10
Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent, 1,5 Carole Nicco, 1 Christiane Che ´reau, 1 Claire Goulvestre, 1 Je ´ro ˆme Alexandre, 1,3 Arnaud Alves, 4 Eva Le ´vy, 1 Francois Goldwasser, 3 Yves Panis, 4 Olivier Soubrane, 2 Bernard Weill, 1 and Fre ´de ´ric Batteux 1 1 Laboratoire d’Immunologie and 2 Laboratoire de Recherche Chirurgicale, Unite´ Propre de Recherche de l’Enseignement Supe´rieur 1833, Faculte´ Cochin, Universite´ Paris V; 3 Service d’Oncologie Me ´dicale, Ho ˆpital Cochin; 4 Service de Chirurgie Digestive, Centre Hospitalier Universitaire Lariboisie`re, Universite´ Paris VII, Paris, France; and 5 Service de Chirurgie Digestive, Centre Hospitalier Universitaire Henri-Mondor, Universite´ Paris XII, Cre ´teil, France Abstract Paradoxically, reactive oxygen species (ROS) can promote normal cellular proliferation and carcinogenesis, and can also induce apoptosis of tumor cells. In this report, we study the contribution of ROS to various cellular signals depending on the nature and the level of ROS produced. In nontransformed NIH 3T3 cells, ROS are at low levels and originate from NADPH oxidase. Hydrogen peroxide (H 2 O 2 ), controlled by the gluta- thione system, is pivotal for the modulation of normal cell proliferation. In CT26 (colon) and Hepa 1-6 (liver) tumor cells, high levels of ROS, close to the threshold of cytotoxicity, are produced by mitochondria and H 2 O 2 is controlled by catalase. N -acetylcysteine, which decreases H 2 O 2 levels, inhibits mito- gen-activated protein kinase and normal cell proliferation but increases tumor cell proliferation as H 2 O 2 concentration drops from the toxicity threshold. In contrast, antioxidant molecules, such as mimics of superoxide dismutase (SOD), increase H 2 O 2 levels through superoxide anion dismutation, as well as in vitro proliferation of normal cells, but kill tumor cells. CT26 tumors were implanted in mice and treated by oxaliplatin in association with one of the three SOD mimics manganese(III)- tetrakis(4-benzoic acid) porphyrin, copper(II)(3,5-diisopropyl- salicylate)2, or manganese dipyridoxyl diphosphate. After 1 month, the volumes of tumors were respectively 35%, 31%, and 63% smaller than with oxaliplatin alone (P < 0.001). Similar data were gained with Hepa 1-6 tumors. In conclusion, antioxidant molecules may have opposite effects on tumor growth. SOD mimics can act in synergy with cytotoxic drugs to treat colon and liver cancers. (Cancer Res 2005; 65(3): 948-56) Introduction Reactive oxygen species (ROS) are natural by-products of aerobic metabolism and their production correlates with normal cell proliferation through activation of growth-related signaling path- ways (1). Indeed, exposure to low levels of ROS can increase growth of many types of mammalian cells, whereas scavengers of ROS suppress normal cell proliferation in human and rodent fibroblasts (2–4). Furthermore, growth factors trigger hydrogen peroxide (H 2 O 2 ) production that leads to mitogen-activated protein kinase activation and DNA synthesis, a phenomenon inhibited by antioxidant molecules (5, 6). Several observations suggest that ROS also participate in carcinogenesis. First, ROS production is increased in cancer cells and an oxidative stress can induce DNA damages that lead to genomic instability and possibly stimulate cancer progression (7). Second, elevated ROS levels are responsible for constant activation of transcription factors, such as nuclear factor nB and activator protein 1, during tumor progression (8). Finally, the transforming capacity of ROS is illustrated by the oncogenic transformation of NIH 3T3 cells by the NADPH oxidase homologue MOX-1, which induces the production of superoxide anions (O 2 j - ; refs. 9, 10). Whereas, under certain circumstances, ROS promote cell proliferation, they can also induce apoptosis. Indeed, most anticancer drugs kill their target cells at least in part through the generation of elevated amounts of intracellular ROS. ROS can stimulate proapoptotic signal molecules, such as apoptosis signal regulating kinase 1, c-Jun-NH 2 -kinase, and p38 (11, 12); activate the p53 protein pathway; or engage the mitochondrial apoptotic cascade (13). The various ROS can exert different effects according to their nature and to their intracellular level, which is determined by both their production rate and the activity of antioxidant enzymes. Using pharmacologic modulators of the ROS pathways, we first investigated the source and nature of ROS produced in nontransformed cells and in established tumor cell lines and then determined their respective contribution to various cellular signals. Finally, we investigated the consequences of ROS modulation in association with cytolytic drugs in vitro and in vivo in mice with implanted tumors. Materials and Methods Animals. BALB/c female mice (for CT-26 tumors) or C57/BL6 female mice (for Hepa 1-6 tumors) between 6 and 8 weeks of age were used in all experiments (Iffa Credo, L’Arbresles, France). Animals received humane care in compliance with institutional guidelines. Chemicals and Cell Lines. All chemicals were from Sigma (Saint Quentin Fallavier, France) except for mangafodipir (Teslascan, Amersham Health, Amersham, United Kingdom), oxaliplatin (Eloxatine, Sanofi-Pharma, Paris, France), and paclitaxel (Taxol, Bristol-Myers Squibb, Rueil Malmaison, France). CT26 (mouse colon carcinoma), Hepa 1-6 (mouse liver hepatoma), and NIH 3T3 (mouse fibroblast) were obtained from American Type Culture Collection (Manassas, VA). Cells were cultured in DMEM/Glutamax-I supplemented with 10% heat-inactivated FCS and antibiotics (Life Technologies, Cergy Pontoise, France). All cell lines were routinely tested to rule out Mycoplasma infection of cells. Cellular Production of O 2 j - and H 2 O 2 . Cells (2 10 4 per well) were seeded in 96-well plates (Costar, Corning, Inc., Corning, NY) and incubated Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Fre ´de ´ric Batteux, Laboratoire d’Immunologie, Pavillon Hardy, Ho ˆpital Cochin, 75679 Paris cedex 14, France. Phone: 33-1-58-41-20-07; Fax: 33- 1-58-41-20-08; E-mail: [email protected]. I2005 American Association for Cancer Research. Cancer Res 2005; 65: (3). February 1, 2005 948 www.aacrjournals.org Research Article Research. on June 12, 2020. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

Controlling Tumor Growth by Modulating Endogenous

Production of Reactive Oxygen Species

Alexis Laurent,1,5Carole Nicco,

1Christiane Chereau,

1Claire Goulvestre,

1Jerome Alexandre,

1,3

Arnaud Alves,4Eva Levy,

1Francois Goldwasser,

3Yves Panis,

4Olivier Soubrane,

2

Bernard Weill,1and Frederic Batteux

1

1Laboratoire d’Immunologie and 2Laboratoire de Recherche Chirurgicale, Unite Propre de Recherche de l’Enseignement Superieur 1833,Faculte Cochin, Universite Paris V; 3Service d’Oncologie Medicale, Hopital Cochin; 4Service de Chirurgie Digestive, Centre HospitalierUniversitaire Lariboisiere, Universite Paris VII, Paris, France; and 5Service de Chirurgie Digestive, Centre Hospitalier UniversitaireHenri-Mondor, Universite Paris XII, Creteil, France

Abstract

Paradoxically, reactive oxygen species (ROS) can promotenormal cellular proliferation and carcinogenesis, and can alsoinduce apoptosis of tumor cells. In this report, we study thecontribution of ROS to various cellular signals depending onthe nature and the level of ROS produced. In nontransformedNIH 3T3 cells, ROS are at low levels and originate from NADPHoxidase. Hydrogen peroxide (H2O2), controlled by the gluta-thione system, is pivotal for the modulation of normal cellproliferation. In CT26 (colon) and Hepa 1-6 (liver) tumor cells,high levels of ROS, close to the threshold of cytotoxicity, areproduced by mitochondria and H2O2 is controlled by catalase.N-acetylcysteine, which decreases H2O2 levels, inhibits mito-gen-activated protein kinase and normal cell proliferation butincreases tumor cell proliferation as H2O2 concentration dropsfrom the toxicity threshold. In contrast, antioxidant molecules,such as mimics of superoxide dismutase (SOD), increase H2O2

levels through superoxide anion dismutation, as well as in vitroproliferation of normal cells, but kill tumor cells. CT26 tumorswere implanted in mice and treated by oxaliplatin inassociation with one of the three SOD mimics manganese(III)-tetrakis(4-benzoic acid) porphyrin, copper(II)(3,5-diisopropyl-salicylate)2, or manganese dipyridoxyl diphosphate. After1 month, the volumes of tumors were respectively 35%, 31%,and 63% smaller than with oxaliplatin alone (P < 0.001).Similar data were gained with Hepa 1-6 tumors. In conclusion,antioxidant molecules may have opposite effects on tumorgrowth. SOD mimics can act in synergy with cytotoxic drugs totreat colon and liver cancers. (Cancer Res 2005; 65(3): 948-56)

Introduction

Reactive oxygen species (ROS) are natural by-products of aerobicmetabolism and their production correlates with normal cellproliferation through activation of growth-related signaling path-ways (1). Indeed, exposure to low levels of ROS can increase growthof many types of mammalian cells, whereas scavengers of ROSsuppress normal cell proliferation in human and rodent fibroblasts(2–4). Furthermore, growth factors trigger hydrogen peroxide(H2O2) production that leads to mitogen-activated protein kinase

activation and DNA synthesis, a phenomenon inhibited byantioxidant molecules (5, 6). Several observations suggest thatROS also participate in carcinogenesis. First, ROS production isincreased in cancer cells and an oxidative stress can induce DNAdamages that lead to genomic instability and possibly stimulatecancer progression (7). Second, elevated ROS levels are responsiblefor constant activation of transcription factors, such as nuclearfactor nB and activator protein 1, during tumor progression (8).Finally, the transforming capacity of ROS is illustrated by theoncogenic transformation of NIH 3T3 cells by the NADPH oxidasehomologue MOX-1, which induces the production of superoxideanions (O2j

-; refs. 9, 10).Whereas, under certain circumstances, ROS promote cell

proliferation, they can also induce apoptosis. Indeed, mostanticancer drugs kill their target cells at least in part through thegeneration of elevated amounts of intracellular ROS. ROS canstimulate proapoptotic signal molecules, such as apoptosis signalregulating kinase 1, c-Jun-NH2-kinase, and p38 (11, 12); activatethe p53 protein pathway; or engage the mitochondrial apoptoticcascade (13). The various ROS can exert different effects accordingto their nature and to their intracellular level, which is determinedby both their production rate and the activity of antioxidantenzymes. Using pharmacologic modulators of the ROS pathways,we first investigated the source and nature of ROS produced innontransformed cells and in established tumor cell lines and thendetermined their respective contribution to various cellular signals.Finally, we investigated the consequences of ROS modulation inassociation with cytolytic drugs in vitro and in vivo in mice withimplanted tumors.

Materials and Methods

Animals. BALB/c female mice ( for CT-26 tumors) or C57/BL6 female

mice ( for Hepa 1-6 tumors) between 6 and 8 weeks of age were used in all

experiments (Iffa Credo, L’Arbresles, France). Animals received humane carein compliance with institutional guidelines.

Chemicals and Cell Lines. All chemicals were from Sigma (Saint

Quentin Fallavier, France) except for mangafodipir (Teslascan, Amersham

Health, Amersham, United Kingdom), oxaliplatin (Eloxatine, Sanofi-Pharma,

Paris, France), and paclitaxel (Taxol, Bristol-Myers Squibb, Rueil Malmaison,

France). CT26 (mouse colon carcinoma), Hepa 1-6 (mouse liver hepatoma),

and NIH 3T3 (mouse fibroblast) were obtained from American Type Culture

Collection (Manassas, VA). Cells were cultured in DMEM/Glutamax-I

supplemented with 10% heat-inactivated FCS and antibiotics (Life

Technologies, Cergy Pontoise, France). All cell lines were routinely tested

to rule out Mycoplasma infection of cells.

Cellular Production of O2j- and H2O2. Cells (2 � 104 per well) were

seeded in 96-well plates (Costar, Corning, Inc., Corning, NY) and incubated

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).

Requests for reprints: Frederic Batteux, Laboratoire d’Immunologie, PavillonHardy, Hopital Cochin, 75679 Paris cedex 14, France. Phone: 33-1-58-41-20-07; Fax: 33-1-58-41-20-08; E-mail: [email protected].

I2005 American Association for Cancer Research.

Cancer Res 2005; 65: (3). February 1, 2005 948 www.aacrjournals.org

Research Article

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

for 48 hours with various concentrations of copper(II)(3,5-diisopropylsali-cylate)2 (CuDIPS), manganese(III)tetrakis(4-benzoic acid) porphyrin

(MnTBAP), N-acetyl-L-cysteine (NAC), reduced glutathione (GSH), amino-

triazol (ATZ), buthionine sulfoximine (BSO), catalase, or culture medium

alone. Cellular production of O2j- was evaluated with the use of nitroblue

tetrazolium reduction technique (14). The level of intracellular H2O2 was

assessed spectrofluorimetrically (Victor2, Perkin-Elmer, Paris, France) by

oxidation of 2V,7V-dichlorodihydrofluorescein diacetate (Molecular Probes,

Leiden, the Netherlands). O2j- and H2O2 productions were reported to the

amount of proteins in each sample (bovine serum albumin microbiuret

assay, Pierce, Bezons, France).

Determination of Enzymatic Activities. The SOD activities of tumor or

normal cells and tissues were evaluated with the use of nitrobluetetrazolium reduction technique as previously described by Beauchamp

and Fridovich (14). The catalase activities of tumor or normal cells and

tissues were determined at 25jC by UV spectroscopy at 240 nm accordingto Aebi (15). Levels of GSH were measured by the method of Baker et al.

(16). Antioxidant enzyme activities and GSH content of cells and tissues

were reported to the amount of proteins in each sample.

In vitro Cell Proliferation and Viability Assays. Cells (2 � 104 per well)were seeded in 96-well plates (Costar) and incubated for 48 hours with

various amounts of pharmacologic modulators of antioxidant enzymes asindicated in the figure captions. Cell proliferation was determined by

pulsing the cells with [3H]thymidine (1 Ci/well) during the last 16 hours of

culture. Cell viability was evaluated by the reduction of 3-(4,5-dimethylth-

iazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan. The absorbance at550 nm was recorded in each well with the use of an ELISA microplate

reader. Results are expressed as % viable cells F SE versus cells in culture

medium alone (100% viability). Cell viability was controlled in all cases by

the crystal violet assay.In vitro Cytostatic and Cytotoxic Effects of Exogenous H2O2. Cells

(5 � 104 cells/well) were seeded as above in culture medium supplemented

or not with 400 mol/L NAC, or 400 mol/L ATZ (a catalase inhibitor) or 400

mol/L BSO, which depletes GSH. H2O2 oxidative stress was elicited by theaddition of serial dilutions of H2O2 (Sigma) to the cells for 48 hours. The

cytostatic and the cytotoxic effects of H2O2 were evaluated by thymidine

incorporation and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-mide reduction.

Measurement of O2j- Production by Flow Cytometry. Cells were

treated with either 10 Amol/L rotenone (an inhibitor of mitochondrial

complex I), 10 Amol/L antimycin (an inhibitor of mitochondrial complexIII), 10 Amol/L diphenyleneiodonium (an inhibitor of NADPH oxidase), or

Figure 1. Production of O2j- and H2O2 in nontransformed and tumor cells. Dotted line, basal level of H2O2. Note the change of scale for NIH 3T3. O2j

- andH2O2 productions were reported to the amount of proteins in each sample and expressed as means F SE pmol/min/Ag for O2j

- and arbitrary units/Ag for H2O2

(A and B ). Antioxidant enzyme activity and glutathione content of tumor and nontransformed cells and tissues were measured and reported to the amount of proteinsin each sample (C). Data from at least three independent experiments have been pooled. Statistical significance: *, versus culture medium alone (basal level); c,versus NIH 3T3 cells.

ROS Control Cancer Growth

www.aacrjournals.org 949 Cancer Res 2005; 65: (3). February 1, 2005

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

10 Amol/L allopurinol (an inhibitor of xanthine oxidase) for 30 minutes in

culture medium. Cells were washed in HBSS and resuspended in HBSS

containing 10 Amol/L DHE (Molecular Probes) at 37jC for 30 minutes. Aftertwo washes, cells were resuspended in 500 AL HBSS. Cells (20,000 events per

sample) were analyzed by flow cytometry (FACScalibur, Becton Dickinson,

Mountain View, CA).

Immunoblotting of Cell Lysates. Cells were treated or not with 400

Amol/L NAC for 24 hours then lysed in ice-cold 10 mmol/L Tris buffer, pH 7.5,

with protease inhibitors, 25 mmol/L NaF, 0.5 mmol/L sodium orthovanadate,

and 1% Triton X-100. Forty micrograms of cell lysate were analyzed by

immunoblotting after 10% SDS-PAGEwith the use of anti–extracellular signal-

regulated kinase 2 monoclonal antibody from Santa Cruz (Le Perray en

Yvelines, France), and anti p-ERK monoclonal antibody from New England

Biolabs (Saint Quentin en Yvelines, France).

DNA Strand Break Analysis. The pcDNA3.1 plasmid DNA (Invitrogen)

was incubated with oxaliplatin at a molar ratio of 0.50 in a final volume of

50 AL. MnTBAP (5 Amol/L), CuDIPS (5 Amol/L), MnDPDP (5 Amol/L), or

NAC (5 mmol/L) was added into the mixture. O2j- was generated with the

use of 200 Amol/L xanthine (Sigma) and 1 unit of xanthine oxidase (Sigma) in

the dark at 37jC for 24 hours. Then, 10 AL aliquots were electrophoresed on

a 0.8% agarose gel and revealed by ethidium bromide staining. The gels were

analyzed with the use of a scanner densitometer (Vilber Lourmat, Marnes-la-

Vallee, France).

In vivo Antitumor Activity of Antioxidant Treatments. CT-26 or Hepa

1-6 tumor cells (2 � 106) were injected s.c. into the back of the neck of

BALB/c or C57/BL6 mice, respectively. When the tumors reached a mean

size of 200 to 500 mm3, the animals received a single injection of either 20

mg/kg oxaliplatin (Eloxatine) or of vehicle alone. Mice were then injected

or not with 10 mg/kg MnDPDP, 10 mg/kg MnTBAP, or 10 mg/kg CuDIPS or

with 150 mg/kg NAC i.p. (three injections weekly at the same dosages for 1

month). Tumor size was measured with a vernier caliper every 3 days.

Tumor volume was calculated as follows: TV (mm3) = (L �W2)/2, where L

is the longest and W the shortest radius of the tumor in millimeters. Results

are expressed as means of tumor volumes F SE (n = 15 in each group). In

five mice implanted with CT26 cells, biopsies have been done every 15 days

for 45 days. Tumor cells were then compared in vitro with cells from the

Figure 2. Measurement of intracellulargeneration of O2j

- by flow cytometry.Cells were treated with either rotenone,antimycin, diphenylene indonium, orallopurinol for 30 minutes in culturemedium. Generation of O2j

- was assayedby dihydroethium staining. Data wereconfirmed in two other independentexperiments.

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 950 www.aacrjournals.org

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

original CT26 line in terms of ROS production, proliferation rate,

intracellular GSH content, and sensitivity to oxaliplatin.

Statistical Analysis. The statistical significance of differences betweenexperimental treated groups and untreated controls was analyzed by m2

test for incidence data and by Student’s t test for comparison of means. Alevel of P < 0.05 was accepted as significant. * or c: P < 0.05; ** or cc: P <0.02; *** or ccc: P < 0.01; **** or cccc: P < 0.001 versus controls.

ResultsLevels of ROS and of Antioxidant Enzymes in Normal and

Tumor Cells. Both CT-26 and Hepa 1-6 cell lines produced higheramounts of O2j

- and H2O2 than NIH 3T3 fibroblasts (P < 0.001; Fig.1A and B). SOD activity was 3-fold lower in CT26 and Hepa 1-6, andcatalase activity 2-fold (CT26) and 3-fold (Hepa 1-6) lower than inNIH 3T3. The level of GSH was 2- to 3-fold higher in NIH 3T3 cellsthan in Hepa 1-6 and CT26 cells, respectively (Fig. 1C). Differences interms of antioxidant enzyme concentrations between tumor andnontransformed cells were still greater whenHepa 1-6 and CT-26 celllines were compared with normal liver (SOD, 94.0 F 4.2 units/mg;catalase, 99.1F 12.2 units/mg; GSH, 6.5F 0.2 nmol/mg) and normal

colon (SOD, 44.0F 4.4 units/mg; catalase, 43.3F 2.0 units/mg; GSH,2.4 F 0.1 nmol/mg).Contribution of the Various Antioxidant Pathways to ROS

Production. MnTBAP and CuDIPS, mimics of MnSOD (17) and ofCu/ZnSOD (18), respectively, decreased O2j

- production in all thecell types tested (P < 0.001); however, they increased intracellularH2O2 (P < 0.001). This effect resulted from the dismutation of O2j

-

into H2O2 that accumulated because of the weak catalase-likeactivity of the SOD mimics (Fig. 1A and B).H2O2 production decreased in a dose-dependentmanner in all cell

types incubated with the thiol-related compound NAC that displays

a catalase-like and a glutathione reductase–like activity (Fig. 1B ;

ref. 19). The concentration of H2O2 decreased in NIH 3T3 cells

treated by GSH and increased in NIH 3T3 cells treated with BSO, an

agent that depletes the intracellular pool of GSH (Fig. 1B). By

contrast, inhibition of catalase by ATZ increased H2O2 production in

both tumor cell lines. ATZ had a milder effect (50% increase) in NIH

3T3 cells. Adding exogenous catalase decreased H2O2 level by 37% in

CT26 and 33% in Hepa 1-6 tumor cells but had no effect in

nontransformed cells (Fig. 1B).

Figure 3. Effects of exogenous H2O2 onthe proliferation and the viability ofnontransformed and tumor cells. Cellproliferation was expressed as %cpmF SE. Viability was expressed as percentF SE versus cells in culture medium alone(100% viability). Data from at least threeindependent experiments have beenpooled. Differences between untreatedand antioxidant-treated cells werealways significant with at least threeconcentrations of H2O2 except forBSO-treated CT26, BSO-treatedHepa 1-6 tumor cells, and ATZ-treatednontransformed NIH 3T3 cells (notsignificant) (A ). Effects of antioxidantmolecules on the in vitro proliferative rateof nontransformed and tumor cells.Cell proliferation expressedas %cpm F SE versus (dotted line ) cellsin culture with medium alone. Data from atleast three independent experiments havebeen pooled. Statistical significance isindicated for the highest concentrationsof antioxidant molecules (B). Inhibitionof mitogen-activated protein kinaseexpression by NAC. Cells were treated (+)or not (�) with 400 Amol/L NAC for 24hours. In the presence of NAC, pERK,the phosphorylated form of ERK,is decreased (C). Model proposed toexplain the opposite effects of variousconcentrations of intracellular H2O2 oncellular proliferation of nontransformedand tumor cells. In normal cells, the basallevel of H2O2 is low and its increase is firstassociated with cell growth. In tumor cells,the high level of H2O2 is associated withrapid cell growth. However, any furtherincrease in intracellular H2O2 inhibitstumor cell proliferation, whereas anydecrease in intracellular ROS increasestumor cell growth (D ).

ROS Control Cancer Growth

www.aacrjournals.org 951 Cancer Res 2005; 65: (3). February 1, 2005

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

Origin of ROS in Normal and Tumor Cells. Mitochondria arethe main source of ROS in nonphagocytic cells (20) but othercytosolic enzymatic systems, such as NADPH oxidase (9) orxanthine oxidase (21), can generate O2j

-. Rotenone andantimycin increased the production of O2j

- in CT26 or Hepa1-6 tumor cells but not in NIH 3T3 cells (Fig. 2). By contrast,neither the NADPH-dependent oxidase inhibitor diphenyleneio-donium nor the xanthine oxidase inhibitor allopurinol couldinhibit the production of O2j

- by tumor cells, whereasdiphenyleneiodonium inhibited ROS production in NIH 3T3fibroblasts (Fig. 2).ROS Level Controls the Proliferative Rate of Normal and

Tumor Cells. Treating nontransformed NIH 3T3 fibroblasts withlow amounts of exogenous H2O2 increased their proliferative rate(P < 0.01 versus untreated cells; Fig. 3A). In contrast, incubationwith increasing amounts of exogenous H2O2 or with molecules thatincrease intracellular H2O2 production, such as BSO or SOD

mimics, resulted in growth arrest and cell death (Fig. 3A).Inhibition of the basal production of intracellular H2O2 by NAC,GSH, and, to a lesser extent, catalase, also decreased theproliferative rate (Fig. 3B). This phenomenon was associated withthe inhibition of the p42/p44 mitogen-activated protein kinasepathway by NAC in NIH 3T3 cells (Fig. 3C).Adding increasing amounts of exogenous H2O2 to CT26 or

Hepa 1-6 tumor cells in culture led to a dose-dependentdecrease in proliferation and to cell death (Fig. 3A). The cy-tostatic and cytotoxic effects of H2O2 were amplified by thecatalase inhibitor ATZ but not by BSO. In addition, theincubation of tumor cells with SOD mimics consistentlyincreased intracellular H2O2 concentration through the dismuta-tion of O2j

- and abrogated cell proliferation (Fig. 3B). Bycontrast, NAC and catalase, the two most potent inhibitors ofH2O2 production, significantly increased the proliferation rates ofHepa 1-6 and CT26 tumor cells. Among the other antioxidant

Figure 4. Production of O2j- and H2O2,

and intracellular levels of GSH in CT26tumor cells exposed to increasing amountsof oxaliplatin. Differences betweenoxaliplatin-treated and untreated cells werealways significant (P < 0.01). MnTBAP andCuDIPS significantly decreased O2j

-

production (P < 0.001). Antioxidanttreatment always affected significantly theproduction of H2O2 versus untreated cells(P < 0.01). Differences in GSH levelsbetween antioxidant-treated and untreatedcells were always significant (P < 0.01)except for ATZ and chloramphenicolacetyltransferase. Similar data wereobserved with Hepa 1-6 cells (not shown)(A). Effects of antioxidant treatment on thecytostatic properties of oxaliplatin. CT26cells were cultured as above with oxaliplatinalone or in association with CuDIPS orMnTBAP at dosages of 0 Amol/L (D0), 100Amol/L (D1), 50 Amol/L (D2), and 25 Amol/L(D3); or of NAC, GSH, ATZ, and BSO at thedosages of 0 Amol/L (D0), 400 Amol/L (D1),200 Amol/L (D2), and 100 Amol/L (D3); or ofcatalase at dosages of 0 units (D0), 100units (D1), 50 units (D2), and 25 units (D3).Cell proliferation was determined bythymidine incorporation. Data from atleast three independent experiments havebeen pooled. At 5 Amol/L oxaliplatin,differences between antioxidant-treated(D1) and untreated (D0) cells were alwayssignificant (P < 0.001) except for ATZand chloramphenicol acetyltransferase.Similar data were observed with Hepa 1-6cells (not shown) (B).

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 952 www.aacrjournals.org

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

molecules tested, mannitol and, to a lesser extent, seleno-L-methionine also stimulated the growth of tumor cell lines(Fig. 3B). Pro-proliferative effects of H2O2 were also observed onnontransformed human umbilical vein endothelial cells, andantiproliferative effects on A549 human lung carcinoma cell line(Supplementary Figs. 1 and 2) and on tumor cell lines withvarious mutations of the p53 gene (Supplementary Fig. 3).The Effects of Anticancer Drugs Are Modulated by H2O2. In

tumor cells cultured with various concentrations of the platinumantitumoral compound oxaliplatin (22), a dose-dependent increasein ROS production associated with a decrease in proliferation wasobserved upon addition of SOD mimics and BSO. Opposite effectswere induced by NAC and GSH (Fig. 4). The catalase pathway wasnot involved in the antitumoral activity of oxaliplatin. Theinhibition of the cytostatic and cytotoxic activities of oxaliplatinby NAC and their increase by SOD mimics was further investigatedwith the use of other anticancer molecules known to augmentintracellular levels of H2O2 in tumor cells, such as paclitaxel (23)and 5-fluorouracil (5-FU; ref. 24). Incubating tumor cells with bothdrugs in association with increasing concentrations of NACresulted in a dose-dependent decrease in the cytostatic andcytotoxic effects of paclitaxel or 5-FU on tumor cells versusincubation with paclitaxel or 5-FU alone (Fig. 5A). Reciprocal

effects of SOD mimics MnDPDP, MnTBAP, and CuDIPS wereobserved.H2O2 Is Responsible for DNA Damages. Oxaliplatin alters

DNA structure as shown by the loss of the supercoiled form( form I DNA) and by the increase in the nicked circular form( form II DNA). H2O2 generated by oxaliplatin in association withthat produced by metalloporphyrin SOD mimics through O2j

-

dismutation can increase the alteration of DNA structure aspreviously described (25). Indeed, the damages caused to DNAby oxaliplatin in vitro were increased by the addition of SODmimics, whereas the association of NAC and oxaliplatin wasantagonistic (Fig. 5B).ROS Modulate Antitumoral Activity of Cytotoxic Drugs

In vivo . When MnTBAP, CuDIPS, or MnDPDP (another SODmimic;ref. 26) was repeatedly injected into BALB/c mice with CT26tumors, the volumes of tumors were respectively 59% (P < 0.01), 28%(not significant), and 54% (P < 0.01) smaller than in untreatedcontrols after 1 month (Fig. 6A). In animals treated by theassociation of oxliplatin and either MnTBAP, CuDIPS, or MnDPDP,the volumes of tumors were respectively 35%, 31%, and 63% smallerafter 1 month than with oxaliplatin alone (P < 0.001). Injecting NACalone resulted in 44% increase in tumor volumes after 1 monthcompared with untreated mice (P < 0.01). NAC infusion into

Figure 5. Effects of antioxidantmolecules on the cytostatic and cytotoxicproperties of oxaliplatin, paclitaxel, and5-FU. CT26 cells or Hepa 1-6 cells wereincubated for 48 hours in culture mediumwith either 10 Amol/L oxaliplatin, 10 Amol/LPaclitaxel, or 10 Amol/L 5-FU alone or inthe presence of CuDIPS or MnTBAPat the dosages of 100 Amol/L (D1), 50Amol/L (D2), 25 Amol/L (D3), and 12.5Amol/L (D4); or in the presence of NACor MnDPDP at dosages of 400 Amol/L (D1),200 Amol/L (D2), 100 Amol/L (D3), and 50Amol/L (D4). Data from at least threeindependent experiments havebeen pooled. Differences betweenantioxidant-treated (D1) and untreated (D0)tumor cells were always significantwhatever the drug considered exceptfor NAC-treated CT26 and Hepa 1-6incubated with 5-FU, and except forNAC-treated CT26 incubated withPaclitaxel (viability) (A ). DNA strandbreak analysis. O2j

- anions weregenerated by the xanthine/xanthineoxidase (X/XO ) system. After theincubation time, aliquots were subjected toelectrophoresis and revealed by ethidiumbromide staining. DNA damages resulted inloss of the supercoiled form and in increasein the nicked circular form of DNA. Datawere confirmed in two other independentexperiments (B ).

ROS Control Cancer Growth

www.aacrjournals.org 953 Cancer Res 2005; 65: (3). February 1, 2005

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

oxaliplatin-treated mice completely abrogated the beneficial effectof oxaliplatin because, at 1 month, the volumes of tumors weresimilar to those in the absence of oxaliplatin. The same effects wereobserved in immunocompetent C57/BL6 mice injected with Hepa 1-6 cells (Fig. 6B). In implanted tumors iteratively biopsied for 45 days,time-dependent increases in GSH content (P < 0.01), O2j

- and H2O2

production (P < 0.001 in both cases), and a time-dependent decreasein proliferation rate (P < 0.001) were observed at day 45 versusoriginal cell line.

Discussion

Tumor cells are under a higher oxidative stress than normal cells(2, 27). In this report, we first show that tumor cells differ fromnontransformed cells not only in the level but also in the origin ofROS produced and in the pathways involved in their control.In nontransformed cells, we observed that ROS essentially

originate from the cytosolic NADPH and are controlled by the GSHsystem. Our data are in agreement with the previous observationthat Mox-1, a homologue of the gp91 phox, the catalytic moiety ofthe NADPH oxidase, increases O2j

- and H2O2 generation in NIH3T3 cells, the latter species being responsible for an increasedmitotic rate, cell transformation, and tumorigenicity (9, 10).Similarly, as previously described (28), treating nontransformedfibroblasts or human umbilical vein endothelial cells with lowamounts of exogenous H2O2 increased the proliferative rate of NIH3T3 cells. The intracellular targets of H2O2 involved in cell growthare multiple. They include p42/p44 and p38 mitogen-activatedprotein kinase, p70S6K, signal transducers and activators oftranscription, Atk/protein kinase B and phospholipase D signalingpathways, as well as direct inhibition of protein tyrosinephosphatase-1B (1, 8). The effects of ROS on the growth ofnontransformed NIH 3T3 cells are finely regulated becausedetoxification of intracellular H2O2 by NAC decreased theproliferative rate. This already observed phenomenon is relatedto the suppression of cell cycle progression into G1 phase byinhibition of the mitogen-activated protein kinase pathway (29).The situation is quite different in established tumor cell lines,

such as CT26 and Hepa 1-6, in which the increased generation ofROS resulted from both an elevated mitochondrial production anda profound decrease in the activity of antioxidant enzymes. Noincrease in the proliferative rates was observed whatever theamounts of exogenous H2O2 added to the cultures. On the contrary,adding increasing amounts of exogenous H2O2 or increasing itsintracellular levels with the use of SOD mimics led to a dose-dependent decrease in proliferation and to cell death. Similar datahave been obtained in A549 human lung carcinoma cell line and incell lines with various p53 gene mutations, suggesting that ROSmodulation induced by SOD mimics or NAC are not linked with aparticular p53 status. Conversely, detoxification of H2O2, especiallythrough the catalase pathway, stimulated tumor cell proliferation.Those data are in line with previous observations that MnSOD orCu/ZnSOD gene transfection inhibits growth of glioma (30) andpancreatic tumor cell lines (31), a phenomenon that can bereverted by cotransfection of the catalase gene (32, 33). Our dataare also in agreement with the report by Hussain et al. (34),showing that p53 is associated with up-regulation of MnSODresulting in ROS overproduction and apoptosis, a phenomenoninhibited by overexpression of catalase.Although H2O2 directly controls tumor cell proliferation, OHj,

which results from the conversion of H2O2 via the Fenton reaction,

is also involved in that control. Indeed, addition of mannitol, acompound known to detoxify OHj (35), stimulated the growth oftumor cell lines. Along with catalase, the glutathione pathwaycould also be involved in the regulation of the intracellular

Figure 6. In vivo antitumor activity of antioxidant molecules. Mice bearing CT26 orHepa 1-6 tumors received a single injection of either oxaliplatin or vehicle alone.Mice were then injected or not with MnDPDP, MnTBAP, or CuDIPS, or with NAC i.p.for 1 month (three injections weekly). Tumor size was measured every 3 days at thetime of injections. Mean of tumor volumes F SE (n = 15 in each group).

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 954 www.aacrjournals.org

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

concentration of H2O2 and tumor cell growth. Indeed, addingseleno-L-methionine, a molecule that stimulates glutathioneperoxidase activity, augmented the proliferative rate of the coloncarcinoma cell lines as previously described (36). However, addingreduced GSH or depleting intracellular GSH with BSO had weakereffects on the proliferation of tumor cells than exogenous catalase,probably because of the indirect effects of those molecules on theintracellular concentration of H2O2.Cytotoxic drugs induce cellular stress responses and the

generation of ROS. However, whether this phenomenon partic-ipates in the antitumoral activity of those drugs has remainedunclear thus far. To address this issue, we have examined thecombined effects of cytotoxic molecules with several modulators ofROS pathways. As already observed with cisplatin (37), oxaliplatinincreased ROS production and decreased CT26 and Hepa 1-6 cellproliferation in a dose-dependent manner. Whereas the GSHsystem was not involved in the control of the basal proliferativerate of tumor cells, it modulated the ROS production and theantitumoral activity of oxaliplatin as shown by the decrease in ROSlevels and the increase in tumor cell proliferation followingaddition of exogenous GSH. Several lines of evidence suggest thatthe antagonistic effects of GSH and oxaliplatin involve intracellularmechanisms and do not result from extracellular interaction ofGSH with oxaliplatin. Indeed, in our study, tumor cells depleted ofGSH by BSO had an increased sensitivity to oxaliplatin. Moreover,when exogenous GSH was added with oxaliplatin to tumor cells inculture, the initial increase in ROS levels was similar to thatobserved when cells were incubated with oxaliplatin alone. Thosedata confirm that platinum-induced oxidative stress involvesoxidation of cellular components and depletion of intracellularGSH (37, 38). The inhibition of the cytostatic and cytotoxicactivities of oxaliplatin by NAC and their increase by SOD mimicswas further investigated with the use of other anticancer moleculesknown to augment intracellular levels of H2O2 in tumor cells.Incubating CT26 and Hepa 1-6 cells with the taxane-relatedcompound paclitaxel (23) or with 5-FU (24), with increasingconcentrations of NAC, resulted in a dose-dependent decrease inthe cytostatic and cytotoxic effects of paclitaxel and 5-FU on tumorcells versus incubation with individual drugs alone. Reciprocaleffects were observed with SOD mimics MnTBAP and CuDIPS thatgenerate H2O2 through O2j

- dismutation.

DNA is the main target of platinum-related antitumor drugs.H2O2 generated by those drugs in association with that producedby metalloporphyrin SOD mimics can alter DNA structure (25).Indeed, the damages caused to DNA by oxaliplatin in vitro wereincreased by the addition of SOD mimics, whereas the associationof NAC and oxaliplatin was antagonistic. The same was observedin vivo in murine models of colon cancers. During tumor growth,H2O2 production increased whereas ex vivo proliferation ratesdecreased, and the GSH content augmented whereas an ex vivoresistance to oxaliplatin developed. Consequently, the three SODmimics studied decreased, whereas NAC increased tumor growthwhen given alone in mice with implanted tumors. In addition,when oxaliplatin was associated, the three SOD mimics potenti-ated, whereas NAC abrogated the antitumoral effect of oxaliplatin.This observation should stimulate further clinical investigationsbecause NAC is widely used after surgery as a mucolytic agent inpatients operated for cancers. Our observation is in line with aprevious large human study that showed an increased incidence inlung cancer in heavy smokers treated with antioxidant molecules(39). In conclusion, the growth of normal cells is triggered whenthose cells are submitted to oxidant signals directed to growth-related genes up to a critical threshold beyond which ROS becomecytotoxic. Because in tumor cells, the level of endogenous ROS isclose to that threshold, the simultaneous exposure to ROS-generating agents and to cytotoxic drugs dramatically increasesthe rate of cell death. Those data have clinical implications inhumans. Indeed, antioxidant molecules probably play a protectiverole against cancer in healthy individuals by preventing DNAdamages linked to the oxidative stress. However, once cancer cellshave emerged, a cancer-promoting effect can result from theadministration of agents that decrease intracellular H2O2 levels,whereas an anticancer activity is exerted by agents that tend toincrease that level.

Acknowledgments

Received 5/21/2004; revised 11/2/2004; accepted 11/29/2004.Grant support: French Ministry for Research, Association pour la Recherche sur le

Cancer, National Agency for Valorization of Research (ANVAR), Association Trans-hepate, Association Benoıt Malassagne, and Paris-Biotech.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

References1. Benhar M, Engelberg D, Levitzki A. ROS, stress-activated kinases and stress signaling in cancer. EMBORep 2002;3:420–5.

2. Burdon RH. Superoxide and hydrogen peroxide inrelation to mammalian cell proliferation. Free RadicBiol Med 1995;18:775–94.

3. Murrell GA, Francis MJ, Bromley L. Modulation offibroblast proliferation by oxygen free radicals. BiochemJ 1990;265:659–65.

4. Pluthero FG, Axelrad AA. Superoxide dismutase as aninhibitor of erythroid progenitor cell cycling. Ann N YAcad Sci 1991;628:222–32.

5. Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T.Requirement for generation of H2O2 for platelet-derivedgrowth factor signal transduction. Science 1995;270:296–9.

6. Bae YS, Kang SW, Seo MS, et al. Epidermal growthfactor (EGF)-induced generation of hydrogen peroxide.Role in EGF receptor-mediated tyrosine phosphoryla-tion. J Biol Chem 1997;272:217–21.

7. Jackson AL, Loeb LA. The contribution of endogenoussources of DNA damage to the multiple mutations incancer. Mutat Res 2001;477:7–21.

8. Gupta A, Rosenberger SF, Bowden GT. Increased ROSlevels contribute to elevated transcription factor andMAP kinase activities in malignantly progressed mousekeratinocyte cell lines. Carcinogenesis 1999;20:2063–73.

9. Suh YA, Arnold RS, Lassegue B, et al. Cell transfor-mation by the superoxide-generating oxidase Mox1.Nature 1999;401:79–82.

10. Arnold RS, Shi J, Murad E, et al. Hydrogen peroxidemediates the cell growth and transformation caused bythe mitogenic oxidase Nox1. Proc Natl Acad Sci U S A2001;98:5550–5.

11. Benhar M, Dalyot I, Engelberg D, Levitzki A. EnhancedROS production in oncogenically transformed cellspotentiates c-Jun N-terminal kinase and p38 mitogen-activated protein kinase activation and sensitization togenotoxic stress. Mol Cell Biol 2001;21:6913–26.

12. Tobiume K, Matsuzawa A, Takahashi T, et al. ASK1 isrequired for sustained activations of JNK/p38 MAPkinases and apoptosis. EMBO Rep 2001;2:222–8.

13. Jabs T. Reactive oxygen intermediates as mediatorsof programmed cell death in plants and animals.Biochem Pharmacol 1999;57:231–45.

14. Beauchamp C, Fridovich I. Superoxide dismutase:improved assays and an assay applicable to acrylamidegels. Anal Biochem 1971;44:276–87.

15. Aebi H. Catalase in vitro . Methods Enzymol1984;105:121–6.

16. Baker MA, Cerniglia GJ, Zaman A. Microtiterplate assay for the measurement of glutathioneand glutathione disulfide in large numbers of biologicalsamples. Anal Biochem 1990;190:360–5.

17. Patel M, Day BJ. Metalloporphyrin class oftherapeutic catalytic antioxidants. Trends PharmacolSci 1999;20:359–64.

18. Leuthauser SW, Oberley LW, Oberley TD, SorensonJR, Ramakrishna K. Antitumor effect of a coppercoordination compound with superoxide dismutase-like activity. J Natl Cancer Inst 1981;66:1077–81.

19. Cotgreave IA. N -acetylcysteine: pharmacological con-siderations and experimental and clinical applications.Adv Pharmacol 1997;38:205–27.

ROS Control Cancer Growth

www.aacrjournals.org 955 Cancer Res 2005; 65: (3). February 1, 2005

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

20. Chen Q, Vazquez EJ, Moghaddas S, Hoppel CL,Lesnefsky EJ. Production of reactive oxygen species bymitochondria: central role of complex III. J Biol Chem2003;278:36027–31.

21. Corda S, Laplace C, Vicaut E, Duranteau J. Rapidreactive oxygen species production by mitochondria inendothelial cells exposed to tumor necrosis factor-a ismediated by ceramide. Am J Respir Cell Mol Biol 2001;24:762–8.

22. Baker DE. Oxaliplatin: a new drug for the treatmentof metastatic carcinoma of the colon or rectum. RevGastroenterol Disord 2003;3:31–8.

23. Varbiro G, Veres B, Gallyas F, Sumegi B. Direct effectof Taxol on free radical formation and mitochondrialpermeability transition. Free Radic Biol Med 2001;31:548–58.

24. Hwang PM, Bunz F, Yu J, et al. Ferredoxin reductaseaffects p53-dependent, 5-fluorouracil-induced apoptosisin colorectal cancer cells. Nat Med 2001;7:1111–7.

25. Ohse T, Nagaoka S, Arakawa Y, Kawakami H,Nakamura K. Cell death by reactive oxygen speciesgenerated from water-soluble cationic metalloporphyr-ins as superoxide dismutase mimics. J Inorg Biochem2001;85:201–8.

26. Bedda S, Laurent A, Conti F, et al. Mangafodipirprevents liver injury induced by acetaminophen in themouse. J Hepatol 2003;39:765–72.

27. Szatrowski TP, Nathan CF. Production of largeamounts of hydrogen peroxide by human tumor cells.Cancer Res 1991;51:794–8.

28. Kim BY, Han MJ, Chung AS. Effects of reactive oxygenspecies on proliferation of Chinese hamster lungfibroblast (V79) cells. Free Radic Biol Med 2001;30:686–98.

29. Sekharam M, Trotti A, Cunnick JM, Wu J. Suppres-sion of fibroblast cell cycle progression in G1 phase byN -acetylcysteine. Toxicol Appl Pharmacol 1998;149:210–6.

30. Zhang Y, Zhao W, Zhang HJ, Domann FE, Oberley LW.Overexpression of copper zinc superoxide dismutasesuppresses human glioma cell growth. Cancer Res 2002;62:1205–12.

31. Cullen JJ, Weydert C, Hinkhouse MM, et al. The roleof manganese superoxide dismutase in the growthof pancreatic adenocarcinoma. Cancer Res 2003;63:1297–303.

32. Amstad P, Peskin A, Shah G, et al. The balancebetween Cu,Zn-superoxide dismutase and catalaseaffects the sensitivity of mouse epidermal cells to oxi-dative stress. Biochemistry 1991;30:9305–13.

33. Rodriguez AM, Carrico PM, Mazurkiewicz JE,Melendez JA. Mitochondrial or cytosolic catalasereverses the MnSOD-dependent inhibition of prolifer-ation by enhancing respiratory chain activity, net ATP

production, and decreasing the steady state levels ofH(2)O(2). Free Radic Biol Med 2000;29:801–13.

34. Hussain SP, Amstad P, He P, et al. p53-induced up-regulation of MnSOD and GPx but not catalaseincreases oxidative stress and apoptosis. Cancer Res2004;64:2350–6.

35. Zhang C, Gong Y, Ma H, et al. Reactive oxygenspecies involved in trichosanthin-induced apoptosisof human choriocarcinoma cells. Biochem J 2001;355:653–61.

36. Li S, Yan T, Yang JQ, Oberley TD, Oberley LW. Therole of cellular glutathione peroxidase redox regula-tion in the suppression of tumor cell growth by man-ganese superoxide dismutase. Cancer Res 2000;60:3927–39.

37. Godwin AK, Meister A, O’Dwyer PJ, et al. Highresistance to cisplatin in human ovarian cancer celllines is associated with marked increase of glutathi-one synthesis. Proc Natl Acad Sci U S A 1992;89:3070–4.

38. Godbout JP, Pesavento J, Hartman ME, Manson SR,Freund GG. Methylglyoxal enhances cisplatin-inducedcytotoxicity by activating protein kinase Cy. J BiolChem 2002;277:2554–61.

39. Blot WJ. Vitamin/mineral supplementation andcancer risk: international chemoprevention trials. ProcSoc Exp Biol Med 1997;216:291–6.

Cancer Research

Cancer Res 2005; 65: (3). February 1, 2005 956 www.aacrjournals.org

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Controlling Tumor Growth by Modulating Endogenous ...Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species Alexis Laurent,1,5 Carole Nicco,1 Christiane

2005;65:948-956. Cancer Res   Alexis Laurent, Carole Nicco, Christiane Chéreau, et al.   Production of Reactive Oxygen SpeciesControlling Tumor Growth by Modulating Endogenous

  Updated version

  http://cancerres.aacrjournals.org/content/65/3/948

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2005/02/22/65.3.948.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/3/948.full#ref-list-1

This article cites 37 articles, 14 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/3/948.full#related-urls

This article has been cited by 35 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/65/3/948To request permission to re-use all or part of this article, use this link

Research. on June 12, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from