WO2006128173A2 - Selective apoptotic induction in cancer cells including activation of procaspase-3 - Google Patents

Selective apoptotic induction in cancer cells including activation of procaspase-3 Download PDF

Info

Publication number
WO2006128173A2
WO2006128173A2 PCT/US2006/020910 US2006020910W WO2006128173A2 WO 2006128173 A2 WO2006128173 A2 WO 2006128173A2 US 2006020910 W US2006020910 W US 2006020910W WO 2006128173 A2 WO2006128173 A2 WO 2006128173A2
Authority
WO
WIPO (PCT)
Prior art keywords
procaspase
compound
pac
cell
cells
Prior art date
Application number
PCT/US2006/020910
Other languages
French (fr)
Other versions
WO2006128173A3 (en
Inventor
Paul J. Hergenrother
Karson S. Putt
Grace W. Chen
Jennifer M. Pearson
Original Assignee
The Board Of Trustees Of The University Of Illinois
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 The Board Of Trustees Of The University Of Illinois filed Critical The Board Of Trustees Of The University Of Illinois
Priority to ES06771588T priority Critical patent/ES2763156T3/en
Priority to AU2006251978A priority patent/AU2006251978A1/en
Priority to EP06771588.8A priority patent/EP1893210B1/en
Priority to DK06771588.8T priority patent/DK1893210T3/en
Priority to MX2007014843A priority patent/MX2007014843A/en
Priority to CA002610366A priority patent/CA2610366A1/en
Priority to JP2008513829A priority patent/JP5513741B2/en
Publication of WO2006128173A2 publication Critical patent/WO2006128173A2/en
Publication of WO2006128173A3 publication Critical patent/WO2006128173A3/en
Priority to IL187603A priority patent/IL187603A0/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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/145Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/15Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96466Cysteine endopeptidases (3.4.22)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2510/00Detection of programmed cell death, i.e. apoptosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • Apoptosis or programmed cell death, plays a central role in the development and homeostasis of all multicellular organisms (Shi Y, 2002, Molecular Cell 9:459-470).
  • a frequent hallmark of cancer is resistance to natural apoptotic signals. Depending on the cancer type, this resistance is typically due to up- or down-regulation of key proteins in the apoptotic cascade or to mutations in genes encoding these proteins. Such changes occur in both the intrinsic apoptotic pathway, which funnels through the mitochondria and caspase-9, and the extrinsic apoptotic pathway, which involves the action of death receptors and caspase-8.
  • alterations in proper levels of proteins such as p53, Bim, Bax, Apaf-1 , FLIP and many others have been observed in cancers.
  • the alterations can lead to a defective apoptotic cascade, one in which the upstream pro-apoptotic signal is not adequately transmitted to activate the executioner caspases, caspase-3 and caspase-7.
  • upstream genetic abnormalities are effectively "breaks" in the apoptotic circuitry, and as a result such cells proliferate atypically.
  • efforts have been made to develop therapeutics that target specific proteins in the apoptotic cascade.
  • peptidic or small molecule binders to cascade members such as p53 and proteins in the BcI family or to the inhibitor of apoptosis (IAP) family of proteins have pro-apoptotic activity, as do compounds that promote the oligomerization of Apaf-1.
  • cascade members such as p53 and proteins in the BcI family or to the inhibitor of apoptosis (IAP) family of proteins have pro-apoptotic activity, as do compounds that promote the oligomerization of Apaf-1.
  • IAP apoptosis
  • procaspase-3 is a homodimer of heterodimers and is produced by proteolysis of procaspase-3. In vivo, this proteolytic activation typically occurs through the action of caspase-8 or caspase-9.
  • procaspase- 3 has a 12 amino acid "safety catch” that blocks access to the IETD site (amino acid sequence, ile-glu-thr-asp) of proteolysis. See Roy, S. et al.; Maintenance of caspase-3 proenzyme dormancy by an intrinsic "safety catch” regulatory tripeptide, Proc. Natl. Acad. Sci. 98, 6132-6137 (2001).
  • This safety catch enables procaspase-3 to resist autocatalytic activation and proteolysis by caspase-9. Mutagenic studies indicate that three consecutive aspartic acid residues appear to be the critical components of the safety catch. The position of the safety catch is sensitive to pH; thus, upon cellular acidification (as occurs during apoptosis) the safety catch is thought to allow access to the site of proteolysis, and active caspase-3 can be produced either by the action of caspase-9 or through an autoactivation mechanism.
  • procaspase-3 is upregulated.
  • procaspase-3 is upregulated in certain neuroblastomas, lymphomas, and liver cancers (Nakagawara, A. et al., 1997, Cancer Res. 57:4578-4584; Izban, K. F. ⁇ t al., Am. J. Pathol. 154:1439-1447; Persad, R. et al., Modem Patholo. 17:861-867).
  • procaspases include in particular activators capable of converting procaspase-3 to its effector form. Furthermore, we demonstrate that certain small drug molecules can directly and immediately activate procaspase-3 to achieve a proapoptotic effect in cancer cells in vivo. We believe these are the first small molecules known to directly activate procaspase-3. The direct activation of executioner caspases represents a novel and valuable anti-cancer strategy.
  • IAP inhibitor of apoptosis
  • PAC-1 procaspase activating compound 1
  • PARP Poly(ADP-ribose) polymerase.
  • the invention broadly provides compounds, methods of therapeutic treatment, methods of screening for compounds, and methods of screening for cell and patient suitability for treatment in connection with modifiers of procaspases.
  • the modifiers are inhibitors or activators.
  • the invention provides such compounds and methods in connection with activators of procaspase-3 and procaspase-7.
  • the inventions are applicable in the context of a variety of cancer diseases and cancer cell types such as breast, lymphoma, adrenal, renal, melanoma, leukemia, neuroblastoma, lung, brain, and others known in the art.
  • compounds of the invention may act via the mechanism of modulation of apoptosis or programmed cell death to be effective in the treatment of cancer cells.
  • the modulation of apoptosis is by induction of apoptosis.
  • the modulation of apoptosis is by inhibition of apoptosis.
  • the invention provides a method of selectively inducing apoptosis in a cancer cell, comprising: (a) administering to said cancer cell a compound capable of modifying a procaspase-3 molecule of said cancer cell; and (b) modifying said procaspase-3 molecule so as to induce apoptosis.
  • said cancer cell is in a patient in need of treatment.
  • said compound is of formula ZZ
  • said compound is selected from the group consisting of formula ZZ, PAC-1 , and Structure 5.
  • said compound is PAC-1.
  • the compound is selected from those having formula ZZ wherein R4 and R5 are both N, A is oxygen, and other variable groups are as defined above. In an embodiment, the compound is selected from those having formula ZZ wherein R4 and R5 and both N, A is oxygen, R2 is hydrogen, and other variable groups are as defined above. In an embodiment, the compound is selected from those having formula ZZ wherein R4 and R5 and both N, A is oxygen, R2 is hydrogen, R3 is allyl, and other variable groups are as defined above.
  • the method further comprises the step of assessing a procaspase-3 or caspase-3 parameter in a cancer cell; wherein said parameter is one or more of a semi-quantitative or quantitative amount, a functional amount, and an activity level of said procaspase-3 or caspase-3.
  • the invention provides a method of direct in vitro screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a purified procaspase-3; (c) exposing the test compound to the purified procaspase-3; (d) measuring a procaspase-3 activity following exposure to the test compound; (e) identifying a modifying compound by comparing a test activity upon the exposure to the test compound with an unmodified activity in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule.
  • the method further comprises comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure of procaspase-3 to a compound selected from the group consisting of structural formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5.
  • the invention provides a method of screening for a compound capable of activating procaspase-3 comprising: a) providing procaspase- 3; providing a test compound, preferably a small molecule; b) reacting the procaspase-3 with the test compound, thereby putatively generating caspase-3; and c) measuring caspase-3 activity.
  • the measuring caspase-3 activity employs a substrate, Ac-DEVD-pNA.
  • the measuring uses a wavelength readout parameter of about 410 nm.
  • the screening is carried out in parallel using multiple test compounds.
  • the invention provides a method of screening which uses the detection of a subunit of procaspase-3 as an indicator that the full length (inactive) procaspase-3 is processed to caspase-3.
  • the subunit has a molecular weight of about 19kD as measured by a protein gel migration technique, for example in a Western blot.
  • the invention provides a method of in cellular screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a cell, wherein the cell putatively expresses procaspase-3; (c) exposing the cell to the test compound; (d) measuring a cell parameter following exposure to the test compound; wherein said parameter comprises one or more of cell viability, apoptotic indicator, and other parameters; (e) identifying a modifying compound by comparing a tested cell parameter upon the exposure to the test compound with an unmodified cell parameter in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule.
  • the method further comprises comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure to a compound selected from the group consisting of formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5.
  • the invention provides a method of identifying or diagnosing a potential susceptibility to treatment for a cancer cell with a procaspase activator compound, comprising (a) assessing a procaspase parameter in said cancer cell; and (b) determining if said parameter allows an increased susceptibility to activation of a procaspase.
  • said procaspase parameter is a procaspase-3 level and said procaspase is procaspase-3.
  • said procaspase parameter is a procaspase-7 level and said procaspase is procaspase- 7.
  • a level can be a semi-quantitative or quantitative amount, or functional amount (e.g. an activity-based amount, e.g. a standardized unit or international unit).
  • the invention provides a method of treating a cancer cell, comprising (a) identifying a potential susceptibility to treatment of a cancer cell with a procaspase activator compound; and (b) exposing said cancer cell to an effective amount of the procaspase activator compound.
  • the procaspase activator compound is selected from the group consisting of formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5.
  • the method of claim 16 wherein said procaspase activator compound is capable of activating procaspase-3, procaspase-7, or both procaspase-3 and procaspase-7.
  • the invention provides a method of synthesizing PAC-1 , comprising the steps of Scheme 1.
  • the invention provides a method of synthesizing Compound 5, comprising the steps of Scheme 1 with appropriate modification.
  • the invention provides a method of synthesizing compounds of the formula ZZ as disclosed herein and as would be understood in the art. [0027] In an embodiment, the invention provides compounds of the formula ZZ:
  • the invention provides compounds of the formula ZZ excluding PAC-1 , wherein the structure of PAC-1 is:
  • the invention provides a compound of Structure 5, wherein the structure is:
  • a composition of the invention is a chemotherapeutic agent.
  • the invention provides compounds and methods involving effective concentrations preferably from about 10 nM to about 100 ⁇ M of the disclosed structural formulas. In another preferred embodiment, the effective concentrations are from about 200 nM to about 5 ⁇ M. In an embodiment, the effective concentration is considered to be a value such as a 50% activity concentration in a direct procaspase activation assay, in a cell apoptosis induction assay, or in an animal clinical therapeutic assessment. In a preferred embodiment, such value is less than about 200 ⁇ M. In a preferred embodiment, the value is less than about 10 ⁇ M.
  • Compounds of the invention and compounds useful in the methods of this invention include those of the disclosed formulas and salts and esters of those compounds, including preferably pharmaceutically-acceptable salts and esters.
  • the invention provides prodrug forms of compositions.
  • Prodrugs of the compounds of the invention are useful in the methods of this invention. Any compound that will be converted in vivo to provide a biologically, pharmaceutically or therapeutically active form of a compound of the invention is a prodrug.
  • Various examples and forms of prodrugs are well known in the art.
  • a biomolecule such as a precursor protein or precursor nucleic acid can be a prodrug. Examples of prodrugs are found, inter alia, in Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985), Methods in Enzymology, Vol. 42, at pp. 309-396, edited by K. Widder, et. al.
  • Molecules disclosed herein may contain one or more ionizable groups [groups from which a proton can be removed (e.g., -OH, -COOH, etc.) or added (e.g., amines) or which can be quaternized (e.g., amines)]. All possible ionic forms of such molecules and salts thereof are intended to be included individually in the disclosure herein. With regard to salts of the compounds herein, one of ordinary skill in the art can select from among a wide variety of available counterions those that are appropriate for preparation of salts of this invention for a given application. For example, in general any anions can be employed in the formation of salts of compounds herein; e.g.
  • halide sulfate, carboxylate, acetate, phosphate, nitrate, trifluoroacetate, glycolate, pyruvate, oxalate, malate, succinate, fumarate, tartarate, citrate, benzoate, methanesulfonate, ethanesulfonate, p-toluenesulfonate, salicylate and others.
  • the invention can encompass all such isomers, individual enantiomers, as well as mixtures of cis and trans isomers, mixtures of diastereomers; non-racemic and racemic mixtures of enantiomers (optical isomers); and the foregoing mixtures enriched for one or more forms; except as stated otherwise herein.
  • the configuration (cis, trans or R or S) of a compound (or of an asymmetric carbon) then any one of the isomers or a mixture of more than one isomer is intended.
  • the processes for preparation can use racemates, enantiomers, or diastereomers as starting materials.
  • enantiomeric or diastereomeric products When enantiomeric or diastereomeric products are prepared, they can be separated by conventional methods, for example, by chromatographic or fractional crystallization.
  • the inventive compounds may be in the free or hydrate form.
  • the invention provides a therapeutic composition comprising one or more compounds and for each compound a pharmaceutically acceptable salt or ester thereof; wherein the compounds are present in the composition in an amount or in a combined amount effective for obtaining the desired therapeutic benefit.
  • the therapeutic compositions of this invention optionally further comprise one or more pharmaceutically acceptable components, for example carriers and excipients as known in the art.
  • the invention provides a compound having the formula ZZ2:
  • R1 and R2 each independently is hydrogen, halogen, alkyl, allyl, haloalkyl, alkenyl, alkenol, alkanol, or haloalkenyl.
  • R1 and R2 each independently is hydrogen, halogen, allyl, or short alkyl.
  • the invention provides a compound selected from the group consisting of a PAC- 1 derivative combinatorial library comprising a hydrazide compound combined with an aldehyde compound.
  • the hydrazide compound is selected from the group consisting of hydrazides generated from AX compounds described herein.
  • the aldehyde compound is selected from the group consisting of BX compounds described herein.
  • the hydrazide compound is selected from the group consisting of AX compounds described herein and the aldehyde compound is selected from the group consisting of BX compounds described herein.
  • the invention provides a method of synthesizing a PAC- 1 derivative compound comprising providing a hydrazide compound, providing an aldehyde compound, and reacting the hydrazide compound with the aldehyde compound, thereby synthesizing a PAC- 1 derivative compound.
  • the hydrazide compound has the formula ZZ3:
  • the aldehyde compound has the formula ZZ4:
  • the hydrazide compound has the formula ZZ3 and the aldehyde compound has the formula ZZ4.
  • the invention provides a compound selected from the group consisting of: L01 R06, L02R03, L02R06, L08R06, L09R03, L09R06, and L09R08.
  • the invention provides a method of screening a candidate cancer patient for possible treatment with a procaspase activator by identifying an elevated level of a procaspase in the candidate, comprising obtaining a cell or tissue test sample from the candidate, assessing the procaspase level in the test sample, and determining whether the procaspase level is elevated in the test sample relative to a reference level, thereby screening a candidate cancer patient for possible treatment with a procaspase activator.
  • the procaspase is selected from the group consisting of procaspase-2, -3, -6-, -7, -8, and -9.
  • the procaspase is procaspase-3.
  • an elevated level of the test sample is at least about 2- fold greater than the reference level. In an embodiment, an elevated level of the test sample is at least about 4-fold greater than the reference level. In an embodiment, the reference level is from a second test sample from the same patient. In an embodiment, the reference level is from a normal cell or tissue sample. The reference level can be from a cell line, such as a cancer cell line or a normal cell line. In an embodiment, the reference level is an absolute threshold amount. See Svingen, P.A. et a!., Clin. Cancer Res. 10:6807-6820 which describes various amounts of levels of procaspases including numbers of molecules per cell.
  • the invention provides a method of inducing death in a cancer cell, comprising administering to said cancer cell a compound capable of activating a procaspase molecule of said cancer cell.
  • the procaspase is one or more of procasepas-3 and procaspase-7.
  • the procaspase is procaspase 3.
  • the compound has structural formula ZZ.
  • the compound has structural formula ZZ2.
  • FIG. 1 A) Activation of mutants in the "safety catch" region of procaspase-3 by PAC-1.
  • PAC-1 has an EC 50 for activation of 0.22 ⁇ M on wild type procaspase-3 (DDD), and corresponding EC 5O values of 2.77 ⁇ M (DAD), 113 ⁇ M (DDA), and 131 ⁇ M (ADD) for certain mutants.
  • DDD wild type procaspase-3
  • DDAD 113 ⁇ M
  • ADD 131 ⁇ M
  • B) PAC-1 activates procaspase-7 with an ECs 0 of 4.5 ⁇ M.
  • FIG. 3 PAC-1 induces apoptosis in HL-60 cells.
  • Figure 4. A) Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 10 ⁇ M etoposide.
  • C) PAC-1 treatment induces a rapid decrease in cellular PARP activity in HL-60 cells, consistent with an immediate activation of cellular caspase-3/- 7. In contrast, etoposide (10 ⁇ M) treated cells show a decrease in PARP activity at much later time points.
  • Fig. 5A illustrates relative procaspase-3 levels in normal and cancerous cells from several patients.
  • Fig. 5B illustrates IC 50 levels for PAC-1 in a variety of cell types having a range of relative procaspase-3 levels.
  • Fig. 5C illustrates the effect of treating animals with PAC-1 on outcomes of tumor growth.
  • Fig. 5D illustrates the effect of oral treatment of animals with PAC-1 on outcomes of tumor growth.
  • Fig. 5E illustrates results of progression of cancer in a lung cancer model for control, PAC-1 , and gefitinib (IressaTM; AstraZeneca) treatment groups.
  • Tumor cells were injected into mice by i.v. administration; Iressa and PAC-1 were given orally at 100 mg/kg.
  • Fig. 6A illustrates relative procaspase-3 levels in normal and cancerous cells of three patients.
  • Fig. 6B illustrates the sensitivity of normal and cancerous cells from Patient 3 to treatment with PAC-1.
  • Fig. 7 illustrates results of administering PAC-1 intraperitoneal ⁇ in the context of a mouse model of lung cancer.
  • Fig. 8A and 8B illustrate structures for compounds of PAC-1 derivatives and a combinatorial library.
  • Fig. 9 illustrates a nucleotide sequence for Homo sapiens caspase 3, apoptosis-related cysteine peptidase (CASP3), transcript variant alpha, mRNA (Accession No. NM_004346; 2689 bp mRNA linear; obtained from http://www.ncbi.nlm.nih.gov/entrez).
  • CAP3 apoptosis-related cysteine peptidase
  • Fig. 10 illustrates a nucleotide sequence for Homo sapiens caspase 7, apoptosis-related cysteine peptidase (CASP7), transcript variant alpha, mRNA (Accession No. NM_001227; 2605 bp; mRNA linear; obtained from http://www.ncbi.nlm.nih.gov/entrez).
  • chemotherapeutic agent refers to any substance capable of reducing or preventing the growth, proliferation, or spread of a cancer cell, a population of cancer cells, tumor, or other malignant tissue.
  • the term is intended also to encompass any antitumor or anticancer agent.
  • the term "effective amount” is intended to encompass contexts such as a pharmaceutically effective amount or therapeutically effective amount.
  • the amount is capable of achieving a beneficial state, beneficial outcome, functional activity in a screening assay, or improvement of a clinical condition.
  • cancer cell is intended to encompass definitions as broadly understood in the art.
  • the term refers to an abnormally regulated cell that can contribute to a clinical condition of cancer in a human or animal.
  • the term can refer to a cultured cell line or a cell within or derived from a human or animal body.
  • a cancer cell can be of a wide variety of differentiated cell, tissue, or organ types as is understood in the art.
  • alkyl refers to a monoradical branched or unbranched saturated hydrocarbon chain preferably having from 1 to 22 carbon atoms and to cycloalkyl groups having one or more rings having 3 to 22 carbon atoms.
  • Short alkyl groups are those having 1 to 6 carbon atoms including methyl, ethyl, propyl, butyl, pentyl and hexyl groups, including all isomers thereof.
  • Long alkyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 and those having 16-18 carbon atoms.
  • cycloalkyl refers to cyclic alkyl groups of from 3 to 22 carbon atoms having a single cyclic ring or multiple condensed rings.
  • Cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.
  • alkenyl refers to a monoradical of a branched or unbranched unsaturated hydrocarbon group preferably having from 2 to 22 carbon atoms and to cycloalkenyl groups having one or more rings having 3 to 22 carbon atoms wherein at least one ring contains a double bond.
  • Preferred alkenyl groups are those having 1 or 2 double bonds.
  • Short alkenyl groups are those having 2 to 6 carbon atoms including ethylene (vinyl) propylene, butylene, pentylene and hexylene groups, including all isomers thereof.
  • Long alkenyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 carbon atoms and those having 16-18 carbon atoms.
  • Cycloalkenyl groups include, by way of example, single ring structures such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclooctenyl, cylcooctadienyl and cyclooctatrienyl.
  • alkynyl refers to a monoradical of an unsaturated hydrocarbon preferably having from 2 to 22 carbon atoms and having one or more triple bonds (CDC).
  • Alkynyl groups include ethynyl, propargyl, and the like.
  • Short alkynyl groups are those having 2 to 6 carbon atoms, including all isomers thereof,
  • Long alkynyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 carbon atoms and those having 16-18 carbon atoms.
  • aryl refers to a group containing an unsaturated aromatic carbocyclic group of from 6 to 22 carbon atoms having a single ring (e.g., phenyl), one or mroe rings (e.g., biphenyl)or multiple condensed (fused) rings, wherein at least one ring is aromatic (e.g., naphthyl, dihydrophenanthrenyl, fluorenyl, or anthryl).
  • Aryls include phenyl, naphthyl and the like.
  • Aryl groups may contain portions that are alkyl, alkenyl or akyriyl in addition to the the unsaturated aromatic ring(s).
  • alkaryl refers to the aryl groups containing alkyl portions, i.e., -alkylene-aryl and -substituted alkylene-aryly. Such alkaryl groups are exemplified by benzyl, phenethyl and the like.
  • Alkyl, alkenyl, alkynyl and aryl groups are optionally substituted as described herein (the term(s) can include substituted variations) and may contain 1-8 non-hydrogen substituents dependent upon the number of carbon atoms in the group and the degree of unsaturation of the group.
  • alkylene refers to a diradical of a branched or unbranched saturated hydrocarbon chain, preferably having from 1 to 10 carbon atoms, more preferably having 1 -6 carbon atoms, and more preferably having 2-4 carbon atoms; the term can include substituted variations. This term is exemplified by groups such as methylene (-CH2-), ethylene (-CH2CH2-), more generally -(CH2)n-, where n is 1-
  • Alkylene groups may be branched. Alkylene groups may be optionally substituted. Alkylene groups may have up to two non-hydrogen substituents per carbon atoms. Perferred substituted alkylene groups have 1 , 2, 3 or 4 non-hydrogen substituents.
  • arylene refers to the diradical derived from aryl (including substituted aryl) as defined above and is exemplified by 1 ,2-phenylene, 1 ,3- phenylene, 1 ,4-phenylene, 1 ,2-naphthylene and the like.
  • amino refers to the group -NH2 or to the group -NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic provided that both R's are not hydrogen.
  • Alkyl groups are optionally substituted as discussed herein and may, dependent upon the size of the alkyl group, have preferably from 1-10 substituent groups.
  • Substituted alkyl groups include those that carry 1 to 8 substituents, 1 to 5 substituents, 1 to 3 substituents, and 1 or 2 substituents.
  • Haloalkyl refers to alkyl as defined herein substituted by one or more halo groups as defined herein, which may be the same or different.
  • Representative haloalkyl groups include, by way of example, trifluoromethyl, 3-fluorododecyl, 12,12,12-trifluorododecyl, 2-bromooctyl, 3-bromo-6-chloroheptyl, and the like.
  • heteroaryl refers to an aromatic group of from 2 to 22carbon atoms having 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur within at least one ring (if there is more than one ring). Heteroaryl groups may be optionally substituted.
  • heterocycle or “heterocyclic” refers to a monoradical saturated or unsaturated group having a single ring or multiple condensed rings, from 2-22 carbon atoms and from 1 to 6 hetero atoms, preferably 1 to 4 heteroatoms, selected from nitrogen, sulfur, phosphorus, and/or oxygen within at least one ring. Heterocyclic groups may be substituted.
  • ester refers to chemical entities as understood in the art and in particular can include groups of the form (RCO-).
  • any of the above groups which contain one or more substituents it is understood, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible.
  • the compounds of this invention include all novel stereochemical isomers arising from the substitution of disclosed compounds. [0088] The invention may be further understood by the following non-limiting examples.
  • PAC-1 In contrast to many known anti-cancer drugs, cells treated with PAC-1 show an immediate activation of procaspase-3, and the toxicity of PAC-1 is shown to be directly proportional to the amount of procaspase-3 contained in a cell. Thus PAC-1 directly activates procaspase-3 to caspase-3 in vivo, allowing this compound to induce apoptosis even in cells that have defective apoptotic machinery.
  • PAC-1 is the first small molecule known to directly activate procaspase-3; the direct activation of executioner caspases is a novel anti-cancer strategy that may prove beneficial in a variety of cancers, including the many cancers in which procaspase-3 is u p regulated.
  • Procaspase-3 was expressed and purified in E. coli (Roy et al., 2001). Procaspase-3 (at a concentration of 50 ng/mL) was added to the wells of a 384-well plate, and the compounds were added to a final concentration of approximately 40 ⁇ M. Each plate was then incubated for two hours at 37°C, after which the caspase-3 peptidic substrate Ac-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNa) was added to a concentration of 200 ⁇ M.
  • Ac-DEVD-pNa the caspase-3 peptidic substrate Ac-Asp-Glu-Val-Asp-p-nitroanilide
  • Procaspase-3 has an N-terminal pro domain (residues 1-28), followed by a large subunit (17 kDa) and a small subunit (12 kDa) that are separated by an intersubunit linker (Pop et al., 2003).
  • procaspase-3 monomers assemble to form a catalytically inactive homodimer that can be activated by cleavage at D175 in the intersubunit linker.
  • the precise role of the pro domain is unclear, and it has been shown that cleavage in the intersubunit region alone is sufficient for full catalytic activity (Stennicke, H. R. et al., 1998).
  • procaspase-3 Although procaspase-3 is catalytically competent, it is highly resistant to autoactivation due to the presence of the 12 amino acid safety catch; however, when the safety catch is mutated significant autoactivation of procaspase-3 is observed (Roy et al., 2001). Compounds that interact with this important regulatory region or at other positions can allow the autoactivation of procaspase-3.
  • caspase-7 also exists as an inactive zymogen that is activated by proteolysis.
  • Caspase-3 and caspase-7 are both executioner caspases and have considerable sequence and structural homology (Denault, J.-B. et al., 2003).
  • Procaspase-7 may also have a similar safety catch region, although it has only two aspartic acids in the key triad (Asp-Thr-Asp), instead of three.
  • PAC-1 can also activate procaspase-7, although in a less efficient manner than its activation of procaspase-3 (EC 50 of 4.5 ⁇ M versus 0.22 ⁇ M for procaspase-3 activation).
  • the white blood cells derived from the bone marrow of a healthy human donor are among those with the lowest amount of procaspase-3, and PAC-1 is comparatively less toxic to these cells.
  • PAC-1 is most potent versus the lung cancer cell line NCI-H226, with an IC 50 of 0.35 ⁇ M. In accordance with data in the literature (Svingen et al., 2004), we found this cell line to have a concentration of procaspase-3 that is greater than five times that of the non-cancerous control.
  • etoposide showed no such correlation between potency in cell culture and cellular levels of procaspase-3.
  • etoposide was ineffective (IC 50 >50 ⁇ M) in inducing death in three of the melanoma cell lines (UACC-62, CRL-1872, and B16-F10), the breast cancer cell line (Hs 578t), and the lung cancer cell line (NCI-H226); these cell lines have procaspase-3 levels of 1.0, 2.4, 1.9, 3.7, and 5.3, respectively.
  • Etoposide was effective (IC 50 ⁇ 1 ⁇ M) versus HL-60, U-937, SK-N-SH and PC-12, which have procaspase-3 levels of 4.3, 4.0, 4.7, and 4.4, respectively. Thus, overall there is no correlation between procaspase-3 levels and IC 5 O for etoposide.
  • Cancerous cells typically have a reduced sensitivity to proapoptotic signals due to the mutation or aberrant expression of an assortment of proteins in the apoptotic cascade. As such, many types of cancer are notoriously resistant to not only the endogenous signals for apoptotic cell death, but also to chemotherapeutic agents that act through similar mechanisms.
  • the paradoxical upregulation of procaspase-3 expression levels in certain cancers provides an opportunity to use this existing intracellular pool of protein to directly induce apoptosis, thus bypassing the often non-functional or compromised upstream portion of the cascade.
  • procaspase-3 is notorious for its relative inability to undergo autoactivation, it is dependent upon a 12 amino acid safety catch to keep itself in the inactive state. PAC-1 induces the autoactivation of procaspase-3 in vitro, and this activation is greatly diminished by mutation of the key tri-aspartate region of the safety catch.
  • PAC-1 treatment induces rapid caspase-3-like activity. It is likely that the caspase-3 mediated cleavage of anti-apoptotic proteins (Bcl-2, BcI-XL, etc.) then induces depolarization of the mitochondrial membrane and amplifies apoptosis. Further, the potency of PAC-1 toward a variety of cancer cell lines is directly proportional to the concentration of procaspase-3 in the cell.
  • Ni-NTA resin and anti-Penta His Alexa Fluor 647 antibody was purchased from Qiagen (Valencia, CA).
  • Bradford dye was purchased from Bio-Rad (Hercules, CA).
  • Pin transfer devices were purchased from V & P Scientific (San Diego, CA).
  • the reagent z-vad-fmk was purchased from Calbiochem (San Diego, CA).
  • Rosetta E. coli was purchased from Novagen (Madison, Wl).
  • Anti-caspase-3 antibody was purchased from Sigma (St. Louis, MO).
  • Annexin V Alexa Fluor 488 conjugate, JC-9, and propidium iodide were purchased from Molecular Probes (Eugene, OR).
  • IPTG and MTS/PMS CellTiter 96 Cell Proliferation Assay reagent were purchased from Promega (Madison, Wl). Fetal Bovine Serum was purchased from Biomeda (Foster City, CA). 96 and 384-well microtiter plates, microscope slides, microscope coverslips, horse serum and all other reagents were purchased from Fisher (Chicago, IL).
  • the cell lysate was incubated for 1 hour at 4°C.
  • the resin was loaded on a column, washed with 10 mL NTA binding buffer followed by 10 mL NTA wash buffer (150 mM NaCI, 50 mM Tris, 20 mM Imidazole, pH 7.9).
  • the proteins were eluted in 1 mL fractions with 10 mL of NTA elution buffer (150 mM NaCI, 50 mM Tris, 250 mM Imidazole, pH 7.9). Fractions containing protein were pooled and the amount of protein was determined using the Bradford assay.
  • Isolated procaspase-3 was diluted to 50 ng/mL in caspase assay buffer (50 mM HEPES, 100 mM NaCI, 10 mM DTT, 0.1 mM EDTA 1 0.1 % CHAPS and 10% glycerol, pH 7.4). 45 ⁇ L of the procaspase-3 solution was added to each well of a Nunc 384-well flat bottom microtiter plate. Approximately 20,000 compounds were screened. About 6,000 of the compounds were collected from various sources within the department of chemistry at the University of Illinois; their structures are available at: http://www.scs.uiuc.edu/ ⁇ phgroup/comcollections.html. The other approximately 14,000 compounds were purchased from Chembridge Corporation (San Diego, California). PAC-1 was a member of the compounds purchased from Chembridge Corporation.
  • Activation curves The dose dependence of procaspase-3 activators was determined by adding various concentrations of compound to 90 ⁇ L of a 50 ng/mL procaspase-3, active caspase-3, procaspase-7 or active caspase-7 in caspase assay buffer in a 96-well plate. The plate was then incubated for 12 hours at 37 0 C. 10 ⁇ L of a 2 mM solution of Ac-DEVD-pNA in caspase assay buffer was then added to each well. The plate was read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 well plate reader. The slope of the linear portion for each well was determined and the fold increase in activation from non-treated control wells was calculated.
  • Procaspase-3 was expressed and isolated exactly as above. Procaspase-3 was diluted to about 50 ⁇ g/mL in caspase assay buffer. The procaspase-3 was then incubated in the presence or absence of 100 ⁇ M PAC-1 for varying times at 37°C. After this incubation, an equal volume of load buffer (150 mM NaCI, 50 mM Tris, 2% SDS, 20% glycerol, pH 8.0) was added to each procaspase-3 sample. All samples were then stored at -80 0 C until the time-course was completed. All samples were then incubated at 95 0 C for 5 minutes and run on a 12% SDS- PAGE gel.
  • load buffer 150 mM NaCI, 50 mM Tris, 2% SDS, 20% glycerol, pH 8.0
  • Proteins were then transferred to nitrocellulose paper overnight. Blots were washed in TTBS (150 mM NaCI, 50 mM Tris, 0.1 % Tween-20, pH 7.4) and blocked with a 10% milk solution for 2 hours. Blots were then incubated in a 1 :5000 dilution of anti-Penta His Alexa Fluor 647 antibody for 2 hours. The blot was then washed with TTBS and scanned on a Typhoon fluorescence scanner (Amersham Biosciences, SunnyVale CA).
  • the DDD procaspase-3 safety catch (SEQ ID NO: 1 ; SEQ ID NO:2; SEQ ID NO:9) was mutated to ADD (SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:10), DAD (SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:11) and DDA (SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:12) using the quickchange strategy with the following primers, gacagacagtggtgttgCGgatgacatggcgtgtcataaatacc (SEQ ID NO: 13), gacagacagtggtgttgatgCtgacatggcgtgtcataaatacc (SEQ ID NO: 14) and gacagacagtggtgttgatgatgCcatggcgtgtcataaatacc (SEQ ID NO: 14) and gacagacagtggtgttgatgatgCcatggcgtgtcataaatacc (
  • the plate was then incubated for 12 hours at 37°C. 10 ⁇ L of a 2 mM solution of Ac- DEVD-pNA in caspase assay buffer was then added to each well. The plate was read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA). The slope of the linear portion for each well was determined and the fold increase in activation for each pH value was calculated.
  • Annexin V staining 500 ⁇ L of media containing 200 ⁇ M PAC-1 or only DMSO as a control was added to the wells of a 24-well plate. 500 ⁇ L HL-60 cells at a concentration of 2 x 10 6 cells/mL were then added to the 24-well plate. The plate was incubated for 20 hours at 37°C. Cells were harvested by centrifugation and washed twice in PBS. The cells were then washed in AnnexinV binding buffer (10 mM HEPES, 140 mM NaCI, 2.5 mM CaCI2, pH 7.4) and resuspended in 100 ⁇ L of Annexin V binding buffer.
  • AnnexinV binding buffer 10 mM HEPES, 140 mM NaCI, 2.5 mM CaCI2, pH 7.4
  • annexin V Alexa Fluor 488 conjugate was added and the tubes were incubated at room temperature for 15 minutes protected from light. 400 ⁇ L of Annexin V binding buffer was then added, followed by the addition of 1 ⁇ L of a 1 mg/mL solution of propidium iodide. The fluorescent intensity of each cell was determined by flow cytometry at 525 nm (green channel) and 675 nm (red channel). At least 50,000 cells were analyzed in each experiment.
  • the fluorescent intensity of each cell was determined by flow cytometry at 525 nm (green channel) and 675 nm (red channel). 50,000 cells were analyzed in each experiment. The shift in the red channel was then used to determine the amount of mitochondrial membrane depolarization.
  • caspase-3 like activity In vivo determination of caspase-3 like activity.
  • the amount of caspase-3 like protease activity was determined by the amount of Ac-DEVD-pNA (N-acetyl- ASP-Glu-Val-Asp-p-nitroanilide) cleaved per minute by cell lysates.
  • 50 ⁇ L of media containing varying concentrations of PAC-1 was added to the wells of a 96-well plate.
  • 50 ⁇ L of HL-60 cells at a concentration of 5 x 10 6 cells/mL were added to the plate and incubated for various times. After the incubation period, the plate was spun at 1000 x g for 5 minutes to pellet the cells.
  • the cells were then washed with 100 ⁇ L of PBS and resuspended in 150 ⁇ L of ice cold Caspase Assay Buffer. Each well was then sonicated to lyse the cells. 90 ⁇ l_ of cell lysate was transferred from each well into a new plate. Ac-DEVD-pNA was added into each well to give a final concentration of 200 ⁇ M. The plate was then read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA). The slope of the linear portion for each well was determined and the amount of Ac-DEVD-pNA cleaved per minute was calculated.
  • the amount of PARP cleavage was determined by using an in vivo PARP activity assay. To accomplish this, 50 ⁇ L of media containing 200 ⁇ M NAD + was added to the control wells of a 96-well plate. 50 ⁇ L of media containing 200 ⁇ M PAC-1 and 200 ⁇ M NAD + was then added to the experimental wells. 25 ⁇ L of HL-60 cells at a concentration of 5 x 10 6 cells/mL were then added to each well. The cells were incubated for various times and then spun at 1000 x g for 5 minutes.
  • the cell media was removed and replaced with 50 ⁇ L Lysing PARP Buffer (50 mM Tris, 10 mM MgCI2, pH 8.0, 1% Triton X-100) containing 25 mM H2O2.
  • the plate was then incubated for 60 minutes at 37°C.
  • 20 ⁇ L of 2 M KOH and 20 ⁇ L of a 20% (v/v) acetophenone (in ethanol) solution was added to each well of the 96-well plate.
  • the plate was then incubated for 10 minutes at 4 0 C. 90 ⁇ L of an 88% (v/v) formic acid solution was added to each well of the 96-well plate.
  • the plate was then incubated for 5 min.
  • the cells were then resuspended in 1 ml_ of a 1:10,000 dilution of anti-mouse Ab Cy3 labeled antibody for 2 hours at room temperature protected from light.
  • the cells were washed five times with PBS and resuspended in 500 ⁇ l_ of PBS.
  • the fluorescent intensity of each cell was determined by flow cytometry at 675 nm (red channel). At least 20,000 cells were analyzed in each experiment. The median of the population was used to determine the relative concentration of procaspase-3 in each cell line.
  • PAC-1 and other compounds are prepared according to the following schemes, e.g., Scheme 1 and/or Scheme 2. Further variations are prepared according to methods known in the art.
  • PAC-1 is prepared according to Scheme 2:
  • Analog compounds of PAC-1 were prepared and assessed for the capability to directly activate purified procaspase-3 in vitro. Table 2. Activity of PAC-1 and analog compounds.
  • the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity, or to organ dysfunctions, etc. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (in light of or precluding toxicity aspects).
  • the magnitude of an administered dose in the management of the disorder of interest can vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, can also vary according to circumstances, e.g. the age, body weight, and response of the individual patient. A program comparable to that discussed above also may be used in veterinary medicine.
  • Suitable agents may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in Alfonso and Gennaro (1995) and elsewhere in the art. Suitable routes may include, for example, oral, rectal, transdermal, vaginal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, or intramedullary injections, as well as intraocular, intrathecal, intravenous, or intraperitoneal administration.
  • agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, water for injection, physiological saline buffer, or other solution.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, water for injection, physiological saline buffer, or other solution.
  • penetrants appropriate to the barrier to be permeated can be used in the formulation. Such penetrants are generally known in the art.
  • compositions of the present invention may be administered parenterally, such as by intravenous injection, or other routes.
  • Appropriate compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, elixirs, solutions, suspensions and the like, e.g. for oral ingestion by a patient to be treated.
  • formulations can be prepared for creams, ointments, lotions, and the like.
  • Agents intended to be administered intracellular ⁇ may be administered using techniques well known to those of ordinary skill in the art. For example, such agents may be encapsulated into liposomes, other membrane translocation facilitating moieties, or other targeting moieties; then administered as described above.
  • Liposomes can include spherical lipid bilayers with aqueous interiors. All molecules present in an aqueous solution at the time of liposome formation can be incorporated into the aqueous interior. The liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm. Additionally, due to hydrophobicity attributes, small organic molecules may be directly administered intracellular ⁇ .
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the disclosure provided herein and other information in the art.
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions, including those formulated for delayed release or only to be released when the pharmaceutical reaches the small or large intestine.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, suspending, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping, lyophilizing, and other processes.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are optionally provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • EXAMPLE 5 Direct Induction of Apoptosis in Cancer Cells with a Small Molecule Activator of Procaspase-3.
  • ABSTRACT Mutation or aberrant expression of proteins in the apoptotic cascade is a hallmark of cancer. These changes prevent proapoptotic signals from being transmitted to the executioner caspases, thus preventing apoptotic cell death and allowing cellular proliferation.
  • Caspase-3 and caspase-7 are the key executioner caspases, existing as inactive zymogens that are activated by upstream signals. Importantly, levels of procaspase-3 are significantly higher in certain cancerous cells relative to non-cancerous controls.
  • PAC-1 small molecule
  • PAC-1 a small molecule that directly activates procaspase-3 to active caspase-3 in vitro with an EC 50 of 220 nanomolar, and induces apoptosis in a variety of cancer cell lines.
  • cells treated with PAC-1 show an immediate activation of procaspase-3, and the efficacy of PAC-1 is shown to be proportional to the amount of procaspase-3 contained in a cell.
  • Derivatives of PAC-1 that do not activate procaspase-3 in vitro also have no proapoptotic activity.
  • Cancerous cells isolated from primary colon tumors are considerably more sensitive to apoptotic induction by PAC-1 than the cells from adjacent non-cancerous tissue from the same patient; these cancerous cells contain on average about 7-fold more procaspase-3 than the cells from the adjacent non-cancerous primary tissue.
  • the sensitivity to PAC-1 of the primary cells from the colon cancer tumors strongly correlates with the level of the procaspase-3 target.
  • PAC-1 as a single entity was shown as active to retard the growth of tumors in three different mouse models, including two models where PAC-1 was administered orally.
  • PAC-1 directly activates procaspase-3 to caspase-3 in vivo, thereby allowing this compound to induce apoptosis even in cells that have defective apoptotic machinery.
  • PAC-1 is the first small molecule known to directly activate procaspase-3; the direct activation of executioner caspases is an anti-cancer strategy that may prove beneficial in the many cancers in which procaspase-3 levels are elevated.
  • INTRODUCTION A hallmark of cancer is its resistance to natural apoptotic signals. Depending on the cancer type, this resistance is typically due to either up- or down-regulation of key proteins in the apoptotic cascade, or to mutations in genes encoding these proteins. Such changes occur in both the intrinsic apoptotic pathway, which funnels through the mitochondria and caspase-9, and the extrinsic apoptotic pathway, which involves the action of death receptors and caspase-8.
  • procaspase-3 results in the generation of the active "executioner” caspase that subsequently catalyzes the hydrolysis of a multitude of protein substrates.
  • Active caspase-3 is a homodimer of heterodimers and is produced by proteolysis of procaspase-3. In vivo, this proteolytic activation typically occurs through the action of caspase-8 or caspase-9. To ensure that this zymogen is not prematurely activated, procaspase-3 has a tri-aspartic acid "safety catch" that blocks access to the IETD site of proteolysis. This safety catch enables procaspase-3 to resist autocatalytic activation and proteolysis by caspase-9.
  • the position of the safety catch is sensitive to pH; thus, upon cellular acidification (as occurs during apoptosis) the safety catch is thought to allow access to the site of proteolysis, and active caspase-3 can be produced either by the action of caspase-9 or through an autoactivation mechanism.
  • procaspase-3 Cells from certain types of cancerous tissue have elevated levels of procaspase-3.
  • procaspase-3 levels are elevated in certain neuroblastomas, lymphomas, and liver cancers.
  • a systematic evaluation of procaspase-3 levels in the 60 cell-line panel used by the NCI revealed that particular lung, melanoma, renal, and breast cancers show greatly enhanced levels of procaspase-3.
  • Procaspase-3 was expressed and purified in E. coli according to standard procedures. Procaspase-3 was added to the wells of a 384-well plate, and the compounds were added to a final concentration of about 40 ⁇ M (the final concentration of procaspase-3 was 50 ng/mL). Each plate was then incubated for two hours at 37 0 C, after which the caspase-3 peptidic substrate Ac-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNa) was added to a concentration of 200 ⁇ M.
  • Ac-DEVD-pNa Ac-DEVD-pNa
  • Procaspase-3 consists of a N-terminal pro domain (residues 1-28), followed by a large subunit (17 kDa) and a small subunit (12 kDa) that are separated by an intersubunit linker. 22 In vivo, two procaspase-3 monomers assemble to form a homodimer that can be activated by cleavage at D175 in the intersubunit linker. The precise role of the pro domain is unclear, and it has been shown that cleavage in the intersubunit region alone is sufficient for full catalytic activity. Although procaspase-3 has enough catalytic activity to drive its own proteolytic maturation, it is highly resistant to this autoactivation due to the presence of the three amino acid safety catch.
  • procaspase-3 when the safety catch is mutated significant autoactivation of procaspase-3 is observed.
  • the procaspase-3 protein was incubated with 100 ⁇ M of PAC-1 for time points ranging from one to five hours.
  • PAC-1 induces the cleavage of procaspase-3 in a time-dependant fashion, with >50% processing observed after 4 hours.
  • procaspase-3 incubated in buffer shows virtually no autoactivation over that same time span.
  • PAC-1 was also effective in this assay at a concentration of 5 ⁇ M.
  • Procaspase-7 is also predicted to have a similar safety catch region, although it has only two aspartic acids in the key triad (Asp-Thr-Asp), instead of three.
  • PAC-1 can also activate procaspase-7, although in a less efficient manner than its activation of procaspase-3 (EC 50 of 4.5 ⁇ M versus 0.22 ⁇ M for procaspase-3 activation).
  • the effect of PAC-1 is abolished at low pH values where procaspase-3 undergoes rapid autoactivation (Figure 2C).
  • PAC-1 was found to induce apoptosis in a variety of cancer cell lines.
  • addition of PAC-1 leads to considerable phosphatidylserine exposure on the cell membrane accompanied by significant chromatin condensation ( Figures 3A, 3B).
  • the compound induces cleavage of the caspase substrate PARP-1 (as assessed by an in vivo PARP activity assay) and causes mitochondrial membrane depolarization (see below).
  • Significant cellular blebbing of PAC-1 treated cells was also observed by microscopy.
  • the toxicity of PAC-1 could be abolished in the presence of the caspase inhibitor z-VAD-fmk.
  • PAC-1 is indeed inducing apoptosis via direct activation of procaspase-3, then the time course of apoptotic events should be altered relative to that observed with standard proapoptotic agents.
  • Etoposide is well known to induce apoptosis through the intrinsic pathway; thus, mitochondrial membrane depolarization is followed by procaspase-3 activation in etoposide-treated cells. Indeed, in HL-60 cells treated with 10 ⁇ M etoposide, mitochondrial membrane depolarization is observed, followed by detection of caspase-3-like activity ( Figure 4A). In contrast, treatment of cells with PAC-1 gives a markedly different result.
  • PAC-1 is most potent versus the lung cancer cell line NCI-H226, with an IC 50 of 0.35 ⁇ M.
  • MCF-7 cancer cell line
  • PAC-1 has virtually no effect on MCF-7 cells, inducing death with an ICso>75 ⁇ M.
  • etoposide showed no such correlation between potency in cell culture and cellular levels of procaspase-3.
  • etoposide was ineffective (IC ⁇ o>5O ⁇ M) in inducing death in three of the melanoma cell lines (UACC-62, CRL- 1872, and B16-F10), the breast cancer cell line (Hs 578t), and the lung cancer cell line (NCI-H226); these cell lines have procaspase-3 levels of 1.0, 2.4, 1.9, 3.7, and 5.3, respectively.
  • Etoposide was effective (IC 50 ⁇ 1 ⁇ M) versus HL-60, U-937, SK-N- SH and PC-12, which have procaspase-3 levels of 4.3, 4.0, 4.7, and 4.4, respectively. Thus, overall there is no correlation between procaspase-3 levels and IC5 0 for etoposide.
  • PAC-1 induced cell death in the primary cancerous cells with IC 50 values from 0.007-1.41 ⁇ M
  • PAC-1 induced cell death in the adjacent non-cancerous tissue with IC 50 values from 5.02-9.98 ⁇ M
  • the cancerous tissue that had elevated levels of procaspase-3 was extremely sensitive to PAC-1.
  • PAC-1 induced death in the cancer cells from patient 17 with an IC5 0 of 7 nM, and these cells were over 700-fold more sensitive to PAC-1 than cells from the adjacent normal tissue.
  • Figure 6A showing relative procaspase-3 concentrations in normal and cancerous samples from Patients 1 , 2, and 3 over a period of time of about 54 days;
  • Figure 6B illustrates that cells in cancerous tissue can be greater than about 80-fold more sensitive to PAC-1 in comparison with normal tissue.
  • PAC-1 was also evaluated against four other non-cancerous cell types: white blood cells isolated from the bone marrow of a healthy donor, Hs888Lu (lung fibroblast cells), MCF-10A (breast fibroblast cells), and Hs578Bst (breast epithelial cells).
  • the non-cancerous cell types are among those with the lowest amount of procaspase-3, and PAC-1 is comparatively less able to induce death in these cells, with IC 50 values of 3.2 - 8.5 ⁇ M ( Figure 5B, green diamonds).
  • PAC-1 induces death in a wide variety of cell types (non-cancerous cell lines, non-cancerous primary cells, cancerous cell lines, primary cancerous cells) in a manner directly related to the level of procaspase-3.
  • the elevation of procaspase- 3 in cancerous cells allows PAC-1 to selectively induce death in these cell types.
  • PAC-1 was evaluated in a mouse xenograft model using a slow release mode of drug delivery. In this model, subcutaneous tumors were formed in ovariectomized female athymic BALB/c (nude) mice using the ACHN (renal cancer) cell line.
  • PAC-1 was evaluated in a second mouse xenograft model, this one using oral administration as the drug delivery mode.
  • subcutaneous xenograft tumors were formed in male athymic BAlBlc-nu/nu mice (5 weeks old, SLC, Hamamaysu, Japan) using the NCI-H226 (lung cancer) cell line, eight mice per group, three tumors per mouse.
  • the mice were treated with PAC-1 via oral gavage once a day for 21 days at a concentration of 0, 50, or 100 mg/kg and sacrificed 1 week later.
  • oral administration of PAC-1 significantly retards tumor growth in a dose-dependent manner.
  • PAC-1 was evaluated in a mouse model where the NCI-H226 cells were injected into male athymic BALB/c-nu/nu mice via tail vein injection. The total experiment lasted 28 days; the mice were treated once a day with PAC-1 (100 mg/kg) via oral gavage on days 1-4 and 7-11. On other days the mice did not receive PAC-1. A second group of mice received only vehicle. After 28 days the mice were sacrificed, and their lungs were examined. As shown in Figure 5E, there is a clear difference between the lung of the control mouse (with obvious gray tumor mass) and the lung of the PAC-1 treated mouse. Results are also shown in a panel from an animal treated with gefitinib (IressaTM; AstraZeneca).
  • Cancerous cells typically have a reduced sensitivity to proapoptotic signals due to the mutation or aberrant expression of an assortment of proteins in the apoptotic cascade. As such, many types of cancer are notoriously resistant to not only the endogenous signals for apoptotic cell death, but also to chemotherapeutic agents that act through similar mechanisms.
  • the paradoxical elevation of procaspase-3 levels in certain cancers provides an opportunity to use this existing intracellular pool of protein to directly induce apoptosis, thus bypassing the often non-functional upstream portion of the cascade.
  • PAC-1 induces the autoactivation of procaspase-3 in vitro. In cell culture, PAC-1 treatment induces rapid caspase-3- like activity.
  • PAC-1 caspase-3 mediated cleavage of anti-apoptotic proteins (Bcl-2, BcI-XL, etc.) then induces depolarization of the mitochondrial membrane and amplifies apoptosis.
  • the potency of PAC-1 toward a variety of cancerous and non-cancerous cell types is proportional to the concentration of procaspase-3 in the cell. As the primary cancerous cells isolated from resected colon tumors have elevated levels of procaspase-3, these cells are considerably more sensitive to PAC-1 than cells from adjacent non-cancerous tissue.
  • PAC-1 is effective in three different mouse models of cancer, including two where PAC-1 is administered orally.
  • procaspase-3 activating compounds can be exceedingly effective against a variety of common cancers in which procaspase-3 levels are aberrantly high.
  • Assessment of procaspase-3 levels in cancer biopsies is simple and rapid; as such, the potential effectiveness of a compound such as PAC-1 can be assessed a priori with a high degree of accuracy.
  • Such personalized medicine strategies can be preferential to therapies that rely on general cytotoxins and can be valuable in anti-cancer therapy.
  • Fig. 1B Cleavage of procaspase-3 to active caspase-3 as induced by PAC-1. Procaspase-3 was recombinantly expressed in E. coli with an N-terminal His-6 tag and purified, lmmunoblotting was performed with an anti-His-6 antibody. In the absence of PAC-1 no maturation of procaspase-3 is observed.
  • PAC-1 In the presence of 100 ⁇ M PAC-1, cleavage to generate the p19 fragment is observed within 1 h, and >50% cleavage is observed after 4 h. PAC-1 is also effective at 5 ⁇ M in this assay.
  • Fig, 2A Activation of mutants in the "safety catch" region of procaspase-3 by PAC-1.
  • PAC-1 has an EC 50 for activation of 0.22 ⁇ M on wild type procaspase-3 (DDD), and corresponding EC 50 values of 2.77 ⁇ M (DAD), 113 ⁇ M (DDA), and 131 ⁇ M (ADD) for the mutants.
  • Fig. 2B PAC-1 activates procaspase-7 with an EC 50 of 4.5 ⁇ M.
  • Fig. 2C Dependence of PAC-1 activation of procaspase-3 on pH. At low pH the safety catch is off and procaspase-3 is essentially maximally activated. Error bars represent standard deviations from the mean
  • FIG. 3A Phosphatidylserine exposure (as measured by Annexin-V staining) after a 20 h treatment with 100 ⁇ M PAC-1. PAC-1 is also effective at 5 ⁇ M in this assay (see Supporting Figure 2).
  • Fig. 3B Chromatin condensation as visualized by Hoescht staining after a 20 h treatment with 100 ⁇ M PAC-1.
  • Fig. 4A Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 10 ⁇ M etoposide.
  • MMP Mitochondrial membrane depolarization
  • PAC-1 Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 100 ⁇ M PAC-1.
  • Fig. 4C PAC-1 treatment (100 ⁇ M) induces a rapid decrease in cellular PARP activity in HL-60 cells, consistent with an immediate activation of cellular caspase-3/-7.
  • etoposide (10 ⁇ M) treated cells show a decrease in PARP activity at much later time points.
  • Fig. 4D and Fig. 4E PAC-1 induces cell death in a procaspase-3 dependant manner.
  • the procaspase-3 levels were determined (by flow cytometry with an antibody to procaspase-3) and the IC 50 of PAC-1 was measured (through a 72 h treatment with a range of PAC-1 concentrations and quantitation using the MTS assay).
  • FIG. 5A Procaspase-3 levels are elevated in cells derived from freshly resected colon cancer tissue. Freshly resected primary colon tumors (together with adjacent non-cancerous tissue) were obtained from 18 different patients, the cancerous and non-cancerous tissue were separated, and the procaspase-3 levels were measured for each using an antibody to procaspase-3 and flow cytometry. On average, cells from the cancerous tissue have a 7.6-fold elevation in procaspase-3 as compared to the cells derived from the adjacent noncancerous tissue from the same patient.
  • Fig. 5B PAC-1 induces cell death in a manner proportional to the cellular level of procaspase-3.
  • the red circles represent the primary cancerous cells from the 18 colon tumors.
  • the black triangles represent the same cancer cell lines depicted in Figure 4D.
  • the green diamonds are four noncancerous cell types: Hs888Lu (lung fibroblast cells), MCF-10A (breast fibroblast cells), Hs578Bst (breast epithelial cells), and white blood cells isolated from the bone marrow of a healthy donor.
  • the blue squares are the primary non-cancerous cells isolated from the tumor margins of the 18 patients.
  • Table 4 Cells derived from primary colon cancer tissue are considerably more sensitive to death induction by PAC-1 than are cells derived from adjacent non-cancerous tissue from the same patient. Fig.
  • PAC-1 reduces the growth of tumors in a xenograft model of cancer. Tumors were formed with the ACHN (renal cancer) cell line by subcutaneous injection, with six mice in each group, and four tumors per mouse. Once the tumors grew to about 30 mm 2 , PAC-1 was implanted as a cholesterol pellet. Error bars represent standard error from the mean. Fig. 5D) Oral administration of PAC-1 significantly retards tumor growth in a mouse xenograft model. Tumors were formed using the NCI-H226 (lung cancer) cell line by subcutaneous injection, eight mice in each group, and three tumors per mouse. PAC-1 or vehicle was administered once a day by oral gavage on days 1-21.
  • ACHN renal cancer
  • Fig. 5E Oral administration of PAC-1 significantly retards tumor growth in an i.v. injection model. Mice were injected i.v. with the NCI-H226 (lung cancer) cell line. The mice were treated with PAC-1 (100 mg/kg) via oral gavage following the protocol as described in the text. Images show the lungs of the mice that did not receive PAC-1 and have a large amount of gray tumor mass on the lung. In contrast, the mice that did receive PAC-1 have almost no visible gray matter.
  • EXAMPLE 6 Testing of PAC-1 in mouse model of lung cancer.
  • a xenograft model was employed using NCI-H226 (lung cancer) cells.
  • PAC-1 was given intraperitoneal ⁇ (i.p.) at 10 mg/kg.
  • a comparison of efficacy was performed with gefitinib (IressaTM; AstraZeneca, Wilmington, Delaware) at 40 mg/kg using 5 mice per group. Results are shown in Figure 7, indicating that PAC-1 was associated with reducing growth in tumor volume.
  • EXAMPLE 7 Combinatorial derivatives, synthesis, and therapeutic use.
  • a number of compounds are prepared as derivatives of the PAC-1 structure.
  • a hydrazide group is reacted with an aldehyde group to yield a combinatorial library of derivative compounds.
  • Any one of hydrazide precursor groups (AX) designated L1-L20 are used to generate hydrazides which are reacted with any one of aldehyde groups (BX) designated 1-28, thus yielding 560 PAC-1 derivative compounds.
  • a derivative compound is synthesized using methods as described herein and according to knowledge available in the art. See the scheme and component structures below in addition to Figure 8A and 8B.
  • such derivative compounds are further modified, e.g. to alter a property such as activity, solubility, toxicity, stability, and/or other properties in connection with pharmaceutical applications.
  • the derivative compounds are used as anti-cancer agents.
  • Compounds are validated as capable of having antineoplastic activity, apoptosis regulation, and/or procaspase-3 activation.
  • primary isolates of freshly removed colon cancer are used fo assess procaspase-3 levels and sensitivity of cells to test .
  • Compounds are classified regarding a propensity to induce cell death in cancerous cells versus normal cells.
  • EXAMPLE 8 Activity of certain derivatives relative to PAC-1.
  • VAD. FMK inhibits apoptosis by blocking the processing of CPP32, Biochem J. 1996
  • FLIP FLICE inhibitory protein
  • NCBI National Center for Biotechnology Information
  • Hergenrother PJ Obtaining and screening compound collections: a user's guide and a call to chemists.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Food Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)

Abstract

Compounds and related methods for synthesis, and the use of compounds in therapy for the treatment of cancer and selective induction of apoptosis in cells are disclosed. Compounds are disclosed in connection with modification of procaspases such as procaspase-3, and particular embodiments are capable of direct activation of procaspase-3 and procaspase-7 to the effector forms of caspase-3 and caspase-7. Procaspase-3 levels can vary among cancer cell types; several types have relatively high levels and can have increased susceptibility to chemotherapy by compounds and methods herein. Therapeutic applications are relevant for a variety of cancer conditions and cell types, e.g. breast, lung, brain, colon, renal, adrenal, melanoma, and others.

Description

Selective Apoptotic Induction in Cancer Cells Including Activation of Procaspase-3
CROSS-REFERENCES TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 USC 119(e) of U.S. Provisional Application Serial 60/684,807 filed May 26, 2005; and U.S. Provisional Application Serial 60743878 filed March 28, 2006; each of which are incorporated by reference in entirety.
STATEMENT ON FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with government support under NSF Grant/Contract CHE-0134779 awarded by the National Science Foundation. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Apoptosis, or programmed cell death, plays a central role in the development and homeostasis of all multicellular organisms (Shi Y, 2002, Molecular Cell 9:459-470). A frequent hallmark of cancer is resistance to natural apoptotic signals. Depending on the cancer type, this resistance is typically due to up- or down-regulation of key proteins in the apoptotic cascade or to mutations in genes encoding these proteins. Such changes occur in both the intrinsic apoptotic pathway, which funnels through the mitochondria and caspase-9, and the extrinsic apoptotic pathway, which involves the action of death receptors and caspase-8. For example, alterations in proper levels of proteins such as p53, Bim, Bax, Apaf-1 , FLIP and many others have been observed in cancers. The alterations can lead to a defective apoptotic cascade, one in which the upstream pro-apoptotic signal is not adequately transmitted to activate the executioner caspases, caspase-3 and caspase-7. [0004] As most apoptotic pathways ultimately involve the activation of procaspase- 3, upstream genetic abnormalities are effectively "breaks" in the apoptotic circuitry, and as a result such cells proliferate atypically. Given the central role of apoptosis in cancer, efforts have been made to develop therapeutics that target specific proteins in the apoptotic cascade. For instance, peptidic or small molecule binders to cascade members such as p53 and proteins in the BcI family or to the inhibitor of apoptosis (IAP) family of proteins have pro-apoptotic activity, as do compounds that promote the oligomerization of Apaf-1. However, because such compounds target early (or intermediate to high) positions on the apoptotic cascade, cancers with mutations in proteins downstream of those members can still be resistant to the possible beneficial effects of those compounds.
[0005] For therapeutic purposes it would be advantageous to identify a small molecule that directly activates a proapoptotic protein far downstream in the apoptotic cascade. The approach to our invention involves such a relatively low position in the cascade, thus enabling the killing of even those cells that have mutations in their upstream apoptotic machinery. Moreover, the therapeutic strategies disclosed herein can have a higher likelihood of success if that proapoptotic protein were upregulated in cancer cells. In the present invention, our efforts to identify small molecules began with targeting the significant downstream effector protein of apoptosis, procaspase-3.
[0006] The conversion or activation of procaspase-3 to caspase-3 results in the generation of the active "executioner" caspase form that subsequently catalyzes the hydrolysis of a multitude of protein substrates. Active caspase-3 is a homodimer of heterodimers and is produced by proteolysis of procaspase-3. In vivo, this proteolytic activation typically occurs through the action of caspase-8 or caspase-9. To ensure that the proenzyme or zymogen is not prematurely activated, procaspase- 3 has a 12 amino acid "safety catch" that blocks access to the IETD site (amino acid sequence, ile-glu-thr-asp) of proteolysis. See Roy, S. et al.; Maintenance of caspase-3 proenzyme dormancy by an intrinsic "safety catch" regulatory tripeptide, Proc. Natl. Acad. Sci. 98, 6132-6137 (2001).
[0007] This safety catch enables procaspase-3 to resist autocatalytic activation and proteolysis by caspase-9. Mutagenic studies indicate that three consecutive aspartic acid residues appear to be the critical components of the safety catch. The position of the safety catch is sensitive to pH; thus, upon cellular acidification (as occurs during apoptosis) the safety catch is thought to allow access to the site of proteolysis, and active caspase-3 can be produced either by the action of caspase-9 or through an autoactivation mechanism.
[0008] In particular cancers, the expression of procaspase-3 is upregulated. A study of primary isolates from 20 colon cancer patients revealed that on average, procaspase-3 was upregulated six-fold in such isolates relative to adjacent noncancerous tissue (Roy et al., 2001). In addition, procaspase-3 is upregulated in certain neuroblastomas, lymphomas, and liver cancers (Nakagawara, A. et al., 1997, Cancer Res. 57:4578-4584; Izban, K. F. θt al., Am. J. Pathol. 154:1439-1447; Persad, R. et al., Modem Patholo. 17:861-867). Furthermore, a systematic evaluation was performed of procaspase-3 levels in the 60 cell-line panel used for cancer screening by the National Cancer Institute (NCI) Developmental Therapeutics Program. The evaluation revealed that certain lung, melanoma, renal, and breast cancers show greatly enhanced levels of procaspase-3 expression (Svingen, P.A. et al., Clin. Cancer Res. 10:6807-6820).
[0009] Due to the role of active caspase-3 in achieving apoptosis, the relatively high expression levels of procaspase-3 in certain cancerous cell types, and the intriguing safety catch-mediated suppression of its autoactivation, we reasoned that small molecules that directly modify procaspase-3 could be identified and that such molecules could have great applicability in targeted cancer therapy.
[0010] Herein we disclose small molecule modifiers of procaspases, including in particular activators capable of converting procaspase-3 to its effector form. Furthermore, we demonstrate that certain small drug molecules can directly and immediately activate procaspase-3 to achieve a proapoptotic effect in cancer cells in vivo. We believe these are the first small molecules known to directly activate procaspase-3. The direct activation of executioner caspases represents a novel and valuable anti-cancer strategy. SUMMARY OF THE INVENTION
[0011] In general the terms and phrases used herein have their art-recognized meaning, which can be found by reference to standard texts, journal references and contexts known to those skilled in the art.
[0012] The following abbreviations are applicable. IAP, inhibitor of apoptosis; PAC-1 , procaspase activating compound 1 ; PARP, Poly(ADP-ribose) polymerase.
[0013] The invention broadly provides compounds, methods of therapeutic treatment, methods of screening for compounds, and methods of screening for cell and patient suitability for treatment in connection with modifiers of procaspases. In an embodiment, the modifiers are inhibitors or activators. In an embodiment, the invention provides such compounds and methods in connection with activators of procaspase-3 and procaspase-7. In embodiments, the inventions are applicable in the context of a variety of cancer diseases and cancer cell types such as breast, lymphoma, adrenal, renal, melanoma, leukemia, neuroblastoma, lung, brain, and others known in the art.
[0014] As a further introduction, we have discovered compounds capable of activating an enzyme that is often overexpressed in its inactive form in cancer cells. The compound induces programmed cell death (apoptosis) in cancer cells, including those that have upregulated procaspase-3. Cancer is a large and growing problem, and is now the number one killer of in the United States. Many cancers resist standard chemotherapy. Compounds of the invention can take advantage of a biological target that may be upregulated in cancer cells and thus can prove effective even in cells with defects in their apoptotic machinery. These compounds can also be successful in targeted cancer therapy, where there can be advantages of selectivity in the killing of cancer cells with comparably reduced toxicity to noncancerous cells having lower levels of procaspase-3.
[0015] Without wishing to be bound by a particular theory, it is believed that compounds of the invention may act via the mechanism of modulation of apoptosis or programmed cell death to be effective in the treatment of cancer cells. In a preferred embodiment, the modulation of apoptosis is by induction of apoptosis. In another embodiment, the modulation of apoptosis is by inhibition of apoptosis. [0016] In an embodiment, the invention provides a method of selectively inducing apoptosis in a cancer cell, comprising: (a) administering to said cancer cell a compound capable of modifying a procaspase-3 molecule of said cancer cell; and (b) modifying said procaspase-3 molecule so as to induce apoptosis. In an embodiment, said cancer cell is in a patient in need of treatment.
[0017] In an embodiment, said compound is of formula ZZ;
Figure imgf000007_0001
wherein n = 1 or 2; R, independently of other R, is hydrogen, halogen, ally!, or short alkyl; R2 = hydrogen, short alkyl, ester, or other moiety that is removable under physiological conditions; R3 = hydrogen, halogen, alkyl, haloalkyl, allyl, alkenyl, alkenol, alkanol, or haloalkenyl; R4 and R5 are N; or R4=N and R5=C; or R4 and R5 = C; and A = oxygen or sulfur. In an embodiment, said compound is selected from the group consisting of formula ZZ, PAC-1 , and Structure 5. In an embodiment, said compound is PAC-1.
[0018] In an embodiment, the compound is selected from those having formula ZZ wherein R4 and R5 are both N, A is oxygen, and other variable groups are as defined above. In an embodiment, the compound is selected from those having formula ZZ wherein R4 and R5 and both N, A is oxygen, R2 is hydrogen, and other variable groups are as defined above. In an embodiment, the compound is selected from those having formula ZZ wherein R4 and R5 and both N, A is oxygen, R2 is hydrogen, R3 is allyl, and other variable groups are as defined above.
[0019] In an embodiment, the method further comprises the step of assessing a procaspase-3 or caspase-3 parameter in a cancer cell; wherein said parameter is one or more of a semi-quantitative or quantitative amount, a functional amount, and an activity level of said procaspase-3 or caspase-3. [0020] In an embodiment, the invention provides a method of direct in vitro screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a purified procaspase-3; (c) exposing the test compound to the purified procaspase-3; (d) measuring a procaspase-3 activity following exposure to the test compound; (e) identifying a modifying compound by comparing a test activity upon the exposure to the test compound with an unmodified activity in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule. In an embodiment, the method further comprises comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure of procaspase-3 to a compound selected from the group consisting of structural formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5.
[0021] In an embodiment, the invention provides a method of screening for a compound capable of activating procaspase-3 comprising: a) providing procaspase- 3; providing a test compound, preferably a small molecule; b) reacting the procaspase-3 with the test compound, thereby putatively generating caspase-3; and c) measuring caspase-3 activity. In a particular embodiment, the measuring caspase-3 activity employs a substrate, Ac-DEVD-pNA. In a particular embodiment, the measuring uses a wavelength readout parameter of about 410 nm. In a particular embodiment, the screening is carried out in parallel using multiple test compounds.
[0022] In an embodiment, the invention provides a method of screening which uses the detection of a subunit of procaspase-3 as an indicator that the full length (inactive) procaspase-3 is processed to caspase-3. In a particular embodiment, the subunit has a molecular weight of about 19kD as measured by a protein gel migration technique, for example in a Western blot.
[0023] In an embodiment, the invention provides a method of in cellular screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a cell, wherein the cell putatively expresses procaspase-3; (c) exposing the cell to the test compound; (d) measuring a cell parameter following exposure to the test compound; wherein said parameter comprises one or more of cell viability, apoptotic indicator, and other parameters; (e) identifying a modifying compound by comparing a tested cell parameter upon the exposure to the test compound with an unmodified cell parameter in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule. In an embodiment, the method further comprises comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure to a compound selected from the group consisting of formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5.
[0024] In an embodiment, the invention provides a method of identifying or diagnosing a potential susceptibility to treatment for a cancer cell with a procaspase activator compound, comprising (a) assessing a procaspase parameter in said cancer cell; and (b) determining if said parameter allows an increased susceptibility to activation of a procaspase. In an embodiment, said procaspase parameter is a procaspase-3 level and said procaspase is procaspase-3. In an embodiment, said procaspase parameter is a procaspase-7 level and said procaspase is procaspase- 7. A level can be a semi-quantitative or quantitative amount, or functional amount (e.g. an activity-based amount, e.g. a standardized unit or international unit).
[0025] In an embodiment, the invention provides a method of treating a cancer cell, comprising (a) identifying a potential susceptibility to treatment of a cancer cell with a procaspase activator compound; and (b) exposing said cancer cell to an effective amount of the procaspase activator compound. In an embodiment, the procaspase activator compound is selected from the group consisting of formula ZZ or subsets of compounds of such formula, PAC-1 , and Structure 5. In an embodiment, the method of claim 16 wherein said procaspase activator compound is capable of activating procaspase-3, procaspase-7, or both procaspase-3 and procaspase-7.
[0026] In an embodiment, the invention provides a method of synthesizing PAC-1 , comprising the steps of Scheme 1. In an embodiment, the invention provides a method of synthesizing Compound 5, comprising the steps of Scheme 1 with appropriate modification. In an embodiment, the invention provides a method of synthesizing compounds of the formula ZZ as disclosed herein and as would be understood in the art. [0027] In an embodiment, the invention provides compounds of the formula ZZ:
Figure imgf000010_0001
wherein n = 1 or 2; R, independently of other R, is hydrogen, halogen, allyl, or short alkyl; R2 = hydrogen, short alkyl, ester, or other moiety that is removable under physiological conditions; R3 = hydrogen, halogen, alkyl, haloalkyl, allyl, alkenyl, alkenol, alkanol, or haloalkenyl; R4 and R5 are N; or R4=N and R5=C; or R4 and R5 = C; and A = oxygen or sulfur.
[0028] In an embodiment, the invention provides compounds of the formula ZZ excluding PAC-1 , wherein the structure of PAC-1 is:
Figure imgf000010_0002
[0029] In an embodiment, the invention provides a compound of Structure 5, wherein the structure is:
Figure imgf000010_0003
[0030] In an embodiment, a composition of the invention is a chemotherapeutic agent.
[0031] In an embodiment, the invention provides compounds and methods involving effective concentrations preferably from about 10 nM to about 100 μM of the disclosed structural formulas. In another preferred embodiment, the effective concentrations are from about 200 nM to about 5 μM. In an embodiment, the effective concentration is considered to be a value such as a 50% activity concentration in a direct procaspase activation assay, in a cell apoptosis induction assay, or in an animal clinical therapeutic assessment. In a preferred embodiment, such value is less than about 200 μM. In a preferred embodiment, the value is less than about 10 μM.
[0032] Compounds of the invention and compounds useful in the methods of this invention include those of the disclosed formulas and salts and esters of those compounds, including preferably pharmaceutically-acceptable salts and esters.
[0033] In an embodiment, the invention provides prodrug forms of compositions. Prodrugs of the compounds of the invention are useful in the methods of this invention. Any compound that will be converted in vivo to provide a biologically, pharmaceutically or therapeutically active form of a compound of the invention is a prodrug. Various examples and forms of prodrugs are well known in the art. A biomolecule such as a precursor protein or precursor nucleic acid can be a prodrug. Examples of prodrugs are found, inter alia, in Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985), Methods in Enzymology, Vol. 42, at pp. 309-396, edited by K. Widder, et. al. (Academic Press, 1985); A Textbook of Drug Design and Development, edited by Krosgaard-Larsen and H. Bundgaard, Chapter 5, "Design and Application of Prodrugs," by H. Bundgaard, at pp. 113-191, 1991); H. Bundgaard, Advanced Drug Delivery Reviews, Vol. 8, p. 1-38 (1992); H. Bundgaard, et al., Journal of Pharmaceutical Sciences, Vol. 77, p. 285 (1988); and Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392).
[0034] When a group of substituents is disclosed herein, it is understood that all individual members of that group and all subgroups, including any isomers and enantiomers of the group members, are disclosed separately. When a Markush group or other grouping is used herein, all individual members of the group and all combinations and subcombinations possible of the group are intended to be individually included in the disclosure. It is intended that any one or more members of any Makush group or listing provided in the specification can be excluded from the invention if desired. When a compound is described herein such that a particular isomer or enantiomer of the compound is not specified, for example, in a formula or in a chemical name, that description is intended to include each isomers and enantiomer of the compound described individual or in any combination. Additionally, unless otherwise specified, all isotopic variants of compounds disclosed herein are intended to be encompassed by the disclosure. For example, it will be understood that any one or more hydrogens in a molecule disclosed can be replaced with deuterium or tritium. Isotopic variants of a molecule are generally useful as standards in assays for the molecule and in chemical and biological research related to the molecule or its use. Specific names of compounds are intended to be exemplary, as it is known that one of ordinary skill in the art can name the same compounds differently.
[0035] Molecules disclosed herein may contain one or more ionizable groups [groups from which a proton can be removed (e.g., -OH, -COOH, etc.) or added (e.g., amines) or which can be quaternized (e.g., amines)]. All possible ionic forms of such molecules and salts thereof are intended to be included individually in the disclosure herein. With regard to salts of the compounds herein, one of ordinary skill in the art can select from among a wide variety of available counterions those that are appropriate for preparation of salts of this invention for a given application. For example, in general any anions can be employed in the formation of salts of compounds herein; e.g. halide, sulfate, carboxylate, acetate, phosphate, nitrate, trifluoroacetate, glycolate, pyruvate, oxalate, malate, succinate, fumarate, tartarate, citrate, benzoate, methanesulfonate, ethanesulfonate, p-toluenesulfonate, salicylate and others.
[0036] Compounds of the present invention, and salts or esters thereof, may exist in their tautomeric form, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention. Additionally, the compounds may have trans and cis isomers and may contain one or more chiral centers, therefore existing in enantiomeric and diastereomeric forms. The invention can encompass all such isomers, individual enantiomers, as well as mixtures of cis and trans isomers, mixtures of diastereomers; non-racemic and racemic mixtures of enantiomers (optical isomers); and the foregoing mixtures enriched for one or more forms; except as stated otherwise herein. When no specific mention is made of the configuration (cis, trans or R or S) of a compound (or of an asymmetric carbon), then any one of the isomers or a mixture of more than one isomer is intended. The processes for preparation can use racemates, enantiomers, or diastereomers as starting materials. When enantiomeric or diastereomeric products are prepared, they can be separated by conventional methods, for example, by chromatographic or fractional crystallization. The inventive compounds may be in the free or hydrate form.
[0037] Every formulation or combination of components described or exemplified herein can be used to practice the invention, unless otherwise stated.
[0038] Whenever a range is described in the present application, for example, a temperature range, a time range, or a composition or concentration range, all intermediate ranges and subranges, as well as all individual values included in the ranges given are intended to be included in the disclosure.
[0039] Information in any references disclosed herein can in some cases indicate the state of the art, for example for patent documents as of their effective filing dates; it is intended that such information can be employed herein, if needed, to exclude specific embodiments that are actually found to be in the prior art. For example, when a compound is disclosed and/or claimed, it should be understood that compounds qualifying as prior art with regard to the present invention, including compounds for which an enabling disclosure is provided in the references, are not intended to be included in the composition of matter claims herein.
[0040] Some references provided herein are incorporated by reference to provide details concerning sources of starting materials, additional starting materials, additional reagents, additional methods of synthesis, additional methods of analysis, and additional uses of the invention. One of ordinary skill in the art will appreciate that starting materials, reagents, solid substrates, synthetic methods, purification methods, and analytical methods other than those specifically exemplified can be employed in the practice of the invention based on knowledge in the art and without resort to undue experimentation.
[0041] In an embodiment, the invention provides a therapeutic composition comprising one or more compounds and for each compound a pharmaceutically acceptable salt or ester thereof; wherein the compounds are present in the composition in an amount or in a combined amount effective for obtaining the desired therapeutic benefit. The therapeutic compositions of this invention optionally further comprise one or more pharmaceutically acceptable components, for example carriers and excipients as known in the art.
[0042] In an embodiment, the invention provides a compound having the formula ZZ2:
Figure imgf000014_0001
wherein R1 and R2 each independently is hydrogen, halogen, alkyl, allyl, haloalkyl, alkenyl, alkenol, alkanol, or haloalkenyl. In an embodiment, R1 and R2 each independently is hydrogen, halogen, allyl, or short alkyl.
[0043] In an embodiment, the invention provides a compound selected from the group consisting of a PAC- 1 derivative combinatorial library comprising a hydrazide compound combined with an aldehyde compound. In an embodiment, the hydrazide compound is selected from the group consisting of hydrazides generated from AX compounds described herein.
[0044] In an embodiment, the aldehyde compound is selected from the group consisting of BX compounds described herein. In an embodiment, the hydrazide compound is selected from the group consisting of AX compounds described herein and the aldehyde compound is selected from the group consisting of BX compounds described herein.
[0045] In an embodiment, the invention provides a method of synthesizing a PAC- 1 derivative compound comprising providing a hydrazide compound, providing an aldehyde compound, and reacting the hydrazide compound with the aldehyde compound, thereby synthesizing a PAC- 1 derivative compound. [0046] In an embodiment, the hydrazide compound has the formula ZZ3:
Figure imgf000015_0001
[0047] In an embodiment, the aldehyde compound has the formula ZZ4:
Figure imgf000015_0002
[0048] In an embodiment, the hydrazide compound has the formula ZZ3 and the aldehyde compound has the formula ZZ4.
[0049] In an embodiment, the invention provides a compound selected from the group consisting of: L01 R06, L02R03, L02R06, L08R06, L09R03, L09R06, and L09R08.
[0050] In an embodiment, the invention provides a method of screening a candidate cancer patient for possible treatment with a procaspase activator by identifying an elevated level of a procaspase in the candidate, comprising obtaining a cell or tissue test sample from the candidate, assessing the procaspase level in the test sample, and determining whether the procaspase level is elevated in the test sample relative to a reference level, thereby screening a candidate cancer patient for possible treatment with a procaspase activator. In an embodiment, the procaspase is selected from the group consisting of procaspase-2, -3, -6-, -7, -8, and -9. In a particular embodiment, the procaspase is procaspase-3.
[0051] In an embodiment, an elevated level of the test sample is at least about 2- fold greater than the reference level. In an embodiment, an elevated level of the test sample is at least about 4-fold greater than the reference level. In an embodiment, the reference level is from a second test sample from the same patient. In an embodiment, the reference level is from a normal cell or tissue sample. The reference level can be from a cell line, such as a cancer cell line or a normal cell line. In an embodiment, the reference level is an absolute threshold amount. See Svingen, P.A. et a!., Clin. Cancer Res. 10:6807-6820 which describes various amounts of levels of procaspases including numbers of molecules per cell. [0052] In an embodiment, the invention provides a method of inducing death in a cancer cell, comprising administering to said cancer cell a compound capable of activating a procaspase molecule of said cancer cell. In an embodiment the procaspase is one or more of procasepas-3 and procaspase-7. In a preferred embodiment the procaspase is procaspase 3. In an embodiment, the compound has structural formula ZZ. In an embodiment, the compound has structural formula ZZ2.
[0053] It is recognized that regardless of the ultimate correctness of any mechanistic explanation or hypothesis believed or disclosed herein, an embodiment of the invention can nonetheless be operative and useful.
BRIEF DESCRIPTION OF THE FIGURES
[0054] Figure 1. A) In vitro activation of procaspase-3 and active caspase-3 by PAC-1. PAC-1 activates procaspase-3 with an EC50 = 0.22 μM. Error bars represent standard deviations from the mean. B) Cleavage of procaspase-3 to active caspase-3 as induced by PAC-1. Procaspase-3 was recombinantly expressed in E. coli with an N-terminal His-6 tag and purified, lmmunoblotting was performed with an anti-His-6 antibody. In the absence of PAC-1 , no maturation of procaspase-3 is observed. In the presence of 100 μM PAC-1 , cleavage to generate the p19 fragment is observed within 1 hour, and >50% cleavage is observed after 4 hours.
[0055] Figure 2. A) Activation of mutants in the "safety catch" region of procaspase-3 by PAC-1. PAC-1 has an EC50 for activation of 0.22 μM on wild type procaspase-3 (DDD), and corresponding EC5O values of 2.77 μM (DAD), 113 μM (DDA), and 131 μM (ADD) for certain mutants. B) PAC-1 activates procaspase-7 with an ECs0 of 4.5 μM. C) Dependence of PAC-1 activation of procaspase-3 on pH. At low pH the safety catch is "off1, and procaspase-3 is essentially maximally activated. Error bars represent standard deviations from the mean.
[0056] Figure 3. PAC-1 induces apoptosis in HL-60 cells. A) Phosphatidylserine exposure (as measured by Annexin-V staining) after a 20 hour treatment with 100 μM PAC-1. B) Chromatin condensation as visualized by Hoescht staining after a 20 hour treatment with 100 μM PAC-1. [0057] Figure 4. A) Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 10 μM etoposide. B) Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 100 μM PAC-1. C) PAC-1 treatment (100 μM) induces a rapid decrease in cellular PARP activity in HL-60 cells, consistent with an immediate activation of cellular caspase-3/- 7. In contrast, etoposide (10 μM) treated cells show a decrease in PARP activity at much later time points. D) PAC-1 induces cell death in a procaspase-3 dependent manner. For a number of diverse cancer cell lines, the procaspase-3 levels were determined (by flow cytometry) and the IC50 of PAC-1 was measured (R2=0.9822). PAC-1 is quite potent (IC5O=O.35 μM) in the NCI-H226 lung cancer cell line known to have high levels of procaspase-3, but markedly less potent in normal white blood cells derived from the bone marrow of a healthy human donor.
[0058] Fig. 5A illustrates relative procaspase-3 levels in normal and cancerous cells from several patients.
[0059] Fig. 5B illustrates IC50 levels for PAC-1 in a variety of cell types having a range of relative procaspase-3 levels.
[0060] Fig. 5C illustrates the effect of treating animals with PAC-1 on outcomes of tumor growth.
[0061] Fig. 5D illustrates the effect of oral treatment of animals with PAC-1 on outcomes of tumor growth.
[0062] Fig. 5E illustrates results of progression of cancer in a lung cancer model for control, PAC-1 , and gefitinib (Iressa™; AstraZeneca) treatment groups. Tumor cells were injected into mice by i.v. administration; Iressa and PAC-1 were given orally at 100 mg/kg.
[0063] Fig. 6A illustrates relative procaspase-3 levels in normal and cancerous cells of three patients.
[0064] Fig. 6B illustrates the sensitivity of normal and cancerous cells from Patient 3 to treatment with PAC-1. [0065] Fig. 7 illustrates results of administering PAC-1 intraperitoneal^ in the context of a mouse model of lung cancer.
[0066] Fig. 8A and 8B illustrate structures for compounds of PAC-1 derivatives and a combinatorial library.
[0067] Fig. 9 illustrates a nucleotide sequence for Homo sapiens caspase 3, apoptosis-related cysteine peptidase (CASP3), transcript variant alpha, mRNA (Accession No. NM_004346; 2689 bp mRNA linear; obtained from http://www.ncbi.nlm.nih.gov/entrez).
[0068] Fig. 10 illustrates a nucleotide sequence for Homo sapiens caspase 7, apoptosis-related cysteine peptidase (CASP7), transcript variant alpha, mRNA (Accession No. NM_001227; 2605 bp; mRNA linear; obtained from http://www.ncbi.nlm.nih.gov/entrez).
DETAILED DESCRIPTION OF THE INVENTION
[0069] The following definitions are provided to clarify their specific use in the context of the invention.
[0070] When used herein, the term "chemotherapeutic agent" refers to any substance capable of reducing or preventing the growth, proliferation, or spread of a cancer cell, a population of cancer cells, tumor, or other malignant tissue. The term is intended also to encompass any antitumor or anticancer agent.
[0071] When used herein, the term "effective amount" is intended to encompass contexts such as a pharmaceutically effective amount or therapeutically effective amount. For example, in embodiments the amount is capable of achieving a beneficial state, beneficial outcome, functional activity in a screening assay, or improvement of a clinical condition.
[0072] When used herein, the term "cancer cell" is intended to encompass definitions as broadly understood in the art. In an embodiment, the term refers to an abnormally regulated cell that can contribute to a clinical condition of cancer in a human or animal. In an embodiment, the term can refer to a cultured cell line or a cell within or derived from a human or animal body. A cancer cell can be of a wide variety of differentiated cell, tissue, or organ types as is understood in the art.
[0073] The term "alkyl" refers to a monoradical branched or unbranched saturated hydrocarbon chain preferably having from 1 to 22 carbon atoms and to cycloalkyl groups having one or more rings having 3 to 22 carbon atoms. Short alkyl groups are those having 1 to 6 carbon atoms including methyl, ethyl, propyl, butyl, pentyl and hexyl groups, including all isomers thereof. Long alkyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 and those having 16-18 carbon atoms.
[0074] The term "cycloalkyl" refers to cyclic alkyl groups of from 3 to 22 carbon atoms having a single cyclic ring or multiple condensed rings. Cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.
[0075] The term "alkenyl" refers to a monoradical of a branched or unbranched unsaturated hydrocarbon group preferably having from 2 to 22 carbon atoms and to cycloalkenyl groups having one or more rings having 3 to 22 carbon atoms wherein at least one ring contains a double bond. Alkenyl groups may contain one or more double bonds (C=C) which may be conjugated. Preferred alkenyl groups are those having 1 or 2 double bonds. Short alkenyl groups are those having 2 to 6 carbon atoms including ethylene (vinyl) propylene, butylene, pentylene and hexylene groups, including all isomers thereof. Long alkenyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 carbon atoms and those having 16-18 carbon atoms. The term "cycloalkenyl" refers to cyclic alkenyl groups of from 3 to 22 carbon atoms having a single cyclic ring or multiple condensed rings in which at least one ring contains a double bond (C=C). Cycloalkenyl groups include, by way of example, single ring structures such as cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclooctenyl, cylcooctadienyl and cyclooctatrienyl.
[0076] The term "alkynyl" refers to a monoradical of an unsaturated hydrocarbon preferably having from 2 to 22 carbon atoms and having one or more triple bonds (CDC). Alkynyl groups include ethynyl, propargyl, and the like. Short alkynyl groups are those having 2 to 6 carbon atoms, including all isomers thereof, Long alkynyl groups are those having 8-22 carbon atoms and preferably those having 12-22 carbon atoms as well as those having 12-20 carbon atoms and those having 16-18 carbon atoms.
[0077] The term "aryl" refers to a group containing an unsaturated aromatic carbocyclic group of from 6 to 22 carbon atoms having a single ring (e.g., phenyl), one or mroe rings (e.g., biphenyl)or multiple condensed (fused) rings, wherein at least one ring is aromatic (e.g., naphthyl, dihydrophenanthrenyl, fluorenyl, or anthryl). Aryls include phenyl, naphthyl and the like. Aryl groups may contain portions that are alkyl, alkenyl or akyriyl in addition to the the unsaturated aromatic ring(s). The term "alkaryl" refers to the aryl groups containing alkyl portions, i.e., -alkylene-aryl and -substituted alkylene-aryly. Such alkaryl groups are exemplified by benzyl, phenethyl and the like.
[0078] Alkyl, alkenyl, alkynyl and aryl groups are optionally substituted as described herein (the term(s) can include substituted variations) and may contain 1-8 non-hydrogen substituents dependent upon the number of carbon atoms in the group and the degree of unsaturation of the group.
[0079] The term "alkylene" refers to a diradical of a branched or unbranched saturated hydrocarbon chain, preferably having from 1 to 10 carbon atoms, more preferably having 1 -6 carbon atoms, and more preferably having 2-4 carbon atoms; the term can include substituted variations. This term is exemplified by groups such as methylene (-CH2-), ethylene (-CH2CH2-), more generally -(CH2)n-, where n is 1-
10 or more preferably 1-6 or n is 2, 3 or 4. Alkylene groups may be branched. Alkylene groups may be optionally substituted. Alkylene groups may have up to two non-hydrogen substituents per carbon atoms. Perferred substituted alkylene groups have 1 , 2, 3 or 4 non-hydrogen substituents.
[0080] The term "arylene" refers to the diradical derived from aryl (including substituted aryl) as defined above and is exemplified by 1 ,2-phenylene, 1 ,3- phenylene, 1 ,4-phenylene, 1 ,2-naphthylene and the like. [0081] The term "amino" refers to the group -NH2 or to the group -NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic provided that both R's are not hydrogen.
[0082] Alkyl groups are optionally substituted as discussed herein and may, dependent upon the size of the alkyl group, have preferably from 1-10 substituent groups. Substituted alkyl groups include those that carry 1 to 8 substituents, 1 to 5 substituents, 1 to 3 substituents, and 1 or 2 substituents.
[0083] Haloalkyl" refers to alkyl as defined herein substituted by one or more halo groups as defined herein, which may be the same or different. Representative haloalkyl groups include, by way of example, trifluoromethyl, 3-fluorododecyl, 12,12,12-trifluorododecyl, 2-bromooctyl, 3-bromo-6-chloroheptyl, and the like.
[0084] The term "heteroaryl" refers to an aromatic group of from 2 to 22carbon atoms having 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur within at least one ring (if there is more than one ring). Heteroaryl groups may be optionally substituted.
[0085] The term "heterocycle" or "heterocyclic" refers to a monoradical saturated or unsaturated group having a single ring or multiple condensed rings, from 2-22 carbon atoms and from 1 to 6 hetero atoms, preferably 1 to 4 heteroatoms, selected from nitrogen, sulfur, phosphorus, and/or oxygen within at least one ring. Heterocyclic groups may be substituted.
[0086] The term "ester" refers to chemical entities as understood in the art and in particular can include groups of the form (RCO-).
[0087] As to any of the above groups which contain one or more substituents, it is understood, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. The compounds of this invention include all novel stereochemical isomers arising from the substitution of disclosed compounds. [0088] The invention may be further understood by the following non-limiting examples.
EXAMPLE 1. Procaspase activating compounds.
[0089] Mutation or aberrant expression of proteins in the apoptotic cascade is a frequent hallmark of cancer. These changes can prevent proapoptotic signals from being transmitted to the executioner caspases, thus preventing apoptotic cell death and allowing cellular proliferation. Caspase-3 and caspase-7 are key executioner caspases, existing as inactive zymogens that are activated by upstream signals. Importantly, expression levels of procaspase-3 are significantly higher in certain cancerous cells relative to non-cancerous controls. Here we report the identification of small molecules that directly activate procaspase-3 to active caspase-3. A particular compound, PAC-1 , effects activation in vitro with an EC50 on the order of 220 nanomolar and induces apoptosis in a multitude of cancerous cell lines.
[0090] In contrast to many known anti-cancer drugs, cells treated with PAC-1 show an immediate activation of procaspase-3, and the toxicity of PAC-1 is shown to be directly proportional to the amount of procaspase-3 contained in a cell. Thus PAC-1 directly activates procaspase-3 to caspase-3 in vivo, allowing this compound to induce apoptosis even in cells that have defective apoptotic machinery. PAC-1 is the first small molecule known to directly activate procaspase-3; the direct activation of executioner caspases is a novel anti-cancer strategy that may prove beneficial in a variety of cancers, including the many cancers in which procaspase-3 is u p regulated.
[0091] A collection of about 20,000 structurally diverse small molecules was screened for the ability to activate procaspase-3 in vitro. Procaspase-3 was expressed and purified in E. coli (Roy et al., 2001). Procaspase-3 (at a concentration of 50 ng/mL) was added to the wells of a 384-well plate, and the compounds were added to a final concentration of approximately 40 μM. Each plate was then incubated for two hours at 37°C, after which the caspase-3 peptidic substrate Ac-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNa) was added to a concentration of 200 μM. The formation of the p-nitroaniline chromophore was followed at 405 nm over the course of two hours. [0092] Of the compounds evaluated, four induced a significant increase over background in the hydrolysis of the peptidic caspase-3 substrate. Of those four, one showed a strong dose-dependent effect on in vitro procaspase-3 activation. As shown in Figure 1A, this first procaspase activating compound (PAC-1) gives half- maximal activation of procaspase-3 at a concentration of 0.22 μM. This compound is not simply increasing the activity of caspase-3 itself, as it has no effect on the catalytic activity of the fully processed caspase-3 enzyme (Figure 1A).
[0093] Procaspase-3 has an N-terminal pro domain (residues 1-28), followed by a large subunit (17 kDa) and a small subunit (12 kDa) that are separated by an intersubunit linker (Pop et al., 2003). In vivo, two procaspase-3 monomers assemble to form a catalytically inactive homodimer that can be activated by cleavage at D175 in the intersubunit linker. The precise role of the pro domain is unclear, and it has been shown that cleavage in the intersubunit region alone is sufficient for full catalytic activity (Stennicke, H. R. et al., 1998). Although procaspase-3 is catalytically competent, it is highly resistant to autoactivation due to the presence of the 12 amino acid safety catch; however, when the safety catch is mutated significant autoactivation of procaspase-3 is observed (Roy et al., 2001). Compounds that interact with this important regulatory region or at other positions can allow the autoactivation of procaspase-3.
[0094] To directly assess the ability of PAC-1 to catalyze the autoactivation of procaspase-3, the procaspase-3 protein was incubated with 100 μM of PAC-1 for time points ranging from one to five hours. As shown by the Western blot in Figure 1 B, PAC-1 induces the cleavage of procaspase-3 in a time- dependent fashion, with >50% processing observed after 4 hours. In contrast, procaspase-3 incubated in buffer shows virtually no autoactivation over that same time span. In an attempt to pinpoint the region of procaspase-3 with which PAC-1 is interacting, alanine substitutions were made in the key aspartic acid triad in the safety catch region, residues Asp179, Asp180 and Asp181. Mutations at these positions all dramatically decreased the ability of PAC-1 to activate procaspase-3, with certain mutations more detrimental to activation of procaspase-3 by PAC-1 (Figure 2A).
[0095] Like caspase-3, caspase-7 also exists as an inactive zymogen that is activated by proteolysis. Caspase-3 and caspase-7 are both executioner caspases and have considerable sequence and structural homology (Denault, J.-B. et al., 2003). Procaspase-7 may also have a similar safety catch region, although it has only two aspartic acids in the key triad (Asp-Thr-Asp), instead of three. As indicated by the data in Figure 2B, PAC-1 can also activate procaspase-7, although in a less efficient manner than its activation of procaspase-3 (EC50 of 4.5 μM versus 0.22 μM for procaspase-3 activation). The potency of procaspase-7 activation by PAC-1 is similar to its effect on the Asp-Ala-Asp mutant of procaspase-3 (EC5O = 2.77 μM). The effect of PAC-1 is abolished at low pH values where procaspase-3 undergoes rapid autoactivation (Figure 2C).
[0096] The ability for a small molecule that activates procaspase-3 to induce apoptosis in human cell lines was tested, and PAC-1 was found to induce apoptosis in a variety of cancer cell lines. In HL-60 cells addition of PAC-1 leads to considerable phosphatidylserine exposure on the cell membrane accompanied by significant chromatin condensation (Figures 3A and B). In addition, the compound induces cleavage of PARP-1 (as assessed by an in vivo PARP activity assay; Putt KS et al., 2005) and causes mitochondrial membrane depolarization (see below). Significant cellular blebbing was also observed by microscopy. Furthermore, the toxicity of PAC-1 could be abolished in the presence of the caspase inhibitor z-VAD- fmk (data not shown; see Slee et al., 1996).
[0097] If PAC-1 is indeed inducing apoptosis via direct activation of procaspase-3, the time course of apoptotic events should be altered relative to that observed with standard proapoptotic agents. Etoposide is well known to induce apoptosis through the intrinsic pathway; thus, mitochondrial membrane depolarization is followed by procaspase-3 activation in etoposide-treated cells. Indeed, in HL-60 cells treated with 10 μM etoposide, mitochondrial membrane depolarization is observed, followed by detection of caspase-3-like activity (Figure 4A). In contrast, treatment of cells with PAC-1 gives a markedly different result. With PAC-1 , the first observed biochemical hallmark of apoptosis is caspase-3-like enzymatic activity. This activity is noted within minutes of compound addition, and 50% activation takes place in just over 2 hours and well before any significant mitochondrial membrane depolarization (Figure 4B). In addition, PARP activity is rapidly reduced in cells treated with PAC-1 , whereas this reduction is observed at later time points in etoposide treated cells (Figure 4C). Control experiments show that PAC-1 does not directly inhibit enzymatic activity of PARP-1. In the typical sequence of apoptotic events, the mitochondrial membrane depolarizes, caspases are activated, and caspase substrates (such as PARP-1) are cleaved. The observation that cells treated with PAC-1 show a rapid activation of caspase-3/-7 (before mitochondrial membrane depolarization) and a rapid cleavage of a caspase substrate is indicative of this compound exerting its cellular toxicity through the direct activation of procaspase-3.
[0098] To further define the potency of PAC-1 , the ability of this compound to induce cell death in cancer cell lines with varying levels of procaspase-3 was assessed. A determination was made of the amount of procaspase-3 present in multiple cancer cell lines (leukemia, lymphoma, melanoma, neuroblastoma, breast cancer, lung cancer and renal cancer) and in the white blood cells isolated from the bone marrow of a healthy donor. The IC50 values for cell death induction were obtained for PAC-1 in these cell lines. The combined data shows a strong correlation between cellular concentration of procaspase-3 and sensitivity to PAC-1 (Figure 4D). Notably, the white blood cells derived from the bone marrow of a healthy human donor are among those with the lowest amount of procaspase-3, and PAC-1 is comparatively less toxic to these cells. PAC-1 is most potent versus the lung cancer cell line NCI-H226, with an IC50 of 0.35 μM. In accordance with data in the literature (Svingen et al., 2004), we found this cell line to have a concentration of procaspase-3 that is greater than five times that of the non-cancerous control.
[0099] In contrast to these experiments with PAC-1 , etoposide showed no such correlation between potency in cell culture and cellular levels of procaspase-3. For instance, etoposide was ineffective (IC50 >50 μM) in inducing death in three of the melanoma cell lines (UACC-62, CRL-1872, and B16-F10), the breast cancer cell line (Hs 578t), and the lung cancer cell line (NCI-H226); these cell lines have procaspase-3 levels of 1.0, 2.4, 1.9, 3.7, and 5.3, respectively. Etoposide was effective (IC50 <1 μM) versus HL-60, U-937, SK-N-SH and PC-12, which have procaspase-3 levels of 4.3, 4.0, 4.7, and 4.4, respectively. Thus, overall there is no correlation between procaspase-3 levels and IC5O for etoposide.
[00100] Cancerous cells typically have a reduced sensitivity to proapoptotic signals due to the mutation or aberrant expression of an assortment of proteins in the apoptotic cascade. As such, many types of cancer are notoriously resistant to not only the endogenous signals for apoptotic cell death, but also to chemotherapeutic agents that act through similar mechanisms. The paradoxical upregulation of procaspase-3 expression levels in certain cancers provides an opportunity to use this existing intracellular pool of protein to directly induce apoptosis, thus bypassing the often non-functional or compromised upstream portion of the cascade. Although procaspase-3 is notorious for its relative inability to undergo autoactivation, it is dependent upon a 12 amino acid safety catch to keep itself in the inactive state. PAC-1 induces the autoactivation of procaspase-3 in vitro, and this activation is greatly diminished by mutation of the key tri-aspartate region of the safety catch.
This data is consistent with the notion that PAC-1 is directly interfering with the ability of the safety catch to maintain procaspase-3 dormancy.
[00101] In cell culture, PAC-1 treatment induces rapid caspase-3-like activity. It is likely that the caspase-3 mediated cleavage of anti-apoptotic proteins (Bcl-2, BcI-XL, etc.) then induces depolarization of the mitochondrial membrane and amplifies apoptosis. Further, the potency of PAC-1 toward a variety of cancer cell lines is directly proportional to the concentration of procaspase-3 in the cell. It is worth noting that several of the cell lines that PAC-1 is effective against have faulty apoptotic pathways that make them resistant to apoptosis; for instance, Apaf-1 expression is dramatically decreased in SK-MEL-5 cells, and Bcl-2 is overexpressed in the NCI-H226 lung cancer cell line.
[00102] Data presented herein fully support the notion that procaspase-3 activating compounds can be exceedingly effective against common cancers. The effectiveness can be enhanced for situations in which procaspase-3 levels are aberrantly high.
[00103] Assessment of procaspase-3 levels in cancer biopsies can be simple and rapid; as such, the potential effectiveness of a compound such as PAC-1 can be assessed a priori with a high degree of accuracy. Procaspase-3 activators and methods herein thus provide personalized medicine strategies that can be preferential to therapies that rely on general cytotoxins in the realm of anti-cancer treatments. MATERIALS AND METHODS
[00104] Materials: Ni-NTA resin and anti-Penta His Alexa Fluor 647 antibody was purchased from Qiagen (Valencia, CA). Bradford dye was purchased from Bio-Rad (Hercules, CA). Pin transfer devices were purchased from V & P Scientific (San Diego, CA). The reagent z-vad-fmk was purchased from Calbiochem (San Diego, CA). Rosetta E. coli was purchased from Novagen (Madison, Wl). Anti-caspase-3 antibody was purchased from Sigma (St. Louis, MO). Annexin V Alexa Fluor 488 conjugate, JC-9, and propidium iodide were purchased from Molecular Probes (Eugene, OR). IPTG and MTS/PMS CellTiter 96 Cell Proliferation Assay reagent were purchased from Promega (Madison, Wl). Fetal Bovine Serum was purchased from Biomeda (Foster City, CA). 96 and 384-well microtiter plates, microscope slides, microscope coverslips, horse serum and all other reagents were purchased from Fisher (Chicago, IL).
[00105] Methods: Cell Culture Conditions. U-937, HL-60, CRL-1872, ACHN, NCI- H226, SK-MEL-5 and UACC-62 cells were grown in RPMI 1640 media supplemented with 10% FBS. SK-N-SH, B16-F10 and Hs 578t cells were grown in Eagle's minimal essential medium with Earle's BSS, 1.5 g/L sodium bicarbonate and supplemented with 10% FBS. PC-12 cells were grown in RPMI 1640 media supplemented with 5% FBS and 10% horse serum. Human bone marrow was grown in IDMEM supplemented with 40% FBS. All cell lines were incubated at 37°C in a 5% CO2, 95% air atmosphere. U-937 and HL-60 cells were split every two to three days as needed. Human bone marrow was thawed from frozen stock and immediately diluted and used for experiments. All other cells were split when they reached approximately 80% confluency.
[00106] Protein Expression and Purification. 1 mL of an overnight culture of Rosetta E. coli containing the procaspase-3 or procaspase-7 expression plasmid was seeded into 1 L of LB media containing proper antibiotic. Cells were induced with 1 mM IPTG for 30 minutes. Cells were then spun down and re-suspended in NTA binding buffer (150 mM NaCI, 50 mM Tris, 10 mM Imidazole, pH 7.9). The cells were lysed by passing twice through a French press. The cell lysate was then spun at 14,000 x g for 30 min. The supernatant was decanted and 1 mL of nickel-NTA resin was added. The cell lysate was incubated for 1 hour at 4°C. The resin was loaded on a column, washed with 10 mL NTA binding buffer followed by 10 mL NTA wash buffer (150 mM NaCI, 50 mM Tris, 20 mM Imidazole, pH 7.9). The proteins were eluted in 1 mL fractions with 10 mL of NTA elution buffer (150 mM NaCI, 50 mM Tris, 250 mM Imidazole, pH 7.9). Fractions containing protein were pooled and the amount of protein was determined using the Bradford assay.
[00107] Library Screen. Isolated procaspase-3 was diluted to 50 ng/mL in caspase assay buffer (50 mM HEPES, 100 mM NaCI, 10 mM DTT, 0.1 mM EDTA1 0.1 % CHAPS and 10% glycerol, pH 7.4). 45 μL of the procaspase-3 solution was added to each well of a Nunc 384-well flat bottom microtiter plate. Approximately 20,000 compounds were screened. About 6,000 of the compounds were collected from various sources within the department of chemistry at the University of Illinois; their structures are available at: http://www.scs.uiuc.edu/~phgroup/comcollections.html. The other approximately 14,000 compounds were purchased from Chembridge Corporation (San Diego, California). PAC-1 was a member of the compounds purchased from Chembridge Corporation.
[00108] The compounds, made up as 10 mM stock solutions in DMSO, were transferred into the wells using a 384-pin transfer apparatus that transfers 0.2 μL of compound. This yielded a final compound concentration of about 40 μM. Controls were performed in which only DMSO (containing no compound) was pin-transferred. The plates were then incubated for 2 hours at 370C. 5 μL of a 2 mM solution of Ac- DEVD-pNA (N-acetyl-ASP-Glu-Val-Asp-p-nitroanilide) in caspase assay buffer was added to each well. The plate was then read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA). The slope of the linear portion for each well was used to determine the activity of caspase-3.
[00109] Activation curves. The dose dependence of procaspase-3 activators was determined by adding various concentrations of compound to 90 μL of a 50 ng/mL procaspase-3, active caspase-3, procaspase-7 or active caspase-7 in caspase assay buffer in a 96-well plate. The plate was then incubated for 12 hours at 370C. 10 μL of a 2 mM solution of Ac-DEVD-pNA in caspase assay buffer was then added to each well. The plate was read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 well plate reader. The slope of the linear portion for each well was determined and the fold increase in activation from non-treated control wells was calculated.
[00110] PAC-1 activation gel. Procaspase-3 was expressed and isolated exactly as above. Procaspase-3 was diluted to about 50 μg/mL in caspase assay buffer. The procaspase-3 was then incubated in the presence or absence of 100 μM PAC-1 for varying times at 37°C. After this incubation, an equal volume of load buffer (150 mM NaCI, 50 mM Tris, 2% SDS, 20% glycerol, pH 8.0) was added to each procaspase-3 sample. All samples were then stored at -800C until the time-course was completed. All samples were then incubated at 950C for 5 minutes and run on a 12% SDS- PAGE gel. Proteins were then transferred to nitrocellulose paper overnight. Blots were washed in TTBS (150 mM NaCI, 50 mM Tris, 0.1 % Tween-20, pH 7.4) and blocked with a 10% milk solution for 2 hours. Blots were then incubated in a 1 :5000 dilution of anti-Penta His Alexa Fluor 647 antibody for 2 hours. The blot was then washed with TTBS and scanned on a Typhoon fluorescence scanner (Amersham Biosciences, SunnyVale CA).
[00111] Safety catch mutations. The DDD procaspase-3 safety catch (SEQ ID NO: 1 ; SEQ ID NO:2; SEQ ID NO:9) was mutated to ADD (SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:10), DAD (SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:11) and DDA (SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:12) using the quickchange strategy with the following primers, gacagacagtggtgttgCGgatgacatggcgtgtcataaaatacc (SEQ ID NO: 13), gacagacagtggtgttgatgCtgacatggcgtgtcataaaatacc (SEQ ID NO: 14) and gacagacagtggtgttgatgatgCcatggcgtgtcataaaatacc (SEQ
ID NO:15) respectively. See also Figure 9 and Figure 10. Mutated bases are underlined and capitalized. All mutant plasmids were sequenced to ensure proper sequence throughout the gene. All mutant plasmids were expressed exactly as wild- type procaspase-3 as described above. The ability of PAC-1 to activate each procaspase-3 mutant was determined by adding various concentrations of PAC-1 to 90 μl_ of a 50 ng/mL wild-type procaspase-3 and mutant procaspase-3 in caspase assay buffer in a 96-well plate. The plate was then incubated for 12 hours at 37°C. 10 μL of a 2 mM solution of Ac-DEVD-pNA in caspase assay buffer was then added to each well. The plate was read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 well plate reader. The slope of the linear portion for each well was determined and the fold increase in activity for each mutant was calculated.
[00112] Effect of pH on PAC-1 activation of procaspase-3. The effect of pH on procaspase-3 activation by PAC-1 was determined by diluting procaspase-3 in pH caspase assay buffer (25 mM MES, 25 mM Tris, 25 mM HEPES, 25 mM PIPES, 100 mM NaCI, 10 mM DTT, 0.1 mM EDTA, 0.1% CHAPS and 10% glycerol) to a concentration of 50 ng/mL The buffer was then changed to various pH values and 90 μL was added to each well of a 96-well plate. PAC-1 was added to a concentration of 100 μM or DMSO was added as a control for each pH value. The plate was then incubated for 12 hours at 37°C. 10 μL of a 2 mM solution of Ac- DEVD-pNA in caspase assay buffer was then added to each well. The plate was read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA). The slope of the linear portion for each well was determined and the fold increase in activation for each pH value was calculated.
[00113] Annexin V staining. 500 μL of media containing 200 μM PAC-1 or only DMSO as a control was added to the wells of a 24-well plate. 500 μL HL-60 cells at a concentration of 2 x 106 cells/mL were then added to the 24-well plate. The plate was incubated for 20 hours at 37°C. Cells were harvested by centrifugation and washed twice in PBS. The cells were then washed in AnnexinV binding buffer (10 mM HEPES, 140 mM NaCI, 2.5 mM CaCI2, pH 7.4) and resuspended in 100 μL of Annexin V binding buffer. 5 μL of annexin V, Alexa Fluor 488 conjugate was added and the tubes were incubated at room temperature for 15 minutes protected from light. 400 μL of Annexin V binding buffer was then added, followed by the addition of 1 μL of a 1 mg/mL solution of propidium iodide. The fluorescent intensity of each cell was determined by flow cytometry at 525 nm (green channel) and 675 nm (red channel). At least 50,000 cells were analyzed in each experiment.
[00114] Condensed chromatin staining. 500 μL of media containing 200 μM PAC-1 or only DMSO as a control was added to the wells of a 24-well plate. 500 μL HL-60 cells at a concentration of 2 x 10s cells/mL were then added to the 24-well plate. The cells were incubated for 20 hours and harvested by centrifugation. The cells were then washed in PBS buffer followed by the addition of ice-cold 100% ethanol. The cells were fixed overnight at 40C. Fixed cells were incubated with 2 μg/mL Hoechst- 33258 for 30 minutes at room temperature. A drop of cells was then added to a microscope slide and covered with a No. 1 thickness coverslip. Condensed chromatin was observed at 40Ox magnification on a Zeiss Axiovert 100 microscope.
[00115] Cell death inhibition by z-vad-fmk. 100 μl_ HL-60 cells at a concentration of 5 x 105 cells/mL were added to the wells of a 96-well plate. The cells were then incubated for 1 hour in the presence or absence of 100 μM z-vad-fmk, a cell- permeable pan caspase inhibitor. PAC-1 was then added at various concentrations, and the cells were incubated for an additional 24 hours. Cell death was quantitated by the addition of 20 μL of the MTS/PMS CellTiter 96 Cell Proliferation Assay reagent to each well. The plates were incubated at 370C for approximately 45 minutes until the colored product formed. The absorbance was then measured at 490 nm in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA).
[00116] In vivo determination of mitochondrial membrane potential. 1 ml_ of HL-60 cells at a concentration of 1 x 106 cells/mL were added to the wells of a 24-well plate. PAC-1 was then added to a concentration of 100 μM or only DMSO was added as a control. The cells were incubated for various times, and the cells then were harvested by centrifugation. The cells were washed in PBS and resuspended in 1 mL of PBS. 10 μg of the JC-9 dye was added and the cells were incubated at room temperature for 10 minutes protected from light. The cells were then washed two times with PBS and brought up in 500 μL PBS. The fluorescent intensity of each cell was determined by flow cytometry at 525 nm (green channel) and 675 nm (red channel). 50,000 cells were analyzed in each experiment. The shift in the red channel was then used to determine the amount of mitochondrial membrane depolarization.
[00117] In vivo determination of caspase-3 like activity. The amount of caspase-3 like protease activity was determined by the amount of Ac-DEVD-pNA (N-acetyl- ASP-Glu-Val-Asp-p-nitroanilide) cleaved per minute by cell lysates. To accomplish this, 50 μL of media containing varying concentrations of PAC-1 was added to the wells of a 96-well plate. 50 μL of HL-60 cells at a concentration of 5 x 106 cells/mL were added to the plate and incubated for various times. After the incubation period, the plate was spun at 1000 x g for 5 minutes to pellet the cells. The cells were then washed with 100 μL of PBS and resuspended in 150 μL of ice cold Caspase Assay Buffer. Each well was then sonicated to lyse the cells. 90 μl_ of cell lysate was transferred from each well into a new plate. Ac-DEVD-pNA was added into each well to give a final concentration of 200 μM. The plate was then read every 2 minutes at 405 nm for 2 hours in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA). The slope of the linear portion for each well was determined and the amount of Ac-DEVD-pNA cleaved per minute was calculated.
[00118] In vivo determination of PARP cleavage. The amount of PARP cleavage was determined by using an in vivo PARP activity assay. To accomplish this, 50 μL of media containing 200 μM NAD+ was added to the control wells of a 96-well plate. 50 μL of media containing 200 μM PAC-1 and 200 μM NAD+ was then added to the experimental wells. 25 μL of HL-60 cells at a concentration of 5 x 106 cells/mL were then added to each well. The cells were incubated for various times and then spun at 1000 x g for 5 minutes. The cell media was removed and replaced with 50 μL Lysing PARP Buffer (50 mM Tris, 10 mM MgCI2, pH 8.0, 1% Triton X-100) containing 25 mM H2O2. The plate was then incubated for 60 minutes at 37°C. To determine the amount of NAD+ still present, 20 μL of 2 M KOH and 20 μL of a 20% (v/v) acetophenone (in ethanol) solution was added to each well of the 96-well plate. The plate was then incubated for 10 minutes at 40C. 90 μL of an 88% (v/v) formic acid solution was added to each well of the 96-well plate. The plate was then incubated for 5 min. in an oven set to 1100C. The plate was allowed to cool and then read on a Criterion Analyst AD (Molecular Devices, Sunnyvale, CA) with an excitation of 360 nm, an emission of 445 nm and a 400 nm" cutoff dichroic mirror. The fluorophore was excited using a 1000 W continuous lamp for 1.6 x 105 μs with 5 reads performed per well. The number of moles of NAD+ cleaved per minute was then calculated and the remaining PARP activity as compared to control wells was determined.
[00119] Relative concentration of procaspase-3 in various cell lines. U-937, HL-60 and human bone marrow cells were harvested by centrifugation while all other cell lines were first trypsinized to release the cells and then harvested by centrifugation. All cells were washed in PBS and resusupended in 1 mL of ice-cold 100% ethanol. Cells were fixed overnight at 40C. The cells were spun at 1000 x g for 5 minutes, washed with PBS and 100 μL of a 1:100 dilution of anti-caspase-3 antibody in PBS was then added. The cells were incubated for 2 hours at room temperature followed by five PBS washes. The cells were then resuspended in 1 ml_ of a 1:10,000 dilution of anti-mouse Ab Cy3 labeled antibody for 2 hours at room temperature protected from light. The cells were washed five times with PBS and resuspended in 500 μl_ of PBS. The fluorescent intensity of each cell was determined by flow cytometry at 675 nm (red channel). At least 20,000 cells were analyzed in each experiment. The median of the population was used to determine the relative concentration of procaspase-3 in each cell line.
[00120] Determination of IC5O values in various cell lines. 50 μL of media containing various concentrations of PAC-1 or etoposide was added to each well of a 96-well plate except control wells, which contained only DMSO. U-937, HL-60 and human bone marrow cells were harvested by centrifugation, while all other cell lines were first trypsinized before centrifugation. Cells were then resuspended in media and diluted to either 1 x 106 cells/mL for U-937, HL-60 and human bone marrow cells or 50,000 cells/mL for all other cell lines. 50 μL of the cell solutions were then added to each well and the plates were incubated for either 24 or 72 hours for etoposide and PAC-1 respectively. Cell death was quantitated by the addition of 20 μL of the MTS/PMS CellTiter 96 Cell Proliferation Assay reagent to each well. The plates were then incubated at 37°C for approximately one hour until the colored product formed. The absorbance was measured at 490 nm in a Spectra Max Plus 384 plate reader (Molecular Devices, Sunnyvale CA).
[00121] Data Analysis: The data from all flow cytometry experiments was analyzed using Summit Software (Cytomation, Fort Collins CO). All graphs were analyzed using Table Curve 2D.
[00122] Professor Ronald Hoffman (University of Illinois-Chicago Cancer Center) provided human bone marrow. Professor Guy Salvesen (Burnham Institute) provided the procaspase-3 and procaspase-7 expression vectors.
[00123] REFERENCE TO SEQUENCE LISTING - Appendix A. The separately accompanying sequence listing information, designated Appendix A, is to be considered and incorporated as part of the specification herewith. Table 1. Overview of Sequence Listing information.
Figure imgf000034_0001
EXAMPLE 2. Synthesis of procaspase activating compounds.
[00124] PAC-1 and other compounds are prepared according to the following schemes, e.g., Scheme 1 and/or Scheme 2. Further variations are prepared according to methods known in the art.
Figure imgf000035_0002
[00125] In a particular example, PAC-1 is prepared according to Scheme 2:
Figure imgf000035_0001
EXAMPLE 3. Analogs of PAC-1.
[00126] Analog compounds of PAC-1 were prepared and assessed for the capability to directly activate purified procaspase-3 in vitro. Table 2. Activity of PAC-1 and analog compounds.
Figure imgf000036_0001
EXAMPLE 4. Pharmaceutical embodiments.
[00127] The following describes information relevant to pharmaceutical and pharmacological embodiments and is further supplemented by information in the art available to one of ordinary skill. The exact formulation, route of administration and dosage can be chosen by an individual physician in view of a patient's condition (see e.g. Fingl et. al., in The Pharmacological Basis of Therapeutics, 1975, Ch. 1 p. 1).
[00128] It should be noted that the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity, or to organ dysfunctions, etc. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (in light of or precluding toxicity aspects). The magnitude of an administered dose in the management of the disorder of interest can vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, can also vary according to circumstances, e.g. the age, body weight, and response of the individual patient. A program comparable to that discussed above also may be used in veterinary medicine.
[00129] Depending on the specific conditions being treated and the targeting method selected, such agents may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in Alfonso and Gennaro (1995) and elsewhere in the art. Suitable routes may include, for example, oral, rectal, transdermal, vaginal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, or intramedullary injections, as well as intraocular, intrathecal, intravenous, or intraperitoneal administration.
[00130] For injection or other routes, agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, water for injection, physiological saline buffer, or other solution. For transmucosai administration, penetrants appropriate to the barrier to be permeated can be used in the formulation. Such penetrants are generally known in the art.
[00131] Use of pharmaceutically acceptable carriers to formulate the compounds herein disclosed for the practice of the invention into dosages suitable for systemic or other administration is within the scope of the invention. With proper choice of carrier and suitable manufacturing practice, the compositions of the present invention, in particular those formulated as solutions, may be administered parenterally, such as by intravenous injection, or other routes. Appropriate compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, elixirs, solutions, suspensions and the like, e.g. for oral ingestion by a patient to be treated. For other routes, formulations can be prepared for creams, ointments, lotions, and the like.
[00132] Agents intended to be administered intracellular^ may be administered using techniques well known to those of ordinary skill in the art. For example, such agents may be encapsulated into liposomes, other membrane translocation facilitating moieties, or other targeting moieties; then administered as described above. Liposomes can include spherical lipid bilayers with aqueous interiors. All molecules present in an aqueous solution at the time of liposome formation can be incorporated into the aqueous interior. The liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm. Additionally, due to hydrophobicity attributes, small organic molecules may be directly administered intracellular^.
[00133] Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the disclosure provided herein and other information in the art.
[00134] In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions, including those formulated for delayed release or only to be released when the pharmaceutical reaches the small or large intestine.
[00135] The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, suspending, granulating, dragee-making, levitating, emulsifying, encapsulating, entrapping, lyophilizing, and other processes. [00136] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
[00137] Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[00138] Dragee cores are optionally provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[00139] Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added.
EXAMPLE 5. Direct Induction of Apoptosis in Cancer Cells with a Small Molecule Activator of Procaspase-3.
[00140] ABSTRACT: Mutation or aberrant expression of proteins in the apoptotic cascade is a hallmark of cancer. These changes prevent proapoptotic signals from being transmitted to the executioner caspases, thus preventing apoptotic cell death and allowing cellular proliferation. Caspase-3 and caspase-7 are the key executioner caspases, existing as inactive zymogens that are activated by upstream signals. Importantly, levels of procaspase-3 are significantly higher in certain cancerous cells relative to non-cancerous controls. Here we report the identification of a small molecule (PAC-1) that directly activates procaspase-3 to active caspase-3 in vitro with an EC50 of 220 nanomolar, and induces apoptosis in a variety of cancer cell lines. In contrast to many known anti-cancer drugs, cells treated with PAC-1 show an immediate activation of procaspase-3, and the efficacy of PAC-1 is shown to be proportional to the amount of procaspase-3 contained in a cell. Derivatives of PAC-1 that do not activate procaspase-3 in vitro also have no proapoptotic activity. Cancerous cells isolated from primary colon tumors are considerably more sensitive to apoptotic induction by PAC-1 than the cells from adjacent non-cancerous tissue from the same patient; these cancerous cells contain on average about 7-fold more procaspase-3 than the cells from the adjacent non-cancerous primary tissue. In addition, the sensitivity to PAC-1 of the primary cells from the colon cancer tumors strongly correlates with the level of the procaspase-3 target. Finally, PAC-1 as a single entity was shown as active to retard the growth of tumors in three different mouse models, including two models where PAC-1 was administered orally. Thus PAC-1 directly activates procaspase-3 to caspase-3 in vivo, thereby allowing this compound to induce apoptosis even in cells that have defective apoptotic machinery. PAC-1 is the first small molecule known to directly activate procaspase-3; the direct activation of executioner caspases is an anti-cancer strategy that may prove beneficial in the many cancers in which procaspase-3 levels are elevated.
[00141] INTRODUCTION. A hallmark of cancer is its resistance to natural apoptotic signals. Depending on the cancer type, this resistance is typically due to either up- or down-regulation of key proteins in the apoptotic cascade, or to mutations in genes encoding these proteins. Such changes occur in both the intrinsic apoptotic pathway, which funnels through the mitochondria and caspase-9, and the extrinsic apoptotic pathway, which involves the action of death receptors and caspase-8. For example, alterations in proper levels of p53, Bim, Bax, Apaf-1 , FLIP and many others have been observed in cancers and lead to a defective apoptotic cascade, one in which the upstream pro-apoptotic signal is not properly transmitted to activate the executioner caspases, caspase-3 and caspase-7. As most apoptotic pathways ultimately involve the activation of procaspase-3, these genetic abnormalities are effectively "breaks" in the apoptotic circuitry, and as a result such cells proliferate uncontrolled.
[00142] Given the central role of apoptosis in cancer, efforts have been made to develop therapeutics that target specific proteins in the apoptotic cascade. For instance, peptidic or small molecule binders to p53, proteins in the BcI family, or to the IAPs have pro-apoptotic activity, as do compounds that promote the oligomerization of Apaf-1. However, because many of these compounds target early or intermediate positions on the apoptotic cascade, cancers with mutations in downstream proteins will likely be resistant to their effects. For therapeutic purposes it would be ideal to identify a small molecule that directly activates a proapoptotic protein far downstream in the apoptotic cascade. In addition, such a therapeutic strategy would have a higher likelihood of success if levels of that proapoptotic protein were elevated in cancer cells.
[00143] The conversion of procaspase-3 to caspase-3 results in the generation of the active "executioner" caspase that subsequently catalyzes the hydrolysis of a multitude of protein substrates. Active caspase-3 is a homodimer of heterodimers and is produced by proteolysis of procaspase-3. In vivo, this proteolytic activation typically occurs through the action of caspase-8 or caspase-9. To ensure that this zymogen is not prematurely activated, procaspase-3 has a tri-aspartic acid "safety catch" that blocks access to the IETD site of proteolysis. This safety catch enables procaspase-3 to resist autocatalytic activation and proteolysis by caspase-9. The position of the safety catch is sensitive to pH; thus, upon cellular acidification (as occurs during apoptosis) the safety catch is thought to allow access to the site of proteolysis, and active caspase-3 can be produced either by the action of caspase-9 or through an autoactivation mechanism.
[00144] Cells from certain types of cancerous tissue have elevated levels of procaspase-3. A study of primary isolates from 20 colon cancer patients revealed that on average procaspase-3 was elevated six-fold in such isolates relative to adjacent non-cancerous tissue. In addition, procaspase-3 levels are elevated in certain neuroblastomas, lymphomas, and liver cancers. In fact, a systematic evaluation of procaspase-3 levels in the 60 cell-line panel used by the NCI revealed that particular lung, melanoma, renal, and breast cancers show greatly enhanced levels of procaspase-3. Given the central importance of active caspase-3 to successful apoptosis, the high levels of procaspase-3 in certain cancerous cell types, and the intriguing safety catch-mediated suppression of its autoactivation, we reasoned that small molecules that directly activate procaspase-3 could be identified and that such molecules could have great potential in targeted cancer therapy. In this manuscript we report the in vitro identification of a small molecule activator of procaspase-3, PAC-1. PAC-1 is powerfully proapoptotic in cancer cell lines in a manner proportional to procaspase-3 levels, its proapoptotic effect is due to its direct and immediate activation of procaspase-3, and it is effective against primary colon cancer isolates and in three different mouse models of cancer.
[00145] Approximately 20,500 structurally diverse small molecules were screened for the ability to activate procaspase-3 in vitro. Procaspase-3 was expressed and purified in E. coli according to standard procedures. Procaspase-3 was added to the wells of a 384-well plate, and the compounds were added to a final concentration of about 40 μM (the final concentration of procaspase-3 was 50 ng/mL). Each plate was then incubated for two hours at 370C, after which the caspase-3 peptidic substrate Ac-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNa) was added to a concentration of 200 μM. The formation of the p-nitroaniline chromophore was followed at 405 nm over the course of two hours. Of the -20,500 compounds evaluated, four induced a significant increase over background in the hydrolysis of the peptidic caspase-3 substrate. Of those four, one showed a strong dose dependent effect on in vitro procaspase-3 activation. As shown in Figure 1A, this first procaspase-activating compound (PAC-1) gives half-maximal activation of procaspase-3 at a concentration of 0.22 μM. This compound is not simply increasing the activity of caspase-3 itself, as it has no effect on the catalytic activity of the fully processed caspase-3 enzyme (Figure 1A).
[00146] Procaspase-3 consists of a N-terminal pro domain (residues 1-28), followed by a large subunit (17 kDa) and a small subunit (12 kDa) that are separated by an intersubunit linker.22 In vivo, two procaspase-3 monomers assemble to form a homodimer that can be activated by cleavage at D175 in the intersubunit linker. The precise role of the pro domain is unclear, and it has been shown that cleavage in the intersubunit region alone is sufficient for full catalytic activity. Although procaspase-3 has enough catalytic activity to drive its own proteolytic maturation, it is highly resistant to this autoactivation due to the presence of the three amino acid safety catch. However, when the safety catch is mutated significant autoactivation of procaspase-3 is observed. To directly assess the ability of PAC-1 to catalyze the maturation of procaspase-3 to the active caspase-3, the procaspase-3 protein was incubated with 100 μM of PAC-1 for time points ranging from one to five hours. As shown by the Western blot in Figure 1B, PAC-1 induces the cleavage of procaspase-3 in a time-dependant fashion, with >50% processing observed after 4 hours. In contrast, procaspase-3 incubated in buffer shows virtually no autoactivation over that same time span. PAC-1 was also effective in this assay at a concentration of 5 μM.
[00147] Alanine substitutions were then made in the key aspartic acid triad in the safety catch region of procaspase-3, residues Asp179, Asp180 and Asp181. Mutations at these positions all dramatically decreased the ability of PAC-1 to activate procaspase-3, with certain mutations more detrimental to activation of procaspase-3 by PAC-1 (Figure 2A). Like caspase-3, caspase-7 also exists as an inactive zymogen that is activated by proteolysis. Caspase-3 and caspase-7 are both executioner caspases and have considerable structural homology. Procaspase-7 is also predicted to have a similar safety catch region, although it has only two aspartic acids in the key triad (Asp-Thr-Asp), instead of three. As indicated by the data in Figure 2B, PAC-1 can also activate procaspase-7, although in a less efficient manner than its activation of procaspase-3 (EC50 of 4.5 μM versus 0.22 μM for procaspase-3 activation). The potency of procaspase-7 activation by PAC-1 is similar to its effect on the Asp-Ala-Asp mutant of procaspase-3 (ECsO= 2.77 μM). As expected, the effect of PAC-1 is abolished at low pH values where procaspase-3 undergoes rapid autoactivation (Figure 2C).
[00148] PAC-1 was found to induce apoptosis in a variety of cancer cell lines. In HL-60 cells addition of PAC-1 leads to considerable phosphatidylserine exposure on the cell membrane accompanied by significant chromatin condensation (Figures 3A, 3B). In addition, the compound induces cleavage of the caspase substrate PARP-1 (as assessed by an in vivo PARP activity assay) and causes mitochondrial membrane depolarization (see below). Significant cellular blebbing of PAC-1 treated cells was also observed by microscopy. Furthermore, the toxicity of PAC-1 could be abolished in the presence of the caspase inhibitor z-VAD-fmk.
[00149] If PAC-1 is indeed inducing apoptosis via direct activation of procaspase-3, then the time course of apoptotic events should be altered relative to that observed with standard proapoptotic agents. Etoposide is well known to induce apoptosis through the intrinsic pathway; thus, mitochondrial membrane depolarization is followed by procaspase-3 activation in etoposide-treated cells. Indeed, in HL-60 cells treated with 10 μM etoposide, mitochondrial membrane depolarization is observed, followed by detection of caspase-3-like activity (Figure 4A). In contrast, treatment of cells with PAC-1 gives a markedly different result. With this compound, the first observed biochemical hallmark of apoptosis is caspase-3-like enzymatic activity, with activity noted within minutes of PAC-1 addition and 50% activation taking place in just over 2 hours and well before any significant mitochondrial membrane depolarization (Figure 4B). In addition, PARP-1 activity is rapidly reduced in cells treated with PAC-1, whereas this reduction is observed at later time points in etoposide treated cells (Figure 4C); control experiments show that PAC-1 does not directly inhibit enzymatic activity of PARP-1. In the typical sequence of apoptotic events the mitochondrial membrane depolarizes, caspases are activated, and caspase substrates (such as PARP-1) are cleaved. The observation that cells treated with PAC-1 show a rapid activation of caspase-3/-7 (before mitochondrial membrane depolarization) and a rapid cleavage of a caspase substrate (PARP-1) is indicative of PAC-1 exerting its cellular toxicity through the direct activation of procaspase-3. [00150] To further define the potency of PAC-1, the ability of this compound to induce cell death in cancer cell lines with varying levels of procaspase-3 was assessed. A determination was first made of the levels of procaspase-3 present in multiple cancer cell lines (leukemia, lymphoma, melanoma, neuroblastoma, breast cancer, lung cancer, adrenal cancer and renal cancer). The IC50 values for cell death induction were obtained for PAC-1 versus these cell lines. The combined data shows a strong correlation between cellular concentration of procaspase-3 and sensitivity to PAC-1 (Figure 4D, Figure 4E). PAC-1 is most potent versus the lung cancer cell line NCI-H226, with an IC50 of 0.35 μM. We found this cell line to have a concentration of procaspase-3 that is greater than five times that of baseline levels. Importantly, there is one cancer cell line (MCF-7, breast cancer cells) that is known to have no expression of procaspase-3. PAC-1 has virtually no effect on MCF-7 cells, inducing death with an ICso>75 μM.
[00151] In contrast, etoposide showed no such correlation between potency in cell culture and cellular levels of procaspase-3. For instance, etoposide was ineffective (ICδo>5O μM) in inducing death in three of the melanoma cell lines (UACC-62, CRL- 1872, and B16-F10), the breast cancer cell line (Hs 578t), and the lung cancer cell line (NCI-H226); these cell lines have procaspase-3 levels of 1.0, 2.4, 1.9, 3.7, and 5.3, respectively. Etoposide was effective (IC50<1 μM) versus HL-60, U-937, SK-N- SH and PC-12, which have procaspase-3 levels of 4.3, 4.0, 4.7, and 4.4, respectively. Thus, overall there is no correlation between procaspase-3 levels and IC50 for etoposide.
[00152] Several derivatives of PAC-1 were synthesized and evaluated for both their procaspase-3 activating properties and their effects on cancer cells in cell culture (Table 3). The PAC-1 derivative that lacks the ally! group (de-allyl PAC-1) is able to induce procaspase-3 activation and cell death at levels similar to PAC-1. However, all other derivatives showed no activity in either assay. Thus, while it appears the allyl group is dispensable for biological activity, the phenolic hydroxyl and aromatic rings are all critical for PAC-1 activity. This data is also consistent with the proposed mechanism of action of PAC-1; compounds that do not activate procaspase-3 in vitro have no proapoptotic effect on cancer cells in culture. [00153] To test this direct, small molecule-mediated procaspase-3 activation strategy in clinical isolates of cancer, we obtained freshly resected colon tumors (together with adjacent non-cancerous tissue) from 18 patients from Carle Foundation Hospital (Urbana, IL). The cancerous and non-cancerous tissue was separated, and the cells derived from these were evaluated for their levels of procaspase-3 and their sensitivity to PAC-1. As shown in Figure 5A, in all cases the cancerous cells had elevated levels (1.7- to 17.2-fold, with an average of 7.6-fold elevation) of procaspase-3 relative to the cells from the adjacent non-cancerous tissue from the same patient. Further, these cancerous cells were quite susceptible to death induction by PAC-1. PAC-1 induced cell death in the primary cancerous cells with IC50 values from 0.007-1.41 μM, while PAC-1 induced cell death in the adjacent non-cancerous tissue with IC50 values from 5.02-9.98 μM (Figure 5B and Table 4). The cancerous tissue that had elevated levels of procaspase-3 was extremely sensitive to PAC-1. For example, PAC-1 induced death in the cancer cells from patient 17 with an IC50 of 7 nM, and these cells were over 700-fold more sensitive to PAC-1 than cells from the adjacent normal tissue. See also Figure 6A showing relative procaspase-3 concentrations in normal and cancerous samples from Patients 1 , 2, and 3 over a period of time of about 54 days; Figure 6B illustrates that cells in cancerous tissue can be greater than about 80-fold more sensitive to PAC-1 in comparison with normal tissue.
[00154] In addition to cells from the non-cancerous tissue of the 18 patients, PAC-1 was also evaluated against four other non-cancerous cell types: white blood cells isolated from the bone marrow of a healthy donor, Hs888Lu (lung fibroblast cells), MCF-10A (breast fibroblast cells), and Hs578Bst (breast epithelial cells). Notably, the non-cancerous cell types are among those with the lowest amount of procaspase-3, and PAC-1 is comparatively less able to induce death in these cells, with IC50 values of 3.2 - 8.5 μM (Figure 5B, green diamonds). As is apparent from Figure 5B, PAC-1 induces death in a wide variety of cell types (non-cancerous cell lines, non-cancerous primary cells, cancerous cell lines, primary cancerous cells) in a manner directly related to the level of procaspase-3. The elevation of procaspase- 3 in cancerous cells allows PAC-1 to selectively induce death in these cell types. [00155] PAC-1 was evaluated in a mouse xenograft model using a slow release mode of drug delivery. In this model, subcutaneous tumors were formed in ovariectomized female athymic BALB/c (nude) mice using the ACHN (renal cancer) cell line. Once the tumors were measured to be greater than about 30 mm2, drug was administered via the implantation of a pellet of PAC-1 and cholesterol, providing for slow and steady levels of compound release. Three groups of mice were used, with pellets containing 0 mg, 1 mg, and 5 mg of PAC-1, six mice per group, with four tumors per mouse. Tumor sizes were monitored for about 8 weeks. As shown in Figure 5C, tumor growth is significantly retarded in the mice that were implanted with the pellet containing 5 mg of PAC-1. Food intake evaluation in the last week of the experiment showed no difference in food consumption between the three groups of mice. After the mice were sacrificed, plasma samples were taken from each mouse, and the PAC-1 content of each was analyzed. For mice that received a 5 mg pellet of PAC-1 , this analysis revealed PAC-1 to be present at a concentration of 5 nM in the plasma after the 54 day experiment.
[00156] PAC-1 was evaluated in a second mouse xenograft model, this one using oral administration as the drug delivery mode. In this model, subcutaneous xenograft tumors were formed in male athymic BAlBlc-nu/nu mice (5 weeks old, SLC, Hamamaysu, Japan) using the NCI-H226 (lung cancer) cell line, eight mice per group, three tumors per mouse. After formation of the tumors in the mice, the mice were treated with PAC-1 via oral gavage once a day for 21 days at a concentration of 0, 50, or 100 mg/kg and sacrificed 1 week later. As clearly indicated by the graph in Figure 5D, oral administration of PAC-1 significantly retards tumor growth in a dose-dependent manner.
[00157] Finally, PAC-1 was evaluated in a mouse model where the NCI-H226 cells were injected into male athymic BALB/c-nu/nu mice via tail vein injection. The total experiment lasted 28 days; the mice were treated once a day with PAC-1 (100 mg/kg) via oral gavage on days 1-4 and 7-11. On other days the mice did not receive PAC-1. A second group of mice received only vehicle. After 28 days the mice were sacrificed, and their lungs were examined. As shown in Figure 5E, there is a clear difference between the lung of the control mouse (with obvious gray tumor mass) and the lung of the PAC-1 treated mouse. Results are also shown in a panel from an animal treated with gefitinib (Iressa™; AstraZeneca).
[00158] Cancerous cells typically have a reduced sensitivity to proapoptotic signals due to the mutation or aberrant expression of an assortment of proteins in the apoptotic cascade. As such, many types of cancer are notoriously resistant to not only the endogenous signals for apoptotic cell death, but also to chemotherapeutic agents that act through similar mechanisms. The paradoxical elevation of procaspase-3 levels in certain cancers provides an opportunity to use this existing intracellular pool of protein to directly induce apoptosis, thus bypassing the often non-functional upstream portion of the cascade. PAC-1 induces the autoactivation of procaspase-3 in vitro. In cell culture, PAC-1 treatment induces rapid caspase-3- like activity. It is likely that the caspase-3 mediated cleavage of anti-apoptotic proteins (Bcl-2, BcI-XL, etc.) then induces depolarization of the mitochondrial membrane and amplifies apoptosis. Further, the potency of PAC-1 toward a variety of cancerous and non-cancerous cell types is proportional to the concentration of procaspase-3 in the cell. As the primary cancerous cells isolated from resected colon tumors have elevated levels of procaspase-3, these cells are considerably more sensitive to PAC-1 than cells from adjacent non-cancerous tissue. It is worth noting that several of the cell lines against which PAC-1 is effective have faulty apoptotic pathways that make them resistant to apoptosis; for instance, Apaf-1 expression is dramatically decreased in SK-MEL-5 cells, and Bcl-2 is overexpressed in the NCI-H226 lung cancer cell line. Finally, PAC-1 is effective in three different mouse models of cancer, including two where PAC-1 is administered orally.
[00159] Data presented herein support the notion that procaspase-3 activating compounds can be exceedingly effective against a variety of common cancers in which procaspase-3 levels are aberrantly high. Assessment of procaspase-3 levels in cancer biopsies is simple and rapid; as such, the potential effectiveness of a compound such as PAC-1 can be assessed a priori with a high degree of accuracy. Such personalized medicine strategies can be preferential to therapies that rely on general cytotoxins and can be valuable in anti-cancer therapy.
[00160] Professor Guy Salvesen (Burnham Institute) provided the procaspase-3 and procaspase-7 expression vectors. FIGURE LEGENDS
[00161] Figures 1 and 2. The structure of PAC-1 is shown elsewhere in the specification. Fig. 1A) In vitro activation of procaspase-3 and active caspase-3 by PAC-1. PAC-1 activates procaspase-3 with an EC50= 0.22 μM. Fig. 1B) Cleavage of procaspase-3 to active caspase-3 as induced by PAC-1. Procaspase-3 was recombinantly expressed in E. coli with an N-terminal His-6 tag and purified, lmmunoblotting was performed with an anti-His-6 antibody. In the absence of PAC-1 no maturation of procaspase-3 is observed. In the presence of 100 μM PAC-1, cleavage to generate the p19 fragment is observed within 1 h, and >50% cleavage is observed after 4 h. PAC-1 is also effective at 5 μM in this assay. Fig, 2A) Activation of mutants in the "safety catch" region of procaspase-3 by PAC-1. PAC-1 has an EC50 for activation of 0.22 μM on wild type procaspase-3 (DDD), and corresponding EC50 values of 2.77 μM (DAD), 113 μM (DDA), and 131 μM (ADD) for the mutants. Fig. 2B) PAC-1 activates procaspase-7 with an EC50 of 4.5 μM. Fig. 2C) Dependence of PAC-1 activation of procaspase-3 on pH. At low pH the safety catch is off and procaspase-3 is essentially maximally activated. Error bars represent standard deviations from the mean.
[00162] Figures 3 and 4. PAC-1 induces apoptosis in HL-60 cells. Fig. 3A) Phosphatidylserine exposure (as measured by Annexin-V staining) after a 20 h treatment with 100 μM PAC-1. PAC-1 is also effective at 5 μM in this assay (see Supporting Figure 2). Fig. 3B) Chromatin condensation as visualized by Hoescht staining after a 20 h treatment with 100 μM PAC-1. Fig. 4A) Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 10 μM etoposide. Fig. 4B) Mitochondrial membrane depolarization (MMP) and caspase-3 like activity in HL-60 cells treated with 100 μM PAC-1. Fig. 4C) PAC-1 treatment (100 μM) induces a rapid decrease in cellular PARP activity in HL-60 cells, consistent with an immediate activation of cellular caspase-3/-7. In contrast, etoposide (10 μM) treated cells show a decrease in PARP activity at much later time points. Fig. 4D and Fig. 4E) PAC-1 induces cell death in a procaspase-3 dependant manner. For a number of diverse cancerous cell lines, the procaspase-3 levels were determined (by flow cytometry with an antibody to procaspase-3) and the IC50 of PAC-1 was measured (through a 72 h treatment with a range of PAC-1 concentrations and quantitation using the MTS assay). PAC-1 is quite potent (IC50=O.35 μM) in the NCI-H226 lung cancer cell line known to have high levels of procaspase-3. Error bars represent standard deviations from the mean.
[00163] Table 3. PAC-1 and de-allyl PAC-1 activate procaspase-3 in vitro and induce death in cancer cells in cell culture, but other structural analogues have no procaspase-3 activating effect in vitro and give no induction of death in cell culture.
[00164] Figure 5. Fig. 5A) Procaspase-3 levels are elevated in cells derived from freshly resected colon cancer tissue. Freshly resected primary colon tumors (together with adjacent non-cancerous tissue) were obtained from 18 different patients, the cancerous and non-cancerous tissue were separated, and the procaspase-3 levels were measured for each using an antibody to procaspase-3 and flow cytometry. On average, cells from the cancerous tissue have a 7.6-fold elevation in procaspase-3 as compared to the cells derived from the adjacent noncancerous tissue from the same patient. Fig. 5B) PAC-1 induces cell death in a manner proportional to the cellular level of procaspase-3. The red circles represent the primary cancerous cells from the 18 colon tumors. The black triangles represent the same cancer cell lines depicted in Figure 4D. The green diamonds are four noncancerous cell types: Hs888Lu (lung fibroblast cells), MCF-10A (breast fibroblast cells), Hs578Bst (breast epithelial cells), and white blood cells isolated from the bone marrow of a healthy donor. The blue squares are the primary non-cancerous cells isolated from the tumor margins of the 18 patients. Table 4) Cells derived from primary colon cancer tissue are considerably more sensitive to death induction by PAC-1 than are cells derived from adjacent non-cancerous tissue from the same patient. Fig. 5C) PAC-1 reduces the growth of tumors in a xenograft model of cancer. Tumors were formed with the ACHN (renal cancer) cell line by subcutaneous injection, with six mice in each group, and four tumors per mouse. Once the tumors grew to about 30 mm2, PAC-1 was implanted as a cholesterol pellet. Error bars represent standard error from the mean. Fig. 5D) Oral administration of PAC-1 significantly retards tumor growth in a mouse xenograft model. Tumors were formed using the NCI-H226 (lung cancer) cell line by subcutaneous injection, eight mice in each group, and three tumors per mouse. PAC-1 or vehicle was administered once a day by oral gavage on days 1-21. Error bars represent standard error from the mean. Fig. 5E) Oral administration of PAC-1 significantly retards tumor growth in an i.v. injection model. Mice were injected i.v. with the NCI-H226 (lung cancer) cell line. The mice were treated with PAC-1 (100 mg/kg) via oral gavage following the protocol as described in the text. Images show the lungs of the mice that did not receive PAC-1 and have a large amount of gray tumor mass on the lung. In contrast, the mice that did receive PAC-1 have almost no visible gray matter.
Table 3. Selected compounds indicating activity levels.
EC50 (μM) for IC50 (μM) for Compound procaspase-3 death induction activation in HL-60 cells
Figure imgf000051_0001
Table 4. Concentration levels of PAC-1 activity in patients.
Figure imgf000052_0001
EXAMPLE 6. Testing of PAC-1 in mouse model of lung cancer.
[00165] A xenograft model was employed using NCI-H226 (lung cancer) cells. PAC-1 was given intraperitoneal^ (i.p.) at 10 mg/kg. A comparison of efficacy was performed with gefitinib (Iressa™; AstraZeneca, Wilmington, Delaware) at 40 mg/kg using 5 mice per group. Results are shown in Figure 7, indicating that PAC-1 was associated with reducing growth in tumor volume.
EXAMPLE 7. Combinatorial derivatives, synthesis, and therapeutic use.
[00166] A number of compounds are prepared as derivatives of the PAC-1 structure. A hydrazide group is reacted with an aldehyde group to yield a combinatorial library of derivative compounds.
[00167] Any one of hydrazide precursor groups (AX) designated L1-L20 are used to generate hydrazides which are reacted with any one of aldehyde groups (BX) designated 1-28, thus yielding 560 PAC-1 derivative compounds. A derivative compound is synthesized using methods as described herein and according to knowledge available in the art. See the scheme and component structures below in addition to Figure 8A and 8B.
Figure imgf000053_0001
AX
Figure imgf000053_0002
L15 L20
L5 L10 BX
Figure imgf000054_0001
[00168] In an embodiment, such derivative compounds are further modified, e.g. to alter a property such as activity, solubility, toxicity, stability, and/or other properties in connection with pharmaceutical applications.
[00169] The derivative compounds are used as anti-cancer agents. Compounds are validated as capable of having antineoplastic activity, apoptosis regulation, and/or procaspase-3 activation. For example, primary isolates of freshly removed colon cancer are used fo assess procaspase-3 levels and sensitivity of cells to test . compound levels, where a test compound is PAC-1 or a derivative compound. Compounds are classified regarding a propensity to induce cell death in cancerous cells versus normal cells.
[00170] In further assessing a derivative compound, in vitro and in vivo testing is performed. Stability in connection with exposure to liver microsomes is evaluated.
EXAMPLE 8. Activity of certain derivatives relative to PAC-1.
[00171] PAC-1 and certain derivatives were tested in the HL-60 cell line and IC50 values were determined. The results are indicated in Table 5 (where L- and R- designations refer to structures shown in the AX and BX series above, respectively). Several of the PAC-1 derivatives exhibited an activity level that was generally about one order of magnitude greater than that of the PAC-1 compound.
Figure imgf000056_0001
STATEMENTS REGARDING INCORPORATION BY REFERENCE AND VARIATIONS
[00172] All references throughout this application, for example patent documents including issued or granted patents or equivalents; patent application publications; unpublished patent applications; and non-patent literature documents or other source material; are hereby incorporated by reference herein in their entireties, as though individually incorporated by reference, to the extent each reference is at least partially not inconsistent with the disclosure in this application (for example, a reference that is partially inconsistent is incorporated by reference except for the partially inconsistent portion of the reference).
[00173] Any appendix or appendices hereto are incorporated by reference as part of the specification and/or drawings.
[00174] Where the terms "comprise", "comprises", "comprised", or "comprising" are used herein, they are to be interpreted as specifying the presence of the stated features, integers, steps, or components referred to, but not to preclude the presence or addition of one or more other feature, integer, step, component, or group thereof. Separate embodiments of the invention are also intended to be encompassed wherein the terms "comprising" or "comprise(s)" or "comprised" are optionally replaced with the terms, analogous in grammar, e.g.;
"consisting/consist(s)" or "consisting essentially of/consist(s) essentially of to thereby describe further embodiments that are not necessarily coextensive. For clarification, as used herein "comprising" is synonymous with "having," "including," "containing," or "characterized by," and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. As used herein, "consisting of excludes any element, step, component, or ingredient not specified in the claim element. As used herein, "consisting essentially of does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim (e.g., not affecting an active ingredient). In each instance herein any of the terms "comprising", "consisting essentially of and "consisting of may be replaced with either of the other two terms. The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein.
[00175] The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention. It will be appreciated by one of ordinary skill in the art that compositions, methods, devices, device elements, materials, optional features, procedures and techniques other than those specifically described herein can be applied to the practice of the invention as broadly disclosed herein without resort to undue experimentation. All art-known functional equivalents of compositions, methods, devices, device elements, materials, procedures and techniques described herein; and portions thereof; are intended to be encompassed by this invention. Whenever a range is disclosed, all subranges and individual values are intended to be encompassed. This invention is not to be limited by the embodiments disclosed, including any shown in the drawings or exemplified in the specification, which are given by way of example or illustration and not of limitation. The scope of the invention shall be limited only by the claims.
REFERENCES
[00176] These applications are particularly incorporated by reference in entirety: U.S. Provisional Patent Application No. 60/516556 by Hergenrother et al., filed October 30, 2003; U.S. Provisional Patent Application No. 60/603246 by Hergenrother et al., filed August 20, 2004; U.S. 10/976,186 by Hergenrother et al., filed October 27, 2004.
[00177] US 6,762,045 Membrane derived caspase-3, compositions comprising the same and methods of use therefore; US 6,534,267 Polynucleotides encoding activators of caspases; US 6,403,765 Truncated Apaf-1 and methods of use thereof; US 6,303,329; 6,878,743 by Choong, et al. issued April 12, 2005; US 20040077542 by Wang, Xiaodong; et al., published April 22, 2004; US 20040180828 by Shi, Yigong, published September 16, 2004. Slee EA et al., Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone (Z-
VAD. FMK) inhibits apoptosis by blocking the processing of CPP32, Biochem J. 1996
Apr 1 ;315 ( Pt 1):21-4.
I . Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57-70 (2000).
2. Okada, H. & Mak, T. W. Pathways of apoptotic and non-apoptotic death in tumour cells. Nature Rev. Cancer 4, 592-603 (2004).
3. Roy, S. et al. Maintenance of caspase-3 proenzyme dormancy by an intrinsic "safety catch" regulatory tripeptide. Proc. Natl. Acad. Sci. 98, 6132-6137 (2001).
4. Svingen, P. A. et al. Components of the cell death machine and drug sensitivity of the National Cancer Institute Cell Line Panel. Clin. Cancer Res. 10, 6807-6820
(2004).
5. Lowe, S. W., Cepero, E. & Evan, G. Intrinsic tumor suppression. Nature 432, 307- 315 (2004).
6. Vogelstein, B. & Kinzler, K. W. Achilles' heel of cancer. Nature 412, 865-866 (2001).
7. Traven, A., Huang, D. C. & Lithgow, T. Protein hijacking: key proteins held captive against their will. Cancer Cell 5, 107-108 (2004).
8. Soengas, M. S. et al. Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 409, 207-211 (2001).
9. Wajant, H. Targeting the FLICE inhibitory protein (FLIP) in cancer therapy. MoI. Interv. 3, 124-127 (2003).
10. Denicourt, C. & Dowdy, S. F. Targeting apoptotic pathways in cancer cells. Science 305, 1411-1413 (2004).
I I . Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844-848 (2004).
12. Degterev, A. et al. Identification of small-molecule inhibitors of interaction between the BH3 domain and BcI-XL. Nature Cell Biol. 3, 173-182 (2001). 13. Becattini, B. et al. Rational design and real time, in-cell detection of the proapoptotic activity of a novel compound targeting BcI-XL. Chem. Biol. 11 , 389-395 (2004).
14. Wang, J.-L. et al. Structure-based discovery of an organic compound that binds Bcl-2 protein and induces apoptosis of tumor cells. Proc. Natl. Acad. Sci. 97, 7124-
7129 (2000).
15. Li, L. et al. A small molecule Smac mimic potentiates TRAIL- and TNFa- mediated cell death. Science 305, 1471-1474 (2004).
16. Nguyen, J. T. & Wells, J. A. Direct activation of the apoptosis machinery as a mechanism to target cancer cells. Proc. Natl. Acad. Sci. U.S.A. 100, 7533-7538
(2003).
17. Jiang, X. et al. Distincitive roles of PHAP proteins and prothymosin-α in a death regulatory pathway. Science 299, 223-226 (2003).
18. Boatright, K. M. & Salvesen, G. S. Mechanisms of caspase activation. Curr. Opin. Cell. Biol. 15, 725-731 (2003).
19. Nakagawara, A. et al. High levels of expression and nuclear localization of interleukin-1 β converting enzyme (ICE) and CPP32 in favorable human neuroblastomas. Cancer Res. 57, 4578-4584 (1997).
20. Izban, K. F. et al. Characterization of the interleukin-1 β-converting enzyme/Ced- 3-family protease, caspase-3/CPP32, in Hodgkin's disease. Am. J. Pathol. 154,
1439-1447 (1999).
21. Persad, R. et al. Overexpression of caspase-3 in hepatocellular carcinomas. Modern Patholo. 17, 861-867 (2004).
22. Pop, C, Feeney, B., Tripathy, A. & Clark, A. C. Mutations in the procaspase-3 dimer interface affect the activity of the zymogen. Biochemistry 42, 12311-12320
(2003).
23. Stennicke, H. R. et al. J. Biol. Chem. 273, 27084-27090 (1998). 24. Denault, J.-B. & Salvesen, G. S. Human caspase-7 activity and regulation by its N-terminal peptide. J. Biol. Chem. 278, 34042-24050 (2003).
25. Putt, K. S., Beilman, G. J. & Hergenrother, P. J. Direct quantitation of PoIy(ADP- ribose) polymerase (PARP) activity as a means to distinguish necrotic and apoptotic death in cell and tissue samples. ChemBioChem 6, 53-55 (2005).
26. Liang, Y., Nylander, K. D., Yan, C. & Schor, N. F. Role of caspase 3-dependent Bcl-2 cleavage in potentiation of apoptosis by Bcl-2. MoI. Pharmacol. 61 , 142-149 (2002).
27. Fujita, N., Nagahshi, A., Nagashima, K., Rokudai, S. & Tsuruo, T. Acceleration of apoptotic cell death after the cleavage of BcI-XL protein by caspase-3-like proteases.
Oncogene 17, 1295-1304 (1998).
28. Earnshaw, W. C, Martins, L. M. & Kaufmann, S. H. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu. Rev. Biochem. 68, 383-424 (1999).
29. Koty, P. P., Zhang, H. & Levitt, M. L. Antisense bcl-2 treatment increases programmed cell death in non-small cell lung cancer cell lines. Lung Cancer 23, 115- 127 (1999).
National Center for Biotechnology Information (NCBI) Database of the National Library of Medicine / National Institutes of Health (NIH) website: http://www.ncbi.nlm.nih.gov/ using the Gene database to search for CASP3 (caspase 3, apoptosis-related cysteine protease [Homo sapiens] GenelD: 836 Locus tag: HGNC:1504; MIM: 600636 updated 15-May-2005. Other Aliases: HGNC:1504, APOPAIN, CPP32, CPP32B, SCA-1 ; Other Designations: Human procaspase3 coding sequence; PARP cleavage protease; SREBP cleavage activity 1 ; Yama; caspase 3; cysteine protease CPP32).
[00178] Hergenrother PJ. Obtaining and screening compound collections: a user's guide and a call to chemists. Curr Opin Chem Biol. 2006 Silverman SK, Hergenrother PJ. Combinatorial chemistry and molecular diversity Tools for molecular diversification and their applications in chemical biology. Curr Opin Chem Biol. 2006.
Goode DR, Sharma AK, Hergenrother PJ. Using peptidic inhibitors to systematically probe the SV site of caspase-3 and caspase-7. Org Lett. 2005 Aug 4;7(16):3529-32. PMID: 16048334
Dothager RS, Putt KS, Allen BJ, Leslie BJ, Nesterenko V, Hergenrother PJ. Synthesis and identification of small molecules that potently induce apoptosis in melanoma cells through G1 cell cycle arrest. J Am Chem Soc. 2005 Jun 22;127(24):8686-96. PMID: 15954774
Putt KS, Hergenrother PJ. A nonradiometric, high-throughput assay for poly(ADP- ribose) glycohydrolase (PARG): application to inhibitor identification and evaluation. Anal Biochem. 2004 Oct 15;333(2):256-64. PMID: 15450800 '
Putt KS, Hergenrother PJ. An enzymatic assay for poly(ADP-ribose) polymerase-1 (PARP-1) via the chemical quantitation of NAD(+): application to the high-throughput screening of small molecules as potential inhibitors. Anal Biochem. 2004 Mar 1 ;326(1):78-86. PMID: 14769338
Nesterenko V, Putt KS, Hergenrother PJ. Identification from a combinatorial library of a small molecule that selectively induces apoptosis in cancer cells. J Am Chem Soc. 2003 Dec 3; 125(48): 14672-3. PMID: 14640619

Claims

CLAIMS:
The invention is further described and set forth by the following exemplary claims.
A method of selectively inducing apoptosis in a cancer cell, comprising: (a) administering to said cancer cell a compound capable of modifying a procaspase-3 molecule of said cancer cell; and (b) modifying said procaspase-3 molecule so as to induce apoptosis.
The method of claim 1 wherein said cancer cell is in a patient in need of treatment. 3. The method of claim 1 or 2 wherein said compound is of formula ZZ:
Figure imgf000063_0001
wherein n = 1 or 2; R, independently of other R, is hydrogen, halogen, allyl, or short alkyl; R2 = hydrogen, short alkyl, ester, or other moiety that is removable under physiological conditions; R3 = hydrogen, halogen, alkyl, haloalkyl, allyl, alkenyl, alkenol, alkanol, or haloalkenyl; R4 and R5 are N; or R4=N and
R5=C; or R4 and R5 = C; and A = oxygen or sulfur.
The method of any of claims 1-3 wherein said compound is selected from the group consisting of formula ZZ, PAC-1 , and Structure 5.
5. The method of any of claims 1-3 wherein said compound is PAC-1. 6. The method of any of claims 1-3, further comprising the step of assessing a procaspase-3 or caspase-3 parameter in a cancer cell; wherein said parameter is one or more of a semi-quantitative or quantitative amount, a functional amount, and an activity level of said procaspase-3 or caspase-3.
7. A compound of the structural formula ZZ
Figure imgf000063_0002
wherein n = 1 or 2; R, independently of other R, is hydrogen, halogen, allyl, or short alkyl; R2 = hydrogen, short alkyl, ester, or other moiety that is removable under physiological conditions; R3 = hydrogen, halogen, alkyl, haloalkyl, allyl, alkenyl, alkenol, alkanol, or haloalkenyl; R4 and R5 are N; or R4=N and R5=C; or R4 and R5 = C; and A = oxygen or sulfur.
8. The compound of claim 7, excluding a compound of a structure PAC-1 , wherein the structure of PAC-1 is:
Figure imgf000064_0001
9. A method of direct in vitro screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a purified procaspase-3; (c) exposing the test compound to the purified procaspase-3; (d) measuring a procaspase-3 activity following exposure to the test compound; (e) identifying a modifying compound by comparing a test activity upon the exposure to the test compound with an unmodified activity in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule.
10. The method of claim 9 further comprising comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure to a compound selected from the group consisting of structural formula ZZ, PAC-1 , and Structure 5.
11. A method of in cellular screening for a compound capable of modifying a procaspase-3 molecule, comprising: (a) providing a test compound; (b) providing a cell, wherein the cell putatively expresses procaspase-3; (c) exposing the cell to the test compound; (d) measuring a cell parameter following exposure to the test compound; wherein said parameter comprises one or more of cell viability, apoptotic indicator, and other parameters; (e) identifying a modifying compound by comparing a tested cell parameter upon the exposure to the test compound with an unmodified cell parameter in the absence of exposure to the test compound; thereby screening for a compound capable of modifying a procaspase-3 molecule.
12. The method of claim 11 further comprising comparing said modified activity or said unmodified activity with a reference activity; wherein said reference activity is due to exposure to a compound selected from the group consisting of ZZ, PAC-1 , and Structure 5.
13. A method of identifying or diagnosing a potential susceptibility to treatment for a cancer cell with a procaspase activator compound, comprising (a) assessing a procaspase parameter in said cancer cell; and (b) determining if said parameter allows an increased susceptibility to activation of a procaspase.
14. The method of claim 13 wherein said procaspase parameter is a procaspase- 3 level and said procaspase is procaspase-3.
15. The method of claim 13 wherein said procaspase parameter is a procaspase- 7 level and said procaspase is procaspase-7. 16. A method of treating a cancer cell, comprising (a) identifying a potential susceptibility to treatment of a cancer cell with a procaspase activator compound; and (b) exposing said cancer cell to an effective amount of the procaspase activator compound.
17. The method of claim 16 wherein the procaspase activator compound is selected from the group consisting of formula ZZ, PAC-1 , and Structure 5.
18. The method of claim 16 or 17 wherein said procaspase activator compound is capable of activating procaspase-3, procaspase-7, or both procaspase-3 and procaspase-7.
19. A method of synthesizing PAC-1 , comprising the steps of Scheme 1. 20. A method of synthesizing Compound 5 or compounds of formula ZZ, comprising the steps of Scheme 1 with appropriate modification. 21. A compound of Structure 5, wherein the structural formula is :
Figure imgf000065_0001
22. A compound having the formula ZZ2:
Figure imgf000066_0001
wherein R1 and R2 each independently is hydrogen, halogen, alkyl, allyl, haloalkyl, alkenyl, alkenol, alkanol, or haloalkenyl. 23. The compound of claim 22 wherein R1 and R2 each independently is hydrogen, halogen, allyl, or short alkyl. 24. A compound selected from the group consisting of a PAC-1 derivative combinatorial library comprising a hydrazide compound combined with an aldehyde compound. 25. The compound of claim 24 wherein the hydrazide compound is selected from the group consisting of hydrazides generated from AX compounds described herein and the aldehyde compound is selected from the group consisting of BX compounds described herein.
26. A method of synthesizing a PAC-1 derivative compound comprising providing a hydrazide compound, providing an aldehyde compound, and reacting the hydrazide compound with the aldehyde compound, thereby synthesizing a PAC-1 derivative compound.
27. The method of claim 26 wherein the hydrazide compound has the formula ZZ3:
Figure imgf000066_0002
28. The method of claim 26 or 27 wherein the aldehyde compound has the formula ZZ4:
Figure imgf000066_0003
29. The method of any of claims 26-28 wherein the hydrazide compound has the formula ZZ3 and the aldehyde compound has the formula ZZ4.
30. A compound selected from the group consisting of: L01 R06, L02R03, L02R06, L08R06, L09R03, L09R06, and L09R08.
31. A method of screening a candidate cancer patient for possible treatment with a procaspase activator by identifying an elevated level of a procaspase in the candidate, comprising obtaining a cell or tissue test sample from the candidate, assessing the procaspase level in the test sample, and determining whether the procaspase level is elevated in the test sample relative to a reference level, thereby screening a candidate cancer patient for possible treatment with a procaspase activator.
32. The method of claim 31 wherein the procaspase is selected from the group consisting of procaspase-2, -3, -6-, -7, -8, and -9.
33. The method of any of claims 31-32 wherein the procaspase is procaspase-3.
34. The method of any of claims 31-33 wherein said elevated level of the test sample is at least about 2-fold greater than the reference level.
35. The method of any of claims 31-34 wherein said elevated level of the test sample is at least about 4-fold greater than the reference level;
36. The method of claim any of claims 31-35 wherein the reference level is from a second test sample from the same patient. 37. The method of claim any of claims 31-36 wherein the reference level is from a normal cell or tissue sample.
38. The method of claim any of claims 31-37 wherein the reference level is from a cell line.
39. The method of claim any of claims 31-38 wherein the reference level is from a cancer cell line.
40. The method of claim any of claims 31-39 wherein the reference level is from a normal cell line.
41. The method of claim any of claims 31-40 wherein the reference level is an absolute threshold amount. 42. A method of inducing death in a cancer cell, comprising administering to said cancer cell a compound capable of activating a procaspase-3 molecule of said cancer cell.
43. The method of claim 42 wherein the compound has structural formula ZZ.
44. The method of claim 42 wherein the compound has structural formula ZZ2.
PCT/US2006/020910 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3 WO2006128173A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
ES06771588T ES2763156T3 (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including procaspase-3 activation
AU2006251978A AU2006251978A1 (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3
EP06771588.8A EP1893210B1 (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3
DK06771588.8T DK1893210T3 (en) 2005-05-26 2006-05-26 SELECTIVE APOPTOTIC INDUCTION IN CANCER CELLS, INCLUDING PROCASPASE-3 ACTIVATION
MX2007014843A MX2007014843A (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3.
CA002610366A CA2610366A1 (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3
JP2008513829A JP5513741B2 (en) 2005-05-26 2006-05-26 Selective apoptosis induction in cancer cells, including activation of procaspase-3
IL187603A IL187603A0 (en) 2005-05-26 2007-11-25 Piperazine derivatives and pharmaceutical compositions containing the same

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US68480705P 2005-05-26 2005-05-26
US60/684,807 2005-05-26
US74387806P 2006-03-28 2006-03-28
US60/743,878 2006-03-28
US11/420,425 2006-05-25
US11/420,425 US20070049602A1 (en) 2005-05-26 2006-05-25 Selective Apoptotic Induction in Cancer Cells Including Activation of Procaspase-3

Publications (2)

Publication Number Publication Date
WO2006128173A2 true WO2006128173A2 (en) 2006-11-30
WO2006128173A3 WO2006128173A3 (en) 2007-05-24

Family

ID=37452994

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/020910 WO2006128173A2 (en) 2005-05-26 2006-05-26 Selective apoptotic induction in cancer cells including activation of procaspase-3

Country Status (10)

Country Link
US (1) US20070049602A1 (en)
EP (3) EP2634581A1 (en)
JP (2) JP5513741B2 (en)
AU (1) AU2006251978A1 (en)
CA (1) CA2610366A1 (en)
DK (1) DK1893210T3 (en)
ES (1) ES2763156T3 (en)
IL (1) IL187603A0 (en)
MX (1) MX2007014843A (en)
WO (1) WO2006128173A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008134474A2 (en) * 2007-04-27 2008-11-06 The Board Of Trustees Of The University Of Illinois Compositions and methods including cell death inducers and procaspase activation
WO2010091382A1 (en) 2009-02-09 2010-08-12 The Board Of Trustees Of The University Of Illinois Design, synthesis and evaluation of procaspase activating compounds as personalized anti-cancer drugs
CN101805338A (en) * 2010-04-06 2010-08-18 沈阳药科大学 Oxadiazole-based piperazine derivative and application thereof
WO2010102513A1 (en) * 2009-03-11 2010-09-16 深圳市湘雅生物医药研究院 Homopiperazine acethydrazide derivatives, preparation method and uses thereof
US8916705B2 (en) 2011-10-14 2014-12-23 The Board of Trustees of The University of Illilnois Procaspase-activating compounds and compositions
CN105085421A (en) * 2015-07-24 2015-11-25 深圳市湘雅生物医药研究院 Orbit azine-fumarate, hydrates, crystal forms and preparation methods therefor
AU2013296187B2 (en) * 2012-08-03 2016-09-29 The Board Of Trustees Of The University Of Illinois Enzyme-activating compounds and compositions
WO2017012113A1 (en) * 2015-07-23 2017-01-26 王智民 Compound pac-1 or salt thereof, and pharmaceutical composition comprising same
WO2017015784A1 (en) * 2015-07-24 2017-02-02 深圳市湘雅生物医药研究院 Orbit azine-fumarate, hydrate, crystal form and preparation method therefor
US10350207B2 (en) 2015-06-05 2019-07-16 The Board Of Trustees Of The University Of Illinois PAC-1 combination therapy
US11510919B2 (en) 2017-11-17 2022-11-29 The Board Of Trustees Of The University Of Illinois Cancer therapy by degrading dual MEK signaling

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070049602A1 (en) * 2005-05-26 2007-03-01 The Board Of Trustees Of The University Of Illinois Selective Apoptotic Induction in Cancer Cells Including Activation of Procaspase-3
CN104487059B (en) * 2012-03-02 2017-06-23 伊利诺伊大学评议会 The strong effective active anticancer activated by dual compound
ES2774930T3 (en) * 2012-03-06 2020-07-23 Univ Illinois Activation of procaspase 3 by combination therapy
KR102075811B1 (en) 2012-03-06 2020-02-10 더 보드 오브 트러스티즈 오브 더 유니버시티 오브 일리노이 Procaspase combination therapy for glioblastoma
CN105218399B (en) 2014-05-30 2018-02-09 中国人民解放军军事医学科学院毒物药物研究所 A kind of substituted acethydrazide derivatives, preparation method and the usage

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303329B1 (en) 1998-07-27 2001-10-16 Pharmacia & Upjohn Company Method for autoactivation of procaspase 8
US6403765B1 (en) 1998-06-16 2002-06-11 Thomas Jefferson University Truncated Apaf-1 and methods of use thereof
US6534267B1 (en) 2000-01-06 2003-03-18 Xiaodong Wang Polynucleotides encoding activators of caspases
US20040077542A1 (en) 2000-08-23 2004-04-22 Board Of Regents, The University Of Texas System Apoptotic compounds
US6762045B2 (en) 2000-11-20 2004-07-13 Idun Pharmaceuticals, Inc. Membrane derived caspase-3, compositions comprising the same and methods of use therefor
US20040180828A1 (en) 2003-01-30 2004-09-16 Yigong Shi Caspase-9 : BIR domain of XIAP complexes and methods of use
US6878743B2 (en) 2001-09-18 2005-04-12 Sunesis Pharmaceuticals, Inc. Small molecule inhibitors of caspases

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998053091A1 (en) * 1997-05-22 1998-11-26 The Burnham Institute Screening assays for agents that alter inhibitor of apoptosis (iap) protein regulation of caspase activity
WO2002069995A2 (en) * 2001-02-16 2002-09-12 Medical College Of Georgia Research Institute, Inc. Use of trail and antiprogestins for treating cancer
KR100439425B1 (en) * 2001-03-22 2004-07-05 (주)바이오케어 Compositions Comprising Xanthorrhizol And The Use Thereof
CA2508690A1 (en) * 2002-12-10 2004-06-24 Ottawa Health Research Institute Modulation of stem cell differentiation by modulation of caspase-3 activity
US20070049602A1 (en) * 2005-05-26 2007-03-01 The Board Of Trustees Of The University Of Illinois Selective Apoptotic Induction in Cancer Cells Including Activation of Procaspase-3

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6403765B1 (en) 1998-06-16 2002-06-11 Thomas Jefferson University Truncated Apaf-1 and methods of use thereof
US6303329B1 (en) 1998-07-27 2001-10-16 Pharmacia & Upjohn Company Method for autoactivation of procaspase 8
US6534267B1 (en) 2000-01-06 2003-03-18 Xiaodong Wang Polynucleotides encoding activators of caspases
US20040077542A1 (en) 2000-08-23 2004-04-22 Board Of Regents, The University Of Texas System Apoptotic compounds
US6762045B2 (en) 2000-11-20 2004-07-13 Idun Pharmaceuticals, Inc. Membrane derived caspase-3, compositions comprising the same and methods of use therefor
US6878743B2 (en) 2001-09-18 2005-04-12 Sunesis Pharmaceuticals, Inc. Small molecule inhibitors of caspases
US20040180828A1 (en) 2003-01-30 2004-09-16 Yigong Shi Caspase-9 : BIR domain of XIAP complexes and methods of use

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38
H. BUNDGAARD: "Design of Prodrugs", 1985, ELSEVIER
K. WIDDER, ET. AL.: "Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396
KROSGAARD-LARSEN AND H. BUNDGAARD: "A Textbook of Drug Design and Development", 1991, article "Design and Application of Prodrugs (Chapter 5)", pages: 113 - 191
NOGRADY: "Medicinal Chemistry A Biochemical Approach", 1985, OXFORD UNIVERSITY PRESS, pages: 388 - 392
See also references of EP1893210A4
SVINGEN, P.A. ET AL., CLIN. CANCER RES., vol. 10, pages 6807 - 6820

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9522901B2 (en) 2005-05-26 2016-12-20 The Board Of Trustees Of The University Of Illinois Compositions and methods including cell death inducers and procaspase activation
US10166229B2 (en) 2005-05-26 2019-01-01 The Board Of Trustees Of The University Of Illinois Compounds and methods for the treatment of cancer
US11612598B2 (en) 2005-05-26 2023-03-28 The Board Of Trustees Of The University Of Illinois Compounds and methods for the treatment of cancer
US8592584B2 (en) 2005-05-26 2013-11-26 The Board of Trustees of the of The University of Illinois Compositions and methods including cell death inducers and procaspase activation
WO2008134474A3 (en) * 2007-04-27 2009-01-08 Univ Illinois Compositions and methods including cell death inducers and procaspase activation
WO2008134474A2 (en) * 2007-04-27 2008-11-06 The Board Of Trustees Of The University Of Illinois Compositions and methods including cell death inducers and procaspase activation
US9643960B2 (en) 2009-02-09 2017-05-09 The Board Of Trustees Of The University Of Illinois Procaspase activating compounds
US8778945B2 (en) 2009-02-09 2014-07-15 The Board Of Trustees Of The University Of Illinois Design, synthesis and evaluation of procaspase activating compounds as personalized anti-cancer drugs
WO2010091382A1 (en) 2009-02-09 2010-08-12 The Board Of Trustees Of The University Of Illinois Design, synthesis and evaluation of procaspase activating compounds as personalized anti-cancer drugs
US9102661B2 (en) 2009-02-09 2015-08-11 The Board Of Trustees Of The University Illinois Design, synthesis and evaluation of procaspase activating compounds as personalized anti-cancer drugs
WO2010102513A1 (en) * 2009-03-11 2010-09-16 深圳市湘雅生物医药研究院 Homopiperazine acethydrazide derivatives, preparation method and uses thereof
WO2011124087A1 (en) * 2010-04-06 2011-10-13 沈阳药科大学 Oxadiazole-based piperazine derivatives and applications thereof
CN101805338A (en) * 2010-04-06 2010-08-18 沈阳药科大学 Oxadiazole-based piperazine derivative and application thereof
US8916705B2 (en) 2011-10-14 2014-12-23 The Board of Trustees of The University of Illilnois Procaspase-activating compounds and compositions
US10022371B2 (en) 2011-10-14 2018-07-17 The Board Of Trustees Of The University Of Illinois Procaspase-activating compounds and methods
US10525056B2 (en) 2011-10-14 2020-01-07 The Board Of Trustees Of The University Of Illinois Procaspase-activating compounds and methods
AU2013296187B2 (en) * 2012-08-03 2016-09-29 The Board Of Trustees Of The University Of Illinois Enzyme-activating compounds and compositions
US10350207B2 (en) 2015-06-05 2019-07-16 The Board Of Trustees Of The University Of Illinois PAC-1 combination therapy
US11129830B2 (en) 2015-06-05 2021-09-28 The Board Of Trustees Of The University Of Illinois PAC-1 combination therapy
WO2017012113A1 (en) * 2015-07-23 2017-01-26 王智民 Compound pac-1 or salt thereof, and pharmaceutical composition comprising same
CN105085421B (en) * 2015-07-24 2017-12-26 深圳市湘雅生物医药研究院 Bit piperazine fumarate difficult to understand, hydrate, crystal formation and preparation method thereof
CN105085421A (en) * 2015-07-24 2015-11-25 深圳市湘雅生物医药研究院 Orbit azine-fumarate, hydrates, crystal forms and preparation methods therefor
WO2017015784A1 (en) * 2015-07-24 2017-02-02 深圳市湘雅生物医药研究院 Orbit azine-fumarate, hydrate, crystal form and preparation method therefor
US10561667B2 (en) 2015-07-24 2020-02-18 Shenzhen Zhenxing Medical Technology Co., Ltd. Orbit azine-fumarate, hydrate, crystal form and preparation method therefor
US11510919B2 (en) 2017-11-17 2022-11-29 The Board Of Trustees Of The University Of Illinois Cancer therapy by degrading dual MEK signaling

Also Published As

Publication number Publication date
EP2634581A1 (en) 2013-09-04
EP1893210A2 (en) 2008-03-05
EP1893210B1 (en) 2019-10-02
EP2659884A2 (en) 2013-11-06
EP1893210A4 (en) 2010-09-01
CA2610366A1 (en) 2006-11-30
IL187603A0 (en) 2008-03-20
JP5907852B2 (en) 2016-04-26
US20070049602A1 (en) 2007-03-01
MX2007014843A (en) 2008-02-14
AU2006251978A1 (en) 2006-11-30
DK1893210T3 (en) 2020-01-13
JP2013100285A (en) 2013-05-23
ES2763156T3 (en) 2020-05-27
WO2006128173A3 (en) 2007-05-24
EP2659884A3 (en) 2014-10-01
JP5513741B2 (en) 2014-06-04
JP2008545718A (en) 2008-12-18

Similar Documents

Publication Publication Date Title
US11612598B2 (en) Compounds and methods for the treatment of cancer
EP1893210B1 (en) Selective apoptotic induction in cancer cells including activation of procaspase-3
Schust et al. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization
Kuo et al. BPR0L075, a novel synthetic indole compound with antimitotic activity in human cancer cells, exerts effective antitumoral activity in vivo
Smith et al. Synthetic analogs of green tea polyphenols as proteasome inhibitors
Yuan et al. Olaparib hydroxamic acid derivatives as dual PARP and HDAC inhibitors for cancer therapy
Agarwal et al. Silibinin inhibits constitutive activation of Stat3, and causes caspase activation and apoptotic death of human prostate carcinoma DU145 cells
US11325892B2 (en) Compounds and methods for treating cancer
US7960435B2 (en) Anti-cancer agents and androgen inhibition activity compound
US8367644B2 (en) Methods and compounds useful to induce apoptosis in cancer cells
Hamdoun et al. Ginkgolic acids inhibit migration in breast cancer cells by inhibition of NEMO sumoylation and NF-κB activity
Singh et al. A coumarin derivative (RKS262) inhibits cell-cycle progression, causes pro-apoptotic signaling and cytotoxicity in ovarian cancer cells
CN101184491A (en) Selective apoptotic induction in cancer cells including activation of procaspase-3
CN106946812B (en) Design, synthesis and evaluation of procaspase activating compounds as personalized anticancer agents
Yan et al. Discovery of a PROTAC targeting ALK with in vivo activity
KR20210027382A (en) Activator of the unfolded protein reaction
Draganov et al. Upregulation of p53 through induction of MDM2 degradation: Anthraquinone analogs
Ray et al. Exploiting the HSP60/10 chaperonin system as a chemotherapeutic target for colorectal cancer
KR101400505B1 (en) Small molecules that replace or agonize p53 function
Wei et al. Drug repurposing of propafenone to discover novel anti-tumor agents by impairing homologous recombination to delay DNA damage recovery of rare disease conjunctival melanoma

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680018292.5

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2610366

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 187603

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2008513829

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/014843

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006251978

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 9866/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006771588

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU

ENP Entry into the national phase

Ref document number: 2006251978

Country of ref document: AU

Date of ref document: 20060526

Kind code of ref document: A