<<

Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Molecular Cancer Preclinical Development Therapeutics

Overcoming Erlotinib Resistance in EGFR –Positive Non–Small Cell Cells by Targeting Survivin

Kunio Okamoto1, Isamu Okamoto1, Erina Hatashita1, Kiyoko Kuwata1, Haruka Yamaguchi1, Aya Kita2, Kentaro Yamanaka2, Mayumi Ono3, and Kazuhiko Nakagawa1

Abstract Loss of PTEN was recently shown to contribute to resistance to epidermal receptor (EGFR) inhibitors (TKI) in EGFR mutation–positive non–small cell lung cancer (NSCLC) through activation of the protein kinase AKT. We previously showed that downregulation of the expression of the antiapoptotic protein survivin by EGFR–TKIs contributes to EGFR–TKI-induced in EGFR mutation– positive NSCLC cells. We have now investigated the role of survivin expression in EGFR–TKI resistance induced by PTEN loss. The EGFR–TKI erlotinib did not affect survivin expression or induce apoptosis in EGFR mutation–positive NSCLC cells with PTEN loss. Downregulation of survivin either by transfection with a specific short interfering RNA or by exposure to the small-molecule survivin suppressor YM155 reversed erlotinib resistance in such cells in vitro. Furthermore, combination therapy with YM155 and erlotinib inhibited the growth of tumors formed by EGFR mutation–positive, PTEN-deficient NSCLC cells in nude mice to a greater extent than did treatment with either drug alone. These results thus indicate that persistent activation of signaling by the AKT–survivin pathway induced by PTEN loss underlies a mechanism of resistance to erlotinib-induced apoptosis in EGFR mutation–positive NSCLC. They further suggest that the targeting of survivin has the potential to overcome EGFR–TKI resistance in EGFR mutation–positive NSCLC. Mol Cancer Ther; 11(1); 204–13. 2011 AACR.

Introduction mechanisms is underway (3–7). In addition, some patients are intrinsically resistant to EGFR–TKIs, even though their Approximately 70% of individuals with non–small cell tumors harbor activating of EGFR (8). Further lung cancer (NSCLC) who harbor somatic mutations in characterization of the mechanisms of EGFR–TKI resis- exons of the epidermal (EGFR) tance is thus important to provide a basis for the devel- gene that encode the tyrosine kinase domain of the recep- opment of effective therapies for patients who develop tor experience substantial tumor regression when treated such resistance. with the EGFR tyrosine kinase inhibitors (TKI) gefitinib or The deletion or inactivation of the PTEN gene occurs in erlotinib (1). However, most patients, even those who a variety of tumor types, including melanoma as well as show a marked response to initial treatment, develop lung, bladder, renal, breast, endometrial, and thyroid acquired resistance to EGFR–TKIs after varying periods cancer, and there are no related proteins that can com- of time (2). To date, several major mechanisms of such pensate for the loss of PTEN function (9). The loss of PTEN acquired resistance, including secondary mutation of results in misregulation of AKT-dependent signaling, EGFR, amplification of MET, and overexpression of hepa- which plays a key role in the progression of malignant tocyte growth factor, have been identified, and the devel- cancer (10). Recent studies have shown that PTEN loss opment of pharmaceutical agents that target these contributes to EGFR–TKI resistance in EGFR mutation– positive lung cancer through activation of the protein kinase AKT (11, 12). We recently found that EGFR–TKIs Authors' Affiliations: 1Department of Medical Oncology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka; 2Drug Discovery Research, downregulate survivin expression through inhibition of Inc., Tsukuba-shi, Ibaraki; and 3Department of Pharma- the phosphoinositide 3-kinase (PI3K)–AKT signaling ceutical Oncology, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan pathway and that such downregulation of survivin con- tributed to EGFR–TKI-induced apoptosis in EGFR muta- Note: Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). tion–positive NSCLC cells (13). Survivin is a member of the inhibitor of apoptosis (IAP) family of proteins and has Corresponding Author: Isamu Okamoto, Department of Medical Oncol- ogy, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka- been shown to inhibit caspases and to prevent caspase- Sayama, Osaka 589-8511, Japan. Phone: 81-72-366-0221; Fax: 81-72- mediated cell death (14). Persistent survivin expression 360-5000; E-mail: [email protected] might therefore be expected to result in resistance to doi: 10.1158/1535-7163.MCT-11-0638 EGFR–TKIs in EGFR mutation–positive NSCLC cells with 2011 American Association for Cancer Research. PTEN loss.

204 Mol Cancer Ther; 11(1) January 2012

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming EGFR–TKI Resistance in EGFR-Mutant NSCLC

We now show that erlotinib does not affect survivin globulin G. Immune complexes were finally detected with expression or induce apoptosis in EGFR mutation–posi- chemiluminescence reagents (Perkin-Elmer Life Science). tive NSCLC cells with PTEN loss. We further examined whether persistent survivin expression was associated Gene silencing with the sensitivity of cells to EGFR–TKIs and whether Cells were plated at 50% to 60% confluence in 6-well modulation of survivin expression might overcome resis- plates or 25 cm2 flasks and then incubated for 24 hours tance to these drugs in EGFR mutation–positive NSCLC before transient transfection for 24 hours with short inter- cells with PTEN loss. fering RNAs (siRNA) mixed with the Lipofectamine reagent (Invitrogen). Small interfering RNAs specific for Materials and Methods PTEN (50-UGAACCUGAUCAUUAUAGATT-30) or sur- vivin (50-GAAGCAGUUUGAAGAAUUA-30) mRNAs as Cell culture and reagents well as a corresponding scrambled (control) siRNA were The human NSCLC cell lines HCC827 and H1650 were obtained from Nippon EGT. obtained from American Type Culture Collection. The NSCLC cell line PC9 was obtained as described previously Annexin V binding assay (15). The NSCLC cell lines PC9/GEF1-1 and PC9/GEF2-1 The binding of Annexin V to cells was measured with were generated and characterized as described previously the use of an Annexin V–FLUOS Staining (Roche). (12). We screened all cell lines for the presence of EGFR Cells were harvested by exposure to trypsin-EDTA, mutations by direct DNA sequencing of exons 18 to 21 as washed with PBS, and centrifuged at 200 g for 5 minutes. described previously (15) for this study. All cells were The cell pellets were resuspended in 100 mL of Annexin V– passaged for 3 months or less before the renewal from FLUOS labeling solution, incubated for 10 to 15 minutes at frozen, early-passage stocks obtained from the indicated 15 to 25C, and then analyzed for fluorescence with a flow sources. Cells were regularly screened for mycoplasma cytometer (FACSCalibur) and Cell Quest software (Bec- with the use of a MycoAlert Mycoplasma Detection Kit ton Dickinson). (Lonza). All cells were cultured under a humidified atmo- sphere of 5% CO2 at 37 C in RPMI 1640 medium (Sigma) Cell growth inhibition assay supplemented with 10% FBS. Erlotinib (Supplementary Cells were transferred to 96-well flat-bottomed plates Fig. S1A) was obtained from Kemprotec, and YM155 (Sup- and cultured for 24 hours before exposure for 72 hours to plementary Fig. S1B) was obtained from Astellas Pharma. various concentrations of erlotinib. Tetra Color One (5 mmol/L tetrazolium monosodium salt and 0.2 mmol/L Immunoblot analysis 1-methoxy-5-methyl phenazinium methylsulfate; Seika- Cells were washed twice with ice-cold PBS and then gaku Kogyo) was then added to each well, and the cells lysed in a solution containing 20 mmol/L Tris-HCl (pH were incubated for 3 hours at 37C before measurement of 7.5), 150 mmol/L NaCl, 1 mmol/L EDTA, 1% Triton X- absorbance at 490 nm with a Multiskan Spectrum instru- 100, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L phe- ment (Thermo Labsystems). Absorbance values were nylmethylsulfonyl fluoride, and leupeptin (1 mg/mL). The expressed as a percentage of that for untreated cells. protein concentration of the cell lysates was determined with the use of the Bradford reagent (Bio-Rad) and equal Growth inhibition assay in vivo amounts of protein were subjected to SDS-PAGE on a All animal studies were carried out in accordance with 7.5% gel. The separated proteins were transferred to a the Recommendations for Handling of Laboratory Ani- nitrocellulose membrane, which was then exposed to 5% mals for Biomedical Research compiled by the Committee nonfat dried milk in PBS for 1 hour at room temperature on Safety and Ethical Handling Regulations for Labora- before incubation overnight at 4C with primary antibo- tory Animal Experiments, Kinki University. The ethical dies. Rabbit polyclonal antibodies to human phosphory- procedures followed conformed to the guidelines of the lated EGFR (pY1068), to phosphorylated or total forms of United Kingdom Coordinating Committee on Cancer AKT, to phosphorylated or total forms of extracellular Prevention Research. Tumors cells (5 106) were injected signal-regulated kinase (ERK), to XIAP, to IAP-1, to Bcl-xl, subcutaneously into the axilla of 5- to 6-week-old female to Bcl-2, to Mcl-1, to PARP, to BIM, and to PTEN were athymic nude mice (BALB/c nu/nu; CLEA Japan). Treat- obtained from Cell Signaling Technology; those to survi- ment was initiated when tumors in each group of 6 mice vin were from Santa Cruz Biotechnology; and those to achieved an average volume of 300 to 600 mm3. Treatment b-actin were from Sigma. Mouse monoclonal antibodies to groups consisted of vehicle control, erlotinib alone (10 EGFR were obtained from Invitrogen. All antibodies were mg/kg), YM155 alone (5 mg/kg), and erlotinib plus used at a 1:1,000 dilution, with the exception of those to YM155. Erlotinib was administered by oral gavage daily b-actin (1:200). The nitrocellulose membrane was then for 31 days, with control animals receiving a 0.5% (w/v) washed with PBS containing 0.05% Tween 20 before aqueous solution of hydroxypropylmethylcellulose as incubation for 1 hour at room temperature with horse- vehicle. Continuous infusion of YM155 has been found radish peroxidase–conjugated goat antibodies to rabbit to induce tumor regression and intratumoral survivin (Sigma) or mouse (Santa Cruz Biotechnology) immuno- suppression in established human hormone-refractory

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 205

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Okamoto et al.

prostate cancer, non-Hodgkin , melanoma, cell lines (HCC827, PC9, and H1650) by staining with and NSCLC xenografts (16–20). YM155 was thus admin- Annexin V. HCC827, PC9, and H1650 cells harbor istered over 7 consecutive days (days 1 to 7) with the use of an EGFR allele with an activating mutation, whereas an implanted micro-osmotic pump (Alzet model 1003D; H1650 cells also show homozygous deletion of PTEN. DURECT Cupertino). Erlotinib induced a marked increase in the propor- Tumor volume was determined from caliper measure- tion of apoptotic cells among HCC827 and PC9 cells, ments of tumor length (L) and width (W) according to the whereas it was without effect in H1650 cells, despite formula LW2/2. Both tumor size and body weight were these cells harboring an activating mutation in EGFR measured twice per week. (Fig.1A).Immunoblotanalysisofthecleavedformof PARP confirmed that erlotinib did not induce apopto- Statistical analysis sis in H1650 cells (Fig. 1B). We and others previously Quantitative data are presented as means SE from 3 showed that PI3K–AKT–survivin and mitogen-activat- independent experiments or for 6 animals per group. The ed protein (MEK)–ERK–BIM signaling pathways significance of differences in the percentage of Annexin play important roles in EGFR–TKI-induced apoptosis V–positive cells or in tumor size was evaluated with the (13, 21–23). We therefore next compared the effects of unpaired 2-tailed Student t test. P < 0.05 was considered erlotinib on the expression level or phosphorylation statistically significant. status of EGFR, AKT, ERK, PTEN, survivin, and BIM in the 3 NSCLC cell lines by immunoblot analysis. Erlo- Results tinib induced the dephosphorylation of EGFR as well as that of AKT and ERK in the erlotinib-sensitive cell Differential apoptotic responses of EGFR mutation– lines HCC827 and PC9 (Fig. 1C). In addition, erlotinib positive NSCLC cell lines to erlotinib induced downregulation of survivin expression and We first examined the effects of the EGFR–TKI erlo- upregulation of BIM expression in HCC827 and PC9 tinib on apoptosis in EGFR mutation–positive NSCLC cells, consistent with our previous observations (13).

A C EGFR 60 mutation positive

HCC827 PC9 H1650 40 Erlotinib (µmol/L): 010.1 0 10.10 10.1 pEGFR

20 EGFR

PTEN pAKT Annexin V–positive cells (%) V–positive Annexin 0 Erlotinib : – + –+ –+ AKT

Survivin HCC827 PC9 H1650 XIAP

IAP-1

B pERK HCC827 PC9 H1650 ERK

Erlotinib : – +–+–+ BIM

PARP Bcl-xI cl-PARP Bcl-2

Actin MCL

Actin

Figure 1. Differential apoptotic responses of EGFR mutation–positive NSCLC cell lines to erlotinib. A, HCC827, PC9, or H1650 cells were incubated in complete medium with or without erlotinib (100 nmol/L) for 48 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry. Data are means SE from 3 independent experiments. B, cells were incubated in complete medium with or without erlotinib (100 nmol/L) for 48 hours, lysed, and subjected to immunoblot analysis with antibodies to PARP or to b-actin (loading control). Bands corresponding to the intact and cleaved (cl) forms of PARP are indicated. C, cells were incubated in complete medium containing the indicated concentrations of erlotinib for 24 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of EGFR, AKT, or ERK or to the indicated proteins. Data in B and C are representative of 3 independent experiments.

206 Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming EGFR–TKI Resistance in EGFR-Mutant NSCLC

However, whereas erlotinib induced the dephos- EGFR, AKT, and survivin in these cell lines by immuno- phorylation of EGFR and ERK as well as the upregula- blot analysis. Similar to our observations with H1650 tion of BIM expression in H1650 cells, it had no effect cells, erlotinib did not affect the levels of AKT phosphor- of the levels of AKT phosphorylation or survivin ylation or survivin expression in PC9/GEF1-1 and PC9/ expression (Fig. 1C). The expression of other IAP or GEF2-1 cells, whereas it still induced the dephosphory- Bcl family members, including XIAP, IAP-1, Bcl-2, lation of EGFR and ERK as well as the upregulation of Bcl-xl, and Mcl-1, was not substantially affected by BIM expression (Fig. 2C). These findings thus suggested erlotinib in any of the cell lines examined. Given that that persistent expression of survivin in the presence of PTEN inhibits AKT activation by catalyzing the erlotinib contributes to the resistance to this drug associ- dephosphorylation of phosphatidylinositol 3,4,5-tri- ated with PTEN loss in EGFR mutation–positive NSCLC sphosphate, the product of class I PI3K action, these cells. results suggested that the resistance of some EGFR mutation–positive cells to erlotinib-induced apoptosis Knockdown of PTEN induces persistent survivin may be associated with persistent activation of the expression and reduces erlotinib sensitivity in EGFR AKT–survivin pathway. mutation–positive cells To investigate the contribution of PTEN to survivin The effect of erlotinib on survivin expression expression and erlotinib resistance, we examined the correlates with that on apoptosis effects of siRNA-mediated depletion of PTEN in PC9 and We have previously established EGFR–TKI (gefitinib)- HCC827 cells. Transfection of these cells with PTEN resistant sublines (PC9/GEF1-1 and PC9/GEF2-1) of PC9 siRNA attenuated the dephosphorylation of AKT and cells that harbor the activating EGFR mutation of the downregulation of survivin expression, without affecting parental cells but which have also lost PTEN expression, the dephosphorylation of ERK and upregulation of BIM, with the loss of PTEN contributing to their EGFR–TKI induced by erlotinib (Fig. 3A). Staining with Annexin V resistance (12). Both staining with Annexin V and immu- also revealed that depletion of PTEN resulted in inhibition noblot analysis of PARP revealed that erlotinib did not of erlotinib-induced apoptosis (Fig. 3B). Moreover, knock- induce a substantial level of apoptosis in PC9/GEF1-1 or down of PTEN reduced the sensitivity of HCC827 and PC9/GEF2-1 cells (Fig. 2A and B), consistent with our PC9 cells to the inhibitory effect of erlotinib on cell growth previous observations (12). To investigate whether loss of (Fig. 3C). These results suggested that PTEN loss contri- PTEN expression affects the modulation of AKT–survivin butes to persistent survivin expression in the presence of signaling by erlotinib, we examined the effects of erlotinib erlotinib and to erlotinib resistance in EGFR mutation– on the expression level or phosphorylation status of positive cells.

Figure 2. Effect of erlotinib on survivin expression correlates with A C PC9 sensitivity to erlotinib in EGFR 50 mutation–positive NSCLC cells. A, Parent GEF1-1 GEF2-1 PC9, PC9/GEF1-1, or PC9/GEF2-1 40 µ cells were incubated in complete Erlotinib ( mol/L): 010.1 0 10.10 10.1 30 medium with or without erlotinib (100 pEGFR nmol/L) for 48 hours, after which the 20 EGFR proportion of apoptotic cells was determined by staining with 10 PTEN fl – uorescein isothiocyanate pAKT

conjugated Annexin V and propidium cells (%) V–positive Annexin 0 fl AKT iodide followed by ow cytometry. Erlotinib : –+ –+ –+ Data are means SE from 3 Survivin independent experiments. B, cells Parent GEF1-1 GEF2-1 were incubated in complete medium XIAP with or without erlotinib (100 nmol/L) PC9 IAP-1 for 48 hours, lysed, and subjected to immunoblot analysis with antibodies B pERK b PC9 to PARP or to -actin. C, cells were ERK incubated in complete medium Parent GEF1-1 GEF2-1 containing the indicated BIM concentrations of erlotinib for 24 Erlotinib : –+–+–+ Bcl-xI hours, after which cell lysates were subjected to immunoblot analysis Bcl-2 PARP with antibodies to the indicated cl-PARP MCL-1 proteins. Data in B and C are representative of 3 independent Actin Actin experiments.

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 207

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Okamoto et al.

A B HCC827 PC9 40 * Control siRNA: + + – – + + – – PTEN siRNA: – – + + – – + + Erlotinib : – + – + – + – + 30 * pEGFR EGFR 20

PTEN pAKT 10 AKT Annexin V–positive cells (%) V–positive Annexin Survivin 0

pERK Control siRNA: + + – – + + – – ERK PTEN siRNA: – – + + – – + + Erlotinib : – + – + – + – + BIM HCC827 PC9 Actin C HCC827 control siRNA PC9 control siRNA HCC827 PTEN siRNA PC9 PTEN siRNA

100 100

75 75

50 50

25 25

Cell viability (% control) 0 Cell viability (% control) 0 0 0.01 0.1 1 10 0 0.01 0.1 1 10 100 Erlotinib (µmol/L) Erlotinib (µmol/L)

Figure 3. Knockdown of PTEN induces persistent survivin expression and reduces erlotinib sensitivity in EGFR mutation–positive cells. A, HCC827 or PC9 cells were transfected with PTEN or scrambled (control) siRNAs for 24 hours and then incubated in complete medium with or without erlotinib (100 nmol/L) for 24 hours. Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to the indicated proteins. Data are representative of 3 independent experiments. B, cells transfected as in A were incubated with or without erlotinib (100 nmol/L) for 48 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry. Data are means SE from 3 independent experiments. , P < 0.05 for the indicated comparisons. C, cells transfected as in A were cultured in complete medium containing the indicated concentrations of erlotinib for 72 hours, after which cell viability was assessed. The number of viable cells is expressed asa percentage of the corresponding value for cells not exposed to erlotinib. Data are means from 3 independent experiments.

Knockdown of survivin reverses erlotinib resistance the survivin siRNA sensitized PC9/GEF1-1 and PC9/ induced by loss of PTEN in EGFR mutation–positive GEF2-1 cells to the proapoptotic effect of erlotinib (Fig. cells 4B). A second siRNA targeted to a different region of To investigate further the contribution of persistent survivin mRNA yielded similar results (data not shown). survivin expression to erlotinib resistance associated with These observations thus suggested that persistent expres- PTEN loss, we examined the effect of siRNA-mediated sion of survivin contributes to erlotinib resistance in EGFR depletion of survivin on erlotinib-induced apoptosis in mutation–positive cells with loss of PTEN. parental PC9 cells and the EGFR–TKI-resistant sublines PC9/GEF1-1 and PC9/GEF2-1. Whereas erlotinib YM155 reverses erlotinib resistance induced by loss induced downregulation of survivin expression only in of PTEN in EGFR mutation–positive cells in vitro the parental PC9 cells, transfection with survivin siRNA We next examined the effect of the combination of resulted in specific depletion of survivin in both the erlotinib and YM155 on apoptosis in NSCLC cells with parental and EGFR–TKI-resistant NSCLC cells (Fig. EGFR–TKI resistance due to loss of PTEN. YM155 is a 4A). The erlotinib-induced upregulation of BIM expres- small-molecule agent that specifically inhibits survivin sion was apparent in all 3 cell lines in the absence or expression in various types of cancer cell lines in vitro and presence of survivin siRNA (Fig. 4A). The survivin siRNA in vivo (16–20). Treatment of PC9/GEF1-1 or PC9/GEF2-1 had no significant effect on erlotinib-induced apoptosis in cells, or of parental PC9 cells, with YM155 resulted in PC9 cells, as revealed by staining with Annexin V (Fig. downregulation of survivin expression (Fig. 5A). Erlotinib 4B). In contrast, depletion of survivin by transfection with induced upregulation of BIM in all 3 cell lines in the

208 Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming EGFR–TKI Resistance in EGFR-Mutant NSCLC

A PC9 PC9/GEF1-1 PC9/GEF2-1

Control siRNA: + + – – + + – – + + – – Survivin siRNA: – – + + – – + + – – + + Erlotinib : – + – + – + – + – + – + Figure 4. Knockdown of survivin expression reverses erlotinib Survivin resistance induced by loss of PTEN in EGFR mutation–positive NSCLC BIM cells. A, PC9, PC9/GEF1-1, or PC9/ GEF2-1 cells were transfected with Actin survivin or scrambled (control) siRNAs for 24 hours and then B incubated in complete medium with or without erlotinib (100 nmol/L) for * 24 hours. Cell lysates were then prepared and subjected to immunoblot analysis with antibodies 60 * to survivin, BIM, or b-actin. Data are representative of 3 independent experiments. B, cells transfected as in A were incubated with or without 40 erlotinib (100 nmol/L) for 48 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin 20 V and propidium iodide followed by flow cytometry. Data are means SE

from 3 independent experiments. cells (%) V–positive Annexin , P < 0.05 for the indicated comparisons. 0 Control siRNA: + + – – + + – – + + – – Survivin siRNA: – – + + – – + + – – + + Erlotinib : – + – + – + – + – + – +

PC9 PC9/GEF1-1 PC9/GEF2-1

absence or presence of YM155 (Fig. 5A). Whereas YM155 increased the sensitivity of H1650 cells to the inhibitory had no significant effect on erlotinib-induced apoptosis in effect of erlotinib on cell growth in vitro (Fig. 6C). Finally, PC9 cells, it sensitized PC9/GEF1-1 and PC9/GEF2-1 cells to determine whether the enhancement of the proapop- to the proapoptotic effect of erlotinib (Fig. 5B). In addition, totic effect of erlotinib by YM155 in EGFR–TKI-resistant whereas YM155 did not affect the sensitivity of PC9 cells to cells observed in vitro might also be apparent in vivo,we the antiproliferative effect of erlotinib, it increased that of injected H1650 cells into nude mice to elicit the formation PC9/GEF1-1 and PC9/GEF2-1 cells to this action of erlo- of solid tumors. After tumor formation, the mice were tinib (Fig. 5C). These results thus suggested that down- treated with erlotinib, YM155, or both drugs. Combined regulation of survivin expression by YM155 promoted the treatment with erlotinib and YM155 inhibited tumor proapoptotic and antiproliferative effects of erlotinib in growth to a greater extent than did treatment with either EGFR mutation–positive cells with loss of PTEN. drug alone (Fig. 6D and E). These data suggested that YM155 enhances the response of EGFR mutation–positive YM155 reverses erlotinib resistance induced by loss tumor cells with loss of PTEN to erlotinib both in vitro and of PTEN in EGFR mutation–positive cells in vivo in vivo. We also examined the effects of siRNA-mediated deple- tion of survivin and of YM155 on erlotinib-induced apo- Discussion ptosis in H1650 cells. Transfection with the survivin siRNA resulted in specific depletion of survivin and Tyrosine kinases have come to be recognized as key sensitized H1650 cells to the proapoptotic effect of erlo- regulators of cancer cell proliferation and apoptosis as tinib (Fig. 6A). YM155 also downregulated survivin well as of tumor angiogenesis and they are therefore expression and sensitized H1650 cells to the induction considered potential targets for anticancer therapies of apoptosis by erlotinib (Fig. 6B). In addition, YM155 (24). Several strategies for targeting these kinases have

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 209

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Okamoto et al.

A B * PC9 PC9/GEF1-1 PC9/GEF2-1 60 YM155: – – + + – – + + – – + + * Erlotinib : – + – + – + – + – + – + pAKT 40 AKT Survivin pERK 20

ERK

BIM 0 Annexin V–positive cells (%) V–positive Annexin Actin YM155: – – + + – – + + – – + + Erlotinib : – + – + – + – + – + – +

PC9 PC9/GEF1-1 PC9/GEF2-1 C PC9 PC9/GEF1-1 PC9/GEF2-1 PC9+YM155 PC9/GEF1-1+YM155 PC9/GEF2-1+YM155

100 100 100

75 75 75

50 50 50

25 25 25 Cell viability (% control) Cell viability (% control)

Cell viability (% control) 0 0 0 0.01 0.1 1 10 100 0.01 0.1 1 10 100 0.01 0.1 1 10 100 Erlotinib (µmol/L) Erlotinib (µmol/L) Erlotinib (µmol/L)

Figure 5. YM155 reverses erlotinib resistance induced by loss of PTEN in EGFR mutation–positive NSCLC cells in vitro. A, PC9, PC9/GEF1-1, or PC9/GEF2-1 cells were incubated in complete medium with or without erlotinib (100 nmol/L), YM155 (10 nmol/L), or both drugs for 24 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to the indicated proteins. Data are representative of 3 independent experiments. B, cells were incubated in complete medium with or without erlotinib (100 nmol/L), YM155 (10 nmol/L), or both drugs for 48 hours, after which the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry. Data are means SE from 3 independent experiments. , P < 0.05 for the indicated comparisons. C, cells were cultured in complete medium containing the indicated concentrations of erlotinib with or without 10 nmol/L YM155 for 72 hours, after which cell viability was assessed. The number of viable cells is expressed as a percentage of the corresponding value for cells not exposed to erlotinib. Data are means from 3 independent experiments.

been pursued, the most successful of which has been the however. We have now shown that erlotinib did not development of small-molecule TKIs (25–27). However, induce apoptosis or downregulation of survivin expres- increasing evidence has shown the development of sion in H1650 cells, which harbor an EGFR allele with an acquired resistance to these drugs, and extensive preclin- activating mutation and also no longer manifest PTEN ical studies are ongoing to provide insight into the molec- expression. Similar results were obtained with EGFR– ular mechanisms underlying such resistance (28). We and TKI-resistant PC9/GEF sublines of PC9 cells that have others have recently shown that loss of PTEN contributes also lost PTEN expression and manifest persistent phos- to EGFR–TKI resistance in EGFR mutation–positive lung phorylation of AKT. We further found that an siRNA cancer through the activation of AKT (11, 12). Loss of specific for PTEN mRNA inhibited the dephosphoryla- PTEN was also found to reduce responsiveness to EGFR– tion of AKT and downregulation of survivin induced by TKIs in patients with recurrent glioblastoma expressing erlotinib in EGFR mutation–positive NSCLC cells. These EGFR variant type III, a constitutively active mutant form results are consistent with our previous observation that of EGFR (29). In addition, restoration of PTEN expression inhibition of the EGFR–PI3K–AKT pathway contributed was shown to increase the susceptibility to EGFR–TKI- to downregulation of survivin expression by EGFR–TKIs induced apoptosis in EGFR mutation–positive NSCLC in EGFR mutation–positive NSCLC cells (13). Whereas the cells with PTEN loss (11). These observations thus support mechanism of survivin downregulation by EGFR–TKIs the notion that PTEN loss is associated with resistance to remains unclear, previous studies have suggested that the EGFR–TKIs in EGFR-driven tumors. PI3K–AKT pathway regulates survivin expression The precise molecular mechanism by which PTEN loss through modulation of transcriptional factors (30, 31). We induces resistance to EGFR–TKIs has remained unclear, confirmed that erlotinib downregulated the amount of

210 Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming EGFR–TKI Resistance in EGFR-Mutant NSCLC

A CD H1650 * H1650 3,000 Control siRNA: + + – – 40 H1650+YM155 ) Survivin siRNA: – – + + 3 Erlotinib: – + – + 30 Control Erlotinib 10 mg/kg 100 2,000 Survivin 20 YM155 5 mg/kg Erlotinib+YM155 75 BIM 10 50 1,000 Actin 0 Annexin V–positive cells (%) V–positive Annexin Control siRNA: + + – – 25

Survivin siRNA: – – + + (mm volume Tumor Erlotinib: – + – + 0 0 * Cell viability (% control) 0.01 0.1 1 10 100 0 10 20 30 H1650 Erlotinib (µmol/L) Time (days)

BEH1650 Control Erlotinib 10 mg/kg YM155 5 mg/kg Erlotinib+YM155 YM155: – – + + * Erlotinib: – + – + 30 pAKT AKT 20 Survivin pERK 10 ERK 0

BIM cells (%) V–positive Annexin Actin YM155: – – + + Erlotinib: – + – +

H1650

Figure 6. YM155 reverses erlotinib resistance induced by loss of PTEN in EGFR mutation–positive cells in vivo. A, H1650 cells were transfected with survivin or scrambled (control) siRNAs for 24 hours and then incubated in complete medium with or without erlotinib (2 mmol/L) for 24 hours (left) or 48 hours (right). Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to survivin, BIM, or b-actin (left), or the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry (right). B, cells were incubated in complete medium with or without erlotinib (2 mmol/L), YM155 (20 nmol/L), or both drugs for 24 hours (left) or 48 hours (right). Cell lysates were then prepared and subjected to immunoblot analysis with antibodies to the indicated proteins (left), or the proportion of apoptotic cells was determined by staining with fluorescein isothiocyanate–conjugated Annexin V and propidium iodide followed by flow cytometry (right). Data in A and B (left) are representative of or means SE (right) from 3 independent experiments. , P < 0.05 for the indicated comparisons. C, cells were cultured in complete medium containing the indicated concentrations of erlotinib with or without 10 nmol/L YM155 for 72 hours, after which cell viability was assessed. The number of viable cells is expressed as a percentage of the corresponding value for cells not exposed to erlotinib. Data are means from 3 independent experiments. D, nude mice with tumor xenografts established by subcutaneous injection of H1650 cells were treated with vehicle (control), erlotinib (10 mg/kg), YM155 (5 mg/kg), or the combination of erlotinib and YM155 as described in Materials and Methods. Tumor volume was measured at the indicated times after the onset of treatment. Data are means SE for 6 mice per group. , P < 0.05 for the combination of erlotinib plus YM155 versus control or either erlotinib or YM155 alone. E, representative mice treated as in D showing tumors at the end of the 31-day treatment period. survivin mRNA in EGFR mutation–positive NSCLC cells tributes to the persistence of survivin expression in the without PTEN loss (data not shown), suggesting that presence of erlotinib and to EGFR–TKI resistance in EGFR survivin downregulation by erlotinib is mediated, at least mutation–positive NSCLC cells. in part, at the transcriptional level. These results suggest In addition to the PI3K–AKT–survivin pathway, we that the PI3K–AKT pathway and its downstream tran- and others have previously shown that the induction of scriptional factors are targeted by EGFR–TKIs in the BIM expression through inhibition of the MEK–ERK regulation of survivin expression. pathway is important for EGFR–TKI-induced apoptosis In this study, we also showed that downregulation of (21–23, 32). Although erlotinib induced upregulation of survivin expression either by transfection with survivin the proapoptotic protein BIM in EGFR mutation–positive siRNA or by exposure to YM155 reversed in vitro or in vivo cells regardless of PTEN status, the proapoptotic effect of the resistance of EGFR mutation–positive NSCLC cells to erlotinib was not observed in cells with PTEN loss. These erlotinib induced by PTEN loss. On the contrary, similar to results suggest that the PI3K–AKT–survivin signaling EGFR mutation–positive cells without PTEN loss, the pathway might be more important than the MEK–ERK– combination of erlotinib and YM155 did not manifest a BIM pathway in the regulation of survival in EGFR muta- synergistic antiproliferative effect in NSCLC cells harbor- tion–positive cells with loss of PTEN. However, we found ing wild-type EGFR alleles (data not shown), suggesting that the addition of YM155 to erlotinib resulted in both the that sensitization to EGFR–TKIs by YM155 is specific for downregulation of survivin and the upregulation of BIM, EGFR–TKI-resistant cells with both an EGFR mutation leading to a greater increase in the number of apoptotic and PTEN loss. Our observations thus show that PTEN cells compared with that observed with either agent alone, loss activates AKT signaling and that this pathway con- in EGFR mutation–positive cells with PTEN loss. These

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 211

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Okamoto et al.

results suggest that, although the relative contributions of in vitro and in vivo. YM155, a small-molecule inhibitor of the PI3K–AKT–survivin and MEK–ERK–BIM signaling the expression of the antiapoptotic protein survivin, is pathways to cell survival may vary among cells, concom- currently in clinical development as the first survivin itant downregulation of survivin and upregulation of BIM suppressant (18, 19, 34–36). This drug was found to are necessary for the induction of a substantial level of exhibit a favorable safety tolerability profile and mod- apoptosis in EGFR mutation–positive cells. Nuclear fac- erate single-agent activity in a recent phase II trial with tor-kB and FAS receptor signaling were also recently patients with advanced, refractory NSCLC (37). Our shown to regulate EGFR–TKI sensitivity in EGFR muta- results now suggest that further studies of combination tion–positive NSCLC cells (33), suggesting that multiple therapy with YM155 and erlotinib are warranted in mechanisms underlie EGFR–TKI resistance. NSCLC patients with EGFR–TKI resistance induced by We have found that the persistent activation of PTEN loss. AKT–survivin signaling by PTEN loss represents a mechanism of primary or acquired resistance to erloti- Disclosure of Potential Conflicts of Interest nib in EGFR mutation–positive cells. Our results show that erlotinib downregulates survivin expression in No potential conflicts of interest were disclosed. EGFR–TKI-sensitive NSCLC cells but not in EGFR– TKI-resistant NSCLC cells with PTEN loss. Although Grant Support we previously showed that the expression of PTEN was reduced in tumor specimens from NSCLC patients This study was supported by KAKENHI (grants-in-aid for scientific research) from the Ministry of Education, Culture, Sports, Science, and showing acquired resistance to EGFR–TKIs (12), anal- Technology of Japan. ysis of serial tumor samples obtained before and during The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked treatment with these drugs will be required to deter- advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate mine whether survivin expression is affected by such this fact. treatment. Moreover, we found that YM155 significantly enhanced the antitumor effect of erlotinib in EGFR Received August 17, 2011; revised November 2, 2011; accepted mutation–positive NSCLC cells with PTEN loss both November 2, 2011; published OnlineFirst November 10, 2011.

References 1. Gazdar AF. Activating and resistance mutations of EGFR in non-small- 11. Sos ML, Koker M, Weir BA, Heynck S, Rabinovsky R, Zander T, et al. cell lung cancer: role in clinical response to EGFR tyrosine kinase PTEN loss contributes to erlotinib resistance in EGFR-mutant lung inhibitors. Oncogene 2009;28:S24–S31. cancer by activation of Akt and EGFR. Cancer Res 2009;69: 2. Engelman JA, Janne PA. Mechanisms of acquired resistance to 3256–61. inhibitors in non- 12. Yamamoto C, Basaki Y, Kawahara A, Nakashima K, Kage M, Izumi H, small cell lung cancer. Clin Cancer Res 2008;14:2895–9. et al. Loss of PTEN expression by blocking nuclear translocation of 3. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. EGR1 in gefitinib-resistant lung cancer cells harboring epidermal Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is growth factor receptor-activating mutations. Cancer Res 2010;70: associated with a second mutation in the EGFR kinase domain. PLoS 8715–25. Med 2005;2:e225–35. 13. Okamoto K, Okamoto I, Okamoto W, Tanaka K, Takezawa K, Kuwata 4. Koizumi F, Shimoyama T, Taguchi F, Saijo N, Nishio K. Establishment K, et al. Role of survivin in EGFR inhibitor-induced apoptosis in non- of a human non-small cell lung cancer cell line resistant to gefitinib. Int J small cell lung cancers positive for EGFR mutations. Cancer Res Cancer 2005;116:36–44. 2010;70:10402–10. 5. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, 14. Ryan BM, O'Donovan N, Duffy MJ. Survivin: a new target for anti- et al. MET amplification leads to gefitinib resistance in lung cancer by cancer therapy. Cancer Treat Rev 2009;35:553–62. activating ERBB3 signaling. Science 2007;316:1039–43. 15. Okabe T, Okamoto I, Tamura K, Terashima M, Yoshida T, Satoh T, et al. 6. Okamoto W, Okamoto I, Tanaka K, Hatashita E, Yamada Y, Kuwata K, Differential constitutive activation of the epidermal growth factor et al. TAK-701, a humanized monoclonal antibody to hepatocyte receptor in non-small cell lung cancer cells bearing EGFR gene growth factor, reverses gefitinib resistance induced by tumor-derived mutation and amplification. Cancer Res 2007;67:2046–53. HGF in non-small cell lung cancer with an EGFR mutation. Mol Cancer 16. Yamanaka K, Nakahara T, Yamauchi T, Kita A, Takeuchi M, Kiyonaga Ther 2011;9:2785–92. F, et al. Antitumor activity of YM155, a selective small-molecule 7. Yano S, Wang W, Li Q, Matsumoto K, Sakurama H, Nakamura T, et al. survivin suppressant, alone and in combination with docetaxel in induces gefitinib resistance of lung adeno- human malignant melanoma models. Clin Cancer Res 2011;17: carcinoma with epidermal growth factor receptor-activating muta- 5423–31. tions. Cancer Res 2008;68:9479–87. 17. Iwasa T, Okamoto I, Suzuki M, Nakahara T, Yamanaka K, Hatashita E, 8. Takeda M, Okamoto I, Fujita Y, Arao T, Ito H, Fukuoka M, et al. De novo et al. Radiosensitizing effect of YM155, a novel small-molecule survivin resistance to epidermal growth factor receptor-tyrosine kinase inhi- suppressant, in non-small cell lung cancer cell lines. Clin Cancer Res bitors in EGFR mutation-positive patients with non-small cell lung 2008;14:6496–504. cancer. J Thorac Oncol 2010;5:399–400. 18. Iwasa T, Okamoto I, Takezawa K, Yamanaka K, Nakahara T, Kita A, 9. Li L, Ross AH. Why is PTEN an important tumor suppressor?J Cell et al. Marked anti-tumour activity of the combination of YM155, a novel Biochem 2007;102:1368–74. survivin suppressant, and platinum-based drugs. Br J Cancer 10. Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the PI3K 2010;103:36–42. pathway as an integrator of multiple inputs during tumorigenesis. Nat 19. Nakahara T, Takeuchi M, Kinoyama I, Minematsu T, Shirasuna K, Rev Cancer 2006;6:184–92. Matsuhisa A, et al. YM155, a novel small-molecule survivin

212 Mol Cancer Ther; 11(1) January 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming EGFR–TKI Resistance in EGFR-Mutant NSCLC

suppressant, induces regression of established human hormone- 29. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ, refractory prostate tumor xenografts. Cancer Res 2007;67:8014–21. et al. Molecular determinants of the response of glioblastomas to 20. Kita A, Nakahara T, Yamanaka K, Nakano K, Nakata M, Mori M, et al. EGFR kinase inhibitors. N Engl J Med 2005;353:2012–24. Antitumor effects of YM155, a novel survivin suppressant, against 30. Guha M, Plescia J, Leav I, Li J, Languino LR, Altieri DC. Endogenous human aggressive non-Hodgkin lymphoma. Leuk Res 2011;35: tumor suppression mediated by PTEN involves survivin gene silencing. 787–92. Cancer Res 2009;69:4954–8. 21. Costa DB, Halmos B, Kumar A, Schumer ST, Huberman MS, Boggon 31. Lim M, Zhong C, Yang S, Bell AM, Cohen MB, Roy-Burman P. Runx2 TJ, et al. BIM mediates EGFR tyrosine kinase inhibitor-induced apo- regulates survivin expression in prostate cancer cells. Lab Invest ptosis in lung cancers with oncogenic EGFR mutations. PLoS Med 2010;90:222–33. 2007;4:1669–80. 32. Faber AC, Li D, Song Y, Liang MC, Yeap BY, Bronson RT, et al. 22. Cragg MS, Kuroda J, Puthalakath H, Huang DC, Strasser A. Gefitinib- Differential induction of apoptosis in HER2 and EGFR addicted can- induced killing of NSCLC cell lines expressing mutant EGFR requires cers following PI3K inhibition. Proc Natl Acad Sci U S A 2009;106: BIM and can be enhanced by BH3 mimetics. PLoS Med 2007;4: 19503–8. 1681–9. 33. Bivona TG, Hieronymus H, Parker J, Chang K, Taron M, Rosell R, et al. 23. Deng J, Shimamura T, Perera S, Carlson NE, Cai D, Shapiro GI, et al. FAS and NF-kappaB signalling modulate dependence of lung cancers Proapoptotic BH3-only BCL-2 family protein BIM connects death on mutant EGFR. Nature 2011;471:523–6. signaling from epidermal growth factor receptor inhibition to the 34. Nakahara T, Kita A, Yamanaka K, Mori M, Amino N, Takeuchi M, mitochondrion. Cancer Res 2007;67:11867–75. et al. Broad spectrum and potent antitumor activities of YM155, a 24. Robertson SC, Tynan J, Donoghue DJ. RTK mutations and human novel small-molecule survivin suppressant, in a wide variety of syndromes: when good receptors turn bad. Trends Genet 2000;16:368. human cancer cell lines and xenograft models. Cancer Sci 2011; 25. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, 102:614–21. et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the 35. Tolcher AW, Mita A, Lewis LD, Garrett CR, Till E, Daud AI, et al. Phase I growth of Bcr-Abl positive cells. Nat Med 1996;2:561–6. and pharmacokinetic study of YM155, a small-molecule inhibitor of 26. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR survivin. J Clin Oncol 2008;26:5198–203. mutations in lung cancer: correlation with clinical response to gefitinib 36. Satoh T, Okamoto I, Miyazaki M, Morinaga R, Tsuya A, Hasegawa Y, therapy. Science 2004;304:1497–500. et al. Phase I study of YM155, a novel survivin suppressant, in 27. Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. patients with advanced solid tumors. Clin Cancer Res 2009;15: Screening for epidermal growth factor receptor mutations in lung 3872–80. cancer. N Engl J Med 2009;361:958–67. 37. Giaccone G, Zatloukal P, Roubec J, Floor K, Musil J, Kuta M, et al. 28. Sierra JR, Cepero V, Giordano S. Molecular mechanisms of Multicenter phase II trial of YM155, a small-molecule suppressor of acquired resistance to tyrosine kinase . Mol Cancer survivin, in patients with advanced, refractory, non-small-cell lung 2010;9:75. cancer. J Clin Oncol 2009;27:4481–6.

www.aacrjournals.org Mol Cancer Ther; 11(1) January 2012 213

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst November 10, 2011; DOI: 10.1158/1535-7163.MCT-11-0638

Overcoming Erlotinib Resistance in EGFR Mutation−Positive Non− Small Cell Lung Cancer Cells by Targeting Survivin

Kunio Okamoto, Isamu Okamoto, Erina Hatashita, et al.

Mol Cancer Ther 2012;11:204-213. Published OnlineFirst November 10, 2011.

Updated version Access the most recent version of this article at: doi:10.1158/1535-7163.MCT-11-0638

Supplementary Access the most recent supplemental material at: Material http://mct.aacrjournals.org/content/suppl/2011/11/04/1535-7163.MCT-11-0638.DC1

Cited articles This article cites 37 articles, 17 of which you can access for free at: http://mct.aacrjournals.org/content/11/1/204.full#ref-list-1

Citing articles This article has been cited by 4 HighWire-hosted articles. Access the articles at: http://mct.aacrjournals.org/content/11/1/204.full#related-urls

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mct.aacrjournals.org/content/11/1/204. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mct.aacrjournals.org on September 24, 2021. © 2012 American Association for Cancer Research.