME/Tox properties of initial promising lead compounds and for broadening patentability (40). Docking
  • the BH3 peptide-bound Bcl-xL structure, PDB Code: IBXL and unliganded Bcl-xL structure, PDB Code: 1R2D are used as target receptors.
  • the Flexx 2.0.1 (41) docking program a fully automatic docking tool for flexible ligands is used to dock the ligands into their binding sites within the receptor structures. All small molecule compounds are prepared by Corina and MOE. After removing all water molecules, the active sites are dissected from the NMR structure of the protein complexes by including all residues that have at least one single (heavy) atom within a distance of 6.5 A from any heavy atoms of the ligand.
  • Residues are kept fixed in their NMR coordinates in all docking experiments.
  • This docking method consists of three steps: the selection of a part of the molecule, the base fragment, the placement of the base fragment into the active site of a protein, and the subsequent reconstruction of the complete drug molecule by linking the remaining components step by step.
  • two algorithms are in use. The first one superposes triples of interaction centers of a base fragment with triples of compatible interaction points in the active site. If a base fragment has fewer than three interaction centers or if the number of placements is too low, the second algorithm, called line matching, is started. This algorithm matches pairs of interaction centers with pairs of interaction points.
  • VMScore Novel score-function
  • the docking-score is the key component in structure-based drug design.
  • the score- function is used for estimating the binding affinity of the ligand with docked conformation within the binding packet. It will help to decide which compound(s) should be selected for experimental development. Score-functions which derived directly from molecular mechanics are not often used in practice largely due to the time-consuming nature of the process and high demand for computational power for a large set of molecules. In the past years, several (semi) empirically based score-functions have been used in the ranking of docked structures. "Score” (30) and “Chemscore” (31) use contributions from hydrogen bonding, metal-ligand contacts, lipophilic contact area and frozen rotatable bonds.
  • the "HINT Score” use contributions representing different types of surface areas (32). Score- functions based on the probability of pair-wise contact of heavy atoms such as “PMF” (potential of mean force) (33) and “Smog” (34) have reported. However, these score- functions do not consider all possible energy contributions for binding. For examples, “Score” and “Chemscore” do not consider the contribution of the restricted conformation of the bound ligand compared to the free molecule; “PMF” and “Smog” do not consider contributions from hydrogen bonding. In addition, most of the current score-functions do not take desolvation energy into account.
  • ⁇ G A* ⁇ G e]e + B* ⁇ G vd w + C* ⁇ G hb + D* ⁇ G CO nf + E* ⁇ G deS oi + F* ⁇ G ent
  • ⁇ G e e is free energy contribution from electrostatic interaction.
  • ⁇ G vdw is free energy contribution from van de waal's interaction.
  • ⁇ Ghb is free energy contribution from hydrogen bonding,
  • ⁇ G CO nf is free energy contribution from conformation change,
  • ⁇ G deso ⁇ is free energy contribution from desolvation energy difference between ligand, protein and their complex,
  • ⁇ Gem is entropy contribution from translation and rotation, and A to F are coefficients.
  • VMScore we have taken a well- described data set containing 53 complex (protein bound with compound) crystal structures with experimental binding affinity (Ki) data ( ⁇ G oc logKi) as training set (32).
  • Table-4 The prediction results of different docking score-functions for the same Endothiapepsin complex testing set.
  • the compounds of the present invention are useful for treating cells in which the cell death signal is down-regulated and the affected cell has an inappropriately diminished propensity for cell death, which is referred to herein as being in a "decreased apoptotic state.”
  • the invention further provides methods for the administration to a subject of a therapeutically effective amount of an apoptosis-modulating compound of the invention to treat an apoptosis- associated disease in which it is desirable to induce apoptosis in certain types of cells, such as virus-infected or autoantibody-expressing cells.
  • a method of treating a cancer characterized by the over- expression of a Bcl-2 family member is provided.
  • Examples of cancers known to be associated with over-expression of a Bcl-2 family member and which can be treated according to the methods provided herein are shown in Table 4.
  • Adenoma 65-98% Adenoma - 50% Carcinoma - 46-60% Carcinoma - 60%
  • the treatment of the cancer can include the treatment of solid tumors or the treatment of leukemias.
  • the cancer can be of the skin, breast, brain, cervix, testis, and the like. More particularly, cancers may include, but are not limited to, the following organs or systems: cardiac, lung, gastrointestinal, genitourinary tract, liver, bone, nervous system, gynecological, hematologic, skin, and adrenal glands.
  • the methods herein can be used for treating gliomas (Schwannoma, glioblastoma, astrocytoma), neuroblastoma, pheochromocytoma, paraganlioma, meningioma, adrenal cortical carcinoma, kidney cancer, vascular cancer of various types, osteoblastic osteocarcoma, prostate cancer, ovarian cancer, uterine leiomyomas, salivary gland cancer, choroid plexus carcinoma, mammary cancer, pancreatic cancer, colon cancer, B and T cell lymphomas, acute and chronic myeloid or lymphoid leukemias, and multiple myeloma.
  • gliomas Rosta, glioblastoma, astrocytoma
  • neuroblastoma pheochromocytoma
  • paraganlioma paraganlioma
  • meningioma adrenal cortical carcinoma
  • kidney cancer vascular cancer of various types
  • osteoblastic osteocarcoma prostate
  • treatment may include pre-malignant conditions associated with any of the above cancers (e.g., colon adenomas, myelodysplastic syndrome).
  • methods of treating a neurodegenerative disease characterized by the over-expression of a Bcl-2 family member are provided.
  • Neurodegenerative diseases include Alzheimer's disease', Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis and other diseases linked to degeneration of the brain, such as Creutzfeldt- Jakob disease and expanded polyglutamine repeat diseases.
  • Expanded polyglutamine repeat diseases with which the present invention is concerned include, but are not limited to, Huntington's disease, dentatorubral pallidoluysian atrophy, spinobulbar muscular atrophy, and spinocerebellar ataxia types 1, 2, 3, 6 and 7. See, e.g., US Patent No. 6,632,616 to Burke et al.
  • methods of treating arthritis, inflammation, autoimmune diseases, human immunodeficiency virus (HIV) immunodeficiency syndrome, myelodysplastic syndromes (such as aplastic anemia), ischaemic syndromes (such as myocardial infarction), liver diseases which are induced by toxins (such as alcohol), alopecia, damage to the skin due to UV light, lichen planus, atrophy of the skin, cataract, and graft rejections are provided.
  • the compounds used in embodiments of the invention will be substantially purified prior to administration.
  • the subject can be an animal, including, but not limited to, cows, pigs, horses, chickens, cats, dogs, and the like, and is typically a mammal, and in a particular embodiment human. In another specific embodiment, a non- human mammal is the subject.
  • apoptosis-modulating compounds are administered as therapeutic or pharmaceutical compositions by any suitable route known to the skilled artisan including, for example, intravenous, subcutaneous, intramuscular, intradermal, transdermal, intrathecal, intracerebral, intraperitoneal, intransal, epidural, and oral routes.
  • Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulations.
  • administration can be by injection or infusion into the cerebrospinal fluid (CSF).
  • CSF cerebrospinal fluid
  • administration can be with one or more other components capable of promoting penetration of the derivative across the blood-brain barrier.
  • it can be desirable to introduce a compound into the target tissue by any suitable route, including intravenous and intrathecal injection.
  • Pulmonary administration can . also be employed, such as, for example, by use of an inhaler or nebulizer, and formulation of the compound with an aerosolizing agent.
  • the compound is coadministered with an inhibitor of esterase activity to further stabilize the compound.
  • Pharmaceutical compositions can also be administered orally ⁇ n any orally acceptable dosage form including, but not limited to, capsules, tablets, caplets, lozenges, aqueous suspensions or solutions.
  • carriers which are commonly used, include lactose and corn starch.
  • Lubricating aids such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • the agent can be combined with emulsifying and suspending aids. If desired, certain sweeteners, flavorants, or colorants can also be used.
  • the compounds of the present invention can be combined with any other tumor and/or cancer therapy.
  • the therapy can include, for example and not by way of limitation, surgery, radiation, and chemotherapy either individually or in any combination.
  • Chemotherapy can include any current known or yet to be discovered chemotherapeutic agent including but are not limited to Aceglatone; Aclarubicin; Altretamine; Aminoglutethimide; 5-
  • Antibody Antilymphocyte Immunoglobulins; Antineoplaston AlO; Asparaginase;
  • Pegaspargase Azacitidine; Azathioprine; Batimastat; Benzoporphyrin Derivative; Bicalutamide; Bisantrene Hydrochloride; Bleomycin Sulphate; Brequinar Sodium;
  • Carmofur Carmustine; Chlorambucil; Chlorozotocin; Chromomycin; Cisplatin; Cladribine;
  • Dactinomycin Daunorubicin Hydrochloride; Decitabine; Diaziquone; Dichlorodiethylsulphide; Didemnin B.; Docetaxel; Doxifluridine; Doxorubicin Hychloride;
  • Droloxifene Echinomycin; Edatrexate; Elliptinium; Elmustine; Enloplatin; Enocitabine;
  • Phosphate Fluorouracil; Flutamide; Formestane; Fotemustine; Gallium Nitrate; Gencitabine; Gusperimus; Homoharringtonine; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide;
  • Lithium Gamolenate Lobaplatin; Lomustine; Lonidamine; Mafosfamide; Meiphalan;
  • Menogaril Mercaptopurine; Methotrexate; Methotrexate Sodium; Miboplatin; Miltefosine;
  • Muromonab-CD3 Mustine Hydrochloride; Mycophenolic Acid; Mycophenolate Mofetil;
  • Nedaplatin Nilutamide; Nimustine Hydrochloride; Oxaliplatin; Paclitaxel; PCNU;
  • Penostatin Peplomycin Sulphate; Pipobroman; Pirarubicin; Piritrexim Isethionate;
  • Piroxantrone Hydrochloride Plicamycin; porfimer Sodium; Prednimustine; Procarbazine Hydrochloride; Raltitrexed; Ranimustine; Razoxane; Rogletimide; Roquinimex; Sebriplatin;
  • administration can be desirable to administer the compounds of the invention locally to the area in need of treatment; this administration can be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application (e.g., in conjunction with a wound dressing after surgery), by injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non- porous, or gelatinous material, including membranes such as silastic membranes, or fibers.
  • administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre-neoplastic tissue.
  • the compounds of the invention can be delivered in a vesicle, in particular a liposome (see, e.g., Langer, Science 249:1527-1533, 1990; Treat et al., In Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365, 1989; Lopez-Berestein, supra, pp. 317-327).
  • a liposome see, e.g., Langer, Science 249:1527-1533, 1990; Treat et al., In Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365, 1989; Lopez-Berestein, supra, pp. 317-327).
  • the compounds of the invention can be delivered in a controlled release system.
  • a pump can be used (see, e.g., Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201, 1987; Buchwald et ah, Surgery 88:507, 1980; Saudek et al., N. Engl. J. Med. 321:574, 1989).
  • polymeric materials can be used (see, e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida, 1974; Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York, 1984; Ranger and Peppas, J. Macromol. ScL Rev. Macromol. Chem. 23:61, 1983; see also Levy et al., Science 228:190, 1985; During et al., Ann. Neurol. 25:351, 1989; Howard et al, J. Neurosurg. 71 : 105, 1989).
  • a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, supra, Vol. 2, pp. 115-138, 1984).
  • Other controlled release systems are discussed in, for example, the review by Langer (Science 249:1527-1533, 1990).
  • the methods of treatment and the pharmaceutical compositions described herein can be combined with any other tumor and/or cancer therapy.
  • the therapy can include, for example and not by way of limitation, surgery, radiation, and chemotherapy either individually or in any combination.
  • Chemotherapy can include any current known or yet to be discovered chemotherapeutic agent including, for example, paclitaxel, doxorubicin, etoposide, melphalan, daunorubicin, 5-fluorouracil, cisplatin, paraplatin, and the like, either individually or in any combination.
  • the methods of treatment as described herein can include concurrently administering one or more additional inducer of apoptosis (for example, staurosporine), and compositions as described herein can optionally include one or more additional inducer of apopotosis.
  • additional inducer of apoptosis for example, staurosporine
  • compositions as described herein can optionally include one or more additional inducer of apopotosis.
  • the methods of treatment and compositions as described herein can be used in selectively targeting cancer cells (for example, Bcl-xL overexpressing cells such as TAMH-Bcl-xl cells).
  • compositions comprise a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of the invention.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more typically in humans.
  • carrier refers to a diluent, adjuvant, excipient, stabilizer, vehicle, or any combination thereof, with which the agent is formulated for administration.
  • Pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like.
  • Water is a typical carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose,, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations, and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • the pharmaceutical composition includes an inhibiter of esterase activity as a stabilizing agent.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like.
  • suitable pharmaceutical carriers are described in, for example, Remington 's Pharmaceutical Sciences, by E.W. Martin.
  • Such compositions will contain a therapeutically effective amount of a compound of the invention, typically in purified form, together with a suitable amount of carrier so as to provide a formulation proper for administration to the subject.
  • the formulation should suit the mode of administration.
  • the compound of the present invention is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • pharmaceutical compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition can also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form. For example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, ' procaine, and the like.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • a "pharmaceutically acceptable salt” as used herein refers to a salt form of a compound permitting its use or formulation as a pharmaceutical and which retains the biological effectiveness of the free acid and base of the specified compound and that is not biologically or otherwise undesirable. Examples of such salts are described in Handbook of
  • Pharmaceutical Salts Properties, Selection, and Use, Wermuth, CG. and Stahl, P.H. (eds.), Wiley- Verlag Helvetica Acta, Zurich, 2002 [ISBN 3-906390-26-8].
  • Examples of pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those formed with free carboxyl groups such as those derived from sodium- potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine and the like.
  • salts also include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, xylenesulfonates, phenylacetates, phenylpropionates,
  • prodrugs or "softdrugs” of the compounds may be used in embodiments of the invention.
  • Pharmaceutically acceptable prodrugs as used herein refers to those prodrugs of the active compounds of the present invention 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, commensurate with a reasonable risk/benefit ratio, and effective for their intended use? as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V.
  • Examples include a prodrug that is metabolized in vivo by a subject to an active drug having an activity of active compounds as described herein, wherein the prodrug is an ester of an alcohol or carboxylic acid group, if such a group is present in the compound; an acetal or ketal of an alcohol group, if such a group is present in the compound; an N-Mannich base or an imine of an amine group, if such a group is present in the compound; or a Schiff base, oxime, acetal, enol ester, oxazolidine, or thiazolidine of a carbonyl group, if such a group is present in the compound, such as described in US Patent No. 6,680,324 and US Patent No. 6,680,322.
  • the amount of the compound of the invention that is combined with the carrier to produce a single dosage form will vary, depending upon the nature of that agent and the composition of the dosage form. It should be understood, however, that a specific dosage and treatment regime for any particular patient or disease state will depend upon a variety of factors, including the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the judgment of the treating physician, and the severity of the particular disease being treated. The amount of active agent will also depend upon the 5 specific activity of the compound and whether that agent is co-administered with any other therapeutic or prophylactic ingredients.
  • Determination of therapeutically effective dosages is typically based on animal model studies and is guided by determining effective dosages and administration protocols that significantly reduce the occurrence or severity of the apoptosis- associated disease in model subjects (e.g., in the case of treatment of malignancies, a tumor0 xenograft model in mice can be used (see, e.g., Example 20).
  • a tumor0 xenograft model in mice can be used (see, e.g., Example 20).
  • a non-limiting range for a therapeutically effective amount of the compounds is about 0.001 mg/kg and about 100 mg/kg body weight per day, and in more specific embodiments between about 0.001 mg/kg and about 50 mg/kg, between about 0.01 mg/kg and about 20 mg/kg. >5 between about 0.1 and about 10 mg/kg, or between about 0.1 mg/kg and about 5 mg/kg body weight per day.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form0 prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use, or sale for human administration.
  • Table 5 includes dose dependence of TABX2S cell survival (IC 50 ) and the selective induction of apoptosis in TABX2S cells over the TAMH.neo control.
  • the gain-of-function activity with Bcl-xL inhibitor Sl was illustrated in Figure 1. It shows that Sl binds to the Bcl-xL hydrophobic groove (See Fig. 1, A) and shows an EC50 of ⁇ 6 to 10 ⁇ mol/L for Bcl-xL-overexpressing TAMH cells, without cytotoxicity for control TAMH-neo cells at these concentrations (See Fig. I 5 B). Pretreatment with 2-deoxyglucose sensitized TAMH-Bcl-xL cells to Sl ⁇ See Fig. 1, C). Sl has no detectable effect on mitochondrial oxygen consumption rate at cytotoxic concentrations (See Fig. 1, D).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to compounds for modulating apoptosis in cells over expressing Bcl-2 Family member proteins (e.g., Bcl-2 or Bcl-xL). The present invention also relates to pharmaceutical compositions containing these compounds, and methods of using the compounds for treating apoptosis-associated diseases such as, for example, neoplastic disease (e.g., cancer) or other proliferative diseases associated with the over-expression of a Bcl-2 family member protein.

Description

COMPOSITIONS AND METHODS FOR MODULATING APOPTOSIS IN CELLS OVER-EXPRESSING Bcl-2 FAMILY MEMBER PROTEINS
Pamela S. Schwartz, Michael K. Manion, John S. Fry, David M. Hockenbery
GOVERNMENT SUPPORT
Aspects of this research are supported by the National Cancer Institute/National Institutes of Health (NCI/NIH) Grant No. CA 80416 and UOl CA 91310, and National Institute on Alcohol Abuse and Alcoholism/National Institutes of Health (NIAAA/NIH) GranT No. R03 AAl 5681. The U.S . government may have certain rights in the invention.
CROSS-REFERENCE TO RELATED APPLICATION
This application claims the benefit of United States Provisional Application Number 60/836,918, filed August 10, 2006 (Attorney Docket No. 9498-17PR) and United States Provisional Application Number 60/948,808, filed July 10, 2007(Attorney Docket No. 9498- 17PR2), the disclosure of which are incorporated by reference herein in its entirety.
FIELD OF THE INVENTION
The present invention relates to compounds for modulating apoptosis in cells over expressing Bcl-2 Family member proteins. The present invention also relates to pharmaceutical compositions containing these compounds and methods of using the compounds.
BACKGROUND OF THE INVENTION Many diseases are believed to be related to the down-regulation of apoptosis in the affected cells. For example, neoplasias may result, at least in part, from an apoptosis- resistant state in which cell proliferation signals inappropriately exceed cell death signals. Furthermore, some DNA viruses, such as Epstein-Barr virus, African swine fever virus and adenovirus, parasitize the host cellular machinery to drive their own replication and at the same time modulate apoptosis to repress cell death and allow the target cell to reproduce the virus. Moreover, certain diseases, such as lymphoproliferative conditions, cancer (including drug resistant cancer), arthritis, inflammation, autoimmune diseases, and the like, may result from a down regulation of cell death signals. In such diseases, it would be desirable to promote apoptotic mechanisms.
Most currently available chemotherapeutic agents target cellular DNA and induce apoptosis in tumor cells (Fisher et al, Cell 78:539-542, 1994). A decreased sensitivity to apoptosis induction has emerged as an important mode of drug resistance. Members of the evolutionarily conserved Bcl-2 family are important regulators of apoptotic cell death and survival. The proteins Bcl-2, BCI-XL, Bcl-w, Al and McH are death antagonists while Bax, Bak, Bad, Bcl-xs, Bid, and Bik are death agonists (Kroemer et al., Nature Med. 6:614-620, 1997). Over-expression of Bcl-2 and BCI-XL confers resistance to multiple chemotherapeutic agents, including alkylating agents, antimetabolites, topoisomerase inhibitors, microtubule inhibitors and anti-tumor antibiotics, and may constitute a mechanism of clinical chemoresi stance in certain tumors (Minn et al., Blood 86:1903-1910, 1995; Decaudin et al., Cancer Res. 57:62-67, 1997). Bcl-2/ Bcl-XL-directed therapies, using either anti-sense oligonucleotides or novel protein-targeted drugs, can increase cellular sensitivity to standard agents in vitro or, in some cases, kill cells as single agents (Jansen et al., Nat. Med. 4:232- 234, 1998).
Structure solutions for BCI-XL and Bcl-2 have demonstrated the presence of a hydrophobic cleft at the surface of both proteins (Muchmore et al., Nature 381:335-341, 1996). Functional studies implicate this groove as a binding surface for heterodimenc partners, including the related pro-apoptotic proteins Bax and Bak, and as a regulatory domain for an intrinsic membrane pore function (Sattler et al., Science 275:983-986, 1997). Efforts to design small molecule inhibitors of Bcl-2/ BCI-XL have thus focused on this structural feature.
Two antimycins, antimycin Ai and A3, have been discovered to inhibit the activity of the anti-apoptotic Bcl-2 family member proteins, Bcl-2 or Bcl-xL. These naturally obtained antimycins are toxic, however, because as discussed above, they also inhibit mitochondrial respiration. Therefore, antimycin derivatives that are effective in inducing apoptosis in cells where apoptosis is inappropriately regulated while exhibiting reduced inhibition of mitochondrial respiration have been identified. It would be desirable to identify other compounds that may be effective in inducing apoptosis in cells where apoptosis is inappropriately regulated while minimally inhibiting mitochondrial respiration. Furthermore, it would be desirable to use the identified compounds as to treat diseases related to the down regulation of apoptosis. SUMMARY OF THE INVENTION
The present invention provides methods of treating an apoptosis-associated disease. Embodiments of the present invention provide methods for treating an apoptosis-associated disease (e.g., cancer) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an active compound of Formula I:
Figure imgf000004_0001
or a pharmaceutically acceptable salt or a prodrug thereof; wherein:
X and X1 are each independently O or S;
Ri, R2, R3, R4,' Rs, Re, R7, Rs, R9, Rio, Rn, R12, and R13 are each independently selected from the group s consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-10-alkyl, Ci-io-alkenyl, Ci-io-alkoxy. Ci-10-alkylsulfanyl, Ci.io-haloalkyl, Cs-io-cycloalkyl, Cs-io-cycloalkyl-Ci-io-alkyl, Cs-io-heterocycloalkyl, C3-10- heterocycloalkyl-Ci-10-alkyl, C3-14 aryl, Cβ-π-arylalkoxy, Cβ-M-aryloxyalkoxy, C1-Io- carbonyloxy, Cs-io-heterocycle-Ci.io-alkoxy and C3-14 heteroaryl.
Embodiments of the present invention provide methods for treating an apoptosis- associated disease (e.g., cancer) in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of an active compound of Formula II:
Figure imgf000004_0002
or a pharmaceutically acceptable salt or a prodrug thereof; wherein:
Xi is O or S;
X2 is C or N;
Ri, R2, R3, R4, R5, Re, R7, Rs, R9, Rio, Rn, R12, and RJ3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-10-alkyl, d-10-alkenyl, Ci-10-alkoxy, Ci-10-alkylsulfanyl, Cπo haloalkyl, Ca-io-cycloalkyl, Cs-io-cycloalkyl-Ci-io-alkyl, Ca-io-heterocycloalkyl, C3.10- heterocycloalkyl-Ci-10-alkyI, C3-14 aryl, C<;-i4-arylalkoxy, Cβ-π-aryloxyalkoxy, Ci-10- carbonyloxy, Ca-io-heterocycIe-Ci-to-alkoxy and C3.14 heteroaryl.
In some embodiments, the active compound has the strcutre of:
Figure imgf000005_0001
or
Figure imgf000005_0002
In some embodiments, the method for treating an apoptosis-associated disease further comprising concurrently adminsterting to said subject another inducer of apopotosis.
Embodiments of the present invention further provide pharmaceutical compositions comprising, consisting of or consisting essentially of
Figure imgf000005_0003
or a pharmaceutically acceptable salt, or a prodrug thereof; wherein: X and X1 are each independently O or S;
R1, R2, R3, R4, Rs, R<>> R7, Rs, R9, Rio, Ri 1, R12, and Ri3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen {e.g., chloro, fluoro, bromo, iodo), Ci-io-alkyl, Ci-10-alkenyl, Ci-io-alkoxy, Ci-10-alkylsulfanyl, Cj. 10-haIoalkyl, Cj-io-cycloalkyl, C3-io-cycloalkyl-Ci-io-alkyl, Cs-io-heterocycloalkyl, C3-10- heterocycloalkyl-Ci-10-alkyl, C3-14 aryl, Cβ-π-arylalkoxy, Ce-u-aryloxyalkoxy, Ci-10- carbonyloxy, C3-o-heterocycle-Ci-o-alkoxy and C3-14 heteroaryl; and a pharmaceutically acceptable carrier.
Embodiments of the present invention further provides pharmaceutical compositions comprising, consisting of, or essentially consisting of an active compound of Formula II:
Figure imgf000006_0001
or a pharmaceutically acceptable salt or a prodrug thereof; wherein:
Xi is O or S;
X2 is C or N;
Ri, R2, R3, R»J R-5* ROJ R7, Rs5 Rs>5 Rio, Ri 1, Ri2, and Rn are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Q.io-alkyl, Cj.io-alkenyl, Ci-io-alkoxy, Ci-10-alkylsulfanyl, Ci- 10-haloalkyl, Cs-io-cycloalkyl, Cs-io-cycloalkyl-Ci-io-alkyl, Ca-io-heterocycloalkyl, C3-io- heterocycloalkyl-Ci-io-alkyl, C3-14 aryl, Cβ-u-arylalkoxy, Cβ-H-aryloxyalkoxy, Ci-io- carbonyloxy, C3-io-heterocycle-Ci-io-alkoxy and C3-14 heteroaryl, and a pharmaceutically acceptable carrier.
Embodiments of the present invention provide the use of an active compound of Formula I:
Figure imgf000007_0001
or a pharmaceutically acceptable salt or a prodrug thereof; wherein:
X and X' are each independently O or S;
Ri, R2, R3, R4, Rs, Re, R7, Rs, Rs>, Rio, Ri 1. R12, and Rj3 are each independently selected from the group s consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-10-alkyl, Ci-10-alkenyl, Ci.io-alkoxy, Ci-10-alkylsulfanyl, Ci-io-haloalkyl, C3.io-cycloalkyl, Cs-io-cycloalkyl-Ci.io-alkyl, Cs-io-heterocycloalkyl, C3-10- heterocycloalkyl-Ci-io-alkyl, C3-14 aryl, C6-i4-arylalkoxy, Cβ-u-aryloxyalkoxy, Ci-10- ! - < carbonyloxy, C3_io-heterocycle-Ci-io-alkoxy and C3-14 heteroaryl, in the manufacture of a medicament for the treatment of apoptosis-associated' disease (e.g., cancer) in a subject in need thereof.
Embodiments of the present invention provides the use of an active compound of Formula II:
Figure imgf000007_0002
or a pharmaceutically acceptable salts or a prodrug thereof; wherein: Xi is O or S; X2 is C or N;
Ri, R2, R3, R4, R5, Re, R7, Rs, R9, Rio, Rn, Ri 2, and Ri 3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-io-alkyl, Ci-io-alkenyl, Ci-io-alkoxy, Ci_io-alkylsulfanyl, Cj. io-haloalkyl, Cs-io-cycloalkyl, C3_io-cycloalkyl-Ci-io-aIkyl, Cs-w-heterocycloalkyl, C3.10- heterocycloalkyl-Ci-io-alkyl, C3-14 aryl, Cό-π-arylalkoxy, Cό-H-aryloxyalkoxy, CMO- carbonyloxy, C3.io-heterocycle-Ci-io-alkoxy and C3-14 heteroaryl, in the manufacture of a medicament for the treatment of apoptosis-associated disease (e.g., cancer) in a subject in need thereof.
The foregoing and other objects and aspects of the present invention are explained in greater detail in the specification set forth below.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 depicts a scheme for gain-of- function activity with Bcl-xL inhibitor Sl. A, fluorescence emission spectra of ANS, Bcl-xL, and the ANS-Bcl-xL complex, with Sl inhibition of ANS binding. B, cell viability after treatment with Sl for 24 h in TAMH-neo and TAMH-Bcl-xL cells. Points, mean of at least three separate experiments with quadruplicate assays; bars, SD. C, cell viability for TAMH-Bcl-xL cells treated with 10 mmol/L 2-deoxyglucose for 2 h followed by indicated concentrations of 2-MeAA for. an additional 24 h. Representative of three experiments. Points, mean of quadruplicate samples; bars, SD. D, oxygen consumption rate of TAMH-BcI -xL cells treated with Sl . Arrows, addition of 1 x 107 cells/mL, Sl (final concentration, 10 μmol/L), and KCN (final concentration, 3 mmol/L).
DETAIL DESCRIPTION OF THE INVENTION
The foregoing and other aspects of the present invention will now be described in more detail with respect to embodiments described herein. It should be appreciated that the invention can be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art. The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the embodiments of the invention and the appended claims, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. Also, as used herein, "and/or" refers to and encompasses any and all possible combinations of one or more of the associated listed items. Furthermore, the term "about," as used herein when referring to a measurable value such as an amount of a compound, dose, time, temperature, and the like, is meant to encompass variations of 20%, 10%, 5%, 1%, 0.5%, or even 0.1% of the specified amount. Unless otherwise defined, all terms, including technical and scientific terms used in the description, have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
It will be appreciated that the compounds, as described herein, may be substituted with any number of substituents or functional moieties. In general, the term "substituted1'.. ; whether preceded 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>- i-i radical of a specified substituent. When more than one position in any given structure may ibe substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. For purposes of this invention, heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms.
"Alkyl" as used herein alone or as part of another group, refers to a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms. In some embodiments, the alkyl employed in the invention contains 1 to 6 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2- dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl, n-decyl, and the like. "Lower alkyl" as used herein, is a subset of alkyl, in some embodiments preferred, and refers to a straight or branched chain hydrocarbon group containing from 1 to 4 carbon atoms. Representative examples of lower alkyl include, but are not limited to, methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, and the like. The term "akyl" or "loweralkyl" is intended to include both substituted and unsubstituted alkyl or loweralkyl unless otherwise indicated and these groups may be substituted with groups selected from halo {e.g., haloalkyl), alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocyclo, heterocycloalkyl, hydroxyl, alkoxy (thereby creating a polyalkoxy such as polyethylene glycol), alkenyloxy, alkynyloxy, haloalkoxy, cycloalkoxy, cycloalkylalkyloxy, aryloxy, arylalkyloxy, heterocyclooxy, heterocyclolalkyloxy, mercapto, alkyl-S(O)m, haloalkyl-S(O)m, alkenyl-S(O)m, alkynyl-S(O)m, cycloalkyl-S(O)m, cycloalkylalkyl-S(O)m, aryl-S(O)m, arylalkyl-S(O)m, heterocyclo-S(O)m, heterocycloalkyl-S(O)m, amino, carboxy, alkylamino, alkenylamino, alkynylamino, haloalkylamino, cycloalkylamino, cycloalkylalkylamino, arylamino, arylalkylamino, heterocycloamino, heterocycloalkylamino, disubstituted-amino, acylamino, acyloxy, ester, amide, sulfonamide, urea, alkoxyacylamino, aminoacyloxy, nitro or cyano where m= 0, 1, 2 or 3. ! . .• , "Alkenyl" as used herein alone or as part of another group, refers to a' straight or branched chain hydrocarbon containing from 1 to.lOfcarbon atoms (or in loweralkenyM to 4 • carbon atoms) which include I1 to 4 double bonds in the normal chain. In some embodiments,- the alkenyl employed in the invention contain 1 to 6 carbonatoms. Representative examples of alkenyl include, but are not limited to, vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2,4-heptadiene, and the like. The term "alkenyl" or "loweralkenyl" is intended to include both substituted and unsubstituted alkenyl or loweralkenyl unless otherwise indicated and these groups may be substituted with groups as described in connection with alkyl and loweralkyl above. "Alkynyl" as used herein alone or as part of another group, refers to a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms (or in loweralkynyl 1 to 4 carbon atoms) which include 1 to 4 triple bond in the normal chain. In some embodiments, the alkynyl employed in the invention contain 1 to 6 carbonatoms. Representative examples of alkynyl include, but are not limited to, 2-propynyl, 3-butynyl, 2- butynyl, 4-pentynyl, 3- pentynyl, and the like. The term "alkynyl" or "loweralkynyl" is intended to include both substituted and unsubstituted alkynyl or loweralknynyl unless otherwise indicated and these groups may be substituted with the same groups as set forth in connection with alkyl and loweralkyl above. "Cycloalkyl", as used herein alone or as part of another group, refers to groups having 3 to 10 carbon atoms. In some embodiments, the cycloalkyl employed in the invention have 3 to 8 carbon atoms. Suitable cycloalkyls include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like, which, as in the case of other aliphatic, heteroaliphatic or hetercyclic moieties, may optionally be substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
"Heterocycloalkyl" or "heterocycle", as used herein alone or as part of another group, refers to a non-aromatic 3-, 4-, 5-, 6-, 7-, or 8- membered ring or a polycyclic group, including, but not limited to a bi- or tri-cyclic group comprising fused six-membered rings having between one and four heteroatoms independently selected from oxygen, sulfur and nitrogen, wherein (i) the nitrogen and sulfur heteroatoms may be optionally oxidized, (ii) the nitrogen heteroatom may optionally be quaternized, and (iv) may form a spiro ring or be fused with an cycloalkyl, aryl, heterocyclic ring, benzene or a heteroaromatic ring. In some embodiments, the heterocycle employed in the invention have 3 to 10 carbon atoms. Representative heterocycles include, but are not limited to, l,4-dioxa-8-azaspiro[4.5]decane, morpholine, azetidine, azepine, aziridine, diazepine',- 1 ,3-dioxolane, dioxane, dithiane, furan, * imidazole, imidazoline, imidazolidine, isothiazole, isothiazoline, isothiazolidine, isoxazole, isoxazoline, isoxazolidine, morpholine, oxadiazole, oxadiazoline, oxadiazolidine, oxazole, oxazoline, oxazolidine, piperazine, piperidine, pyran, pyrazine, pyrazole, pyrazoline, pyrazolidine, pyridine, pyrimidine, pyridazine, pyrrole, pyrroline, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, tetrazine, tetrazole, thiadiazole, thiadiazoline, thiadiazolidine, thiazole, thiazoline, thiazolidine, thiophene, thiomorpholine, thiomorpholine sulfone, thiopyran, triazine, triazole, trithiane, benzimidazole, benzothiazole, benzothiadiazole, benzothiophene, benzoxadiazole, benzoxazole, benzofuran, benzopyran, benzothiopyran. benzodioxine, 1 ,3-benzodioxole, cinnoline, indazole, indole, indoline, indolizine, naphthyridine, isobenzofuran, isobenzothiophene, isoindole, isoindoline, isoquinoline, phthalazine, purine, pyranopyridine, quinoline, quinolizine, quinoxaline, quinazoline, tetrahydroisoquinoline, tetrahydroquinoline, thiopyranopyridine, and the like. These rings include quaternized derivatives thereof and may be optionally substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
"Aryl" as used herein alone or as part of another group, refers to a monocyclic carbocyclic ring system or a bicyclic carbocyclic fused ring system having one or more aromatic rings. In some embodiments, the aryl employed in the invention have 3 to 14 carbon atoms. Representative examples of aryl include, azulenyl, indanyl, indenyl, naphthyl, phenyl, tetrahydronaphthyl, and the like. The term "aryl" is intended to include both substituted and unsubstituted aryl unless otherwise indicated and these groups may be optionally substituted with the same groups as set forth in connection with alkyl and loweralkyl above.
"Arylalkyl" as used herein alone or as part of another group, refers to an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein. Representative examples of arylalkyl include, but are not limited to, benzyl, 2- phenylethyl, 3-phenylpropyl, 2-naphth-2-ylethyl, and the like. "Heteroaryl" as used herein alone or as part of another group, refers to a cyclic, aromatic hydrocarbon in which one or more carbon atoms have been replaced with heteroatoms such as O, N, and S. If the heteroaryl group contains more than one heteroatom, the heteroatoms may be the same or different. In some embodiments, the heteroaryl employed in the invention have 3 to 14 carbon atoms. Examples of heteroaryl groups include pyridyl, pyrimidinyl, imidazolyl, thienyl, furyl, pyrazinyl, pyrrolyl, pyranyl, isobenzofufanyl, chromenyl, xanthenyl, 'iridolyl.; isoindolyU iηdoliziήyl, triazplyl, pyridazinyl, indazolyl, .. r purinyl, quinolizinyl, isoquinolyl, quinolyl, phthalazinyl; naphthyridinyl, quinoxalinyl, isothiazolyl, and benzo[b]thienyl. In some embodiments, heteroaryl groups are five and six membered rings and contain from one to three heteroatoms independently selected from O, N, and S. The heteroaryl group, including each heteroatom, can be unsubstituted or substituted with from 1 to 4 substituents, as chemically feasible. For example, the heteroatom N or S may be substituted with one or two oxo groups, which may be shown as =0.
"Alkoxy" (or "alkyloxy"), or "thioalkyl", as used herein alone or as part of another group, refers to an alkyl or loweralkyl group, as defined herein (and thus including substituted versions such as polyalkoxy), appended to the parent molecular moiety through an oxy group, -O-, through a sulfur atom. In some embodiments, the alkoxy or thioalkyl group contains 1-10 carbon atoms. In other embodiments, the alkyl, alkenyl, and alkynyl groups employed in the invention contain 1-8 carbon atoms. In still other embodiments, the alkyl group contains 1-6 carbon atoms. In yet other embodiments, the alkyl group contains 1-4 carbon atoms. Representative examples of alkoxy, include but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, tert-butoxy, neopentoxy and n-hexoxy and the like. Representative examples of thioalkyl include, but are not limited to, methylthio, ethylthio, propylthio, isopropylthio, n-butylthio, and the like.
"Aryloxyalkoxy", as used herein alone or as part of another group, refer to an alkyl or alkoxy radical substituted with an aryl or aryloxy group and includes benzyl, phenethyl, benzyloxy, 2-phenoxyethyl and the like. In some embodiments, the aryloxyalkoxy group contains 6-14 carbon atoms.
"Arylalkoxy" as used herein alone or as part of another group, refers to an alkoxy group as defined above that is further substituted with an aryl, as defined above. In some embodiments, the arylalkoxy group contains 6-14 carbon atoms. "Carbonyloxy", as used herein alone or as part of another group, refers to the group -
C(O)-O-R, where R is hydrogen, loweralkyl, cycloalkyl, heteroclycloalkyl, amino, aryl, heteroaryl or loweraralkyl. In some embodiments, the Carbonyloxy group contains 1-10 carbon atoms.
"Halo", as used herein alone or as part of another group, refers to any suitable halogen, including -F, -Cl, -Br, and —I.
•""Amine" or "amino group", as;used<hereiri alone or as part of another group, refers^to the radical -NH2. An "optionally substituted"' amines refers to -NH2 groups wherein none, one or two of the hydrogens is replaced by a suitable substituent. Disubstituted amines may have substituents that are bridging, i.e., form a heterocyclic ring structure that includes the amine nitrogen.
"Aminoalkyl group" as used herein alone or as part of another group, refers to the radical -NHR3, where R3 is an alkyl group.
"Haloalkyl", as used herein alone or as part of another group, refers to an alkyl group, as defined above, having one, two, or three halogen atoms attached thereto and is exemplified by such groups as chloromethyl, bromoethyl, trifluoromethyl, and the like.
The term "apoptosis" refers to a regulated network of biochemical events which lead to a selective form of cell suicide, and is characterized by readily observable morphological and biochemical phenomena, such as the fragmentation of the deoxyribo-nucleic acid (DNA), condensation of the chromatin, which may or may not be associated with endonuclease activity, chromosome migration, margination in cell nuclei, the formation of apoptotic bodies, mitochondrial swelling, widening of the mitochondrial cristae, opening of the mitochondrial permeability transition pores and/or dissipation of the mitochondrial proton gradient and the like. The term "preferentially induce" apoptosis refers to at least a 5-fold greater stimulation of apoptosis, at a given concentration an agent, including a 2-methoxy antimycin derivative, in cells that over-express a Bcl-2 family member protein as compared with cells that do not over-express the Bcl-2 family member protein (e.g., a 5-fold lower LD50 or ICso). The term "substantially non- toxic" refers to an agent including a 2- methoxyantimycin that induces apoptosis in at least about 50 percent of cells that over- express a Bcl-2 family member protein, but does not induce apoptosis in more than about 5%, more preferably less than 1%, of cells that do not over-express the Bcl-2 family member protein. The term "Bcl-2 family member protein(s)" refers to an evolutionarily conserved family of proteins characterized by having one or more amino acid homology domains, BHl, BH2, BH3, and/or BH4. The Bcl-2 family member proteins include Bcl-2, Bcl-xL, Bcl-w, Al, McI-I, Bax, Bak, Bad, Bcl-xs, Bid and the like. The "Bcl-2 family member proteins" further include those proteins, or their biologically active fragments, that have at least 70%, preferably at least 80%, and more preferably at least 90% amino acid sequence identity with a Bcl-2*. family member protein.. ' . • ^
The term "anti-apoptotic Bcl-2 family member protein" refers to Bcl-2, BCI-XL, BCI- w, Al, McI-I, and other proteins characterized by having one or more amino acid homology domains, BHl, BH2, BH3, and/or BH4, and that promote cell survival by attenuating or inhibiting apoptosis. The "anti-apoptotic Bcl-2 family member proteins" further include those proteins, or their biologically active fragments, that have at least 70%, preferably at least 80%, and more preferably at least 90% amino acid sequence identity with an anti- apoptotic Bcl-2 family member protein.
The terms "identity" or "percent identity" in the context of two or more nucleic acid or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using either a PILEUP or BLAST sequence comparison algorithm (see, e.g., J. MoI. Evol. 35:351-360, 1987; Higgins and Sharp, CABIOS 5:151-153, 1989; Altschul et ai.t J. MoI. Biol. 215:403-410, 1990; Zhang et al.t Nucleic Acid Res. 26:3986-3990, 1998; Altschul et al, Nucleic Acid Res. 25:3389-
33402, 1997). Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman, Adv. Appl. Math. 2:482, 1981, by the homology alignment algorithm of Needleman and Wunsch, J. MoI. Biol. 48:443, 1970, by the search for similarity method of Pearson and Lipman, Proc. Nat. Acad. ScL USA 85:2444, 1988, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see, generally, Ausubel et al., supra). In the context of Bcl-2 family member proteins, "correspondence" of one polypeptide sequence to another sequence (e.g., regions, fragments, nucleotide or amino acid positions, or the like) is based on the convention of numbering according to nucleotide or amino acid position number, and then aligning the sequences in a manner that maximizes the number of nucleotides or amino acids that match at each position, as determined by visual inspection or by using a sequence comparison algorithm such as, for example, PELEUP (see, e.g., supra; Higgins & Sharp, supra) or BLAST (see, e.g., Altschul et al, supra; Zhang et at, supra; Altschul et al., supra). For example, a mutant Bcl-2 family member amino acid sequence having one or more amino acid substitutions^ additions, or deletions as compared to the wild-type protein may correspond to a second Bcl-2 family member amino acid sequence (e.g., the wild-type sequence or a functionally equivalent variant thereof) according to the convention for numbering the second Bcl-2 family member sequence, whereby the mutant sequence is aligned with the second Bcl-2 family member sequence such that at least 50%, typically at least 60%, more typically at least 70%, preferably at least 80%, more preferably at least 90%, and even more preferably at least 95% of the amino acids in a given sequence of at least 20 consecutive amino acids are identical. Because not all positions with a given "corresponding region" need be identical, non-matching positions within a corresponding region are herein regarded as "corresponding positions."
As used herein, a single amino acid substitution in one ("first") mutant Bcl-2 family member protein "corresponds" to a single amino acid substitution in a second mutant Bcl-2 family member protein (e.g., BCI-XL) where the corresponding substituted amino acid positions of the first and second mutant proteins are identical.
In the context of Bcl-2 family member protein mutants, the phrase "no substantial effect on tertiary protein structure relative to the corresponding wild-type Bcl-2 family member protein" or "no substantial alteration of tertiary protein structure relative to the corresponding wild-type Bcl-2 family member protein" means that, when a Ca trace providing a position for each Ca carbon of the mutant protein is superimposed onto a Ca trace of the corresponding wild-type protein and an α carbon root mean square (RMS) difference root mean square deviation (RMSD) is calculated; i.e., the deviation of the mutant structure from that of the wild-type structure), the RMSD value is no more than about 1.0 A when calculated using the same structural modeling method, typically no more than about 0.75A, even more typically no more than about 0.5 A, preferably no more than about 0.35 A, and even more preferably no more than about 0.25 A. The terms "biologically active" or "biological activity" refer to the ability of a molecule to modulate apoptosis, such as by binding to a Bcl-2 family member protein. A biologically active molecule can modulate apoptosis by causing a change in the mitochondrial protonmotive force gradient {see, e.g., Example 2); by causing a change in mitochondrial swelling or the morphological characteristics of mitochondria (see, e.g., Example 2); by affecting the release of a reporter molecule, such as, for example, rhodamine 123 or calcein, from mitochondria or vesicles (see, e.g., Examples 4 and 8) comprising a pore-forming anti-apoptotic Bcl-2 family member protein (see, e.g., Example 8); or by causing any other morphological change associated with apoptosis.
The term "effective amount" or "effective" is intended to designate a dose that causes a relief of symptoms of a disease or disorder as noted through clinical testing and evaluation, patient observation, and/or the like. "Effective amount" or "effective" further can further designate a dose that causes a detectable change in biological or chemical activity. The detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process. Moreover, "effective amount" or "effective" can designate an amount that maintains a desired physiological state, i.e.. reduces or prevents significant decline and/or promotes improvement in the condition of interest. For example, an amount of an agent that effectively modulates the apoptotic state of an individual cell such that apoptosis is induced and/or the inappropriately regulated cell death cycle in the cell returns to a normal state. As is generally understood in the art, the dosage will vary depending on the administration routes, symptoms and body weight of the patient but also depending upon the compound being administered.
The terms "therapeutically useful" and "therapeutically effective" refer to an amount of an agent that effectively modulates the apoptotic state of an individual cell such that apoptosis is induced and/or the inappropriately regulated cell death cycle in the cell returns to a normal state.
The terms "diagnostically useful" and "diagnostically effective" refer to an agent (e.g., an antimycin derivative) for detecting the induction or inhibition of apoptosis in a subject. These terms further include molecules useful for detecting diseases associated with apoptosis, or the susceptibility to such diseases, and for detecting over-expression or under- expression of a Bcl-2 family member protein.
The terms "over-expression" and "under-expression" refer to an increase or decrease, respectively, in the levels of a Bcl-2 family member protein in a cell relative to the level of such a protein found in the same cell or a closely related non-malignant cell under normal physiological conditions.
The term "apoptosis-associated disease" includes diseases, disorders, and conditions that are linked to an increased or decreased state of apoptosis in at least some of the cells of a subject. Such diseases include neoplastic disease (e.g., cancer and other proliferative diseases), tumor formation, arthritis, inflammation, autoimmune disease, human immunodeficiency virus (HIV) immunodeficiency syndrome, neurodegenerative diseases, myelodysplastic syndromes (such as aplastic anemia), ischaemic syndromes (such as myocardial infarction), liver diseases which are induced by toxins (such as alcohol), alopecia, damage to the skin due to UV light, lichen planus, atrophy of the skin, cataract, and graft rejections and other premalignant and noneoplastic hyperproliferative disorders. Apoptosis- "associated diseases further include drug resistance associated with increased or decreased levels of an anti-apoptotic Bcl-2 family member protein as well as multiple chemotherapeutic drug resistance.
"Concurrently administer " as used herein means that the two compounds or agents are administered closely enough in time to produce a combined effect (that is, concurrently may be simultaneously, or it may be two or more events occurring within a short time period before or after each other, e.g., sequentially). Simultaneous administration may be carried out by mixing the compounds prior to administration, or by administering the compounds at the same point in time but at different anatomic sites and/or by using different routes of administration.
A "combinatorial library" is a collection of compounds in which the compounds comprising the collection are composed of one or more types of subunits. The subunits can be selected from natural or unnatural moieties, including dienes, benzene compounds, cycloalkanes, lactones, dilactones, amino acids, alkanes, and the like. The compounds of the combinatorial library differ in one or more ways with respect to the number, order, type or types of modifications made to one or more of the subunits comprising the compounds. Alternatively, a combinatorial library may refer to a collection of "core molecules" which vary as to the number, type or position of R groups they contain and/or the identity of molecules composing the core molecule. The collection of compounds is generated in a systematic way. Any method of systematically generating a collection of compounds differing from each other in one or more of the ways set forth above is a combinatorial library. A combinatorial library can be synthesized on a solid support from one or more solid phase-bound resin starting materials. The library can contain five (5) or more, preferably ten (10) or more, organic molecules, which are different from each other (i.e., five (5) different molecules and not five (5) copies of the same molecule). Each of the different molecules (different basic structure and/or different substituents) will be present in an amount such that its presence can be determined by some means (e.g., can be isolated, analyzed, detected with a binding partner or suitable probe). The actual amounts of each different molecule needed so that its presence can be determined can vary due to the actual procedures used and can change as the technologies for isolation, detection and analysis advance. When the molecules are present in substantially equal molar amounts, an amount of about 100 picomoles or more can be detected. Preferred libraries comprise substantially equal molar vamounts of each desired reaction product and do not include relatively large or small amounts of any given molecules so that the presence of such molecules dominates or is completely . suppressed in any assay.
Combinatorial libraries are generally prepared by derivatizing a starting compound onto a solid-phase support (such as a bead). In general, the solid support has a commercially available resin attached, such as a Rink or Merrifield Resin, and the like. After attachment of the starting compound, substituents are attached to the starting compound. For example, the starting compound can comprise the dilactone moiety, or a precursor thereof. Substituents are added to the starting compound, and can be varied by providing a mixture of reactants comprising the substituents. Examples of suitable substituents include, but are not limited to, the following:
(1) hydrocarbon substituents, that is, aliphatic (e.g., alkyl or alkenyl), alicyclic (e.g., cycloalkyl, cycloalkenyl) substituents, aromatic, aliphatic and alicyclic-substituted aromatic nuclei, and the like, as well as cyclic substituents; (2) substituted hydrocarbon substituents, that is, those substituents containing nonhydrocarbon radicals which do not alter the predominantly hydrocarbon substituent; those skilled in the art will be aware of such radicals (e.g., halo (especially chloro and fluoro), alkoxy, mercapto, alkylmercapto, nitro, nitroso, sulfoxy, and the like); (3) hetero substituents, that is, substituents which will, while having predominantly hydrocarbyl character, contain other than carbon atoms. Suitable heteroatoms will be apparent to those of ordinary skill in the art and include, for example, sulfur, oxygen, nitrogen, and such substituents as pyridyl, furanyl, thiophenyl, imidazolyl, and the like. Heteroatoms, and typically no more than one, will be present for each carbon atom in the hydrocarbon-based substituents. Alternatively, there may be no such radicals or heteroatoms in the hydrocarbon-based substituent and it will, therefore, by purely hydrocarbon.
Methods of making combinatorial libraries are known in the art, and include for example, the following: U.S. Patent Nos. 5,958,792; 5,807,683; 6,004,617; 6,077,954.
Active Compounds
In some embodiments of the invention, active compounds are provided. In some embodiments, the active compounds may be used to modulate apoptosis in cells that over- express a Bcl-2 family member protein, which may be used to treat apoptosis-associated diseases. Examples of active compounds include compounds of Formula I and Formula II as set forth below, and including the pharmaceutically acceptable salts and prodrugs>thereof.
Formula I
Figure imgf000019_0001
wherein:
X and X' are each independently O or S;
Ri, R2, R3, R4, R5, Re, R7, Rs, R9, Rio, Riu Ri2» and R13 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-10 alkyl, d.io alkenyl, Ci-10 alkoxy, CM 0 alkylsulfanyl, Ci-10 haloalkyl, C3-10 cycloalkyl, C3.]o cycloalkyl-Ci-io alkyl, C3.10 heterocycloalkyl, C3-10 heterocycloalkyl-Ci-io alkyl, C3-U aryl, C6-i4-arylalkoxy, C6-i4-aryloxyalkoxy, Ci-I0- carbonyloxy, C3-io-heterocycle-Ci-io-alkoxy and C3.i4 heteroaryl; and pharmaceutically acceptable salts and prodrugs thereof.
In some embodiments X is O.
In some embodiments X1 is O.
In some embodiments Ri, R4, Rg, R9, and Rio are each independently preferably Ci-10- alkyl, Ci-10-alkenyl, or Ci.io-alkoxy.
In some embodiments R2, R3, R5, R7, Rs, R12 and Rj3 are each independently preferably H or halo.
In some embodiments Rn is preferably H, Ci-10-alkyl, or Ci-10-alkenyl.
In a particular embodiment of the foregoing:
X and X' are O;
Ri1Rj, Re, R9, and R]0 are each independently selected from the group consisting of Ci-4-alkyl, and Ci-4-alkenyl;
■? R2, R3, R5, R7, R8, Ri2 and Ri3 are each independently selected from the group consisting of H and halo;
Ri 1 is H or Ci-4-alkyl; or a pharmaceutically acceptable salt thereof.
An example of the foregoing is Sl (4-Methoxy-benzoic acid (3,3,6,8-tetramethyl-l- oxo-3,4-dihydro-lH-naphthalen-2-ylidene)-hydrazide), having the structure:
Figure imgf000020_0001
or a pharmaceutically acceptable salt or prodrug thereof.
Formula II
Figure imgf000021_0001
wherein:
Xi is O or S; X2 is C or N;
Ri, R2, R3, Rj, Rs, Re, R7, Rs, R9, Rio, Rn, R12, and Ri3 are each independently selected from the group consisting of: hydrogen, hydroxy, cyano, amine, nitro, halogen (e.g., chloro, fluoro, bromo, iodo), Ci-io-alkyl, Ci_io alkenyl, Ci-10 alkoxy, Cj-io alkylsulfanyl, Ci-10 haloalkyl, C3.10 cycloalkyl, C3-10 cycloalkyl-Ci-io alkyl, C3_io heterocycloalkyl, C3-Ioheterocycloalkyl-Ci-io alkyl, C3-H aryl, Cβ-i4 arylalkoxy, Cβ-u-aryloxyalkoxy, CM O- carbonyloxy, C3-io-heterocycle-Ci_io-alkoxy and C3-i4 heteroaryl; and pharmaceutically acceptable salts and prodrugs thereof.
In some embodiments Xi is O.
In some embodiments X2 is N. In some embodiments, R2 is selected from the group consisting of nitro, halogen, OH, cyano, amine, Ci-4-alkyl and Ci-4-alkenyl.
In some embodiments, Ri, R3, R5, Re, R7, Rs, R9, Rio, Ri2» and Rt3 are each independently H or halo.
In some embodiments R4 and Rn are are each independently H, Ci-10-alkyl, or Ci-10 alkenyl.
In some embodiments:
Xi is O;
X2 is N;
R2 is selected from the group consisting of Nitro, CN, halogen and Ci-4-alkyl; Ri, R3, R5, Re, R7, Rs, R9, Rio, Ri2, and R]3 are each indepently H or halo;
R4 and Rn are each independently H or Ci-4-alkyl; or a pharmaceutically acceptable salt thereof. An example of the foregoing is S2 (2-Nitro-6-(N'-quinolin-2-yl- hydrazinomethylene)-cyclohexa-2,4-dienone), having the structure:
Figure imgf000022_0001
or a pharmaceutically acceptable salt or prodrug thereof.
The compounds used in the methods of the invention may be synthesized by any suitable method known to one of skill in the art. See the examples, infra, for commercial sources for some of the compounds useful in embodiments of the invention.
Methods of Identifying Compounds that Modulate Apoptosis
Compounds that modulate apoptosis and are substantially non-toxic to cells that do not over-express a Bcl-2 family member may be identified by a method generally comprises the steps of contacting a candidate compound with a cell that over-expresses a Bcl-2 family member protein; contacting the compound with another cell that does not over-express the Bcl-2 family member protein; and determining whether the compound modulates the activity of the Bcl-2 family member protein to produce a physiological change in the cell that over- expresses the Bcl-2 family member protein, but does not produce a substantial physiological change in the cell which does not over-express that protein. Physiological changes that are indicative of binding of the candidate compound to the Bcl-2 family member protein (e.g., in the hydrophobic pocket) include an affect on cell death, cell shrinkage, chromosome condensation and migration, mitochondria swelling, and/or disruption of mitochondrial transmembrane potential (i.e., the mitochondrial proton gradient), and/or cell death (e.g., as measured by trypan dye exclusion).
Specific assay methods for identifying apoptosis-modulating compounds may be found in PCT Application No. PCT/USOO/22891, the content of which is herein incorporated by reference in its entirety. Biologically active compounds can also be identified by evaluating the ability of the agents to modulate glucose uptake and/or lactate production in cells expressing an anti-apoptotic Bcl-2 family member protein. Apoptosis-modulating compounds increase cellular glucose uptake or lactate production in proportion to the level of expression of a Bcl-2 family member target protein. Methods for assaying glucose production or lactate production are well-known in the art.
Combinatorial libraries of test compounds can be screened for biological activity using any of the assay methods described above. Combinatorial libraries and processes are described, for example, in PCT/USOO/22891. We deposited chemical structures for commercially available small organic molecules from two-dozen chemical vendors in a single screening database. This database consists of over 5.7 million small organic molecules, with the following filters applied: (a) deprotonation of acids and protonatation of bases; (b) omission of compounds with transition metals; (c) omission compounds with 9- membered rings or larger; (d) omission compounds with d-hybridized atoms. Some of the larger chemical vendors that provide these compounds are Asinex (347,725 compounds), ChemBridge (411,532), ChemDiv (493,524 compounds), SPECS (261,875 compounds), Timtec (216,277 compounds), and CNC-2D (4,062,088 compounds).
Computer-based methods may also be used to identify biologically active compounds by using a "molecular docking" algorithm to score test compounds for binding to each of a* Bcl-2 family member protein and a corresponding mutant protein as described ^ supra. Those test compounds that demonstrate a lower score for binding to the mutant protein relative to the corresponding Bcl-2 family member protein (e.g., a mutant BcI- XL protein (having a E92L, F97W, L130A, A142L, F146L, or Y195G mutation) and the wild- type BcI- XL protein, respectively) can be further evaluated in biological assays as described supra to verify biological activity.
Computer-based techniques for examining potential ligands (e.g., candidate compounds) for binding to target molecules are well-known in the art. (See, e.g., Kuntz et al, J. MoI. Biol. 161:269-288, 1982; Kuntz, Science 257:1078-1082, 1992; Ewing and Kuntz, J. Comput. Chem. 18:1175-1189, 1997). For example, the DOCK suite of programs is designed to find possible orientations of a ligand in a receptor site. See, e.g., Ewing and Kuntz, supra. The orientation of the ligand is evaluated with a shape scoring function (an empirical function resembling the van der Waals attractive energy) and/or a function approximating the ligand-receptor binding energy (which is taken to be approximately the sum of the van der Waals and electrostatic interaction energies). After an initial orientation and scoring evaluation, a grid-based rigid body minimization is carried out for the ligand to locate the nearest local energy minimum within the receptor binding site. The position and conformation of each docked molecule can be optimized, for example, using the single anchor search and torsion minimization method of DOCK4.0. {See, e.g., Ewing and Kuntz, supra; Kuntz, supra.)
In addition, heuristic docking and consensus scoring strategies can be used in the computer-based identification of biologically active compounds (i.e., different docking and scoring methods can be applied to evaluate the screening results). For example, following a primary screening using, e.g. , DOCK4.0 (supra), top-scoring compounds can be re-scored using other docking algorithms such as, for example, GOLD, FlexX, PM (see Muegge and Martin, J. Med. Chem. 42:791, 1999, and/or AutoDock3.0 (see Morris et al, J. Comput. Chem. 19:1639-1662, 1998). Optionally, following a primary and any subsequent screen(s) using individual docking algorithms, a consensus score (Cscore) can be determined by combining results from any of the individual docking programs used to score the candidate compounds (see Clark et al, J. MoI. Graph Model 20:281-295, 2002). Based on the scoring results from a secondary or other subsequent screen, a subset, for example, of the top-scoring molecules from the primary screen can be selected for further analyses (e.g., a tertiary virtual screen or, alternatively or additionally, biological screening assays such as, for example, any of the assays described herein or otherwise known in the art).
Similarity Searching/ Pharmacophore Mapping
2D database similarity searching may be performed using BITMACCS within MOE, and 2Dph, an in-house 2D-pharmacophore-based program, to search for compounds similar to 2-MeAAl, Sl and troglitazone (setting the threshold for the percentage of similarity at >65%) from our screening database. The final "hits" of commercially available compounds may then be saved into the separate databases. Anther approach may focus on partial and whole 3D pharmaophore mapping/ searching with potential bioisosteric replacements of sub- group(s) of our identified initial hits based on a topological pharmacophore description of the fragments. For examples, the 5-benzylthiazolidine-2,4-dione moiety of troglitazone can be substituted by various 2-oxy-3-arylcarboxylic acids, and the 9-membered ring of 2-MeAAl could be replaced by two 5- or 6-merbered rings. The Bioster database (39) (Accelrys, San Diego, CA) can be used to search for the most promising replacements, and focused virtual libraries can be built. This method has been used in the pharmaceutical industry to improve the ADME/Tox properties of initial promising lead compounds and for broadening patentability (40). Docking
The BH3 peptide-bound Bcl-xL structure, PDB Code: IBXL and unliganded Bcl-xL structure, PDB Code: 1R2D are used as target receptors. The Flexx 2.0.1 (41) docking program, a fully automatic docking tool for flexible ligands is used to dock the ligands into their binding sites within the receptor structures. All small molecule compounds are prepared by Corina and MOE. After removing all water molecules, the active sites are dissected from the NMR structure of the protein complexes by including all residues that have at least one single (heavy) atom within a distance of 6.5 A from any heavy atoms of the ligand. Residues are kept fixed in their NMR coordinates in all docking experiments. This docking method consists of three steps: the selection of a part of the molecule, the base fragment, the placement of the base fragment into the active site of a protein, and the subsequent reconstruction of the complete drug molecule by linking the remaining components step by step. For placing base fragments, two algorithms are in use. The first one superposes triples of interaction centers of a base fragment with triples of compatible interaction points in the active site. If a base fragment has fewer than three interaction centers or if the number of placements is too low, the second algorithm, called line matching, is started. This algorithm matches pairs of interaction centers with pairs of interaction points. Because of geometric ambiguity, multiple placements are generated by rotation around the axis defined by the interaction points and centers. Both base placement algorithms generate a large number of solutions. Sampling is done with 400 solutions per partial solution at each iteration of incremental construction. A reduction by clash tests and clustering follows. All FlexX parameters are set to their default standard values, and only the 10 best scored poses by FlexX are used for further ranking. The final ranking of docking results is performed with an in-house novel scoring-function, VMScore (see below).
Novel score-function, VMScore, in target structure-based modeling and prediction.
The docking-score is the key component in structure-based drug design. The score- function is used for estimating the binding affinity of the ligand with docked conformation within the binding packet. It will help to decide which compound(s) should be selected for experimental development. Score-functions which derived directly from molecular mechanics are not often used in practice largely due to the time-consuming nature of the process and high demand for computational power for a large set of molecules. In the past years, several (semi) empirically based score-functions have been used in the ranking of docked structures. "Score" (30) and "Chemscore" (31) use contributions from hydrogen bonding, metal-ligand contacts, lipophilic contact area and frozen rotatable bonds. The "HINT Score" use contributions representing different types of surface areas (32). Score- functions based on the probability of pair-wise contact of heavy atoms such as "PMF" (potential of mean force) (33) and "Smog" (34) have reported. However, these score- functions do not consider all possible energy contributions for binding. For examples, "Score" and "Chemscore" do not consider the contribution of the restricted conformation of the bound ligand compared to the free molecule; "PMF" and "Smog" do not consider contributions from hydrogen bonding. In addition, most of the current score-functions do not take desolvation energy into account.
We have developed a novel score-function -"VMScore" by a method of considering all possible energy contributions including those from electrostatic, van de waal's, hydrogen bonding, conformation change, desolvation and entropy. It is given as,
ΔG = A*ΔGe]e + B*ΔGvdw + C* ΔGhb + D*ΔGCOnf + E*ΔGdeSoi + F*ΔGent
-v;
Where, ΔGe|e is free energy contribution from electrostatic interaction. ΔGvdw is free energy contribution from van de waal's interaction. ΔGhb is free energy contribution from hydrogen bonding, ΔGCOnf is free energy contribution from conformation change, ΔGdesoι is free energy contribution from desolvation energy difference between ligand, protein and their complex, ΔGem is entropy contribution from translation and rotation, and A to F are coefficients. To test and validate our new score-function, "VMScore", we have taken a well- described data set containing 53 complex (protein bound with compound) crystal structures with experimental binding affinity (Ki) data (ΔG oc logKi) as training set (32). Our results, shown in Fig.10, Left, fit the experimental data with a squared correlation coefficient, R2 = 0.77, and with LOO (leave-one-out) cross-validation, Q2 = 0.70. These results are much better than the reported "HINT Score" with R2 = 0.54 for the same training set.
Figure imgf000027_0001
Left: VMScore result for the training set with 53 complex structures, R2 = 0.77, with cross- validation prediction (leave-one-out) Q2 = 0.70.
Right: VMScore prediction for the testing set of Endothiapepsin complex dataset, R2 = ad< 0.62. small molecules in this data set contain multiple rotatable bonds, and both hydrogen bond donors and acceptors and are thus very flexible. This dataset has been widely used as testing, set for currently available score-functions. The prediction results based on currently available score-functions are listed in*Table-4. None of these (squared) correlation coefficients is larger than 0.3. The poor predictions mainly derive from lack of consideration of either conformation change and/or hydrogen bonding. By using our "VMScore" score- function, a (squared) correlation coefficient, R2 = 0.62 Has been achieved for the same dataset (Fig.10, Right and Table-4), which represents a significant improvement over the current available score-functions.
Table-4. The prediction results of different docking score-functions for the same Endothiapepsin complex testing set.
Figure imgf000027_0002
Methods of Using the Apoptosis-Modulating Compounds
The compounds of the present invention are useful for treating cells in which the cell death signal is down-regulated and the affected cell has an inappropriately diminished propensity for cell death, which is referred to herein as being in a "decreased apoptotic state." The invention further provides methods for the administration to a subject of a therapeutically effective amount of an apoptosis-modulating compound of the invention to treat an apoptosis- associated disease in which it is desirable to induce apoptosis in certain types of cells, such as virus-infected or autoantibody-expressing cells.
In some embodiments, a method of treating a cancer characterized by the over- expression of a Bcl-2 family member is provided. Examples of cancers known to be associated with over-expression of a Bcl-2 family member and which can be treated according to the methods provided herein are shown in Table 4.
Table 4. Percentages of common human cancers with elevated levels of Bcl-2 or BCI-XL expression
Tumor Bcl-2 BCI-XL
Lymphoma Hodgkπϊs - 47-65% Hodgkin's - 48-94% NHL - 9-57% NHL - 25-45%
Leukemia AML - 13-20% AML - 38% ALL - 89-92% ALL - 13% CML - 33-54% CLL - 70-95%
Myeloma 43% 29%
Lum NSCLC-squambus - 25% Most NSCLC, SCLC adenoca - 12% SCLC - 83-90%
Colorectal Adenoma - 65-98% Adenoma - 50% Carcinoma - 46-60% Carcinoma - 60%
Breast 80% 43-75%
Pancreas 23% 88%
Urogenital Bladder - 24% Bladder - 80.9% Renal - 53% Renal - 38%
Liver Rare 95+%
Ovary 30-39% 62%
Brain Medulloblastoma - 5-25% Medulloblastoma - 56% Glioma - 28-92% Glioma - 98% Oligodendroglioma -16-60% Oligodendroglioma - <5% Esophagus SCC - 45% Adenocarcinoma - 90%
Adenocarcinoma - 20-40%
In some cases, the treatment of the cancer can include the treatment of solid tumors or the treatment of leukemias. For example, the cancer can be of the skin, breast, brain, cervix, testis, and the like. More particularly, cancers may include, but are not limited to, the following organs or systems: cardiac, lung, gastrointestinal, genitourinary tract, liver, bone, nervous system, gynecological, hematologic, skin, and adrenal glands. More particularly, the methods herein can be used for treating gliomas (Schwannoma, glioblastoma, astrocytoma), neuroblastoma, pheochromocytoma, paraganlioma, meningioma, adrenal cortical carcinoma, kidney cancer, vascular cancer of various types, osteoblastic osteocarcoma, prostate cancer, ovarian cancer, uterine leiomyomas, salivary gland cancer, choroid plexus carcinoma, mammary cancer, pancreatic cancer, colon cancer, B and T cell lymphomas, acute and chronic myeloid or lymphoid leukemias, and multiple myeloma. Further, treatment may include pre-malignant conditions associated with any of the above cancers (e.g., colon adenomas, myelodysplastic syndrome).' In some embodiments, methods of treating a neurodegenerative disease characterized by the over-expression of a Bcl-2 family member are provided. Neurodegenerative diseases include Alzheimer's disease', Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis and other diseases linked to degeneration of the brain, such as Creutzfeldt- Jakob disease and expanded polyglutamine repeat diseases. Expanded polyglutamine repeat diseases with which the present invention is concerned include, but are not limited to, Huntington's disease, dentatorubral pallidoluysian atrophy, spinobulbar muscular atrophy, and spinocerebellar ataxia types 1, 2, 3, 6 and 7. See, e.g., US Patent No. 6,632,616 to Burke et al.
In some embodiments, methods of treating arthritis, inflammation, autoimmune diseases, human immunodeficiency virus (HIV) immunodeficiency syndrome, myelodysplastic syndromes (such as aplastic anemia), ischaemic syndromes (such as myocardial infarction), liver diseases which are induced by toxins (such as alcohol), alopecia, damage to the skin due to UV light, lichen planus, atrophy of the skin, cataract, and graft rejections are provided. Typically, the compounds used in embodiments of the invention will be substantially purified prior to administration. The subject can be an animal, including, but not limited to, cows, pigs, horses, chickens, cats, dogs, and the like, and is typically a mammal, and in a particular embodiment human. In another specific embodiment, a non- human mammal is the subject.
Various delivery systems are known and can be used to administer a compound of the invention, such as, for example, encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of producing the derivative, receptor-mediated endocytosis {see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432, 1987), and the like. The apoptosis-modulating compounds are administered as therapeutic or pharmaceutical compositions by any suitable route known to the skilled artisan including, for example, intravenous, subcutaneous, intramuscular, intradermal, transdermal, intrathecal, intracerebral, intraperitoneal, intransal, epidural, and oral routes. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulations. For treating tissues in the central nervous system, administration can be by injection or infusion into the cerebrospinal fluid (CSF). When it is intended that a compound be administered to cells in the central nervous system, administration can be with one or more other components capable of promoting penetration of the derivative across the blood-brain barrier. In addition, it can be desirable to introduce a compound into the target tissue by any suitable route, including intravenous and intrathecal injection. Pulmonary administration can . also be employed, such as, for example, by use of an inhaler or nebulizer, and formulation of the compound with an aerosolizing agent. In certain embodiments, the compound is coadministered with an inhibitor of esterase activity to further stabilize the compound. Pharmaceutical compositions can also be administered orallyάn any orally acceptable dosage form including, but not limited to, capsules, tablets, caplets, lozenges, aqueous suspensions or solutions. In the case of tablets for oral use, carriers, which are commonly used, include lactose and corn starch. Lubricating aids, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required, the agent can be combined with emulsifying and suspending aids. If desired, certain sweeteners, flavorants, or colorants can also be used. Further, the compounds of the present invention can be combined with any other tumor and/or cancer therapy. The therapy can include, for example and not by way of limitation, surgery, radiation, and chemotherapy either individually or in any combination. Chemotherapy can include any current known or yet to be discovered chemotherapeutic agent including but are not limited to Aceglatone; Aclarubicin; Altretamine; Aminoglutethimide; 5-
Aminogleavulinic Acid; Amsacrine; Anastrozole; Ancitabine Hydrochloride; 17-1 A
Antibody; Antilymphocyte Immunoglobulins; Antineoplaston AlO; Asparaginase;
Pegaspargase; Azacitidine; Azathioprine; Batimastat; Benzoporphyrin Derivative; Bicalutamide; Bisantrene Hydrochloride; Bleomycin Sulphate; Brequinar Sodium;
Broxuridine; Busulphan; Campath-IH; Caracemide; Carbetimer; Carboplatin; Carboquone;
Carmofur; Carmustine; Chlorambucil; Chlorozotocin; Chromomycin; Cisplatin; Cladribine;
Corynebacterium parvum; Cyclophosphamide; Cyclosporin; Cytarabine; Dacarbazine;
Dactinomycin; Daunorubicin Hydrochloride; Decitabine; Diaziquone; Dichlorodiethylsulphide; Didemnin B.; Docetaxel; Doxifluridine; Doxorubicin Hychloride;
Droloxifene; Echinomycin; Edatrexate; Elliptinium; Elmustine; Enloplatin; Enocitabine;
Epirubicin Hydrochloride; Estramustine Sodium Phosphate; Etanidazole; Ethoglucid;
Etoposide; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine
Phosphate; Fluorouracil; Flutamide; Formestane; Fotemustine; Gallium Nitrate; Gencitabine; Gusperimus; Homoharringtonine; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide;
• Ilmofosine; Improsulfan Tosylate; Inolimomab; Interleukin-2; Irinotecan; JM-216; Letrozole;
Lithium Gamolenate; Lobaplatin; Lomustine; Lonidamine; Mafosfamide; Meiphalan;
Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Miboplatin; Miltefosine;
Misonidazole; Mitobronitol; Mitoguazone Dihydrochioride; Mitolactol; Mitomycin; Mitotane; Mitozanetrone Hydrochloride; Mizoribine; Mopidamol; Muitlaichilpeptide;
Muromonab-CD3; Mustine Hydrochloride; Mycophenolic Acid; Mycophenolate Mofetil;
Nedaplatin; Nilutamide; Nimustine Hydrochloride; Oxaliplatin; Paclitaxel; PCNU;
Penostatin; Peplomycin Sulphate; Pipobroman; Pirarubicin; Piritrexim Isethionate;
Piroxantrone Hydrochloride; Plicamycin; porfimer Sodium; Prednimustine; Procarbazine Hydrochloride; Raltitrexed; Ranimustine; Razoxane; Rogletimide; Roquinimex; Sebriplatin;
Semustine; Sirolimus; Sizofiran; Sobuzoxane; Sodium Bromebrate; Sparfosic Acid;
Sparfosate Sodium; Sreptozocin; Sulofenur; Tacrolimus; Tamoxifen; Tegafur; Teloxantrone
Hydrochloride; Temozolomide; Teniposide; Testolactone; Tetrasodium
Mesotetraphenylporphine-sulphonate; Thioguanine; Thioinosine; Thiotepa; Topotecan; Toremifene; Treosulfan; Trimetrexate; Trofosfamide; Tumor Necrosis Factor; Ubenirnex;
Uramustine; Vinblastine Sulphate; Vincristine Sulphate; Vindesine Sulphate; Vinorelbine
Tartrate; Vorozole; Zinostatin; Zolimomab Aritox; and Zorubicin Hydrochloride, and the like, either individually or in any combination. See, e.g., US Patent No. 7,071,158. In some embodiments, it can be desirable to administer the compounds of the invention locally to the area in need of treatment; this administration can be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application (e.g., in conjunction with a wound dressing after surgery), by injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non- porous, or gelatinous material, including membranes such as silastic membranes, or fibers. In one embodiment, administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre-neoplastic tissue.
In another embodiment, the compounds of the invention can be delivered in a vesicle, in particular a liposome (see, e.g., Langer, Science 249:1527-1533, 1990; Treat et al., In Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365, 1989; Lopez-Berestein, supra, pp. 317-327).
In yet another embodiment, the compounds of the invention can be delivered in a controlled release system. In one embodiment, a pump can be used (see, e.g., Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201, 1987; Buchwald et ah, Surgery 88:507, 1980; Saudek et al., N. Engl. J. Med. 321:574, 1989). In another embodiment, polymeric materials can be used (see, e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida, 1974; Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York, 1984; Ranger and Peppas, J. Macromol. ScL Rev. Macromol. Chem. 23:61, 1983; see also Levy et al., Science 228:190, 1985; During et al., Ann. Neurol. 25:351, 1989; Howard et al, J. Neurosurg. 71 : 105, 1989). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, supra, Vol. 2, pp. 115-138, 1984). Other controlled release systems are discussed in, for example, the review by Langer (Science 249:1527-1533, 1990).
The methods of treatment and the pharmaceutical compositions described herein can be combined with any other tumor and/or cancer therapy. The therapy can include, for example and not by way of limitation, surgery, radiation, and chemotherapy either individually or in any combination. Chemotherapy can include any current known or yet to be discovered chemotherapeutic agent including, for example, paclitaxel, doxorubicin, etoposide, melphalan, daunorubicin, 5-fluorouracil, cisplatin, paraplatin, and the like, either individually or in any combination. Furether, in some embodiments, the methods of treatment as described herein can include concurrently administering one or more additional inducer of apoptosis (for example, staurosporine), and compositions as described herein can optionally include one or more additional inducer of apopotosis. Yet, in another embodiment, the methods of treatment and compositions as described herein can be used in selectively targeting cancer cells (for example, Bcl-xL overexpressing cells such as TAMH-Bcl-xl cells).
The present invention also provides pharmaceutical compositions. Such compositions comprise a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of the invention. The term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more typically in humans. The term "carrier" refers to a diluent, adjuvant, excipient, stabilizer, vehicle, or any combination thereof, with which the agent is formulated for administration. Pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water is a typical carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose,, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. Pharmaceutical compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations, and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. In addition, in certain embodiments, the pharmaceutical composition includes an inhibiter of esterase activity as a stabilizing agent.
Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutical carriers are described in, for example, Remington 's Pharmaceutical Sciences, by E.W. Martin. Such compositions will contain a therapeutically effective amount of a compound of the invention, typically in purified form, together with a suitable amount of carrier so as to provide a formulation proper for administration to the subject. The formulation should suit the mode of administration.
In one embodiment, the compound of the present invention is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, pharmaceutical compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition can also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form. For example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration. The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, ' procaine, and the like.
The compounds of the invention can be formulated as neutral or salt forms. A "pharmaceutically acceptable salt" as used herein refers to a salt form of a compound permitting its use or formulation as a pharmaceutical and which retains the biological effectiveness of the free acid and base of the specified compound and that is not biologically or otherwise undesirable. Examples of such salts are described in Handbook of
Pharmaceutical Salts: Properties, Selection, and Use, Wermuth, CG. and Stahl, P.H. (eds.), Wiley- Verlag Helvetica Acta, Zurich, 2002 [ISBN 3-906390-26-8]. Examples of pharmaceutically acceptable salts, without limitation, include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those formed with free carboxyl groups such as those derived from sodium- potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine and the like. Examples of salts also include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogen phosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycollates, tartrates, methanesulfonates, ethane sulfonates, propanesulfonates, toluenesulfonates, naρhthalene-1 -sulfonates, naρhthalene-2-sulfonates, and mandelates. In some embodiments, pharmaceutically acceptable salt includes sodium, potassium, calcium, ammonium, trialkylarylammonium and tetraalkylammonium salts.
Furthermore, pharmaceutically acceptable prodrugs or "softdrugs" of the compounds may be used in embodiments of the invention. Pharmaceutically acceptable prodrugs as used herein refers to those prodrugs of the active compounds of the present invention 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, commensurate with a reasonable risk/benefit ratio, and effective for their intended use? as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Prodrugs as Novel delivery Systems, Vol. 14 of the A.C.S. Symposium Series and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are . incorporated by reference herein. See also US Patent No. 6,680,299 Examples include a prodrug that is metabolized in vivo by a subject to an active drug having an activity of active compounds as described herein, wherein the prodrug is an ester of an alcohol or carboxylic acid group, if such a group is present in the compound; an acetal or ketal of an alcohol group, if such a group is present in the compound; an N-Mannich base or an imine of an amine group, if such a group is present in the compound; or a Schiff base, oxime, acetal, enol ester, oxazolidine, or thiazolidine of a carbonyl group, if such a group is present in the compound, such as described in US Patent No. 6,680,324 and US Patent No. 6,680,322.
The amount of the compound of the invention that is combined with the carrier to produce a single dosage form will vary, depending upon the nature of that agent and the composition of the dosage form. It should be understood, however, that a specific dosage and treatment regime for any particular patient or disease state will depend upon a variety of factors, including the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the judgment of the treating physician, and the severity of the particular disease being treated. The amount of active agent will also depend upon the 5 specific activity of the compound and whether that agent is co-administered with any other therapeutic or prophylactic ingredients. Determination of therapeutically effective dosages is typically based on animal model studies and is guided by determining effective dosages and administration protocols that significantly reduce the occurrence or severity of the apoptosis- associated disease in model subjects (e.g., in the case of treatment of malignancies, a tumor0 xenograft model in mice can be used (see, e.g., Example 20). For treatment of human subjects, such animal model studies are typically followed up by human clinical trials. A non-limiting range for a therapeutically effective amount of the compounds is about 0.001 mg/kg and about 100 mg/kg body weight per day, and in more specific embodiments between about 0.001 mg/kg and about 50 mg/kg, between about 0.01 mg/kg and about 20 mg/kg. >5 between about 0.1 and about 10 mg/kg, or between about 0.1 mg/kg and about 5 mg/kg body weight per day.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form0 prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use, or sale for human administration.
. The following examples are provided merely as illustrative of various aspects of the invention and shall not be construed to limit the invention in any way. 5
EXAMPLES EXAMPLE 1
The selectivity and IC50 for the following compounds was determined (in Example 2): Sl : 4-Methoxy-benzoic acid (3,3,6,8-tetramethyl-l -oxo-3, 4-dihydro-l H-naphthalen-0 2-ylidene)-hydrazide (provided by National Cancer Institute)
Figure imgf000037_0001
S2: 2-Nitro-6-(N'-quinolin-2-yl-hydrazinomethylene)-cyclohexa-2,4-dienone (provided by National Cancer Institute)
Figure imgf000037_0002
Example 2
The effect of the compounds described in Example 1 on TABX2S cells (cells over- expressing BCI-XL) and TAMH.neo (control) cells are summarized below in Table 5. Table 5 includes dose dependence of TABX2S cell survival (IC50) and the selective induction of apoptosis in TABX2S cells over the TAMH.neo control.
Table S
Compound IC50 (TABX2S) Selectivity (IC50 TABX2S/1C50 Neo)
Sl 14 >20
S2 11 3.3
Example 3
The gain-of-function activity with Bcl-xL inhibitor Sl was illustrated in Figure 1. It shows that Sl binds to the Bcl-xL hydrophobic groove (See Fig. 1, A) and shows an EC50 of ~6 to 10 μmol/L for Bcl-xL-overexpressing TAMH cells, without cytotoxicity for control TAMH-neo cells at these concentrations (See Fig. I5 B). Pretreatment with 2-deoxyglucose sensitized TAMH-Bcl-xL cells to Sl {See Fig. 1, C). Sl has no detectable effect on mitochondrial oxygen consumption rate at cytotoxic concentrations (See Fig. 1, D).
The foregoing is illustrative of the present invention and is not to be construed as limiting thereof. Although a few exemplary embodiments of this invention have been described, those skilled in the art will readily appreciate that many modifications are possible in the exemplary embodiments without materially departing from the novel teachings and advantages of this invention. Accordingly, all such modifications are intended to be included within the scope of this invention as defined in the claims. Therefore, it is to be understood that the foregoing is illustrative of the present invention and is not to be construed as limited to the specific embodiments disclosed, and that modifications to the disclosed embodiments, as well as other embodiments, are intended to be included within the scope of the appended claims. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims

WHAT IS CLAIMED IS:
1. A method for treating an apoptosis-associated disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an active compound having the structure of
Figure imgf000039_0001
Figure imgf000039_0002
or a pharmaceutically acceptable salt or prodrug thereof.
2. The method of Claim 1, wherein said compound modulates apopotosis by binding to a Bcl-2 family member protein.
3. The method of Claim 1, wherein said Bcl-2 family member protein is Bcl-2 or BcI-
XL.
4. The method of Claim 1, wherein said apoptosis-associated disease is a neoplastic disease.
5. The method of Claim 1, wherein the neoplastic disease is a cancer.
6. The method of Claim 5, wherein the cancer comprises a solid tumor.
7. The method of Claim 5, wherein the cancer comprises leukemia.
8. The method of Claim 1, further comprising concurrently adminsterting to said subject another inducer of apopotosis.
9. The method of Claim 1, wherein said another inducer of apopotosis trigger BCI-XL dependent apoptotic pathways.
10. A pharmaceutical composition comprising an active compound having the structure of
Figure imgf000040_0001
Figure imgf000040_0002
or a pharmaceutically acceptable salt or prodrug thereof in a pharmaceutically acceptable carrier.
11. The composition of Claim 10, further comprising an inducer of apopotosis.
12. The compsition of Claim 11, wherein said another inducer of apopotosis trigger
BCI-XL dependent apoptotic pathways.
13. The use of an active compound having the structure of
Figure imgf000040_0003
Figure imgf000041_0001
or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of apoptosis-associated disease in a subject in need thereof.
PCT/US2007/017752 2006-08-10 2007-08-10 Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins WO2008021211A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US83691806P 2006-08-10 2006-08-10
US60/836,918 2006-08-10
US94880807P 2007-07-10 2007-07-10
US60/948,808 2007-07-10

Publications (2)

Publication Number Publication Date
WO2008021211A2 true WO2008021211A2 (en) 2008-02-21
WO2008021211A3 WO2008021211A3 (en) 2008-07-31

Family

ID=39082620

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/017752 WO2008021211A2 (en) 2006-08-10 2007-08-10 Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins

Country Status (1)

Country Link
WO (1) WO2008021211A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011041737A2 (en) 2009-10-02 2011-04-07 Fred Hutchinson Cancer Research Center Gain-of-function bcl-2 inhibitors
WO2011041731A2 (en) 2009-10-02 2011-04-07 Fred Hutchinson Cancer Research Center Method of inhibiting bcl-2-related survival proteins
WO2013021384A1 (en) 2011-08-11 2013-02-14 Yeda Research And Development Co. Ltd Compositions and methods for modulating apoptosis

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4997835A (en) * 1987-05-14 1991-03-05 Schaper Klaus Juergen Substituted 2-acylpyridine-α-(N)-hetarlyhydrazones and medicaments containing the same
US5700823A (en) * 1994-01-07 1997-12-23 Sugen, Inc. Treatment of platelet derived growth factor related disorders such as cancers
US6048703A (en) * 1996-11-15 2000-04-11 Cephalon, Inc. Methods for detecting cell apoptosis
US6566395B1 (en) * 1999-05-25 2003-05-20 Biomedicines, Inc. Methods of treating proliferative disorders
US6939979B2 (en) * 2003-11-10 2005-09-06 Scientific Design Co., Inc. Oxidation process and catalyst

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4997835A (en) * 1987-05-14 1991-03-05 Schaper Klaus Juergen Substituted 2-acylpyridine-α-(N)-hetarlyhydrazones and medicaments containing the same
US5700823A (en) * 1994-01-07 1997-12-23 Sugen, Inc. Treatment of platelet derived growth factor related disorders such as cancers
US6048703A (en) * 1996-11-15 2000-04-11 Cephalon, Inc. Methods for detecting cell apoptosis
US6566395B1 (en) * 1999-05-25 2003-05-20 Biomedicines, Inc. Methods of treating proliferative disorders
US6939979B2 (en) * 2003-11-10 2005-09-06 Scientific Design Co., Inc. Oxidation process and catalyst

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011041737A2 (en) 2009-10-02 2011-04-07 Fred Hutchinson Cancer Research Center Gain-of-function bcl-2 inhibitors
WO2011041731A2 (en) 2009-10-02 2011-04-07 Fred Hutchinson Cancer Research Center Method of inhibiting bcl-2-related survival proteins
WO2013021384A1 (en) 2011-08-11 2013-02-14 Yeda Research And Development Co. Ltd Compositions and methods for modulating apoptosis

Also Published As

Publication number Publication date
WO2008021211A3 (en) 2008-07-31

Similar Documents

Publication Publication Date Title
WO2008021250A2 (en) Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins
KR102111704B1 (en) Bivalent inhibitors of iap proteins and therapeutic methods using the same
CN110114075B (en) Disulfide-containing cell penetrating peptides and methods of making and using the same
CN106029639B (en) Treat 2- amino -3,5 of the BACE1 inhibitor of Alzheimer disease, 5- tri- fluoro- 3,4,5,6- tetrahydropyridines
CN105980388B (en) Compound
US9233086B2 (en) Inhibition of WDR5 interaction with its binding partners
AU2014203516A1 (en) Compositions and methods for treatment of leukemia
CN107108637A (en) Triazolopyrimidine compound and application thereof
RU2728789C2 (en) Iap inhibitors
CN105246896A (en) Pyrazolo-pyrrolidin-4-one derivatives and their use in the treatment of disease
US8865901B2 (en) Gain-of-function Bcl-2 inhibitors
WO2010077589A2 (en) Stat3 inhibitors and therapeutic methods using the same
WO2011041737A2 (en) Gain-of-function bcl-2 inhibitors
CN106536524A (en) [1,2,4]triazolo[4,3-b]pyridazines for use in the treatment of proliferative diseases
MX2015002310A (en) Novel phenyl-pyridine/pyrazine amides for the treatment of cancer.
WO2008021211A2 (en) Compositions and methods for modulating apoptosis in cells over-expressing bcl-2 family member proteins
Wu et al. Small molecules targeting protein–protein interactions for cancer therapy
Dessai et al. Design, synthesis, graph theoretical analysis and molecular modelling studies of novel substituted quinoline analogues as promising anti-breast cancer agents
CN112920176B (en) Bifunctional compound capable of inducing PRC2 protein complex core subunit degradation, pharmaceutical composition and application
WO2011041731A2 (en) Method of inhibiting bcl-2-related survival proteins
US8980838B2 (en) Cyclized peptidomimetic small molecule inhibitors of the WDR5 and MLL1 interaction
Li et al. Discovery of triazinone derivatives as novel, specific, and direct NLRP3 inflammasome inhibitors for the treatment of DSS-induced ulcerative colitis
US20160095873A1 (en) Compositions and methods for transient up-regulation of myc in b-cell lymphomas for enhancing p53 independent apoptotic responses to chemotherapy
Barkhudaryan et al. Study of molecular mechanisms of anti-tumor effect of hemorphin-7 in vivo
CN105712945A (en) 2-Substituted-oxy-5-methylsulfonyl aryl piperazine acidamide analogue and preparation method and application 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: 07811230

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: 07811230

Country of ref document: EP

Kind code of ref document: A2