4
 LETTER  doi:10.1038/nature10167 Selective killing of cancer cells by a small molecule targeting the stress response to ROS Lakshmi Raj 1 , Takao Ide 1 , Aditi U. Gurkar 1 , Michael Foley 2 , Monica Schenone 2 , Xiaoyu Li 2 , Nicola J. Tolliday 2 , Todd R. Golub 2 , Steven A. Carr 2 , Alykhan F. Shamji 2 , Andrew M. Stern 2 , Anna Mandinova 1,2 , Stuart L. Schreiber 2 & Sam W. Lee 1,2 Malignanttransformation,driven by gain-of- functionmutations in oncogenes and loss-of-function mutations in tumour suppressor genes, results in cell deregulation that is frequently associa ted with enhanced cellular stress (for example, oxidative, replicative, meta- boli c and prot eoto xic stre ss,and DNA damag e) 1 . Ada pta ti on to thi s str ess phenoty pe is requ ired for canc er cells to surv ive, and con- seq uent ly cance r cells may beco me depen dent upon non-onco gene s that do not ord inari ly per for m suc h a vit al fun ctionin norma l cel ls. Thus, targeting these non-oncogen e dependencies in the context of a transformed genotype may result in a synthetic lethal interaction and the selective death of cancer cells 2 . Here we used a cell-based small-molecule screening and quantitative proteomics approach that res ulte d in the unbi ased iden tifi cati on of a small mole cule that selectively kills cancer cells but not normal cells. Piperlongumine increases the level of reactive oxygen species (ROS) and apoptotic cel l dea th in bot h cancer cel ls and nor mal cel ls eng ineere d to hav e a cancer genotype, i rrespectiv e of p53 stat us, but it has little effect on either rapidly or slowly dividing primary normal cells. Significant antitumour effects are observed in piperlongumine-tr eated mouse xenograft tumour models, with no apparent toxicity in normal mice. Moreover, piperlongumine potently inhibits the growth of spontaneously formed malignant breast tumours and their asso- ciated metastases in mice. Our results demonstrate the ability of a small molecule to induce apoptosis selectively in cells that have a cancer geno type, by targ eti ng a non- oncogene co-d epen dency acq uir ed thr oug h the exp ressi on of the can cer gen ot ype in res pons e to transfor mation- induced oxidative stress 3–5 . Using a luciferase reporter gene fused with the  CDIP  (cell death involved p53 target, also known as 5730403B10Rik) promoter 6 , we performed a small-molecule screen (Supplementary Fig. 1) to identify compou nds acting through novel pro-apoptotic mechanisms. The compound with the highest composite  Z  value was piperlongumine (Supplementary Fig. 2a), which increased luciferase activity from the reporter gene at levels comparable to the positive control, etoposide (Supplementary Figs 2b and 3). Piperlongumine is a natural product isolated from the plant species  Piper longum L. (Fig. 1a) and it was pre vio uslyshown to hav e cyto toxi c effe cts 7 . Weexaminedtheeffectsof piperlongumine on the viability of cultured cancer cells and normal cells (Fig. 1b and Supplementary Figs 4 and 6). Piperlongumine treat- ment markedly induced cell death in cancer cells with both wild-type p53and mut ant p53.When pri mar y normalcellsandnon -tra nsfo rme d immortalized cells with diverse proliferative capacities were incubated with highly purified piper longu mine (Supplementar y Fig. 5) for 24 h (under the indicat ed condit ions, which avoid spontaneous transfo rma- tion and minimize stress), there was little apparent reduction in cell  viabil ity, even at the h ighes t conce ntration te sted (15 mM, a concentra- tion of piperlongumine that approaches its solubility limit). This indi- cated that piperlo ngumin e may have a cance r-cell -sele ctive killing property, and that sensitivity to piperlongumine may result from the pro cess of mal ign ant trans for mat ion. To test this hyp oth esi s, we use d a define d model 8 of oncog enic conversion of normalcells throu gh ectop ic exp res sio n of the telo mer ase cat aly tic subunit(hTERT ) in combin ation with small T antigen and an oncogenic allele of  HRAS (Fig. 1c), and 1 Cuta neousBiolo gyResearchCenter , Massa chuse ttsGenera l Hosp italandHarvardMedica l Schoo l,Building149 13 th Street,Charlestown, Massachusetts02129, USA. 2 Broa d Inst ituteofHarvardandMIT, 7 Cambridge Center, Massachusetts 02142, USA. 0 5 1 10 15 Piperlongumine ( μM) PUMA p53  PUMA p53 β-actin a b β-actin 100 N O 80 60 20 0    P   e   r   c   e   n    t   a   g   e   c   e    l    l   v    i   a    b    i    l    i    t   y Tu N 40 SW620 DLD-1 BT-474 MDA-MB-231 MDA-MB-435 Panc-1 Mia PaCa-2 U2OS Saos-2 U2OS MCF7 HCT 116 WT-p53 cells p53-null Saos-2 100  80 60 40 20  0 PL: ( μM) PL: ( μM) PL: ( μM) 5 5  5  10 10 10 BJ-hTERT BJ-ST BJ-ELR  A549 H1975 PAE 76N HKC HDF 184B5 MCF 10A EJ HCT 116 E RB B 2 E RBB2 -RAS MCF 10A    V    i   a    b    l   e   c   e    l    l   s    (    %    ) 100  80 60 40 20  0 PL: ( μM)    V    i   a    b    l   e   c   e    l    l   s    (    %    ) 1 2.5 5 10 0 1 2.5 5 10 0 2.5 5 10 0 1 2.5 5 10 1 0 c d O OCH 3 OCH 3 OCH 3 Figure 1 |  Selective killing effect of piperlongumine in cancer cells. a , Structure of piper long umine . b, Piper longu minetreatmen t indu ces celldeath in cancercell s butnot in nor malcell s.Norma l humancells(N),incl udingaorti c endothelial cells (PAE), breast epithelial cells (76N), keratinocytes (HKC) and skin fibroblasts (HDF), as well as two immortalized breast epithelial cell lines (184B5 and MCF 10A), were grown in 12-well or 24-well plates and treated with piperlongumine at 1–15 mM for 24 h. A variety of human cancer cell lines (Tu) were also treated with piperlongumine or DMSO (control) for 24 h. Cytotoxicity was measured by trypan blue exclusion staining (average of three independent experiments). Piperlongumine was HPLC-purified (,99% purity) before the treatment. c, Selective cell death caused by piperlongumine (PL) in oncogenically transformed human BJ skin fibroblasts (left panel) and MCF 10A cell lines (right panel). A representative graph for cell viability is shown (mean 6s.d. of three independent experiments;  *, P ,0.0001). d, The effects of piperlongumine on p53 and its target PUMA were measured by west ern blot analy ses in seve ral cancercell lines . b-actin expre ssion was used as a loading control. 1 4 J U LY 2 0 1 1 | V O L 4 7 5 | N AT U R E | 2 3 1 Macmillan Publishers Limited. All rights reserved ©2011

Nature 10167

Embed Size (px)

Citation preview

Page 1: Nature 10167

5/16/2018 Nature 10167 - slidepdf.com

http://slidepdf.com/reader/full/nature-10167 1/4

LETTERdoi:10.1038/nature1016

Selective killing of cancer cells by a small moleculetargeting the stress response to ROSLakshmi Raj1, Takao Ide1, Aditi U. Gurkar1, Michael Foley2, Monica Schenone2, Xiaoyu Li2, Nicola J. Tolliday2, Todd R. Golub2,Steven A. Carr2, Alykhan F. Shamji2, Andrew M. Stern2, Anna Mandinova1,2, Stuart L. Schreiber2 & Sam W. Lee1,2

Malignanttransformation,driven by gain-of-functionmutations inoncogenes and loss-of-function mutations in tumour suppressor genes, results in cell deregulation that is frequently associated withenhanced cellular stress (for example, oxidative, replicative, meta-bolic and proteotoxic stress,and DNA damage)1. Adaptation to thisstress phenotype is required for cancer cells to survive, and con-sequently cancer cells may become dependent upon non-oncogenesthat do not ordinarily perform such a vital function in normal cells.Thus, targeting these non-oncogene dependencies in the context of 

a transformed genotype may result in a synthetic lethal interactionand the selective death of cancer cells2. Here we used a cell-basedsmall-molecule screening and quantitative proteomics approachthat resulted in the unbiased identification of a small molecule thatselectively kills cancer cells but not normal cells. Piperlongumineincreases the level of reactive oxygen species (ROS) and apoptoticcell death in both cancer cells and normal cells engineered to have a cancer genotype, irrespective of p53 status, but it has little effect oneither rapidly or slowly dividing primary normal cells. Significantantitumour effects are observed in piperlongumine-treated mousexenograft tumour models, with no apparent toxicity in normalmice. Moreover, piperlongumine potently inhibits the growth of spontaneously formed malignant breast tumours and their asso-ciated metastases in mice. Our results demonstrate the ability of a 

small molecule to induce apoptosis selectively in cells that have a cancer genotype, by targeting a non-oncogene co-dependency acquired through the expression of the cancer genotype in responseto transformation-induced oxidative stress3–5.

Using a luciferase reporter gene fused with the CDIP  (cell deathinvolved p53 target, also known as 5730403B10Rik) promoter6, weperformed a small-molecule screen (Supplementary Fig. 1) to identify compounds acting through novel pro-apoptotic mechanisms. Thecompound with the highest composite Z  value was piperlongumine(Supplementary Fig. 2a), which increased luciferase activity from thereporter gene at levels comparable to the positive control, etoposide(Supplementary Figs 2b and 3). Piperlongumine is a natural productisolated from the plant species Piper longum L. (Fig. 1a) and it waspreviouslyshown to have cytotoxic effects7. Weexamined theeffectsof 

piperlongumine on the viability of cultured cancer cells and normalcells (Fig. 1b and Supplementary Figs 4 and 6). Piperlongumine treat-ment markedly induced cell death in cancer cells with both wild-typep53and mutantp53.When primary normalcells and non-transformedimmortalized cells with diverse proliferative capacities were incubatedwith highly purified piperlongumine (Supplementary Fig. 5) for 24 h(under the indicated conditions, which avoid spontaneous transforma-tion and minimize stress), there was little apparent reduction in cell

 viability, even at the highest concentration tested (15mM, a concentra-tion of piperlongumine that approaches its solubility limit). This indi-cated that piperlongumine may have a cancer-cell-selective killing property, and that sensitivity to piperlongumine may result from theprocess of malignant transformation. To test this hypothesis, we used a

defined model8 of oncogenic conversionof normalcells through ectopexpression of the telomerase catalytic subunit (hTERT ) in combinatiowith small T antigen and an oncogenic allele of  HRAS (Fig. 1c), an

1

CutaneousBiologyResearchCenter, MassachusettsGeneral HospitalandHarvardMedical School,Building149 13

th

Street,Charlestown, Massachusetts02129, USA.

2

Broad InstituteofHarvardandM7 Cambridge Center, Massachusetts 02142, USA.

0 51 10 15

Piperlongumine ( μM)

PUMA

p53

PUMA

p53

β-actin

a

b

β-actin

100

N

O

80

60

20

0

   P  e  r  c  e  n   t  a  g  e  c  e   l   l  v   i  a   b   i   l   i   t  y

Tu

N

40SW620DLD-1BT-474MDA-MB-23MDA-MB-4Panc-1Mia PaCa-2U2OSSaos-2

U2OS MCF7 HCT 116

WT-p53 cells p53-null

Saos-2

100

8060

40

20

0PL:

( μM)

PL:

( μM)

PL:

( μM)

– 5 – 5 – 5– 10 – 10 – 10

BJ-hTERT BJ-ST BJ-ELR

 A549H1975

PAE76NHKCHDF

184B5MCF 10A

EJHCT 116

– ERBB2 ERBB2-RA

MCF 10A

   V   i  a   b   l  e  c  e   l   l  s

   (   %   ) 100

80

60

40

20

0PL:

( μM)

   V   i  a   b   l  e  c  e   l   l  s

   (   %   )

1 2.5 5 10 0 1 2.5 5 10 0 2.5 5 10 0 1 2.5 5 1010

c

d

O

OCH3

OCH3

OCH3

Figure 1 | Selective killing effect of piperlongumine in cancer cells.a , Structure of piperlongumine. b, Piperlonguminetreatment induces celldeat

in cancercells butnot in normalcells.Normal humancells(N),includingaortendothelial cells (PAE), breast epithelial cells (76N), keratinocytes (HKC) anskin fibroblasts (HDF), as well as two immortalized breast epithelial cell line(184B5 and MCF 10A), were grown in 12-well or 24-well plates and treatedwith piperlongumine at 1–15 mM for 24 h. A variety of human cancer cell lin(Tu) were also treated with piperlongumine or DMSO (control) for 24 h.Cytotoxicity was measured by trypan blue exclusion staining (average of threindependent experiments). Piperlongumine was HPLC-purified (,99%purity) before the treatment. c, Selective cell death caused by piperlongumin(PL) in oncogenically transformed human BJ skin fibroblasts (left panel) anMCF 10A cell lines (right panel). A representative graph for cell viability isshown (mean6 s.d. of three independent experiments; *, P , 0.0001). d, Theffects of piperlongumine on p53 and its target PUMA were measured by western blot analyses in several cancercell lines. b-actin expression was useda loading control.

1 4 J U L Y 2 0 1 1 | V O L 4 7 5 | N A T U R E | 2 3

Macmillan Publishers Limited. All rights reserved ©2011

Page 2: Nature 10167

5/16/2018 Nature 10167 - slidepdf.com

http://slidepdf.com/reader/full/nature-10167 2/4

observedsensitivity to piperlongumine upononcogenic transformationof normal cells. Similar results were obtained using serial transforma-tion of spontaneously immortalized MCF 10A breast epithelial cells by overexpression of ERBB2 and/or HRAS9 (Fig. 1c).

Western blot analysis showed that wild-type p53 expression wassignificantly enhanced in different types of cancer cells by treatmentwith piperlongumine (Fig. 1d). Moreover, a p53 proapoptotic target,BCL2 binding component 3 (BBC3, also known as PUMA), was sig-

nificantly induced in response to piperlongumine, even in p53-nullSaos-2 cancer cells (Fig. 1d). Piperlongumine treatment was able torepress the expression of severalpro-survival proteins, including B-cellCLL/lymphoma 2 (BCL2), baculoviral IAP repeat containing 5 (alsoknown as survivin) and X-linked inhibitor of apoptosis (XIAP)(Supplementary Fig. 7). Among 55 death- or survival-related genes,we observed increased levels of apoptotic transcripts and decreasedlevels of pro-survival transcripts in cancer cells in the presence of piperlongumine, but no significant changes in these transcripts innormal cells (Supplementary Fig. 8). These results indicate that piper-longumine induces cell death or apoptosis (Supplementary Fig. 4a, e)preferentially in cancer cells by modulating the expression of membersof apoptotic and survival pathways, including p53 targets and p53itself, and that it does not require p53 for this activity.

We next tested piperlongumine in established tumour xenografts inmice (human bladder, breast and lung tumours in nude mice, andmouse melanoma in C57BL/6 mice; Supplementary Fig. 9). Markedantitumour effects were observed in tumour-bearing mice treated withpiperlongumine, as compared to dimethyl sulphoxide (DMSO)-treatedcontrols (Supplementary Fig. 9). Piperlongumine treatment enhancedthe expression of cyclin-dependent kinase inhibitor 1A (CDKN1A, orp21WAF1/CIP), PUMA and caspase3 in EJ-celltumours (Supplementary Fig. 10a). Moreover, piperlongumine treatment inhibited the forma-tionof blood vessels in xenograft-tumour mice(Supplementary Figs9dand 10b). We also studied piperlongumine in a transgenic mousemodel of spontaneous breast cancer, MMTV-PyVT10. When tumoursizes hadgrownto about 5–6 mmin diameter (infemaleMMTV-PyVTmice, 8–9 weeks of age), piperlongumine was administered intraperi-

toneally (2.4 mg kg 

21

) daily for two weeks and notable antitumoureffects were observed (Fig. 2a, b).Furthermore,therewere no secondary tumours in piperlongumine-treated mice compared to vehicle-treatedcontrols. At day 13, the vehicle-treated control mice showed severemalignant progression indicated by the formation of aggressive adeno-carcinoma (Fig. 2c). In contrast, the mammary glands of piperlongu-mine-treated mice were preserved and the tissue showed ahyperplastic-like, non-malignant phenotype (Fig. 2c). Notably, piper-longumine seemed to be more effective in tumour growth inhibitionthan paclitaxel (Fig. 2d). Piperlongumine also showed excellent oralbioavailability and desirable exposure levels (C max and bioavailability,measured by calculating the area under curve (AUC)) in mice, asobserved after a single oral administration and after intravenousinjection (Supplementary Fig. 11). To examine potential cytotoxic

side-effects of piperlongumine on normal tissues, CD-1 mice wereintraperitoneally treated with piperlongumine (2.4 mg kg 21) orDMSO,daily for 6 days,and wholeblood samplesas well asvitalorganswere collected for haematology and histopathological analyses,respectively. Piperlongumine-treated CD-1 mice remained healthy throughout the treatment time and no notable differences betweenthe vehicle-treated and piperlongumine-treated groups were evident(Supplementary Figs 12 and 13). High-dose acute toxicity studiesdemonstrated that piperlongumine did not cause any obvious clinicalindications(SupplementaryFig. 14 andSupplementary Tables1 and2).Together, these results indicate that treatment with piperlonguminepotently suppresses tumour growth in diverse tissues without affecting normal tissues in mice.

We next used a method combining affinity enrichment with stable-isotope labelling with amino acids in cell culture (SILAC) and quanti-tative proteomics to identify the target proteins and their associated

complexes that bind to piperlongumine11 (Supplementary Figs 15 and16a and Supplementary Methods). Twelve interaction partners of

piperlongumine that were similar in both EJ and U2OS cells wereidentified (Supplementary Fig. 16b). Seven of these, including thetop four high-signal outliers, are known to participate in the cellularresponse to oxidative stress caused by elevated ROS. GlutathioneS-transferase pi 1 (GSTP1) was the highest-confidence hit, followedby carbonyl reductase 1 (CBR1) (Supplementary Fig. 16b). Several ofthese proteins are known to be part of a common complex 12,13, indi-cating that the affinity purification may have identified direct andindirect partners.

These results indicate that, by binding to proteins known to regulateoxidative stress, piperlongumine may modulate redox and ROS home-ostasis. Consistent with this hypothesis, we found that piperlonguminecan interact directly with purified recombinant GSTP1 and inhibit itsactivity (Supplementary Figs 17 and 18), and also that it can lead to adecreasein reduced glutathione(GSH) levels and an increase in oxidizedglutathione (GSSG) levels in cancer cells (Fig. 3a). Piperlonguminetreatment did not increase GSSG levels in normal cells (76N(NMEC)) (Fig. 3a). Furthermore, co-treatment with piperlongumineand the reducing agent N -acetyl-L-cysteine (NAC, 3 mM), whichquenches ROS, prevented piperlongumine-mediated GSH depletion(Fig. 3a).

We next determined the effect of piperlongumine on cellular ROSlevels in several human cancer cells (EJ, MDA-MB-231, U2OS andMDA-MB-435) through flow cytometry using the redox-sensitivefluorescent probe 29-,79-dichlorofluorescein diacetate (DCF-DA)Treatment with piperlongumine for 1 h and 3 h caused a markedincrease in ROS levels in these cancer cells (Fig. 3b and Supplemen-tary Figs 19 and 20). Paclitaxel also caused an increase in DCF-DAfluorescence after 1 h, but piperlongumine enhanced ROS to nearly

MMTV-PyVT (8–12 weeks old)

+ DMSO

(5% v/v)

+ PL

(2.4 mg kg–1 day–1 )

Control PL

Control (carcinoma)

PL (non-malignant)

a c

b d

1 14 48 813 100 0

5

10

15

20

25

4

8

12

16

20

Control

DMSO-treatedPaclitaxel

PL

PL-treated

   T  u  m  o  u  r  s   i  z  e   (  m  m   )

   S   i  n  g   l  e   t  u  m  o  u  r  s   i  z  e   (  m  m   )

Treatment time (days) Treatment time (days)

Figure 2 | In vivo  antitumour effect of piperlongumine. a , Inhibition of mammary tumour growth by piperlongumine treatment in MMTV-PyVTtransgenic tumour-bearing mice. MMTV-PyVT mice spontaneously developed breast adenocarcinoma by ,8 weeks of age. When tumours hadgrown to,5–6 mm in diameter, piperlongumine (2.4mg kg 21), paclitaxel(10mgkg 21) or DMSO (5% v/v) was administered intraperitoneally daily for13 days (n5 12 mice per group). Mice were then euthanized and mammary tumours were excised and processed for histological examination. Arrowsindicate the initial as well as secondary tumour lesions in the DMSO treatedanimals (left panel) versus only a single initial lesion in the piperlonguminetreated animals (right panel). b, Thesizesof thegrossly dissectedtumours weremeasured and plotted. c, Histological morphology of mammary tissue sectionsfrom MMTV-PyVT tumour-bearing mice treated with piperlongumine orDMSO after 13 days, stained with haematoxylin and eosin. d, Size of singletumours after 10 days of piperlongumine or paclitaxel treatment (asterisk indicatesshorter treatmentperiod dueto high toxicity of paclitaxel in animals).Values in bar graphs are mean6 s.d. of three independent experiments.

RESEARCH LETTER

2 3 2 | N A T U R E | V O L 4 7 5 | 1 4 J U L Y 2 0 1 1

Macmillan Publishers Limited. All rights reserved ©2011

Page 3: Nature 10167

5/16/2018 Nature 10167 - slidepdf.com

http://slidepdf.com/reader/full/nature-10167 3/4

twice these levels (Fig. 3b). Co-treatment with NAC fully reversed thepiperlongumine-inducedincrease in ROSand cell death (Fig. 3b, d andSupplementary Fig. 21). Using a series of fluorescent probes specificfor individual species of ROS, we found that hydrogen peroxide andnitric oxide, but not superoxide anion, were among the ROS speciesinduced by piperlongumine in cancer cells (Fig. 3b, c and Sup-plementary Figs 22–24).

In contrast to the results in cancer cells, piperlongumine did notcause an increase in ROS levels in normal cells (Fig. 4a and Sup-plementary Fig. 25). This selective induction of ROS in cancer cells

distinguishes piperlongumine from other small molecules that affectROSlevels, suchas the microtubule-stabilizing agent paclitaxel and theglutathione synthesis inhibitor buthionine sulphoximine (Fig. 4a andSupplementary Fig. 25), and indicates that piperlongumine-inducedROS elevation is a consequence of cell transformation. Engineering normalcellsto have a cancergenotypepotentiated thepiperlongumine-induced increase in ROS (Fig. 4b and Supplementary Figs 26 and 27).Serial transformationitself leads to increased expression of the putativepiperlongumine targets GSTP1 and CBR1 (Supplementary Fig. 28),indicating that these proteins may have a role in enabling the trans-formed cell to adapt to transformation-induced oxidative stress. Wetherefore hypothesized that overexpression of CBR1 or GSTP1 mightrescue transformed cells from both piperlongumine-induced ROSelevation and piperlongumine-induced apoptosis. Stably overexpres-sing CBR1 or GSTP1, and particularly both, in EJ cells markedly reduced piperlongumine-induced ROS levels and partially rescued

the piperlongumine-induced apoptotic phenotype (Supplementar

Fig. 29). In a complementary study, knockdown of GSTP1 or CBRdid not affect piperlongumine-induced ROS levels (SupplementarFig. 30). These results may reflect the fact that other members of thGST family were observed to bind piperlongumine in our affinityenrichment studies (Supplementary Fig. 16b) and may have partialoverlapping functions in the cell. These data indicate that piperlongumine induces apoptosis by interfering with redox and ROS homeostatic regulators such as GSTP1 and CBR1.

The ability of piperlongumine to inhibit the growth of rapidly growing and highly invasive multifocal mammary tumours without genertoxicity indicates that perturbing redox and ROS homeostasis is promising strategy for cancer treatment. Our cell-based experimenindicate that piperlongumine treatment selectively increases ROlevels and induces apoptosis in cancer cells relative to normal cell

This correlates with the selective induction of related phenotypeincluding DNA damage (Fig. 4c and Supplementary Figs 27, 31 an32) and alterations in mitochondrial morphology and function, occuring selectively in cancer cells (Supplementary Fig. 33). The differentiresponse of cancer cells and normal cells to treatment with piperlongumine indicates that piperlongumine targets a dependency associated with ROS homeostasis that arises during transformatioNormal cells, including stem cells, have low basal levels of ROS1,4,5,14–

and therefore a diminished reliance on the ROS stress-response pathway, whereas cancer cells, especially cancer stem cells, have high leveof ROS14 and might therefore be expected to have a strong reliance othe ROS stress-response pathway 1,5,16,18,19. The use of small moleculethat alter levels of ROS such as b-phenylethyl-isothiocyanate anbuthionine sulphoximine4,17 has been suggested for the treatment ocancer. Other small molecules such as curcumin20 and 2-cyano-12-dioxoolean-1,9-dien-28-oic acid (CDDO) derivatives21 have bee

a

b

c d

   C  e   l   l  u   l  a  r   G   S   H

   (   %   c  o  n   t  r  o   l   )

   T  o   t  a   l   G   S   S   G   l  e  v  e   l  s

0

0

0

4

8

12

16

5

10

15

20

25

1.0

2.0

3.0

40

80

100

10 10 10 10

1 1 3

33

PL ( μM)Time (h)

NAC (mM)3

––– – –

60

0

20

DMSO DMSO

EJ 76N (NMEC)

0

2

6

8

10

   M  e  a  n   f  u  o  r  e  s  c  e  n  c  e

   (   R   O   S   l  e  v  e   l   )

   M  e  a  n

   f  u  o  r  e  s  c  e  n  c  e

   (   R   O   S   l  e  v  e   l   )

   M  e  a  n

   f  u  o  r  e  s  c  e  n  c  e

   (   R   O   S   l  e  v  e   l   )

   L   i  v  e  c  e   l   l  s   (   %   )

DMSO

DMSO

PL ( μM)

T (nM)

NAC (mM)

––

– – –– – – –

––3 3 3 3 3

10 10 20 2025 25 50 50

0

40

60

80

100 + PL (24 h) + Paclitaxel (24 h)

20

PL

PL

PL + CAT

PL + CAT

DMSO PL PL PL +

NAC

T T NAC

EJ

U2OS

1 h

3 h

4

PL PL (10 μM)

Figure 3 | Piperlongumine enhances ROS accumulation in cancer cells by targeting the stress response to ROS. a , Piperlongumine-mediatedmodulation of GSH and GSSG. GSH levels were determined after EJ cells wereeithertreated with piperlongumine or pretreated with NAC for1 h, followed by piperlongumine treatment for 1 h or 3 h (left panel). GSSG levels were alsodetermined after EJ cells and 76N (NMEC) cells were treated withpiperlongumine for 3 h (right panel). b, Piperlongumine-induced ROSelevationand reversionby NAC.EJ cells weretreatedwith piperlongumine(PL,10mM), paclitaxel (T, 25 nM) or DMSO for 1 h and 3 h. Cells were alsopretreated with 3mM NAC for 1 h, followed by 10mM piperlongumine for3 h.c, Reversion of piperlongumine-induced ROS accumulation by catalase. EJ orU2OS cells were pretreated with catalase (CAT, 2,000 U ml21) for 2 h, followedby10mM piperlonguminefor 3 h. d, Piperlongumine-induced celldeath can berescued by NAC. EJ cells were treated with piperlongumine for 24 h, or treated

with 3 mM NAC for 1 h followed by piperlongumine or paclitaxel for 24h. Cell viability was measured by trypan blue exclusion staining assay. All values aremean6 s.d. of three independent experiments.

p53 (total)

Ser15p-p53

Ser15p-p

p21

γ -H2AX

β-actin

a

b

c

γ -H2AX

H2AX

β-actin

ETO: – – – 5 10 μMPL: – 7.5 10 –

16N12 h 12 h24 h

76N

12

10

8

6

4

2

0DMSO PL

(10 μM)

Paclitaxel

(25 nM)

C P10 P15 ETO C P10 P15 ETO

   M  e  a  n   D   C   F  -   D   A

   f  u  o  r  e  s  c  e  n  c  e   (     o   l   d   )

   M  e  a  n   D   C   F  -   D   A

   f  u  o  r  e  s  c  e  n  c  e   (     o   l   d   )

BJ-hTERT BJ-ST BJ-ELR

20

16

PL:( μM) – 5 – 55 –

12

8

4

0

– μM

Figure 4 | Piperlongumine does not increase ROS levels and the ROS-induced DNA-damage response in normal and immortalized non-

transformed cells. a , Piperlongumine does not increase ROS levels in normalcells (16N). ROS levels were measured by flow cytometry and shown by quantitative bar graph measured as the fold change over DMSO-treated levels.b, Selective inductionof ROS by piperlonguminein oncogenically transformed Bhuman fibroblasts (BJ-ELR), but not in non-transformed BJ fibroblasts (BJ-hTERT and BJ-ST). ROS levels were measured after treating with DMSO (2) opiperlongumine (PL, 5mM) for 8 h. All values are mean6 s.d. of threeindependent experiments. c, The effect of piperlongumine on stress-responsetargets was determined by westernblot analysis of p53,phosphorylated p53(Ser 1p-p53), p21 and c-H2AX in normal 76N cells, treated with piperlongumine(10mM (P10) or 15mM (P15)), 10 mM etoposide (ETO) or DMSO as a solventcontrol (C) for 12 h and 24 h (left panel). Western blots were performed similaron 16Ncells treated for12 h with piperlongumine (7.5mMor10mM)or etoposid(5mM or 10mM) (right panel). b-actin expression was used as a loading contro

LETTER RESEARCH

1 4 J U L Y 2 0 1 1 | V O L 4 7 5 | N A T U R E | 2 3

Macmillan Publishers Limited. All rights reserved ©2011

Page 4: Nature 10167

5/16/2018 Nature 10167 - slidepdf.com

http://slidepdf.com/reader/full/nature-10167 4/4

reported to promote ROS and reduceGSH levels in cancer cells, in onecase in an oncogene-dependent manner17, and the activation of theKEAP1–NRF2 antioxidant pathway 22,23 has been suggested to beinvolved.

The introduction of a single oncogene (HRAS) leads to increasedlevels of ROS (Fig. 4b and ref. 24), increased expression of GSTP1 andCBR1 (Supplementary Fig. 28), an increased apoptotic response topiperlongumine (Fig. 1c), and notably, to a substantial increase in

levels of ROS after treatment with piperlongumine. In EJ cells,piperlongumine-induced cell death is rescued by the antioxidantNAC (Fig. 3d). The increased dependence of cancer cells on theROS stress-response pathway may be the basis for the selectivity of piperlongumine-induced apoptosis in cancer cells (Figs 1 and 2). Insupport of this hypothesis, the activation of signalling through the JNK(also known as MAPK8) pathway has been implicated as an antitu-morigenic responseto oncogene expression25. Thisresponse is coupledto oncogene-dependent oxidative stress through p53 stabilization, andcould also function independently of p53 through pro-apoptoticcJUN-dependent transcription. In addition to its role in regulating ROS, GSTP1 is also known to be a direct negative regulator of JNK26, providing a possible mechanism for piperlongumine-inducedapoptosis in both p53-wild-type and p53-mutant cancer cells.

A global investigation of the spectrum of cancer genotypes will berequired to identify the range of cancer genotypes that impart piper-longumine sensitivity, but our results alreadyhighlight a novel strategy for cancer therapy that preferentially eradicates cancer cells by target-ing the ROS stress-response pathway.

METHODS SUMMARYApoptotic cell populations were determined by TdT-mediated dUTP nick endlabelling (TUNEL) assay and quantified using flow cytometry. Cell viability wasalso determined by crystal violet staining (0.2% w/v in 2% ethanol), by trypan blueexclusion andby the Alamar bluecell viability assay. For crystalviolet staining, cellswereplatedin 6-well and12-well plates and, after reaching60–70%confluency, thecells were treatedwith piperlongumine for12 h and 24h. Formeasurement of ROSproduction,cells weretreated withpiperlongumine or paclitaxel for 1 h and3 h andthen incubated with 10mM DCF-DA for 30 min at 37uC, washed twice with PBS

and immediately analysed by a FACScan flow cytometer. Cells were treated withpiperlongumine and etoposide for 18–24 h and processed for Comet assay follow-ing the manufacturer’s instructions (Trevigen). For xenograft tumour models,cancer cell lines EJ, A549 and MDA-MB-435 were injected subcutaneously intotheflanks of nude mice. Forthe melanomamouse model,B16-F10 melanomacellswereinjected intothe flanksof C57BL/6J mice. FVB/N-Tg(MMTV-PyVT) 634Mulmales were obtained from the Mouse Models of Human Cancer consortium(MMHCC) at NCI-Frederick and bred with FVB females. Female offspring weregenotyped for the presence of the transgene using the primers published by MMHCC. For piperlongumine target identification, we followed the SILAC-basedaffinity enrichment methodology previously described11,27. For further details seeSupplementary Methods.

Received 17 February 2010; accepted 3 May 2011.

1. Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and

non-oncogene addiction. Cell 136, 823–837 (2009).2. Yap,T. A.,Sandhu,S. K., Carden,C. P.& deBono, J.S. Poly(ADP-ribose)polymerase(PARP) inhibitors: Exploiting a synthetic lethal strategy in the clinic. CA Cancer J.Clin. 61, 31–49 (2011).

3. Poole,L. B.& Nelson,K. J.Discoveringmechanismsof signaling-mediated cysteineoxidation. Curr. Opin. Chem. Biol. 12, 18–24 (2008).

4. Schumacker, P.T. Reactiveoxygenspecies incancercells:liveby thesword,diebythe sword. Cancer Cell 10, 175–176 (2006).

5. Trachootham, D., Alexandre, J. & Huang, P. Targeting cancer cellsby ROS-mediated mechanisms: a radical therapeutic approach? Natl Rev. 8, 579–591(2009).

6. Brown, L. et al. CDIP, a novel pro-apoptotic gene, regulates TNFa-mediatedapoptosis in a p53-dependent manner. EMBO J. 26, 3410–3422 (2007).

7. Bezerra, D. P. et al. Piplartine induces inhibition of leukemia cell proliferationtriggering bothapoptosis and necrosis pathways. Toxicol. In Vitro 21, 1–8 (2007).

8. Hahn, W. C. et al. Creation of human tumour cells with defined genetic elementsNature 400, 464–468 (1999).

9. Ryo, A . et al. PIN1 is an E2F target gene essential for Neu/Ras-inducedtransformation of mammaryepithelial cells.Mol.Cell. Biol.22, 5281–5295(2002).

10. Guy, C.T., Cardiff,R. D. & Muller,W. J. Inductionof mammary tumors byexpressionof polyomavirus middle T oncogene: a transgenic mouse model for metastaticdisease. Mol. Cell. Biol. 12, 954–961 (1992).

11. Ong,S. E. etal. Identifying theproteins to which small-molecule probes and drugsbind in cells. Proc. Natl Acad. Sci. USA 106, 4617–4622 (2009).

12. Bateman, R. L., Rauh, D., Tavshanjian, B. & Shokat, K. M. Human carbonylreductase 1 is an S -nitrosoglutathione reductase. J. Biol. Chem. 283,35756–35762 (2008).

13. Ralat, L. A., Manevich, Y., Fisher, A. B. & Colman,R. F. Direct evidence for theformation of a complex between 1-cysteine peroxiredoxin and glutathioneS -transferase p withactivity changes in bothenzymes. Biochemistry 45, 360–372(2006).

14. Diehn, M. etal. Associationof reactiveoxygenspecies levels andradioresistanceincancer stem cells. Nature 458, 780–783 (2009).

15. Fruehauf, J. P. & Meyskens, F. L. Jr. Reactive oxygen species: a breath of life ordeath? Clin. Cancer Res. 13, 789–794 (2007).

16. Huang, P., Feng, L., Oldham, E. A., Keating, M. J. & Plunkett, W. Superoxidedismutase as a target for theselective killing of cancer cells. Nature 407, 390–395(2000).

17. Trachootham, D. et al. Selective killing of oncogenically transformed cells througha ROS-mediated mechanism by b -phenylethyl isothiocyanate. Cancer Cell 10,241–252 (2006).

18. Gogvadze, V.,Orrenius,S. & Zhivotovsky,B. Mitochondriain cancer cells:what is sospecial about them? Trends Cell Biol. 18, 165–173 (2008).

19. Szatrowski,T. P.& Nathan,C. F. Productionof largeamounts of hydrogenperoxideby human tumor cells. Cancer Res. 51, 794–798 (1991).

20. Ravindran, J., Prasad, S. & Aggarwal, B. B. Curcumin and cancer cells: how manyways can curry kill tumor cells selectively? Am. Assoc. Pharm. Sci. J. 11, 495–510(2009).

21. Yue, P., Zhou, Z., Khuri, F. R. & Sun, S. Y. Depletion of intracellular glutathionecontributes to JNK-mediated death receptor 5 upregulation and apoptosisinduction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). Cancer Biol. Ther. 5, 492–497 (2006).

22. Banning,A., Deubel,S., Kluth,D., Zhou,Z. & Brigelius-Flohe,R. TheGI-GPxgeneis atarget for Nrf2. Mol. Cell Biol. 25, 4914–4923 (2005).

23. Dinkova-Kostova, A. T. et al. An exceptionally potent inducer of cytoprotectiveenzymes: elucidation of the structural features that determine inducer potencyand reactivity with KEAP1. J. Biol. Chem. 285, 33747–33755 (2010).

24. Lee, A.C. et al.Rasproteins inducesenescenceby alteringtheintracellular levels ofreactive oxygen species. J. Biol. Chem. 274, 7936–7940 (1999).

25. Schramek, D. et al. The stress kinase MKK7 couples oncogenic stress to p53stability and tumor suppression. Nature Genet. 43, 212–219 (2011).

26. Wang, T., Arifoglu, P., Ronai, Z. & Tew, K. D. Glutathione S -transferase P1–1(GSTP1–1) inhibits c-Jun N-terminal kinase (JNK1) signaling through interactionwith the C terminus. J. Biol. Chem. 276, 20999–21003 (2001).

27. Margolin, A. A. et al. EmpiricalBayes analysis of quantitative proteomicsexperiments. PLoS ONE 4, e7454 (2009).

Supplementary Information is linked to the online version of the paper atwww.nature.com/nature.

AcknowledgementsWe thank K.Chu, L.Brown-Endres, E.Lernerand F. Nevillefor theirhelp in preparing the manuscript, W. C. Hahn for BJ cell lines, V. Band for 76N cells,D.BeerforH1975cellsandK. Todorova,G.Wei,S.Ong,S.Nortonand F.Anfor technicaassistance. This project has beensupported in part by grants CA142805, CA127247CA085681 and CA080058 from NIH. This research was supported by the NationalCancer Institute’s Initiative for Chemical Genetics Contract (N01-CO-12400) andCancer Target Discovery and Development Network grant (5 RC2 CA148399-02), aswell as the National Institutes of Health Genomics Based Drug Discovery—Target IDProjectGrant (RL1HG004671, which is administratively linked to NationalInstitutes ofHealth Grants RL1CA133834, RL1GM084437 and UL1RR024924). S.L.S. is anInvestigator with the Howard Hughes Medical Institute.

Author Contributions L.R. and T.I. conducted most of the experimental work. A.U.G.,M.S. and X.L. made criticalexperimentalcontributions.N.J.T., A.M.S.,T.R.G.,S.A.C., A.F.Sand M.F. designed the experimental plans, analysed and interpreted the data. A.M.,S.L.S. and S.W.L. designed and directed the project and drafted the manuscript.

Author Information Reprints and permissions information is available atwww.nature.com/reprints . The authors declare competing financial interests: detailsaccompany the full-text HTML version of the paper at www.nature.com/nature.Readers are welcome to comment on the online version of this article atwww.nature.com/nature. Correspondence and requests for materials should beaddressed to S.W.L. ([email protected]) , A.M. ([email protected]) or S.L.S([email protected]) .

RESEARCH LETTER

2 3 4 | N A T U R E | V O L 4 7 5 | 1 4 J U L Y 2 0 1 1

Macmillan Publishers Limited. All rights reserved ©2011