NewSoftAlkylatingAgentswithEnhancedCytotoxicityagainst...

12
New Soft Alkylating Agents with Enhanced Cytotoxicity against Cancer Cells Resistant to Chemotherapeutics and Hypoxia Alexandre Patenaude, 1 Re ´na G. Deschesnes, 1 Jean L.C. Rousseau, 1 E ´ ric Petitclerc, 1 Jacques Lacroix, 1 Marie-France Co ˆte ´, 1 and Rene ´ C.-Gaudreault 1,2 1 Unite´ de Biotechnologie et de Bioinge´nierie, CHUQ, Ho ˆpital Saint-Franc¸ois d’Assise and 2 De´partement de Me ´decine, Faculte´deMe´decine,Universite´Laval,Que´bec,Canada Abstract Chloroethylureas (CEU) are soft alkylating agents that covalently bind to B-tubulin (BTAC) and affect microtubule polymerization dynamics. Herein, we report the identification of a CEU subset and its corresponding oxazolines, which induce cell growth inhibition, apoptosis, and microtubule disruption without alkylating B-tubulin (N-BTAC). Both BTAC and N-BTAC trigger the collapse of mitochondrial potential (#W m ) and modulate reactive oxygen species levels, following activation of intrinsic caspase-8 and caspase-9. Experiments using human fibrosarcoma HT1080 respiratory- deficient cells (R 0 ) and uncoupler of the mitochondrial respiratory chain (MRC) showed that BTAC and N-BTAC impaired the MRC. R 0 cells displayed an increased sensitivity toward N-BTAC as compared with R + cells but, in contrast, were resistant to BTAC or classic chemotherapeutics, such as paclitaxel. Oxazoline-195 (OXA-195), an N-BTAC derivative, triggered massive swelling of isolated mitochondria. This effect was insensitive to cyclosporin A and to Bcl-2 addition. In contrast, adenine nucleotide translocator (ANT) antagonists, bongkrekic acid or atractyloside, diminished swelling induced by OXA-195. The antiproliferative activities of the N-BTACs CEU-025 and OXA-152 were markedly decreased in the presence of atractyloside. Conversely, pretreatment with cyclosporin A enhanced growth inhibition induced by BTAC and N-BTAC. One of the proteins alkylated by N-BTAC was identified as the voltage-dependent anion channel isoform-1, an ANT partner. Our results suggest that BTAC and N-BTAC, despite their common ability to affect the microtubule network, trigger different cytotoxic mechanisms in cancer cells. The role of mitochondria in these mechanisms and the potential of N-BTAC as a new therapeutic approach for targeting hypoxia-resistant cells are discussed. [Cancer Res 2007;67(5):2306–16] Introduction A major challenge of modern chemotherapy is the development of drugs that selectively target cancer cells, overcome chemo- resistant tumor cells, and have limited toxic effects. To that end, we developed over the past decade a new class of antimitotics called N -phenyl-N -2-(chloroethyl)ureas (CEU). CEU inhibits the growth of numerous tumors and drug-resistant cell lines (1–5). N - [4-Iodophenyl]-N -(2-chloroethyl)ureas (CEU-098) and N -[4-(1,1- dimethylethyl)-phenyl]-N -(2-chloroethyl)urea (CEU-022) were found to block the migration of cancer cells in vitro and display antitumor and antiangiogenic properties in vivo (4, 6). CEUs are nonmutagenic monoalkylating agents that are unreactive toward most cellular nucleophiles, such as glutathione and DNA (2, 7). Protein extracts from cells exposed to [ 14 C]CEU-022 showed that the drug covalently binds to a limited number of proteins, notably h-tubulin isoform-2 (8). Competition experiments confirmed that CEU-022 and several derivatives bind to the colchicine-binding site and disrupt the microtubule network (8) to block the cell division in G 2 /M phase (8). These CEUs are members of a molecular subset that hereafter is designated as hTAC. Apoptosis is one of the mechanisms by which chemotherapeutic agents induce cancer cell demise. The mitochondrion acts as a convergent point at which the signaling pathways integrate and trigger apoptosis, as shown with various anticancer agents. The mitochondrial-dependent apoptotic pathway results from a mitochondrial membrane permeability transition of the outer mitochondrial membrane culminating in the release of proapop- totic proteins in the cytosol, such as cytochrome c (9). One of the mechanisms leading to membrane permeability transition is the formation of a permeability transition pore (PTP; ref. 10). PTP opening is known to occur in response to different stimuli, such as increased production of reactive oxygen species (ROS), mitochon- drial calcium overload, thiol oxidation, adenine nucleotide depletion, and collapse of the mitochondrial membrane potential (DW m ; ref. 10). Specific contacts between the voltage-dependent anion channel isoform-1 (VDAC-1), adenine nucleotide trans- locator (ANT), and cyclophilin D are believed to constitute the backbone of PTP (11). Members of the Bcl-2 protein family, which act as checkpoints of apoptosis, play a key role in the membrane permeability transition process (12) and regulate PTP opening/ closing in concert with VDAC and ANT (13). Several molecules such as cyclosporin A have been shown to modulate PTP opening and used to decipher its regulation (14). Chemotherapeutics such as arsenite and lonidamine were found to be direct PTP opening agents (15). For instance, the mitochondrion is recognized as an important target for the development of new anticancer agents (15). Moreover, several lines of evidence suggest that mitochondria of transformed cells exhibit alterations that can be exploited to selectively induce their demise. For example, an increased binding of hexokinase-2 with VDAC-1 is observed in tumor cells, which alleviates survival of cancer cells in hypoxic conditions (16). Inhibition of hexokinase activity by 3-bromopyruvate or decreased binding of hexokinase-2 to VDAC-1 Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Dr. E. Petitclerc is a scholar of the Fonds de la recherche en sante´ du Que ´bec (Junior II level). Requests for reprints: Rene ´ C.-Gaudreault, or Alexandre Patenaude, Unite´ de Biotechnologie et de Bioinge´nierie, Centre de recherche, CHUQ, Ho ˆpital Saint- Franc ¸ois d’Assise, Que´bec, Que´bec, Canada G1L 3L5. Phone: 418-525-4444, ext. 52363 or 53401; Fax: 418-525-4372; E-mail: [email protected], or [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-3824 Cancer Res 2007; 67: (5). March 1, 2007 2306 www.aacrjournals.org Research Article Research. on May 28, 2018. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of NewSoftAlkylatingAgentswithEnhancedCytotoxicityagainst...

New Soft Alkylating Agents with Enhanced Cytotoxicity against

Cancer Cells Resistant to Chemotherapeutics and Hypoxia

Alexandre Patenaude,1Rena G. Deschesnes,

1Jean L.C. Rousseau,

1Eric Petitclerc,

1

Jacques Lacroix,1Marie-France Cote,

1and Rene C.-Gaudreault

1,2

1Unite de Biotechnologie et de Bioingenierie, CHUQ, Hopital Saint-Francois d’Assise and 2Departement de Medecine,Faculte de Medecine, Universite Laval, Quebec, Canada

Abstract

Chloroethylureas (CEU) are soft alkylating agents thatcovalently bind to B-tubulin (BTAC) and affect microtubulepolymerization dynamics. Herein, we report the identificationof a CEU subset and its corresponding oxazolines, whichinduce cell growth inhibition, apoptosis, and microtubuledisruption without alkylating B-tubulin (N-BTAC). BothBTAC and N-BTAC trigger the collapse of mitochondrialpotential (#Wm) and modulate reactive oxygen species levels,following activation of intrinsic caspase-8 and caspase-9.Experiments using human fibrosarcoma HT1080 respiratory-deficient cells (R0) and uncoupler of the mitochondrialrespiratory chain (MRC) showed that BTAC and N-BTACimpaired the MRC. R0 cells displayed an increased sensitivitytoward N-BTAC as compared with R+ cells but, in contrast,were resistant to BTAC or classic chemotherapeutics, such aspaclitaxel. Oxazoline-195 (OXA-195), an N-BTAC derivative,triggered massive swelling of isolated mitochondria. Thiseffect was insensitive to cyclosporin A and to Bcl-2 addition. Incontrast, adenine nucleotide translocator (ANT) antagonists,bongkrekic acid or atractyloside, diminished swelling inducedby OXA-195. The antiproliferative activities of the N-BTACsCEU-025 and OXA-152 were markedly decreased in thepresence of atractyloside. Conversely, pretreatment withcyclosporin A enhanced growth inhibition induced by BTACand N-BTAC. One of the proteins alkylated by N-BTAC wasidentified as the voltage-dependent anion channel isoform-1,an ANT partner. Our results suggest that BTAC and N-BTAC,despite their common ability to affect the microtubulenetwork, trigger different cytotoxic mechanisms in cancercells. The role of mitochondria in these mechanisms and thepotential of N-BTAC as a new therapeutic approach fortargeting hypoxia-resistant cells are discussed. [Cancer Res2007;67(5):2306–16]

Introduction

A major challenge of modern chemotherapy is the developmentof drugs that selectively target cancer cells, overcome chemo-

resistant tumor cells, and have limited toxic effects. To that end, wedeveloped over the past decade a new class of antimitotics calledN-phenyl-N ¶-2-(chloroethyl)ureas (CEU). CEU inhibits the growthof numerous tumors and drug-resistant cell lines (1–5). N-[4-Iodophenyl]-N ¶-(2-chloroethyl)ureas (CEU-098) and N-[4-(1,1-dimethylethyl)-phenyl]-N ¶-(2-chloroethyl)urea (CEU-022) werefound to block the migration of cancer cells in vitro and displayantitumor and antiangiogenic properties in vivo (4, 6). CEUs arenonmutagenic monoalkylating agents that are unreactive towardmost cellular nucleophiles, such as glutathione and DNA (2, 7).Protein extracts from cells exposed to [14C]CEU-022 showed thatthe drug covalently binds to a limited number of proteins, notablyh-tubulin isoform-2 (8). Competition experiments confirmed thatCEU-022 and several derivatives bind to the colchicine-binding siteand disrupt the microtubule network (8) to block the cell divisionin G2/M phase (8). These CEUs are members of a molecular subsetthat hereafter is designated as hTAC.Apoptosis is one of the mechanisms by which chemotherapeutic

agents induce cancer cell demise. The mitochondrion acts as aconvergent point at which the signaling pathways integrate andtrigger apoptosis, as shown with various anticancer agents.The mitochondrial-dependent apoptotic pathway results from amitochondrial membrane permeability transition of the outermitochondrial membrane culminating in the release of proapop-totic proteins in the cytosol, such as cytochrome c (9). One of themechanisms leading to membrane permeability transition is theformation of a permeability transition pore (PTP; ref. 10). PTPopening is known to occur in response to different stimuli, such asincreased production of reactive oxygen species (ROS), mitochon-drial calcium overload, thiol oxidation, adenine nucleotidedepletion, and collapse of the mitochondrial membrane potential(DWm; ref. 10). Specific contacts between the voltage-dependentanion channel isoform-1 (VDAC-1), adenine nucleotide trans-locator (ANT), and cyclophilin D are believed to constitute thebackbone of PTP (11). Members of the Bcl-2 protein family, whichact as checkpoints of apoptosis, play a key role in the membranepermeability transition process (12) and regulate PTP opening/closing in concert with VDAC and ANT (13). Several moleculessuch as cyclosporin A have been shown to modulate PTP openingand used to decipher its regulation (14).Chemotherapeutics such as arsenite and lonidamine were found

to be direct PTP opening agents (15). For instance, themitochondrion is recognized as an important target for thedevelopment of new anticancer agents (15). Moreover, several linesof evidence suggest that mitochondria of transformed cells exhibitalterations that can be exploited to selectively induce their demise.For example, an increased binding of hexokinase-2 with VDAC-1 isobserved in tumor cells, which alleviates survival of cancer cellsin hypoxic conditions (16). Inhibition of hexokinase activity by3-bromopyruvate or decreased binding of hexokinase-2 to VDAC-1

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).Dr. E. Petitclerc is a scholar of the Fonds de la recherche en sante du Quebec

(Junior II level).Requests for reprints: Rene C.-Gaudreault, or Alexandre Patenaude, Unite de

Biotechnologie et de Bioingenierie, Centre de recherche, CHUQ, Hopital Saint-Francois d’Assise, Quebec, Quebec, Canada G1L 3L5. Phone: 418-525-4444, ext. 52363or 53401; Fax: 418-525-4372; E-mail: [email protected], [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-3824

Cancer Res 2007; 67: (5). March 1, 2007 2306 www.aacrjournals.org

Research Article

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

increases the susceptibility of cancer cells to chemotherapeutics(17) and hypoxia (18).In this work, we report the identification of a subset of cytotoxic

CEUs and their corresponding oxazolines that do not alkylate h-tubulin (N-hTAC). N-hTACs have an increased cytotoxicity onhypoxia-resistant fibrosarcoma HT1080 cells generated by mito-chondrial DNA depletion. VDAC-1 was identified as a target of N-hTAC.

Materials and Methods

Reagents and chemicals. Nonradioactive CEU and oxazoline derivatives(Table 1) were kindly provided by Dr. Jean Rousseau from IMOTEP, Inc.

(Quebec, Canada). The synthesis of [14C]CEU-025 and [14C]CEU-027 was

carried out as previously described (19). All other drugs and reagents werepurchased from Sigma (St. Louis, MO). All drugs assessed in this study were

dissolved in DMSO and used at a final concentration of <0.19% (v/v). Thehuman Bcl-2 recombinant protein used (Sigma) is a 68-kDa fusion protein

composed of a maltose binding protein moiety followed by a histidine tag

and a Bcl-2 protein lacking 21 amino acid residues at the COOH terminus.

Cell lines. Human colon carcinoma HT29, breast carcinoma MDA-MB-231, human fibrosarcoma HT1080, and murine melanoma M21 cell

lines were obtained from the American Type Culture Collection

(Manassas, VA) and cultured in DMEM (Hyclone, Road Logan, UT)

supplemented with 5% bovine calf serum. Wild-type (wt) Chinesehamster ovary (CHO) cells (CHO-10001), colchicine/vinblastine–resistant

cells (CHO-VV 3-2), and paclitaxel-resistant cells (CHO-TAX 5-6) were

generously provided by Dr. Fernando Cabral (University of Texas

Medical School, Houston, TX) and cultured as previously described (8).HT1080 cells partially depleted of mitochondrial DNA were obtained by

maintaining wt HT1080 cells for 4 weeks in the presence of 100 ng/mL

ethidium bromide in the culture medium supplemented with 300 Ag/mLuridine and 2 mmol/L Na pyruvate.

Table 1. Growth inhibition of CEU and oxazoline and their relative alkylation potency with chlorambucil

Names Structure GI50 (Amol/L)* P ¶c

HT-29 HT-1080 M21 MDA-MB-231

CEU-022

N-[4-(1,1-Dimethylethyl)-phenyl]-

N ¶-(2-chloroethyl)urea6.4 10.6 8.1 5.0 20

CEU-025

N-[4-Cyclohexyl-phenyl]-

N ¶-(2-chloroethyl)urea36.8 33.3 45.5 81.9 12

CEU-027

N-[4-Heptyl-phenyl]-

N ¶-(2-chloroethyl)urea11.6 17.2 12.0 13.2 15

CEU-091

N-[4-(1,1-Dimethylethyl)-phenyl]-

N ¶-(ethyl)urea>100 >100 >100 >100 2

CEU-098N-[4-Iodo-phenyl]-

N ¶-(2-chloroethyl)urea4.5 9.3 5.6 5.7 10

CEU-107(R)N-[4-Iodo-phenyl]-

N ¶-(2-chloro-1(R)-methyl-ethyl)urea1.7 3.6 2.3 3.0 5

OXA-152

(4-Cyclohexyl-phenyl)-

(4,5-dihydro-oxazol-2-yl)-amine

12.6 16.6 15.5 26.5 35

OXA-195

(4-Heptyl-phenyl)-

(4,5-dihydro-oxazol-2-yl)-amine

7.1 10.1 8.9 7.3 23

CEU-236

N-[3-(5-Hydroxy-pentyl)-phenyl]-

N ¶-(2-chloroethyl)urea0.25 0.47 0.37 0.36 19

NOTE: The growth inhibition induced by different CEUs and oxazolines was determined using the sulforhodamine B assay as described in Materials and

Methods. GI50 is the concentration inhibiting 50% of cell growth. The alkylating potency (P ¶) for each CEU and oxazoline was determined by their abilityto alkylate 4-(4-nitrobenzyl)pyridine and compared with chlorambucil (see Materials and Methods).

*Values are representative of two independent experiments.cP ¶ = [A t2 � A t1 / (t2 � t1) (CEU or OXA) / A t2 � A t1 / (t2 � t1) chlorambucil], where A is absorption at 570 nm and (t2 � t1) is period of incubation.

New Cytotoxic Compounds against Chemoresistant Cells

www.aacrjournals.org 2307 Cancer Res 2007; 67: (5). March 1, 2007

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Growth inhibition and cytotoxicity assays. The sulforhodamine Bgrowth assay was done as previously reported (4). Cell cytotoxicity was

assessed by the resazurin assay. Cells were seeded onto 96-well microtiter

plates and, 24 h later, exposed for 16 or 48 h to the drugs before the addition

of medium containing resazurin (25 Ag/mL final) for 1 h at 37jC; then,fluorescence measurements [excitation: 530 (F35) nm, emission: 590 (F25)nm] were done with a microtiter plate fluorescence reader (Bio-Tek FL600,

Bio-Tek, Winooski, VT). Background fluorescence (BGF) emitted from thecontrol wells containing medium and resazurin without cells was

subtracted from fluorescence values obtained in the presence of cells. The

percentage of cell viability was calculated as follows: % viability = (mean

fluorescence with drug � BGF) / (mean fluorescence without drug � BGF)� 100.Alkylating potency of CEU. The alkylating potency of CEU and

oxazoline versus chlorambucil was determined with the colorimetric assay

previously described (8). The alkylation potency P ¶ refers to the percentkinetics ability of CEU or oxazoline to alkylate 4-(4-nitrobenzyl)pyridine

compared with chlorambucil.

Immunocytochemical analysis of microtubule network integrity.HT1080 cells (8 � 105) were seeded onto fibronectin 16 h before exposure tothe drugs. Cells were washed with PBS and fixed in 3.7% paraformaldehyde-

PBS for 10 min. After two washes with PBS, fixed cells were permeabilized

with 0.1% Triton X-100 for 3 min, washed, then blocked for 30 min (10%goat serum) before h-tubulin antibody (1:200) incubation for 1 h at roomtemperature. After washes, cells were incubated with 1:2,000 dilution of

antimouse immunoglobulin G Alexa 488 (Molecular Probes, Eugene, OR) for45 min at room temperature. All washes were done in PBS containing 10%

goat serum or in PBS alone. Cells were then mounted on a microscope slide

overnight with slow fade reagent (DakoCytomation, Carpinteria, CA) before

analysis under a confocal microscope Eclipse E800 (Nikon, Tokyo, Japan).Evaluation of mitochondrial cytochrome c release. Cytochrome c

release was evaluated by immunocytochemistry and confocal microscopy as

described for h-tubulin analysis (see above) except that a cytochrome cantibody was used (1:1,000 dilution; clone no. 6H2.B4, BD PharMingen, San-

Diego, CA).

Flow cytometric analysis. Following treatments with oxazoline-195(OXA-195) or CEU-236 for different periods of time or with DMSO for 24 h,HT1080 cell pellets (2.5 � 105) were resuspended in DMEM containing

5 Amol/L hydroethidine and 40 nmol/L DiOC6 to measure ROS productionand DWm, respectively, as described elsewhere (20). After treatment of the

cells with the dyes (30 min/37jC), the cells were placed on ice until analysis.DNA content cell cycle analysis was done by propidium iodide staining (8).

One-dimensional and isoelectric focusing/two-dimensional SDS-PAGE analysis. MDA-MB-231 and HT1080 cells were exposed to differentconcentrations of radioactive (or not) CEU or oxazoline. After incubation,

adherent and floating cells were pooled. The cells were pelleted and washed

with PBS before their resuspension in 1� Laemmli buffer containing 5% h-mercaptoethanol. The lysates were sonicated for 5 s and boiled for 5 minbefore one-dimensional electrophoresis. Isoelectric focusing was done with

the Protean IEF Cell apparatus according to the instructions of the

Figure 1. Relationship between h-tubulin alkylation potency and cell growth inhibition of CEUs and oxazolines toward different cancer and antimicrotubule-resistanth-tubulin–mutant CHO cell lines. A, MDA-MB-231 or HT1080 cells were plated and then treated the following day for 48 h with the following concentrations: DMSO,0.19% (v/v); CEU-022 and CEU-98, 30 Amol/L; CEU-25, CEU-027, and CEU-091, 75 Amol/L; CEU-107R, 10 Amol/L; OXA-152 and OXA-195, 50 Amol/L; CEU-236,2 Amol/L. After treatment, the cells were harvested as described in Materials and Methods and protein extracts (30 Ag) were separated by SDS-PAGE, followed byWestern blot with h-tubulin antibody. B, MDA-MB-231 cells were exposed for 24 h to 100 Amol/L of [14C]CEU-022, [14C]CEU-025, or [14C]CEU-027. The cells were thenharvested. Protein extracts, equivalent to 1 � 105 cells, were separated by SDS-PAGE (17.5%), transferred onto nitrocellulose membranes, and autoradiographedfor 13 d. C, MDA-MB-231 cells were plated 24 h before a 48-h treatment with DMSO (0.19%), the indicated CEU or oxazoline (50 Amol/L), colchicine, or vinblastine(VBL ; 5 Amol/L) in the absence or presence of CEU-022 (30 Amol/L). Total protein extracts were separated by SDS-PAGE for Western blot analysis of h-tubulin.The results show that CEU-022–mediated h-tubulin alkylation is blocked by colchicine (COL ) but not by other compounds tested. Similar results were obtained withHT1080 cells (data not shown). D, wt (n), CHO-VV 3-2 (.), and CHO-TAX 5-6 (E) h-tubulin–mutant CHO cell lines were exposed for 48 h with increasingconcentrations of CEU-022, CEU-025, CEU-027, OXA-152, and CEU-195 before cell fixation and sulforhodamine B staining for cell growth comparative analysis,as described in Materials and Methods. Points, mean growth percentage of triplicate 585-nm absorbance for each drug concentration exposure, compared with thoseof an untreated control. Representative of two independent experiments.

Cancer Research

Cancer Res 2007; 67: (5). March 1, 2007 2308 www.aacrjournals.org

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

manufacturer (Bio-Rad, Hercules, CA). Detection of native and alkylated h-tubulin isoforms is based on a one-dimensional electrophoretic shift assay

(8). The detection of the native and alkylated forms of VDAC-1 was done

with a 1:5,000 dilution of monoclonal anti–VDAC-1 [clone 89-173/033 (ab3)

and 89-173/045 (ab4), Calbiochem, San Diego, CA]. All incubations withantibodies were done at room temperature for 2 to 3 h in TBS, 0.1% Tween

20 with 1% milk or in TBS, 0.1% Tween 20 + 5% bovine serum albumin.

Poly(ADP-ribose) polymerase (PARP), caspase-3, caspase-8 (clone no. 1C12),

and caspase-9 antibodies were obtained from Cell Signaling, Inc. (Beverly,MA). Cytochrome oxidase subunit II (clone no. 12C4) and cytochrome

oxidase subunit IV (clone no. 10E8) antibodies were obtained from

Molecular Probes.

Mitochondrial isolation. Rat liver mitochondria were purified bydifferential centrifugation and Percoll gradient according to the procedure

reported by Almeida and Medina (21).

Mitochondrial swelling assay. Swelling of rat liver mitochondria wasmeasured with a spectrophotometer (Shimadzu, Kyoto, Japan) at 540 nm.

The mitochondrial suspension consisting of rat liver mitochondria (0.5 mg/

mL) was incubated in the swelling buffer [70 mmol/L sucrose, 214 mmol/L

mannitol, 5 mmol/L HEPES (pH 7.4), 0.5 mmol/L NaPO4 (pH 7.4)] with orwithout 5 mmol/L glutamate and 2.5 mmol/L sodium malate. The swelling

assays were done at 25jC. Signal decrease at 540 nm is indicative of

mitochondria swelling.

VDAC-1 purification. Transmembrane protein purification was done asdescribed by de Pinto et al. (22) with minor modifications. Briefly, cultured

cells exposed to CEU-025 were resuspended in PB2 buffer [3% Triton X-100,

10 mmol/L Tris (pH 7), 1 mmol/L EDTA, and 1 tablet of Complete mix ofprotease inhibitors (Roche, Penzberg, Germany) per 10 mL of buffer]. The

protein concentration of the extract was adjusted to 5 mg/mL before

applying onto a dry hydroxyapatite/celite (2:1 w/v) column (0.1 g/mg

protein). Two milliliters of PB2 buffer were used for elution of VDAC-1.VDAC-1 oxidation assay. The capacity of CEU and oxazoline to alkylate

the cysteinyl residues on VDAC-1 was verified using diamide-mediated

oxidation, described elsewhere (23). Total cell or rat liver mitochondria

extracts, exposed or not to CEU in Triton 3% buffer, were denatured withSDS 1% (10 min/25jC). All denatured samples were then treated for 30 minwith 250 Amol/L diamide. Subsequently, they were boiled in the presence orabsence of h-mercaptoethanol (5%) for SDS-PAGE analysis.

Results

Comparison of CEU growth inhibition versus alkylatingpotency. To optimize the antiproliferative properties of ourprototypical CEU-022 (1, 2, 4–6), several CEUs were prepared bysubstituting the alkyl group at the third or fourth position of thephenyl ring (Table 1). We selected from a CEU library a subset of9 molecules displaying GI50 ranging from 100 nmol/L to 100Amol/L and included CEU-091 as a negative control for proteinalkylation. CEU-091 has no chlorine atom and is devoid of bothalkylating and growth inhibition properties on solid tumors cells(4). CEU-107(R) bears a methyl group (R-enantiomer) on theethyl linker arm between the urea moiety and the chlorine atom.Interestingly, the S-enantiomer has no growth inhibition activitywhile having the same electrophilicity (24), thus illustrating theimportance of structure-activity relationships in the design ofnovel CEUs. OXA-152 and OXA-195 are ‘‘bioisosteres’’ of CEU-025and CEU-027, respectively. OXA-152 and OXA-195 are thecyclized byproducts of the 2-chloroethylamino moiety of CEUinto a 2-oxazoline heterocycle. As shown in Table 1, theseoxazolines exhibit electrophilic properties and antiproliferativeactivities similar to or even higher than those of their respective2-chloroethylurea counterparts. The 4-iodo derivative CEU-098 isa bioisostere of the 4-tert-butyl moiety designed to improve themetabolic resistance of CEU-022 (6). Finally, CEU-236, which is a3-(5-hydroxypentyl) substituted form of CEU, exhibits the

highest cell growth inhibition capacity on several tumor celllines (7, 25).N-BTAC: new CEUs and oxazolines that inhibit cancer cell

growth without alkylating the B-tubulin colchicine-bindingsite. Several CEUs were earlier shown to disrupt the cytoskeletonthrough their covalent binding to the colchicinoid-binding siteof h-tubulin isoform-2 (8). As shown in Fig. 1A , CEU-022, CEU-098, CEU-107R, and CEU-236 covalently bind to h-tubulin, whichleads to the formation of a protein byproduct exhibiting a fasterelectrophoretic mobility shift than the native h-tubulin (8).

Figure 2. hTAC and N-hTAC induce microtubule depolymerization. HT1080were plated on a microscope slide embedded on a six-well plate overnight andunexposed or exposed to OXA-195 (50 Amol/L) or CEU-236 (1 Amol/L) fordifferent periods of time (A) or for 16 h with CEU-022 (30 Amol/L), CEU-025(75 Amol/L), CEU-027 (75 Amol/L), OXA-152 (60 Amol/L), and OXA-195(50 Amol/L; B). Cells were treated with DMSO (0.19%) for 16 h (A and B). Aftertreatments, cells were fixed and incubated with the h-tubulin antibody forimmunofluorescence analysis by confocal microscopy as described in Materialsand Methods.

New Cytotoxic Compounds against Chemoresistant Cells

www.aacrjournals.org 2309 Cancer Res 2007; 67: (5). March 1, 2007

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

CEU-091 is, as expected, neither cytotoxic nor electrophilic (Table 1).Figure 1A also shows that the electrophoresis of proteins extractedfrom cells treated with CEU-025, CEU-027, OXA-152, or OXA-195 atconcentrations higher than their IG50 did not reveal the formation ofany h-tubulin byproducts, suggesting that N-hTAC does not alkylateh-tubulin. In support of this result, protein extracts from MDA-MB-231 cells exposed to [14C]CEU-022, [14C]CEU-025, or [14C]CEU-027revealed (Fig. 1B) a 50-kDa signal corresponding to the alkylated h-tubulin byproduct, restricted to[14C]CEU-022 labeling. However,N-hTAC still might partly act on h-tubulin as reversible instead ofirreversible antagonists of the colchicine-binding site. To address thisissue, we did competition experiments to determine the capacity ofN-hTAC to compete with CEU-022–mediated h-tubulin alkylation.Colchicine competes with hTAC for the colchicine-binding site (8),as shown in Fig. 1C . In contrast, neither vinblastine nor any of theN-hTACs tested diminished significantly the formation of the CEU-022 h-tubulin byproduct (Fig. 1C), thus confirming that CEU-025,CEU-027, and their corresponding oxazoline bioisosteres are notreversible antagonists of the h-tubulin colchicine-binding site.These observations suggest that N-hTAC does not inhibit cell

growth as hTAC or classic h-tubulin targeting agents such ascolchicinoids and taxoids. To confirm that hypothesis, we did growthinhibition assays using CHO-10001 (wt) and mutant CHO-VV 3-2 andCHO-TAX 5-6 cell lines, which exhibit differential sensitivity to anti–h-tubulin agents through h-tubulin mutations (26, 27). CHO-VV 3-2cells are resistant to molecules such as vinblastine and colchicinoidsand hypersensitive to taxoids such as paclitaxel. Conversely, CHO-TAX 5-6 cells are resistant to paclitaxel and hypersensitive to

vinblastine and colchicinoids. CHO-VV 3-2 and CHO-TAX 5-6 werepreviously found to be resistant and hypersensitive, respectively, tohTAC such as CEU-022 (8, 24) and as shown in this study (Fig. 1D). Incontrast, wt and h-tubulin–mutant cell lines displayed similar growthinhibition sensitivities in response to CEU-025, CEU-027, OXA-152,and OXA-195. These results suggest that N-hTAC agents inhibittumor cell growth through mechanisms unrelated to the microtubuledynamic perturbations triggered by classic microtubule-targetingagents such as colchicine and vinblastine and molecules belonging tothe hTAC subset.BTAC and N-BTAC induce microtubule depolymerization. To

assess whether N-hTAC can lead to the microtubule networkdepolymerization, h-tubulin immunofluorescence analysis wasdone with confocal microscopy following treatment of HT1080cells with N-hTAC. CEU-236 was used as a positive control toinduce microtubule depolymerization and was compared withOXA-195. Unexpectedly, we found that OXA-195, similarly toCEU-236, induced an early (2 h) massive depolymerization ofmicrotubule (Fig. 2A). This suggests that CEU or OXA-mediatedh-tubulin alkylation is not a prerequisite to induce microtubuledepolymerization. Figure 2B shows that other N-hTACs (e.g.,CEU-025 and CEU-152) provoked also substantial microtubuledepolymerization whereas CEU-027 induced weak microtubuledepolymerization in HT1080 cells. Together, this supports thathTAC and N-hTAC interfere with the polymerization process ofmicrotubule to different extents and mechanisms.BTAC and N-BTAC induce apoptosis. We next investigated

whether hTAC and N-hTAC induce cell death through apoptosis.

Figure 3. Cytotoxic CEU and oxazolineinduce apoptosis regardless of theircapacity to alkylate h-tubulin. MDA-MB-231(A) or HT1080 (B ) cells were exposedfor 48 h to different CEUs and oxazolines(A), as described in the legend of Fig. 1A ,or to DMSO (0.19%) for 24 h or for theindicated time periods with OXA-195(50 Amol/L; B). After treatment, cells wereharvested and protein extracts (30 Ag)were separated by SDS-PAGE and thentransferred onto nitrocellulose membranesfor Western blot analysis of PARP (A ),caspase-3, caspase-9, andcaspase-8 cleavages (A and B). B, rightarrows, cleaved protein form recognized bythe antibodies designated on the left,reflecting the activation state for caspases.C, immunocytochemistry analysis ofcytochrome c release by confocal analysisof HT1080 cells exposed to DMSO(0.1%) or OXA-195 (50 Amol/L) for 16 h.D, HT1080 cells were incubated withDMSO (0.1%/4 h) or with OXA-195(50 Amol/L) for 2 and 4 h and thenharvested for DWm and ROS productionanalysis by flow cytometry with DiOC6 andhydroethidine (He ) fluorescence probes,respectively. The percentages of DiOC6

and hydroethidine histograms indicate thelow and high staining cell populations,respectively.

Cancer Research

Cancer Res 2007; 67: (5). March 1, 2007 2310 www.aacrjournals.org

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

We have shown that CEUs and oxazolines, regardless of their h-tubulin alkylating potency, induced PARP cleavage in MDA-MB-231cells (Fig. 3A). In addition, we have shown that DNA fragmentationoccurs following exposure to any of the CEU and oxazoline used inthe present study (data not shown). Kinetic analysis of caspaseactivation in HT1080 cells shows that OXA-195 (50 Amol/L) triggersPARP cleavage after 8 h (Fig. 3B) and the activation of initiatorscaspase-8 and caspase-9 (Fig. 3B), suggesting that both the intrinsicand extrinsic apoptotic pathways are activated in response to OXA-195. As expected, the effector caspase-3 was activated subsequentlyto the activation of caspase-8 and caspase-9 (Fig. 3B). Accordingly,cytochrome c is released from mitochondria following exposure toOXA-195 (Fig. 3C) and occurs simultaneously with the induction ofcaspase-9 activation in response to OXA-195 (e.g., after 8 h; data notshown). Moreover, we observed that other hTACs and N-hTACsinduced also the activation of caspase-3, caspase-8, and caspase-9and the release of cytochrome c from mitochondria in HT1080 andMDA-MB-231 cells (data not shown). Altogether, these observa-tions indicate that CEU and oxazoline, regardless of their capacityto alkylate h-tubulin, induce apoptosis in cancer cells.BTAC and N-BTAC impair the mitochondrial respiratory

chain. ROS production and loss of DWm are common cell death

manifestations occurring during the early phases of apoptosis (28).We assessed the loss of DWm and ROS production changes inresponse to OXA-195 by flow cytometry using the cationic DiOC6and hydroethidine probes, respectively. HT1080 exposed to OXA-195 exhibited an increased percentage of the cell populationshowing time-dependent DWm loss and increased ROS production,both appearing as early as 2 h posttreatment (Fig. 3D). OtherhTACs and N-hTACs did also induce DWm loss (Supplementarydata). However, in the same conditions, these agents triggeredcontrasting ROS production responses (Fig. 4A , q+). For example,OXA-152 and OXA-195 induced substantial increase of ROSproduction, whereas CEU-022 and CEU-025 triggered a decreaseof ROS production compared with DMSO (Fig. 4A , q+). Wehypothesized that the abilities of CEUs and oxazolines to modulateROS production originate from the impairment of the mitochon-drial respiratory chain (MRC). To investigate that possibility, wecompared the ROS production induced by different CEUs andoxazolines in wt HT1080 cells (U+) and HT1080 cells that aredepleted of mitochondrial DNA (U0). U0 cells do not express anumber of essential components of the MRC, hence abrogating theproduction of ROS from MRC (29). Mitochondrial depletion wasconfirmed by Western blot analysis of cytochrome oxidase subunit

Figure 4. Mitochondrial DNA depletioneffects on HT1080 ROS production andviability loss induced by hTAC, N-hTAC, andclassic antineoplastics. A, U+ and U0 cellswere seeded onto multiwell plates andexposed the following day for 4 h in thepresence of DMSO (0.19%), CEU-022(30 Amol/L), CEU-025 (60 Amol/L), CEU-027(60 Amol/L), CEU-098 (20 Amol/L),CEU-107R(10Amol/L),OXA-152 (60Amol/L),OXA-195 (50 Amol/L), and CEU-236(5 Amol/L). After 3 h in the presence ofDMSO,CEUs, or oxazolines, carbonyl cyanidem-chlorophenylhydrazone (CCCP;10 Amol/L) was added or not, as indicated,for 1 h in the culture medium. Cells werethen harvested, washed, and stained withhydroethidine to measure ROS productionlevel by fluorescence-activated cell sorting.Columns, percent increase of hydroethidinefluorescence obtained for the indicatedtreatment compared with DMSO-treated U+

or U0 cells. Representative of two to fourindependent experiments. B and C, U+ andU0 cells were seeded onto 96-well microtiterplates 24 h before exposure to CEU-022(24 Amol/L, 16 h), CEU-025 (24 Amol/L,48 h), CEU-027 (13 Amol/L, 48 h), CEU-098(22 Amol/L, 48 h), CEU-107R (10 Amol/L,16 h), OXA-152 (30 Amol/L, 48 h), OXA-195(20 Amol/L, 48 h), and CEU-236 (3.3 Amol/L,16 h; B); or to 5-FU (67 Amol/L, 48 h),daunorubicin (DNM ; 0.2 Amol/L, 48 h),chlorambucil (CBL ; 130 Amol/L, 16 h);paclitaxel (PCT ; 20 nmol/L, 16 h),and vinblastine (VBL ; 20 nmol/L, 16 h; C).Exposure to 2-deoxy-D-glucose (2-DG; 1.2Amol/L, 48 h) was assessed to compare theglycolytic dependence of U0 and U+ cells (C).The resazurin assay was used to assessthe viability responses (B and C ; seeMaterials and Methods). Columns, percentviability representing the percent of resazurinfluorescence obtained in the presence ofthe drug and compared with DMSO(0.19%)–treated cells for similar timeexposures (n = 3); bars, SD. Representativeof >3 independent experiments.

New Cytotoxic Compounds against Chemoresistant Cells

www.aacrjournals.org 2311 Cancer Res 2007; 67: (5). March 1, 2007

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

II, a mitochondrial DNA–encoded protein (data not shown). Figure4A shows that mitochondrial DNA depletion abrogated themodulating action of CEU or oxazoline tested on the ROSproduction level, thus supporting our hypothesis. Moreover, inthe presence of the respiration uncoupler carbonyl cyanide m-chlorophenylhydrazone, an agent stimulating the electron flow ofMRC, all CEUs and oxazolines increased ROS production in U+ cellsbut not in U0 cells (Fig. 4A). No significant loss of viability wasobserved at all time periods of treatment used in these experimentsas revealed by the forward and side scatter analysis of flowcytometry data (data not shown), thus excluding that the ROS andDWm responses are resulting from extensive cell damage.Altogether, these results suggest that CEU and oxazoline interferewith the MRC early in cell death cascade induced by thesecompounds.Mitochondrial DNA depletion in HT1080 cells induces

opposite effects on the cytotoxic activities of BTAC versusN-BTAC. Interestingly, U0 cells are relatively resistant to hTAC(Fig. 4B ) and classic chemotherapeutic agents such as 5-fluorouracil (5-FU), daunorubicin, chlorambucil, paclitaxel, andvinblastine (Fig. 4C). In contrast, U0 cells show increase sensitivityto N-hTAC (Fig. 4B). U0 cells are alsomuchmore sensitive to 2-deoxy-D-glucose compared with U+ cells (Fig. 4C), thus illustrating theglycolytic metabolic dependence of these cells.OXA-195 induces massive swelling of mitochondria. Our

finding that hTAC and N-hTAC induce early MRC impairment ledus to hypothesize that these compounds may target themitochondrion to initiate apoptosis. We speculated that CEU andoxazoline might be PTP opening agents. To investigate this, westudied the capacity of CEU and oxazoline to induce swelling ofisolated rat liver mitochondria. Ca2+ mitochondrial overloading bycalcium chloride (CaCl2) exposure was used as a positive control

inducing mitochondrial swelling. In our assays, swelling of rat livermitochondria was optimal when using 150 Amol/L Ca2+ (Fig. 5A).None of the CEUs and oxazolines (Table 1) assessed with arelatively high concentration (100 Amol/L) induced swelling, exceptfor OXA-195 (Fig. 5A). Under these conditions, OXA-195 inducedrapid swelling of mitochondria that caused larger amplitudechanges than Ca2+ (Fig. 5A). OXA-195–mediated mitochondrialswelling was also observed on mitochondria of intact MDA-MB-231cells, as confirmed by electron microscope analysis (Supplementarydata).Classical PTP-regulating agents differentially affect the OXA-

195– and Ca2+-induced swelling. To understand the potential roleof PTP-associated proteins in the OXA-195–mediated swellingeffect, we have evaluated the effect of inhibitors known to interferewith the function of these proteins. Cyclosporin A, a classic PTPblocker that interferes with the function of cyclophilin D into PTPopening (14), blocked the swelling induced by Ca2+ (Fig. 5B).However, it had a weak inhibitory activity on the swelling inducedby OXA-195 (Fig. 5B). Bongkrekic acid is an antiapoptotic agentinhibiting PTP opening and converting ANT into its ‘‘closed’’m-conformation (i.e., exposing the adenine-binding site ofANT toward the matrix; ref. 30). Bongkrekic acid pretreatment(25 Amol/L) considerably inhibited the Ca2+-mediated swellingeffect (Fig. 5B). Bongkrekic acid could also inhibit OXA-195–induced swelling but only with higher concentrations (50–100Amol/L; Fig. 5B). Interestingly, cotreatment with cyclosporin A(1 Amol/L) and bongkrekic acid (25 Amol/L) completely blocked theswelling induced by Ca2+, but had no such additive effect on theswelling induced by OXA-195 even at high concentrations (5 and 100Amol/L, respectively; Fig. 5B), as compared with each agent usedseparately at the same concentrations (data not shown). Atractylo-side is also an ANT inhibitor but, in contrast to bongkrekic acid, is a

Figure 5. OXA-195 induces massiveswelling of rat liver mitochondria that iscyclosporin A and Bcl-2 insensitive.Kinetics of rat liver mitochondria swellingwas measured by absorbance at 540 nmusing 0.5 mg/mL rat liver mitochondria asdescribed in Materials and Methods.Arrows, time at which the drugs wereadded as depicted by the sharpabsorbance change due to the opening ofthe spectrophotometer samplecompartment. As indicated, DMSO (0.25%v/v) or CaCl2 (150 Amol/L) was added asnegative and positive control of swellinginduction, respectively. A, the CEU oroxazoline derivatives (100 Amol/L) listed inTable 1 were added. B to D, swellingassays were done as described in (A ) inthe presence of different concentrations(indicated in the text) of cyclosporin A(CsA ) and/or bongkrekic acid (BA ; B ),atractyloside (ATR ; C ), or recombinantBcl-2 (10 Ag/mL; D ) for 2 min beforeexposure to OXA-195 (115 Amol/L) orCaCl2 (150 Amol/L). Arrows on top (A–D )indicate the time at which OXA-195 orCaCl2 was added and left arrow (B–D)marks the addition of other agents. Allswelling responses are representative of atleast three independent experiments.

Cancer Research

Cancer Res 2007; 67: (5). March 1, 2007 2312 www.aacrjournals.org

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

proapoptotic agent that opens PTP. Atractyloside induces thec-conformation of ANT (i.e., exposing the adenine-binding site ofANT toward the cytoplasm; ref. 30). In agreement with other reports(31), we showed that atractyloside itself could induce the swelling ofrat liver mitochondria (Fig. 5C). In our assays, this effect wasobserved at concentrations >2 mmol/L (Fig. 5C). To evaluate thecontribution of atractyloside on other swelling inducers, we used 1mmol/L atractyloside, which per se induced little swelling (Fig. 5C).Pretreatment of rat liver mitochondria with atractyloside slightlyenhanced the kinetics of Ca2+-mediated swelling (Fig. 5C).Surprisingly, atractyloside pretreatment significantly inhibitedOXA-195–mediated rat liver mitochondria swelling (Fig. 5C).Markedly, supplementation of rat liver mitochondria with recom-binant Bcl-2 (10 Ag/mL) almost completely abrogated Ca2+-mediated swelling (Fig. 5D). In contrast, a similar Bcl-2 additiondid not prevent the swelling induced by OXA-195. Altogether, theseresults suggest that OXA-195 can induce mitochondrial swelling viaa nonclassic PTP-dependent mechanism.

PTP-regulating agents modulate CEU- and oxazoline-medi-ated growth inhibition responses.We next investigated the effectof PTP inhibitors on the growth inhibition responses induced byCEU or oxazoline. Unexpectedly, pretreatment of HT1080 cells withthe PTP blocker cyclosporin A increased the growth inhibitionresponses induced by N-hTAC and hTAC (Supplementary data). Incontrast, pretreatment of cells with atractyloside substantiallyreduced the antiproliferative action of N-hTAC but not of hTAC(Supplementary data).CEU and oxazoline bind covalently to the VDAC-1. Previous

experiments conducted by Legault et al. (8) suggested that CEUand oxazoline derivatives can covalently bind to proteins that areunrelated to h-tubulin to induce their growth inhibition andproapoptotic actions. SDS-PAGE autoradiograms of cells treatedwith [14C]CEU-022, [14C]CEU-025, or [14C]CEU-027 (Fig. 1B)revealed the radiolabeling of several proteins, notably a 34-kDa(p34) protein. The identification of p34 was done using isoelectricfocusing/two-dimensional electrophoresis of proteins extracted

Figure 6. Covalent binding of CEU-025 oncysteinyl residues of VDAC-1 inMDA-MB-231 cells. A, MDA-MB-231 cellswere treated for 3 d in the presence of[14C]CEU-25 (100 Amol/L). Protein extractsequivalent to 2 � 105 cells wereprepared as described in Materials andMethods, subjected to isoelectric focusing/two-dimensional SDS-PAGE (10%), andtransferred onto nitrocellulose membranes.The membranes were autoradiographedfor 5 d and analyzed by Western blotwith anti–VDAC-1 (ab3). B, VDAC-1was purified from cell extracts of [14C]CEU-25–treated MDA-MB-231 cells as de-scribed in Materials and Methods. Aneluted hydroxyapatite/celite (2:1) purifiedprotein (10 AL) extract was separated onSDS-PAGE (17.5%) and transferred ontonitrocellulose membrane. The membranewas autoradiographed for detection of [14C]signal. The same membrane was usedfor VDAC-1 detection by Western blotanalysis with anti–VDAC-1. A and B,molecular weight protein standards inkilodaltons are indicated on the right.C, protein extracts from MDA-MB-231 cellswere incubated in the presence or absenceof diamide before boiling in Laemmli bufferwith or without h-mercaptoethanol(b-MeSH ). Protein extracts were thenseparated by SDS-PAGE (12.5%) beforeanalysis by Western blot with anti–DAC-1antibody. D, protein extracts fromMDA-MB-231 cells exposed to CEU-025(100 Amol/L) for different periods of timewere incubated in the presence of diamideand then analyzed with anti–VDAC-1antibody following their electrophoresis bySDS-PAGE (12.5%). The reduced (Re )and oxidized (Ox ) VDAC-1 have apparentmolecular weights of 34 and 30 kDa,respectively.

New Cytotoxic Compounds against Chemoresistant Cells

www.aacrjournals.org 2313 Cancer Res 2007; 67: (5). March 1, 2007

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

from MDA-MB-231 cells treated with [14C]CEU-025. Figure 6Ashows the presence of the [14C]CEU-025 labeled p34 byproductexhibiting an estimated isoelectric point of f8. p34 was localizedby silver staining on a corresponding two-dimensional gel, excisedfrom the gel, trypsinized, and then analyzed by mass spectrometry.It allowed the identification of the voltage-dependence anionchannel isoform-1 (VDAC-1) as the most likely target of CEU-025(data not shown). Western blot analysis with anti–VDAC-1 revealeda spot that perfectly matches the radioactive spot onto thenitrocellulose membrane (Fig. 6A), strongly suggesting that VDAC-1 is a target of [14C]CEU-025. Similar results were observed forprotein extracts labeled with [14C]CEU-022 and [14C]CEU-027 (datanot shown). Furthermore, purified VDAC-1 isoform isolated fromMDA-MB-231 cells treated with [14C]CEU-025 also displayedcorresponding anti–VDAC-1 and radioactive signals (Fig. 6B).CEU-025 alkylates cysteine residue(s) of VDAC-1. The human

VDAC-1 isoform possesses only two conserved cysteinyl residues(Cys127 and Cys232) able to form a disulfide bridge via a redoxreaction, which is impaired by alkylating agents (32). We examinedthe ability of CEU and oxazoline to block the diamide-mediateddisulfide bridge formation. Under nonreducing conditions, thedisulfide bridge of VDAC-1 (oxidized form) generated by theaddition of diamide exhibits an increased electrophoretic mobilityof VDAC-1 easily distinguishable from the fully reduced form, asdescribed by De Pinto et al. (32) and shown in Fig. 6C . Themonothiol alkylating agent N -ethylmaleimide abolished themigration shift of VDAC-1 induced by diamide (data not shown).Figure 6D shows that increasing the time of exposure of MDA-MB-231 cells to CEU-025 leads to a progressive formation of thereduced band of VDAC-1 in the presence of diamide. Similar resultswere obtained in response to CEU-O22 and CEU-027 (data notshown). These results strongly suggest that CEU-022, CEU-025, andCEU-027 alkylate VDAC-1 on at least one of the two cysteinylresidues.

Discussion

The main objective of this work was to investigate themechanism underlying the cytotoxic and growth inhibitionproperties of hTAC and a newly discovered subset of CEUs andoxazolines called N-hTAC. Regardless of their potency to bind to h-tubulin, we found that CEU and oxazoline induced apoptosis incancer cells. The activation of initiator caspase-8 and caspase-9 inresponse to OXA-195 supports the view that the intrinsic andextrinsic apoptotic pathways can be activated by hTAC and N-hTAC.An important finding is that the cytotoxicity of N-hTAC was

increased in U0 cells when compared with U+ cells. Depletion ofmitochondrial DNA reportedly increases the resistance of cancercells toward chemotherapeutic drugs (33). Accordingly, weobserved that HT1080 U0 cells are less sensitive than U+ cells tovarious antineoplastic agents, such as microtubule-targeting agentspaclitaxel and vinblastine. The mechanisms responsible for suchresistance are not fully understood. Mitochondrial DNA depletionmay be involved in the etiology of cancer, notably by increasing theexpression of genes that enhance glycolysis, block apoptosis, andincrease the invasive behavior of cancer cells (34). These geneticchanges might be essential for the survival of cancer cell clusters intumor hypoxic environments. Accordingly, U0 cells are much moresensitive than U+ cells to the glycolytic inhibitor 2-deoxy-D-glucose.Glycolytic inhibitors constitute a new chemotherapeutic strategy to

target hypoxia-resistant tumor cells. Although we do not know yetwhether and how N-hTAC affects glycolysis, our study suggests thatthese agents should be tested for their capacity to selectively targethypoxic chemoresistant tumor cells.Based on time course experiments, both hTAC and N-hTAC

were found to induce early mitochondrial perturbations such ascollapse of DWm and MRC-dependent ROS production, their onset(2 h) preceding the apparent release of cytochrome c (8 h, data notshown). The roles of DWm loss and ROS production in apoptosishave been the subject of controversies, but both were suggested tobe initial events inducing membrane permeability transition andcytochrome c release (28). Our observations suggest that hTAC andN-hTAC stimulate ROS production through impairment of theMRC, but the contribution of those mitochondrial perturbations tothe cytotoxic mechanism and the targets of these compoundsremain to be determined. Mitochondrial ROS stimulation byprooxidants was shown to induce PTP via an ANT dimerization–dependent process (35). Increased ROS production can induce lipidperoxidation and loss of mitochondrial lipid cardiolipin, enablingcytochrome c detachment from cardiolipin (36) and alteringmembrane fluidity (37). Those alterations may in turn uncoupleMRC and further enhance ROS production. In this context, it isnoteworthy that some CEUs were found to interact with lipids andalter membrane fluidity (38, 39).OXA-195 was found to trigger a rapid swelling of rat liver

mitochondria, in contrast to other CEUs and oxazolines tested,involving an atypical mechanism, which is notably insensitive to orpoorly affected by the PTP inhibitor cyclosporin A and Bcl-2supplementation. Intriguingly, the PTP opening agent atractylosidediminished the extent of the rat liver mitochondria swelling andthe growth inhibition induced by some N-hTAC. In contrast,cyclosporin A enhanced the growth inhibition induce by hTAC andN-hTAC. It is unclear how CEU and oxazoline induce thesepharmacologically dependent responses. Yet, it further emphasizesthat hTAC and N-hTAC induce different cytotoxic mechanisms andsuggests that some of these compounds, such as OXA-195, inducean unusual PTP-dependent apoptosis process. Accordingly, wefound that OXA-195 triggers cytochrome c release from isolated ratliver mitochondria (data not shown). However, mitochondrialswelling might not be essential to initiate membrane permeabilitytransition in response to CEU or oxazoline because CEU-025, whichdoes not induce swelling, induced cytochrome c release (data notshown). Mitochondrial swelling before rupture of outer mitochon-drial membrane is not a prerequisite for cytochrome c release (31).We found that VDAC-1 is a potential target of N-hTAC (CEU-025

and CEU-027). VDAC-1 is a channel-forming protein present in theouter mitochondrial membrane, which allows the exchange ofmost metabolites, notably ADP/ATP between the cytosol and theinternal compartments of the mitochondrion (40). VDAC-1 wasproposed to be an intrinsic PTP component that by itself, or inconcert with other proteins such as ANT and proteins of theproapoptotic Bcl-2 family, constitutes a channel allowing thepassage of cytochrome c (13). In that context, the conductanceproperties of VDAC-1 were shown to be important whereas closedconductance states of VDAC-1 stimulate membrane permeabilitytransition. Bcl-2 protein members modulate the conductance stateof VDAC-1 and thus control the onset of membrane permeabilitytransition (41). Interestingly, cytochrome c release, induced by ROS,was found to be VDAC-1 dependent and cyclosporin A indepen-dent (42). The binding of hexokinase-1 or hexokinase-2 tomitochondria was shown to involve interactions with VDAC-1

Cancer Research

Cancer Res 2007; 67: (5). March 1, 2007 2314 www.aacrjournals.org

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

(17, 43). Interestingly, these interactions increase the glycolysis-dependent metabolism of cancer cells and prevent the induction ofthe apoptosis induced by different apoptotic stimuli such asstaurosporine (44–47). In the near future, it will be important toassess whether N-hTAC–dependent VDAC-1 alkylation affects suchinteractions with hexokinase-1 and/or hexokinase-2 to selectivelykill these hypoxia-resistant cells. It should be pointed out that theapparent ability of hTAC compound (CEU-022) to bind to VDAC-1raises issues in this context. Nevertheless, the ability of radiolabeledhTAC and N-hTAC to alkylate various proteins suggests that thecell growth inhibition mechanisms triggered by these compoundsare complex and do not rely per se on the selective and specificalkylation of a single protein. In addition, the alkylation of a proteinor a subset of proteins may modulate or be modulated by thealkylation of other proteins. For example, the covalent binding ofhTAC to h-tubulin could modify the responses of VDAC-1alkylation and vice versa . Thus, despite their inability to bind toh-tubulin, N-hTAC could destabilize the microtubule networkthrough such alkylation interplays. In this context, it is known thatmitochondria are dynamically associated with microtubule andthat VDAC-1 is a binding site for h-tubulin (48). VDAC-1 wasrecently reported to be required for Bim-dependent apoptosis (49),a Bcl-2 proapoptotic member released from microtubule onexposure to some cell death stimuli (50).

In conclusion, we have identified a new class of soft alkylatingagents, called N-hTACs, which induce apoptosis and microtubuledepolymerization of cancer cells. In contrast to hTAC, N-hTACdoes not covalently bind to h-tubulin and shows an increasedcytotoxicity against cells exhibiting mitochondrial DNA depletion,which leads to resistance toward other chemotherapeutics. N-hTAC molecules are promising tools for the design of newantineoplastic agents that target chemoresistant or hypoxia-resistant cancer cells. These molecules may contribute to anunderstanding of the signaling pathways underlying such resis-tance mechanisms. ANT and VDAC-1 might be key players incontrolling PTP-dependent apoptosis, microtubule-mitochondrioninteractions, and energetic balance either as protein bindingpartners or an independent tandem with specific roles.

Acknowledgments

Received 10/16/2006; revised 11/20/2006; accepted 12/8/2006.Grant support: Cancer Research Society of Canada.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.We thank Dr. M.R. Ven Murthy and Marc-Edouard Mirault for critical reading of the

manuscript; Dr. Fernando Cabral for h-tubulin mutant CHO cells; Dr. Maurice Dufourfor helping with the fluorescence-activated cell sorting analysis; and Claude Marquisfor excellent technical assistance.

New Cytotoxic Compounds against Chemoresistant Cells

www.aacrjournals.org 2315 Cancer Res 2007; 67: (5). March 1, 2007

References1. Bechard P, Lacroix J, Poyet P, C.-Gaudreault R.Synthesis and cytotoxic activity of new alkyl-[3-(chlor-oethyl)ureido] benzene derivatives. Eur J Med Chem1994;29:963–6.2. Lacroix J, Gaudreault RC, Page M, Joly LP. In vitro andin vivo activity of 1-aryl-3-(2-chloroethyl) urea deriva-tives as new antineoplastic agents. Anticancer Res 1988;8:595–8.3. Gaudreault RC, Lacroix J, Page M, Joly LP. 1-Aryl-3-(2-chloroethyl) ureas: synthesis and in vitro assay aspotential anticancer agents. J Pharm Sci 1988;77:185–7.4. Petitclerc E, Deschesnes RG, Cote MF, et al. Anti-angiogenic and antitumoral activity of phenyl-3-(2-chloroethyl)ureas: a class of soft alkylating agentsdisrupting microtubules that are unaffected by celladhesion-mediated drug resistance. Cancer Res 2004;64:4654–63.5. Gaudreault RC, Alaui-Jamali MA, Batist G, Bechard P,Lacroix J, Poyet P. Lack of cross-resistance to a newcytotoxic arylchloroethyl urea in various drug-resistanttumor cells. Cancer Chemother Pharmacol 1994;33:489–92.6. Miot-Noirault E, Legault J, Cachin F, et al. Antineo-plastic potency of arylchloroethylurea derivatives inmurine colon carcinoma. Invest New Drugs 2004;22:369–78.7. Mounetou E, Legault J, Lacroix J, C.-Gaudreault R.Antimitotic antitumor agents: synthesis, structure-activity relationships, and biological characterizationof N -aryl-N ¶-(2-chloroethyl)ureas as new selective alky-lating agents. J Med Chem 2001;44:694–702.8. Legault J, Gaulin JF, Mounetou E, et al. Microtubuledisruption induced in vivo by alkylation of h-tubulin by1-aryl-3-(2-chloroethyl)ureas, a novel class of softalkylating agents. Cancer Res 2000;60:985–92.9. Saelens X, Festjens N, Vande Walle L, van Gurp M, vanLoo G, Vandenabeele P. Toxic proteins released frommitochondria in cell death. Oncogene 2004;23:2861–74.10. Halestrap AP, McStay GP, Clarke SJ. The permeabilitytransition pore complex: another view. Biochimie 2002;84:153–66.11. Crompton M. The mitochondrial permeability tran-sition pore and its role in cell death. Biochem J 1999;341:233–49.

12. Sharpe JC, Arnoult D, Youle RJ. Control of mitochon-drial permeability by Bcl-2 family members. BiochimBiophys Acta 2004;1644:107–13.13. Tsujimoto Y, Shimizu S. The voltage-dependent anionchannel: an essential player in apoptosis. Biochimie2002;84:187–93.14. Halestrap AP, Davidson AM. Inhibition of Ca2(+)-induced large-amplitude swelling of liver and heartmitochondria by cyclosporin is probably caused by theinhibitor binding to mitochondrial-matrix peptidyl-prolyl cis-trans isomerase and preventing it interactingwith the adenine nucleotide translocase. Biochem J1990;268:153–60.15. Armstrong JS. Mitochondria: a target for cancertherapy. Br J Pharmacol 2006;147:239–48.16. Pedersen PL, Mathupala S, Rempel A, Geschwind JF,Ko YH. Mitochondrial bound type II hexokinase: a keyplayer in the growth and survival of many cancers andan ideal prospect for therapeutic intervention. BiochimBiophys Acta 2002;1555:14–20.17. Pastorino JG, Hoek JB, Shulga N. Activation ofglycogen synthase kinase 3h disrupts the binding ofhexokinase II to mitochondria by phosphorylatingvoltage-dependent anion channel and potentiates che-motherapy-induced cytotoxicity. Cancer Res 2005;65:10545–54.18. Xu RH, Pelicano H, Zhou Y, et al. Inhibition ofglycolysis in cancer cells: a novel strategy to overcomedrug resistance associated with mitochondrial respira-tory defect and hypoxia. Cancer Res 2005;65:613–21.19. Azim EM, Madelmont JC, Cussac C, et al. MarquagePAR 14C et 13C du terbutylanilino-4 chloroethyluree. JLabelled Comp Radiopharm 1993;XXXIII:1079–82.20. Patenaude A, Ven Murthy MR, Mirault ME. Mito-chondrial thioredoxin system: effects of TrxR2 over-expression on redox balance, cell growth, and apoptosis.J Biol Chem 2004;279:27302–14.21. Almeida A, Medina JM. A rapid method for theisolation of metabolically active mitochondria from ratneurons and astrocytes in primary culture. Brain ResBrain Res Protoc 1998;2:209–14.22. de Pinto V, Prezioso G, Palmieri F. A simple and rapidmethod for the purification of the mitochondrial porinfrom mammalian tissues. Biochim Biophys Acta 1987;905:499–502.

23. Kosower EM, Correa W, Kinon BJ, Kosower NS.Glutathione. VII. Differentiation among substrates bythe thiol-oxidizing agent, diamide. Biochim BiophysActa 1972;264:39–44.24. Mounetou E, Legault J, Lacroix J, C-Gaudreault R. Anew generation of N -aryl-N ¶-(1-alkyl-2-chloroethyl)ureasas microtubule disrupters: synthesis, antiproliferativeactivity, and h-tubulin alkylation kinetics. J Med Chem2003;46:5055–63.25. Moreau E, Fortin S, Desjardins M, Rousseau J,Peticlerc E, C-Gaudreault R. Preparation of a newgeneration of N -phenyl-N ¶-(2-chloroethyl)ureas as po-tential antineoplastic agents: synthesis and cytocidalactivity. Bioorg Med Chem 2005;13:6703–12.26. Cabral F, Sobel ME, Gottesman MM. CHO mutantsresistant to colchicine, colcemid or griseofulvin have analtered h-tubulin. Cell 1980;20:29–36.27. Schibler MJ, Barlow SB, Cabral F. Elimination ofpermeability mutants from selections for drug resis-tance in mammalian cells. FASEB J 1989;3:163–8.28. Fleury C, Mignotte B, Vayssiere JL. Mitochondrialreactive oxygen species in cell death signaling. Bio-chimie 2002;84:131–41.29. Guidot DM, McCord JM, Wright RM, Repine JE.Absence of electron transport (Rho 0 state) restoresgrowth of a manganese-superoxide dismutase-deficient Saccharomyces cerevisiae in hyperoxia.Evidence for electron transport as a major sourceof superoxide generation in vivo . J Biol Chem 1993;268:26699–703.30. Vieira HL, Haouzi D, El Hamel C, et al. Permeabiliza-tion of the mitochondrial inner membrane duringapoptosis: impact of the adenine nucleotide trans-locator. Cell Death Differ 2000;7:1146–54.31. Machida K, Hayashi Y, Osada H. A novel adeninenucleotide translocase inhibitor, MT-21, induces cyto-chrome c release by a mitochondrial permeabilitytransition-independent mechanism. J Biol Chem 2002;277:31243–8.32. De Pinto V, al Jamal JA, Benz R, Genchi G, Palmieri F.Characterization of SH groups in porin of bovine heartmitochondria. Porin cysteines are localized in thechannel walls. Eur J Biochem 1991;202:903–11.33. Singh KK, Russell J, Sigala B, Zhang Y, Williams J,

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Cancer Research

Cancer Res 2007; 67: (5). March 1, 2007 2316 www.aacrjournals.org

Keshav KF. Mitochondrial DNA determines the cellularresponse to cancer therapeutic agents. Oncogene 1999;18:6641–6.34. Lee HC, Yin PH, Lin JC, et al. Mitochondrial GenomeInstability And mtDNA depletion in human cancers.Ann N Y Acad Sci 2005;1042:109–22.35. Costantini P, Belzacq AS, Vieira HL, et al. Oxidationof a critical thiol residue of the adenine nucleotidetranslocator enforces Bcl-2-independent permeabilitytransition pore opening and apoptosis. Oncogene 2000;19:307–14.36. Petrosillo G, Ruggiero FM, Paradies G. Role ofreactive oxygen species and cardiolipin in the releaseof cytochrome c from mitochondria. FASEB J 2003;17:2202–8.37. Morin C, Zini R, Tillement JP. Anoxia-reoxygenation-induced cytochrome c and cardiolipin release from ratbrain mitochondria. Biochem Biophys Res Commun2003;307:477–82.38. Gicquad C, Auger M, Wong TT, Poyet P,Boudreau N, R CG. Interaction of 4-tert -butyl-[3-(2-chloroethyl) ureido] benzene with phosphatidylcho-line bilayers: a differential scanning calorimetry andinfrared spectroscopy study. Arch Biochem Biophys1996;334:193–9.

39. Saint-Laurent A, Boudreau N, Lariviere D, Legault J,Gaudreault RC, Auger M. Membrane interactions of anew class of anticancer agents derived from arylchlor-oethylurea: a FTIR spectroscopic study. Chem PhysLipids 2001;111:163–75.40. Lemasters JJ, Holmuhamedov E. Voltage-dependentanion channel (VDAC) as mitochondrial governator-thinking outside the box. Biochim Biophys Acta 2006;1762:181–90.41. Rostovtseva TK, Tan W, Colombini M. On the role ofVDAC in apoptosis: fact and fiction. J BioenergBiomembr 2005;37:129–42.42. Madesh M, Hajnoczky G. VDAC-dependent perme-abilization of the outer mitochondrial membrane bysuperoxide induces rapid and massive cytochrome crelease. J Cell Biol 2001;155:1003–15.43. Zaid H, Abu-Hamad S, Israelson A, Nathan I,Shoshan-Barmatz V. The voltage-dependent anionchannel-1 modulates apoptotic cell death. Cell DeathDiffer 2005;12:751–60.44. Azoulay-Zohar H, Israelson A, Abu-Hamad S,Shoshan-Barmatz V. In self-defence: hexokinase pro-motes voltage-dependent anion channel closure andprevents mitochondria-mediated apoptotic cell death.Biochem J 2004;377:347–55.

45. Gottlob K, Majewski N, Kennedy S, Kandel E, RobeyRB, Hay N. Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step ofglycolysis and mitochondrial hexokinase. Genes Dev2001;15:1406–18.46. Pastorino JG, Shulga N, Hoek JB. Mitochondrialbinding of hexokinase II inhibits Bax-induced cyto-chrome c release and apoptosis. J Biol Chem 2002;277:7610–8.47. Majewski N, Nogueira V, Bhaskar P, et al. Hexokinase-mitochondria interaction mediated by Akt is required toinhibit apoptosis in the presence or absence of Bax andBak. Mol Cell 2004;16:819–30.48. Carre M, Andre N, Carles G, et al. Tubulin is aninherent component of mitochondrial membranes thatinteracts with the voltage-dependent anion channel. JBiol Chem 2002;277:33664–9.49. Sugiyama T, Shimizu S, Matsuoka Y, Yoneda Y,Tsujimoto Y. Activation of mitochondrial voltage-dependent anion channel by a proapoptotic BH3-onlyprotein Bim. Oncogene 2002;21:4944–56.50. Puthalakath H, Huang DC, O’Reilly LA, King SM,Strasser A. The proapoptotic activity of the Bcl-2 familymember Bim is regulated by interaction with the dyneinmotor complex. Mol Cell 1999;3:287–96.

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:2306-2316. Cancer Res   Alexandre Patenaude, Réna G. Deschesnes, Jean L.C. Rousseau, et al.   Hypoxiaagainst Cancer Cells Resistant to Chemotherapeutics and New Soft Alkylating Agents with Enhanced Cytotoxicity

  Updated version

  http://cancerres.aacrjournals.org/content/67/5/2306

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2007/03/02/67.5.2306.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/5/2306.full#ref-list-1

This article cites 47 articles, 14 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/5/2306.full#related-urls

This article has been cited by 3 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/67/5/2306To request permission to re-use all or part of this article, use this link

Research. on May 28, 2018. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from