Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

13
Cancer Therapy: Preclinical Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma Xianbiao Xie 1,5 , Markus P.H. Ghadimi 1,5 , Eric D. Young 1,5 , Roman Belousov 1,5 , Quan-sheng Zhu 1,5 , Juehui Liu 1,5 , Gonzalo Lopez 2,5,6 , Chiara Colombo 1,5 , Tingsheng Peng 1,5 , David Reynoso 3,5 , Jason L. Hornick 7 , Alexander J. Lazar 4,5,6 , and Dina Lev 2,5,6 Abstract Purpose: Molecular deregulations underlying epithelioid sarcoma (ES) progression are poorly under- stood yet critically needed to develop new therapies. Epidermal growth factor receptor (EGFR) is over- expressed in ES; using preclinical models, we examined the ES EGFR role and assessed anti-ES EGFR blockade effects, alone and with mTOR inhibition. Experimental Design: EGFR and mTOR expression/activation was examined via tissue microarray (n ¼ 27 human ES specimens; immunohistochemistry) and in human ES cell lines (Western blot and quantitative reverse transcriptase PCR). Cell proliferation, survival, migration, and invasion effects of EGFR and mTOR activation treated with erlotinib (anti-EGFR small-molecule inhibitor) alone and combined with rapamycin were assessed in cell culture assays. In vivo growth effects of erlotinib alone or with rapamycin were evaluated using severe combined immunodeficient mouse ES xenograft models. Results: EGFR was expressed and activated in ES specimens and cell lines. EGFR activation increased ES cell proliferation, motility, and invasion and induced cyclin D1, matrix metalloproteinase (MMP) 2, and MMP9 expression. EGFR blockade inhibited these processes and caused significant cytostatic ES growth inhibition in vivo. mTOR pathway activation at varying levels was identified in all tissue microarray– evaluable ES tissues; 88% of samples had no or reduced PTEN expression. Similarly, both ES cell lines showed enhanced mTOR activity; VAESBJ cells exhibited constitutive mTOR activation uncoupled from EGFR signaling. Most importantly, combined erlotinib/rapamycin resulted in synergistic anti-ES effects in vitro and induced superior tumor growth inhibition in vivo versus single agent administration. Conclusions: EGFR and mTOR signaling pathways are deregulated in ES. Preclinical ES model–derived insights suggest that combined inhibition of these targets might be beneficial, supporting evaluations in clinical trials. Clin Cancer Res; 17(18); 5901–12. Ó2011 AACR. Introduction First described almost 50 years ago, epithelioid sarcoma (ES) is a malignancy exhibiting both epithelial (keratins and epithelial membrane antigen) and mesenchymal (most notably vimentin and CD34) differentiation mar- kers and remains a biological and clinical enigma (1). Both the cell lineage and cell of origin of ES are unclear. Recent insights into ES molecular underpinnings suggest that loss of INI1 expression, a component of a chromatin remodeling complex regulating transcription machinery, may be involved in disease inception (2). This distinct and uncommon soft tissue sarcoma (STS) histologic subtype is further classified into the distal and the more aggressive proximal subtypes, typically develops in young adults (third and fourth decades of life), and exhibits a propensity for local recurrence, lymphatic spread, and pulmonary metastasis (3, 4). Although complete surgical resection of localized tumors results in relatively favor- able 5-year outcome rates (75%–88%), local and systemic recurrences tend to occur later in the course of disease and underlie a dismal 10-year disease-specific survival rate of less than 50% (1, 3–5). Unresectable locally advanced and metastatic ES are usually chemoresistant and conse- quentially are generally fatal (3). No major improve- ments in the therapy of ES have been made since the early 1970s and novel therapeutic approaches are therefore critically needed. Authors' Affiliations: Departments of 1 Surgical Oncology, 2 Cancer Bi- ology, 3 Sarcoma Medical Oncology, and 4 Pathology, 5 Adult Sarcoma Research Center, The University of Texas MD Anderson Cancer Center; 6 The University of Texas Graduate School of Biomedical Sciences, Hous- ton, Texas; and 7 Department of Pathology, Brigham and Women's Hos- pital, Harvard Medical School, Boston, Massachusetts Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Dina Lev, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1104, Houston, TX 77030. Phone: 713-792-1637; Fax: 713-563-1185; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-0660 Ó2011 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 5901 on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Transcript of Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Page 1: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Cancer Therapy: Preclinical

Combining EGFR and mTOR Blockade for the Treatmentof Epithelioid Sarcoma

Xianbiao Xie1,5, Markus P.H. Ghadimi1,5, Eric D. Young1,5, Roman Belousov1,5, Quan-sheng Zhu1,5,Juehui Liu1,5, Gonzalo Lopez2,5,6, Chiara Colombo1,5, Tingsheng Peng1,5, David Reynoso3,5,Jason L. Hornick7, Alexander J. Lazar4,5,6, and Dina Lev2,5,6

AbstractPurpose: Molecular deregulations underlying epithelioid sarcoma (ES) progression are poorly under-

stood yet critically needed to develop new therapies. Epidermal growth factor receptor (EGFR) is over-

expressed in ES; using preclinical models, we examined the ES EGFR role and assessed anti-ES EGFR

blockade effects, alone and with mTOR inhibition.

Experimental Design: EGFR and mTOR expression/activation was examined via tissue microarray (n¼27 human ES specimens; immunohistochemistry) and in human ES cell lines (Western blot and

quantitative reverse transcriptase PCR). Cell proliferation, survival, migration, and invasion effects of

EGFR and mTOR activation treated with erlotinib (anti-EGFR small-molecule inhibitor) alone and

combined with rapamycin were assessed in cell culture assays. In vivo growth effects of erlotinib alone

or with rapamycin were evaluated using severe combined immunodeficient mouse ES xenograft models.

Results: EGFR was expressed and activated in ES specimens and cell lines. EGFR activation increased ES

cell proliferation, motility, and invasion and induced cyclin D1, matrix metalloproteinase (MMP) 2, and

MMP9 expression. EGFR blockade inhibited these processes and caused significant cytostatic ES growth

inhibition in vivo. mTOR pathway activation at varying levels was identified in all tissue microarray–

evaluable ES tissues; 88% of samples had no or reduced PTEN expression. Similarly, both ES cell lines

showed enhanced mTOR activity; VAESBJ cells exhibited constitutive mTOR activation uncoupled from

EGFR signaling. Most importantly, combined erlotinib/rapamycin resulted in synergistic anti-ES effects in

vitro and induced superior tumor growth inhibition in vivo versus single agent administration.

Conclusions: EGFR and mTOR signaling pathways are deregulated in ES. Preclinical ES model–derived

insights suggest that combined inhibition of these targets might be beneficial, supporting evaluations in

clinical trials. Clin Cancer Res; 17(18); 5901–12. �2011 AACR.

Introduction

First described almost 50 years ago, epithelioid sarcoma(ES) is a malignancy exhibiting both epithelial (keratinsand epithelial membrane antigen) and mesenchymal(most notably vimentin and CD34) differentiation mar-kers and remains a biological and clinical enigma (1). Both

the cell lineage and cell of origin of ES are unclear. Recentinsights into ES molecular underpinnings suggest thatloss of INI1 expression, a component of a chromatinremodeling complex regulating transcription machinery,may be involved in disease inception (2). This distinctand uncommon soft tissue sarcoma (STS) histologicsubtype is further classified into the distal and the moreaggressive proximal subtypes, typically develops in youngadults (third and fourth decades of life), and exhibits apropensity for local recurrence, lymphatic spread, andpulmonary metastasis (3, 4). Although complete surgicalresection of localized tumors results in relatively favor-able 5-year outcome rates (75%–88%), local and systemicrecurrences tend to occur later in the course of disease andunderlie a dismal 10-year disease-specific survival rate ofless than 50% (1, 3–5). Unresectable locally advancedand metastatic ES are usually chemoresistant and conse-quentially are generally fatal (3). No major improve-ments in the therapy of ES have been made sincethe early 1970s and novel therapeutic approaches aretherefore critically needed.

Authors' Affiliations: Departments of 1Surgical Oncology, 2Cancer Bi-ology, 3Sarcoma Medical Oncology, and 4Pathology, 5Adult SarcomaResearch Center, The University of Texas MD Anderson Cancer Center;6The University of Texas Graduate School of Biomedical Sciences, Hous-ton, Texas; and 7Department of Pathology, Brigham and Women's Hos-pital, Harvard Medical School, Boston, Massachusetts

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Dina Lev, Department of Cancer Biology, TheUniversity of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,Unit 1104, Houston, TX 77030. Phone: 713-792-1637; Fax: 713-563-1185;E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-0660

�2011 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org 5901

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 2: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

The rapidly increasing awareness of malignancy-relatedgenetic and epigenetic deregulations has driven the inclu-sion of biologically based treatment approaches as anintegral and desired component of anticancer therapeuticstrategies (6). Such tumor-tailored, target-orientatedapproaches are aimed specifically at molecules which areinvolved in the initiation and maintenance of a givencancer and are easily "druggable," thereby offering hopefor achieving the overarching goal of cancer therapy whichis to develop drugs that eliminate tumor cells while sparingnormal tissues (7). One molecular target that has attractedconsiderable investigative interest is the epidermal growthfactor receptor (EGFR), an HER receptor family member(HER1/EGFR, HER2/neu, HER3, and HER4; ref. 8). Uponligand binding, EGFR phosphorylation triggers the activa-tion of downstream signaling pathways involved in criticalcellular functions such as proliferation, survival, angiogen-esis (9). Functional EGFR dysregulation is frequently ob-served in human cancers, with overexpression andactivation by mutations or autocrine/paracrine growthfactor loops identified in approximately 50% of epithelialmalignancies, suggesting a pivotal role in tumorigenesisand disease progression (10). Similarly, increased EGFRexpression has been described as occurring in several STShistologic subtypes (11). Consequentially, significantefforts have been directed toward development and eval-uation of anti-EGFR therapeutic approaches, specificallysmall molecule tyrosine kinase inhibitors and receptor-blocking monoclonal antibodies (12). Multiple preclinicaland even clinical studies have shown therapeutic efficacy ofEGFR inhibition especially in lung, head and neck, andcolorectal carcinoma where such therapies have been in-tegrated into the therapeutic armamentarium (12).

Although not yet extensively investigated in the contextof ES, a recent report has identified enhanced EGFR ex-pression in a cohort of human ES tumor samples (13). Thisinitial finding has provided the impetus for the studiespresented here, seeking to ascertain EGFR and activatedEGFR (pEGFR) expression levels in an independent subsetof human ES specimens. Most importantly, the ES-associ-ated function of EGFR and the effects of an EGFR inhibitoron ES growth were evaluated using cellular and xenograftES models. Finally, we sought to also identify an EGFRblockade containing therapeutic combination whose anti-ES effects were superior compared with single agent EGFRinhibition.

Materials and Methods

Cell lines and reagentsThe human ES cell lines VAESBJ (American Type Culture

Collection) and Epi544 (developed in our laboratory) weremaintained and propagated as previously described (4).Several additional human cancer cell lines and normal cellprimary cultures were used as controls (see SupplementaryInformation). Authentication of ES cell lines was con-ducted utilizing short tandem repeat DNA fingerprinting(STR; Supplementary Data). Recombinant human EGF waspurchased from R&D Systems, the EGFR inhibitor erlotinib(Tarceva) from LC Laboratories, and the mTOR inhibitorrapamycin from the University of Texas MD AndersonCancer Center (UTMDACC) pharmacy. Commerciallyavailable antibodies were used for immunoblot or immu-nohistochemical detection of: pEGFR (Tyr1173), pERK(Thr202/Tyr404), ERK, pAKT (Ser473), AKT, p4EBP1(Thr70), 4EBP1, p70S6K (Thr389), 70S6K, pSRP(Ser235/236), CDK2, CDK4, PARP, and PTEN (all fromCell Signaling); MMP2, MMP9 (R&D Systems); Ki67(Thermo/Lab Vision); EGFR, cyclin D1, cyclin B1, p27,and b-actin (Santa Cruz Biotechnology); cyclin A1, cleavedPARP (Abcam); cleaved caspase 3 (Biocare Medical); and,INI1 (BD Transduction Laboratories).

ImmunohistochemistryA recently established tissue microarray (TMA) contain-

ing tissues retrieved from human ES surgical specimens wasused to immunohistochemically evaluate biomarker ex-pression (4). Of note, 8 breast adenocarcinoma specimenswere incorporated into this TMA as controls. TMA immu-nostaining and xenograft-derived specimen immunohis-tochemistry were conducted as previously described (14).For TMA analysis, most biomarkers (see exceptions de-scribed below) were scored by 2 independent observers(A.J. Lazar and X. Xie) after excluding samples with insuf-ficient tumor tissue. Intensity was graded as none (=0),weak/low (=1), or moderate-high (=2). This system wasmodified for Ki67 and cyclin D1, and the percentage ofpositive tumor nuclei was scored. For INI1, the tumors werescored as having nuclear expression intact or absent. As-sessment of PTEN categorized tumors as: 0, no expression;1, greatly reduced; and 2, relatively normal or increased

Translational Relevance

Epithelioid sarcomas (ES), while uncommon, areunfavorable malignancies affecting young adults witha marked propensity for local recurrence and distantmetastasis. Complete surgical resection, the only poten-tially curative therapy, is often not achievable, pointingto a critical need for more effective therapeutic strate-gies. Studies reported here suggest a role for epidermalgrowth factor receptor (EGFR) activation in promotingthe ES aggressive/metastatic phenotype and show EGFRblockade to induce anti-ES effects including abrogatedcell growth, survival, migration, and invasion. Further-more, mTOR pathway activation was found to com-monly occur in ES, possibly mediated, at least in part,by reduced or lost PTEN expression. Most importantly,dual targeting of EGFR and mTOR significantly andsynergistically abrogates ES growth in vitro and in vivo.Taken together, these data offer new insights into ESmolecular deregulations and support further evaluationof therapeutic combinations targeting EGFR and mTORin the ES clinical setting.

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5902

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 3: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

expression; intratumoral blood vessel endothelium wasused as an internal positive control.

Cellular assaysA panel of in vitro cell culture–based assays was used.

These included the following: MTS and clonogenicityassays to determine cell growth; propidium iodide (PI)staining and PI/Annexin V staining fluorescence-activatedcell-sorting (FACS) analyses to evaluate cell-cycle progres-sion and rate of apoptosis, respectively; and migration andinvasion assays to assess these respective cellular pheno-types. Western blot analyses were used to evaluate levels ofprotein expression and phosphorylation, and quantitativereal time PCR (qRT-PCR) was used to determine matrixmetalloproteinase (MMP) 2 and MMP9 mRNA expressionlevels. All these experiments were carried out as we havepreviously described (15); further information is availableas Supplementary Data.

In vivo animal modelsAll animal procedures/care were approved by UTM-

DACC Institutional Animal Care and Usage Committee.Animals received humane care as per the Animal WelfareAct and the NIH "Guide for the Care and Use of LaboratoryAnimals." Animal models were used as previouslydescribed (16). Information about the xenograft modeland therapeutic schemas are provided in SupplementaryData.

StatisticsCell culture–based assays were repeated at least twice;

mean � SD was calculated. Cell lines were examinedseparately. For outcomes that were measured at a singletime point, 2-sample Student’s t tests were used to assess thedifferences. To determine whether the cytotoxic interac-tions of erlotinib and rapamycin in ES cells were synergis-tic, additive, or antagonistic, drug effects were examinedusing the combination index (CI) method of Chou andTalalay (17–19). Briefly, the fraction affected (Fa) wascalculated from cell viability assays, and CIs were generatedusing CalcuSyn software (Biosoft). Differences in xenograftsize, weight, and lung weight in vivo were assessed using a2-tailed Student’s t test. Significance was set at P � 0.05.

Results

EGFR is highly expressed and activated in human ESBefore evaluating the potential utility of EGFR as an ES

therapeutic target, we sought to expand previous observa-tions of enhanced EGFR expression in this STS histologicsubtype (13). A preconstructed TMA containing human ESspecimens retrieved from 27 patients was used for immu-nohistochemical analysis (Fig. 1A and SupplementaryTable S2). Twenty of the evaluable specimens (77%)expressed EGFR (11 moderate to high expression and 9low); only 6 did not express EGFR, corroborating the abovedescribed previously published observation. Of potentialimportance, only 2 of the breast cancer samples included

on the TMA exhibited EGFR expression. To further deter-mine whether ES-expressed EGFR is activated we evaluatedthe expression of pEGFR in ES samples: 95% (19, 20) ofEGFR-expressing specimens exhibited positive staining atvarying levels whereas no EGFR phosphorylation wasobserved in only 1 sample (Fig. 1A). Taken together, theseresults suggest that the EGFR is both expressed andactivated in human ES.

Figure 1. EGFR is highly expressed and activated in human ES. A,representative photographs of ES TMA EGFR and pEGFR (Tyr1173)immunostaining (original images were captured at �200 magnification)depicting low and high expressing tumors. B, Western blot analysesshowing INI1 loss and pEGFR and EGFR expression in protein extracts ofhuman ES cell lines (VAESBJ and Epi544) grown under regular serumcontaining conditions and compared with expression in normal cellcontrols (NHDF and HC-SMC) and other cancer cell lines [SW872(liposarcoma), SKLMS1 (leiomyosarcoma), MESA (uterine sarcoma), andA549 (lung carcinoma)]. C, Western blot analyses depicting enhancedEGFR phosphorylation and downstream signaling in response to EGF.Cells were grown in serum-free media overnight prior to EGF stimulation.Interestingly, a high basal level of pAKT was noted in VAESBJ cells evenunder serum-free conditions. EGF stimulation induced only a minimalincrease in pAKT expression in VAESBJ cells in contrast to the markedpAKT induction observed in Epi544 cells.

Targeted Therapy for Epithelioid Sarcoma

www.aacrjournals.org Clin Cancer Res; 17(18) September 15, 2011 5903

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 4: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Next, we analyzed the expression of EGFR in 2 human EScell lines available to us. Immunoblotting showed loss ofINI1 expression in both of these cell lines as compared withnormal human cells (NHDF and HC-SMC) as well as apanel of randomly selected cancer cell lines representingboth sarcomas (SW872, SKLMS1, and MESA) and carcino-mas (A549), a finding which supports their ES origin(Fig. 1B). Increased EGFR expression was noted in ES cellsas compared with normal controls at levels comparablewith those observed in several other cancer cell lines(Fig. 1B). Increased EGFR expression was similarly notedin 2 human ES primary cultures (early passages; Supple-mentary Fig. S1); both these cell strains exhibit INI1 proteinloss confirming their ES origin. Under serum-containinggrowth conditions, pEGFR expressionwas found in both EScell lines although it was more pronounced in VAESBJ ascompared with Epi544. We evaluated whether ES cell–expressed EGFR can be further activated with the additionof an appropriate ligand. Toward that end, cells werecultured under serum-free conditions overnight and werethen stimulated with EGF; a marked increase in EGFRphosphorylation was observed and a consequent activationof the downstream extracellular signal–regulated kinase(ERK) signaling pathways was found in both cell lines(Fig. 1C). Interestingly, even under serum-free conditions,we noted a high basal level of pAKT in VAESBJ cells andEGF stimulation induced only a minimal additional phos-phorylation in contrast to the marked pAKT inductionobserved in Epi544 cells. These initial findings showedthat EGFR is both highly expressed and functional inhuman ES cell lines, thereby providing a rationale forexamining the effect of EGFR activation and blockade onthe ES protumorigenic phenotype.

EGFR activation induces the proliferation, cell-cycleprogression, migration, and invasion of ES cells

The functional effects of EGFR activation on ES cells werenext examined. A significant (P < 0.05) increase in ES cellgrowth was observed after EGF stimulation as perMTS (100ng/mL/24 h) and clonogenicity (100 ng/mL/14 d) assays(Fig. 2A). EGF induced G1 cell-cycle progression in serum-starved ES cells possibly, at least in part, through increase inthe expression of the cell-cycle regulator cyclinD1 (Fig. 2B).Immunohistochemical analysis of the ES TMA also showedincreased nuclear cyclin D1 expression in human tumors(Fig. 2B and Supplementary Table S2), possibly suggestingthat a functional EGFR signaling network is operative in situalthough additional mechanisms enhancing cyclin D1expression might also be at play. Western blot analysesidentified EGF to affect the expression of several additionalG1 cell-cycle regulators; increased cyclin A1, cyclin B1,cyclin-dependent kinase (CDK) 2, and CDK4 and de-creased p27 expression were noted, especially in theEpi544 cell line (Supplementary Fig. S1)

Modified Boyden chamber assays were conducted toevaluate effects of EGF on cell migration and invasion.To assure that the effects seen were not secondary to EGFimpact on cell proliferation, experiments were limited to 6

hours. As depicted in Figure 2C, EGF enhanced the migra-tion and invasion of both ES cell lines tested (P < 0.05).Metalloproteinases play an important role in cellular in-vasive capacity; consequently, we evaluated the effect ofEGFR activation on both MMP2 and MMP9 expression. Asignificant increase in the mRNA levels of these 2 enzymeswas noted after 24 hours of EGF stimulation (Fig. 2D). As inthe case of cyclin D1, MMP2 and MMP9 were found to becommonly expressed in human ES specimens upon im-munohistochemical staining of TMA (Fig. 2D). In summa-ry, EGFR signaling was found to enhance ES proliferation,cell-cycle progression, migration, and invasion—all func-tions critical for tumor growth and progression.

EGFR blockade inhibits ES cell growth in vitro andin vivo

The orally available small molecule EGFR inhibitorerlotinib was used to evaluate the impact of EGFR blockadein our ES preclinical models. ES cells were treated withincremental drug doses for 4 hours; decreases in EGF-induced pEGFR were observed even at the lowest dose(1 mmol/L) tested (Fig. 3A). Furthermore, EGFR blockaderesulted in a dose-dependent inhibition of pERK in bothcell lines tested (Fig. 3A). Interestingly, while a markedeffect on the phosphorylation of AKT and its downstreammTOR-regulated effectors P70S6 kinase and 4EBP1 wasobserved in Epi544 cells, only a limited effect on thissignaling pathway was seen in VAESBJ cells (Fig. 3A).

Functionally, a dose-dependent decrease in ES cellgrowth in response to erlotinib (96 hours) was observedin both cell lines, although more pronouncedly in Epi544(Fig. 3B). Similarly, EGFR blockade inhibited the colonyformation capacity of ES cells (Fig. 3B). Next, the effects ofEGFR inhibition on cell-cycle progression and apoptosiswere evaluated. Erlotinib treatment (24 hours) resulted in aG1 cell-cycle arrest in both cell lines (Fig. 3B). This effectcould, at least in part, be secondary to the observeddecrease in cyclin D1 expression in treated cells(Fig. 3C). Furthermore, PI/Annexin V staining FACS anal-ysis showed a significant (P < 0.05) increase in apoptosisinduced by erlotinib in the ES cells after 96 hours (Fig. 3C).Concordantly, increased levels of PARP cleavage werenoted in both cell lines in response to EGFR blockade(96 hours) further confirming the presence of treatment-induced apoptosis.

To evaluate the effects of erlotinib on ES cell migrationand invasion while taking into account the effects of thisinhibitor on ES cell growth and survival, assays wereconducted with cells pretreated with erlotinib for 4 hours(only viable cells were used). A significant decrease inEGF-stimulated migration and invasion was identified(P < 0.05); qRT-PCR showed a marked decrease inMMP2 and MMP9 mRNA levels (Fig. 3D). Taken together,these results suggest that EGFRblockade results in decreasedES cell proliferation, G1 cell-cycle arrest, enhanced apopto-sis, and abrogated migration and invasion capacities.

On the basis of the above findings, we next sought toevaluate whether the impact of EGFR blockade can also be

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5904

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 5: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Figure 2. EGFR activation enhances ES cell growth, cell-cycle progression, migration, and invasion. A, a significant increase in ES cell growth wasobserved after EGF stimulation as per MTS (top) and clonogenicity assays (bottom). B, EGF stimulation induced G1 cell-cycle progression in serum-starvedES cells (PI staining FACS analysis; top). Western blotting showed increase in cyclin D1 expression in response to EGF (middle). Representative photographsof ES TMA cyclin D1 immunostaining depicting low and high expressing tumors (bottom). C, enhanced ES cell migration and invasion in responseto EGF stimulation was identified using modified Boyden chamber assays. D, EGF stimulation enhances MMP2 and MMP9 mRNA expression (qRT-PCR)in ES cells (top). Representative photographs of ES TMA MMP2 and MMP9 immunostaining depicting low and high expressing tumors (bottom).Graphs represent the average of at least 2 repeated experiments � SD; *, statistically significant effects (P < 0.05).

Targeted Therapy for Epithelioid Sarcoma

www.aacrjournals.org Clin Cancer Res; 17(18) September 15, 2011 5905

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 6: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Figure 3. EGFR inhibition exerts anti-ES effects in vitro. A, Western blot analysis identified EGFR phosphorylation inhibition in both cell lines response toerlotinib treatment. Furthermore, EGFR blockade resulted in a dose-dependent inhibition of pERK. Interestingly, whereas a marked effect on AKTphosphorylation and its downstream mTOR-regulated effectors p70S6 kinase (p70S6K) and 4EBP1 was observed in Epi544 cells, only a limited effect on thissignaling pathway was seen in VAESBJ cells. B, erlotinib induced a dose-dependent decrease in ES cell growth (MTS; top) which was more pronounced inEpi544 cells. Similarly, EGFR blockade inhibited the colony formation capacity of ES cells (top middle). Furthermore, erlotinib treatment resulted in a G1

cell-cycle arrest in both cell lines (PI staining FACS analysis; bottom middle) and a dose-dependent decrease in cyclin D1 expression (Western blot; bottom).C, PI/Annexin V staining FACS analyses showed a significant increase in ES cell apoptosis induced by erlotinib (top). Concordantly, increased levels ofPARP cleavage were noted in both cell lines (Western blot; bottom). D, a significant decrease in EGF-stimulated ES cell migration and invasion wasobserved in response to erlotinib (top). qRT-PCR showed a marked decrease in MMP2 and MMP9 mRNA levels (bottom). Graphs represent the averageof at least 2 repeated experiments � SD; *, statistically significant effects (P < 0.05).

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5906

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 7: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

observed in vivo. Using a recently published ES xenograftanimal model resulting from the subcutaneous injection ofVAESBJ cells (4), we compared the effect of erlotinib ontumor growth in severe combined immunodeficient micewith the impact of control vehicle administration. Therapywas initiated after tumor establishment (�5mm in greatestdimension). Mice in both groups were followed for tumorsize and toxicity; treatment was terminated when tumors incontrol group reached an average of 1.5 cm in largestdimension. Treatment with erlotinib induced significant(P ¼ 0.002) tumor growth delay as compared with controlvehicle–treated tumors (Fig. 4A). Average tumor weightsrecorded at termination of the study were control group:1.19 � 0.45 g versus erlotinib group: 0.63 � 0.3 g(P < 0.05; Fig. 4A).Next, formalin-fixed, paraffin-embedded tumor sections

from mice of both study arms were immunohistochemi-cally evaluated. Decrease in pEGFR expression in erlotinib-treated tumors confirmed EGFR activation blockade in vivo(Fig. 4B); as expected, no decrease in the total EGFRexpression level was observed. A decrease in the numberof Ki67 (a nuclear marker for proliferation)-expressingtumor cells and an increase in the number of cleavedcaspase 3 (marker for apoptosis)-positive cells were ob-served in erlotinib-treated samples (Fig. 4B). Furthermore,as was found in vitro, erlotinib induced a decrease in cyclinD1, MMP2, and MMP9 expression in vivo (Fig. 4B).

Taken together, the impact of EGFR blockade on ESxenograftsmirrors the effects noted in cell culture.However,taking into account the increasing body of clinical evidence,suggesting only minimal effects of compounds inducingEGFR blockade as single anticancer agents (20, 21), and ourdata showing tumor growthdelay but not tumor abrogationin response to erlotinib suggested that it might be pertinentto identify EGFR blockade containing therapeutic combina-tions that possessed superior anti-ES effects.

mTOR pathway is commonly deregulated in ESWe sought to identify additional targets that could be

blocked in combination with EGFR blockade as a treatmentstrategy for ES. Insights from experiments conducted abovehave suggested evaluating the potential deregulation of theAKT/mTOR in ES. As depicted in Figure 5A, both ES celllines express pAKT and activated mTOR-regulated down-stream targets. However, expression levels are markedlymore pronounced in VAESBJ cells where pAKT expressionis shown to be independent of serum conditions (Fig. 5A).In addition, 1 of the 2 ES cell strains showed pronouncedAKT/mTOR activation (Supplementary Fig. S1). Phosphor-ylation of the AKT/mTOR pathway in Epi544 cells isdecreased under serum-free culture conditions, suggestingthat its activation might possibly occur via upstreamsignaling. VAESBJ-expressed pAKT/p4EBP1/pP70S6K levelsare similar to those observed in sarcoma cells exhibiting

Figure 4. EGFR blockade delays ES growth in vivo. A, VAESBJ xenografts (once average size tumor reached �5 mm in largest dimension) were treatedwith erlotinib (n ¼ 10) or vehicle control (n ¼ 8). Erlotinib-treated mice exhibited slower growth (left graph; P ¼ 0.002) and a significantly decreased tumorweight at study termination (*, P < 0.05; right graph) as compared with control treated mice. B, immunohistochemical staining confirmed decreased pEGFRexpression in erlotinib-treated tumors without significant change in total EGFR expression. Erlotinib-treated tumors exhibited decreased proliferation(measured by Ki67), increased apoptosis [measured via cleaved caspase 3 (CC3)], and decreased cyclin D1, MMP2, and MMP9 expression (originalimages were captured at �200 magnification).

Targeted Therapy for Epithelioid Sarcoma

www.aacrjournals.org Clin Cancer Res; 17(18) September 15, 2011 5907

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 8: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

loss of PTEN (SW872 and MESA; ref. 22) and although notcompletely lost, PTEN expression in VAESBJ cells is muchlower than in Epi544 cells (Fig. 5A).

To confirm that mTOR deregulation is a bona fidecommon event in human ES and that the results above

are not just reflective of cell culture artifact, we deter-mined the expression of p4EBP1 and pSRP (surrogatebiomarkers for mTOR pathway activation) in our TMA(Fig. 5B and Supplementary Table S2). p4EBP1 and pSRPexpression was 100% in both the samples (sample

Figure 5. mTOR pathway is commonly deregulated in ES. A, Western blot analyses showed increased AKT/mTOR activation in ES cells which is morepronounced in VAESBJ cells as compared with Epi544 (top). VAESBJ pAKT expression level is independent of serum conditions (bottom) whereas in Epi544cells, it is decreased under serum-free culture conditions (bottom). VAESBJ-expressed pAKT/p4EBP1/pP70S6K levels are similar to those observed insarcoma cells exhibiting loss of PTEN (SW872 and MESA) and while not completely lost, PTEN expression in VAESBJ cells is lower than in Epi544 cells (top).B, representative photographs of ES TMA pSRP and p4EBP1 immunostaining depicting low and high expressing tumors (top). PTEN immunohistochemistry(bottom) showed decreased to absent PTEN expression in the majority of ES samples as compared with levels observed in normal cells and othercancer tissues included on the TMA (PTEN expression level in a breast sample is depicted for comparison). C, rapamycin blocks mTOR signaling in EScell lines (Western blot; left) and inhibits tumor cell growth (MTS; middle) and colony formation capacity (right).

Figure 6. Combined EGFR and mTOR targeting results in superior anti-ES effects as compared with monotherapy. A, combining erlotinib and rapamycinresulted in inhibition of both EGFR and mTOR signaling (Western blot). B, dual blockade resulted in a markedly superior ES cell growth inhibition as comparedwith each agent alone (MTS, P < 0.05; top). Isobologram analysis (middle) revealed that the growth inhibitory effect of this therapeutic combination is stronglysynergistic in both cell lines, although more pronouncedly in VAESBJ cells, with CI < 0.5 for most combinations tested. Similarly, combination therapy induceda superior inhibitory effect on colony formation as compared with each agent alone (bottom). C, the most pronounced G1 cell-cycle arrest was found tooccur in response to erlotinib and rapamycin combination (PI staining FACS analysis; top) as is also reflected in the most significant decrease in cyclin D1expression observed in response to this regimen (Western blot; top middle). Similarly, erlotinib and rapamycin combination resulted in a more pronouncedapoptotic rate as compared with monotherapy (PI/Annexin V staining FACS analysis; bottommiddle) and PARP cleavage (bottom). D, the impact of combinedtherapy was assessed in vivo using VAESBJ xenografts growing in SCID mice. Both erlotinib and rapamycin as single agents inhibited tumor growth ascompared with control (P < 0.05; left and middle top; v, vehicle; R, rapamycin; E, erlotinib; C, combination). Most importantly, combination therapy resulted insignificant growth abrogation as compared with erlotinib, rapamycin, or control (P < 0.05). Combination-treated mice exhibited the most significant decreasein tumor weight as comparedwith all other therapeutic groups (right top). Immunohistochemical analyses showed a decrease in Ki67-positive staining cells andan increase in cleaved caspase 3 (CC3)-positive cells most pronounced in combination-treated tumors (bottom). *, statistically significant effects (P < 0.05).

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5908

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 9: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

Targeted Therapy for Epithelioid Sarcoma

www.aacrjournals.org Clin Cancer Res; 17(18) September 15, 2011 5909

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 10: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

1: moderate-high ¼ 78%, low ¼ 22%; sample 2: moder-ate-high ¼ 87%, low ¼ 13%). Furthermore, complete lossof PTEN expression in tumor cells was found in 40% of ESspecimens. Interestingly, of those samples expressingPTEN (n ¼ 15), 80% exhibited a relative low stainingintensity whereas in only 20% expression was noted thatwas equivalent to the level observed in intratumoral andsurrounding normal cells (lymphocytes and endothelialcells) used as controls (Fig. 5B and SupplementaryTable S1). These findings suggest that PTEN deregulationis a common molecular aberration in human ES. Similarto ES expression pattern, pSRP and p4EBP1 were com-monly expressed in the breast cancer samples evaluated:all 8 specimens were found to express moderate-highlevels of pSRP; low p4EBP1 expression was noted in 1breast cancer sample whereas moderate-high expressionwas found in 7 samples. None of these samples exhibitedloss of PTEN expression (reduced expression as comparedwith normal stroma was noted in 3 cases), suggesting thatother molecular mechanisms possibly drive AKT/mTORactivation in these tumors.

To further confirm that mTOR activation has a function-al role in ES, we sought to test the effects of the mTORinhibitor rapamycin on tumor cell growth. As shown inFigure 5C, rapamycin blocked mTOR signaling in ES celllines. A rapamycin dose-dependent decrease (one thatplateaus after 10 nmol/L) in tumor cell growth and colonyformation capacity was observed in both cell lines. Of note,VAESBJ cells exhibited amore pronounced sensitivity to thedrug possibly secondary to PTEN loss (Fig. 5C). In sum, ourdata suggest that mTOR pathway activation is a commonmolecular deregulation in human ES contributing to tumorcell growth, thereby providing the rationale for testingtherapeutic combinations that target this pathway in con-junction with EGFR blockade.

Combining EGFR and mTOR blockade results insuperior anti-ES effects

We tested the effects of erlotinib and rapamycin combi-nations on ES cells. As anticipated and depicted in Figure6A, combining these 2 compounds resulted in the inhibi-tion of both EGFR and mTOR signaling. Of importance,this therapeutic combination abrogated rapamycin "feed-back" induction of AKT phosphorylation. To determineeffects on cell growth, ES cells were treated for 96 hourswith increasing doses of erlotinib alone, rapamycin alone,or with different dose combinations of these 2 drugs. Inboth cell lines, combined therapy resulted in markedlysuperior effects as compared with each agent alone(P < 0.05; Fig. 6B). We further determined whether erlo-tinib and rapamycin interactions were additive or syner-gistic. Isobologram analysis revealed that the growthinhibitory effect of these drugs was strongly synergisticin both cell lines although more pronouncedly in VAESBJcells, with CI < 0.5 for most combinations tested (Fig. 6Band Supplementary Table S3). Similarly, combinationtherapy induced a superior inhibitory effect on colonyformation as compared with each agent alone (Fig. 6B).

Although each compound individually leads to increasedES G1 cell-cycle arrest, the most pronounced effect wasobserved after combination therapy (24 hours; Fig. 6C).Similarly, the most significant decrease in cyclin D1expression was noted in combination-treated cells(Fig. 6C). To determine the effect of the therapeutic com-bination on cell death PI/Annexin V staining FACS analyseswere conducted after 96 hours of treatment: no apoptosiswas found in response to rapamycin alone whereas similarto the findings described above, erlotinib did result in EScell apoptosis. However, a significantly higher apoptoticrate was observed after combination therapy (P < 0.05;Fig. 6C). This observation was further strengthened by themarked increase in PARP cleavage also selectively observedafter combination therapy (Fig. 6C).

Finally, a 4-armed therapeutic study was conducted todetermine the comparative impact on tumor growth in vivo,assessing the effect of combination treatment to each drugalone or vehicle control. Both erlotinib and rapamycin assingle agents inhibited VAESBJ xenograft growth as com-pared with control (P < 0.05; Fig. 6D). Most importantly,combination therapy resulted in significant growth abro-gation as compared with erlotinib, rapamycin, or controlalone (P < 0.05). Average tumor weights recorded attermination of the study were control group: 1.21 � 0.4g; erlotinib group: 0.53 � 0.3 g; rapamycin group: 0.38 �0.09 g; and combination group: 0.15 � 0.11 g (Fig. 6D).Immunohistochemical analysis showed a decrease in Ki67-positive staining cells and an increase in cleaved caspase 3–positive cells which was most pronounced in combinationtreatment tumors. These data suggest that EGFR blockadein combination withmTOR inhibition results in significantanti-ES effects in vitro and in vivo, a finding of potentialclinical utility.

Discussion

The current study highlights several ES-associated mo-lecular deregulations of potential translational and clinicalimportance. First, a recent observation (13) identifyingEGFR overexpression as commonly occurring in ES wasvalidated and was further expanded to show receptoractivation in human ES samples. Moreover, a role for EGFRactivation in supporting the malignant phenotype of ESwas shown. Most importantly, EGFR blockade was foundto inhibit the growth of ES cells in vitro and in vivo, inducingarrested cell-cycle progression, enhanced apoptosis, andabrogated tumor cell migration and invasion. While thelimitations of only a small cohort of ES cell lines availablefor testing should be taken into consideration, thesefindings are encouraging and possibly support furtherinvestigation of EGFR blockade in the clinical ES context.ClinicalTrials.gov (http://clinicaltrials.gov/NCT00148109)reports a currently active phase II clinical trial using anEGFR monoclonal antibody for the treatment of metastaticor locally advanced bone and STSs. It is not certain whetherany ES patients were included in this study; however, onthe basis of observations made here, it might be useful to

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5910

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 11: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

establish the response rate of this subcohort, perhaps as aseparate evaluation. Moreover, it is pertinent that a largebody of evidence suggests that despite promising responsesto monotherapy EGFR manipulation in multiple preclin-ical epithelial cancer models in which the receptor is over-expressed (23, 24), the clinical therapeutic benefit isminimal, even with high levels of EGFR expression withinthe tumor, and is mostly observed in tumors harboring agenetic deregulation of the receptor, that is, amplificationsand activating mutations (11, 25). Furthermore, eventumors harboring such genetic modifications are boundto develop resistance to EGFR blockade (26). It is of notethat no EGFR amplifications or gene mutations werepreviously identified in ES specimens (13). These findingssuggest that enhanced EGFR expression and activation perse are not sufficient to predict response to EGFR blockadeand that additional molecular deregulations may possiblydecrease the efficacy of EGFR inhibitors as single agents.In the past few years, we have learned that rationalcombinations of targeted therapeutics may achieve morepotent antitumor effects and help overcome initialtherapeutic resistance (16); by implication, identifyingother ES-associated molecular deregulations that can betargeted in combination with EGFR might be of greatbenefit.The pivotal mTOR pathway, a convergence downstream

node of a large number of molecular processes includingEGFR signaling, has been shown to play a major role incancer progression and metastasis (27). mTOR is known toact as amaster switch for cellular catabolismand anabolism,enhancing cancer cell growth and proliferation (27, 28). Inaddition, mTOR can induce cell-cycle progression, enhancecell survival, and block cell death as dictated by the cellularcontext and specific downstream targets (27, 28). Rapamy-cin and its derivatives that block themTORC1complex havebeen developed and are currently being evaluated in severalclinical trials for a variety of hematologic and solid malig-nancies (29, 30). To the best of our knowledge, no previousstudies have evaluated mTOR deregulation and function inES. Our findings identify ubiquitous phosphoinositide-3-kinase (PI3K)/mTOR pathway activation in human ESspecimens (to levels similar to those found in breast cancersamples) and cell lines possiblymediated, at least in part, byreduced or lost expression of the negative regulator PTEN,although additional molecular mechanisms are potentiallyrelevant. Of potential importance, we found that the ES cellline exhibiting PTEN loss (VAESBJ) wasmost pronouncedlysensitive to rapamycin. To that end, it is possible that PTENloss should be further evaluated as a molecular biomarkerfor appropriate patient selection for inclusion in mTORblockade–based human ES clinical trials. Reduced PTENexpression in cancer can occur through genomic alterationsor epigenetic modifications such as DNAmethylation (31).Loss of INI1, the product of the hSNF5/SMARCB1/BAF47gene, occurs in the vast majority of ES (2). INI1 is acomponent of the hSWI/SNF chromatin remodeling com-plex regulating gene transcription by modulating nucleo-somal structures in an ATP-dependent manner (32). The

possible role this epigenetic modifier plays in the activationof the PI3K/mTOR pathway and in decreased PTEN expres-sion is currently unknown and should be further explored.

Interestingly, response to EGFR inhibitors has beenshown to inversely correlate with PI3K/mTOR signalingoveractivation (33–36). For example, breast cancer cellsexhibiting enhanced mTOR activation secondary to PTENloss were found to be EGFR blockade resistant; PTENreintroduction into these cells results in increased responseto EGFR inhibition (33). Similarly, loss of PTEN has beenfound to predict resistance to EGFR blockade in colorectalcancer, lung cancer, and glioblastoma (34–36); our studiesexpand these observations to now include ES—a tumorlacking effective systemic treatment. Although erlotinibinhibited the growth of both available ES cell lines, a morelimited response was observed in VAESBJ cells. These cellsexhibit uncoupling of EGFR from its downstream PI3K/mTOR signaling, a higher level of constitutive PI3K/mTORactivation, and consequently survive independent of EGFR.Multiple preclinical studies have shown that dual targetingof EGFR and mTOR results in enhanced anticancer effectsin several different cancer types (37–39). Such an approachwas shown to be effective in experimental models exhi-biting relative sensitivity to EGFR blockade as a singleagent and also for targeting resistant cells where mTORblockade restores therapeutic sensitivity (37, 39). In lightof these findings, several phase I/II trials combining EGFRand mTOR blockade for the treatment of a variety ofepithelial cancers have recently been initiated (Clinical-Trials.gov). In the present study, we found EGFR inhibi-tion in combination with mTOR blockade to exert asynergistic anti-ES effect in vitro and in vivo. This combi-nation resulted in significant ES cell growth arrest, inhi-bition of cell-cycle progression, enhanced apoptosis, andblocked migration and invasion. Together, these datasupport the inclusion of ES patients in future EGFR/mTOR blockade clinical trials.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We appreciate the expert assistance provided by Mr. Paul Cuevas in thepreparation and submission of themanuscript andMs. Kim Vu for her aid infigure preparation.

Grant Support

The manuscript was supported in part by an NIH/NCI RO1CA138345 (to D.Lev), an Amschwand Foundation seed grant (to D. Lev), a Chinese scholarshipcouncil grant (to X. Xie), a Deutsche Forschungsgemeinschaft training grant (toM.P.H. Ghadimi), an UTMDACC Physician-Scientist grant (to A.J. Lazar), and anAIRC fellowship grant (to C. Colombo). MDACC cell line characterization CoreFacility was further supported by a National Cancer Institute Cancer Centersupport grant (CA16672).

The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received March 11, 2011; revised July 21, 2011; accepted July 25, 2011;published OnlineFirst August 5, 2011.

Targeted Therapy for Epithelioid Sarcoma

www.aacrjournals.org Clin Cancer Res; 17(18) September 15, 2011 5911

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 12: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

References1. Halling AC, Wollan PC, Pritchard DJ, Vlasak R, Nascimento AG.

Epithelioid sarcoma: a clinicopathologic review of 55 cases. MayoClin Proc 1996;71:636–42.

2. Hornick JL, Dal Cin P, Fletcher CD. Loss of INI1 expression ischaracteristic of both conventional and proximal-type epithelioidsarcoma. Am J Surg Pathol 2009;33:542–50.

3. Wolf PS, Flum DR, Tanas MR, Rubin BP, Mann GN. Epithelioidsarcoma: the University of Washington experience. Am J Surg2008;196:407–12.

4. Sakharpe A, Lahat G, Gulamhusein T, Liu P, Bolshakov S, Nguyen T,et al. Epithelioid sarcoma and unclassified sarcoma with epithelioidfeatures: clinicopathological variables, molecular markers, and a newexperimental model. Oncologist 2011;16:512–22.

5. Fisher C. Epithelioid sarcoma of Enzinger. Adv Anat Pathol 2006;13:114–21.

6. Morse DL, Gillies RJ. Molecular imaging and targeted therapies.Biochem Pharmacol 2010;80:731–8.

7. Blagosklonny MV. Target for cancer therapy: proliferating cells orstem cells. Leukemia 2006;20:385–91.

8. Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growthfactor-related peptides and their receptors in human malignancies.Crit Rev Oncol Hematol 1995;19:183–232.

9. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level.Nat Rev Mol Cell Biol 2006;7:505–16.

10. Hynes NE, Stern DF. The biology of erbB-2/neu/HER-2 and its role incancer. Biochim Biophys Acta 1994;1198:165–84.

11. Dobashi Y, Takei N, Suzuki S, Yoneyama H, Hanawa M, Ooi A.Aberration of epidermal growth factor receptor expression in boneand soft-tissue tumors: protein overexpression, gene amplificationand activation of downstream molecules. Mod Pathol 2004;17:1497–505.

12. Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N EnglJ Med 2008;358:1160–74.

13. Cascio MJ, O’Donnell RJ, Horvai AE. Epithelioid sarcoma expressesepidermal growth factor receptor but gene amplification and kinasedomain mutations are rare. Mod Pathol 2010;23:574–80.

14. Zou C, Smith KD, Liu J, Lahat G, Myers S, Wang WL, et al. Clinical,pathological, and molecular variables predictive of malignant periph-eral nerve sheath tumor outcome. Ann Surg 2009;249:1014–22.

15. Jin Z, Lahat G, Korchin B, Nguyen T, Zhu QS, Wang X, et al. Midkineenhances soft-tissue sarcoma growth: a possible novel therapeutictarget. Clin Cancer Res 2008;14:5033–42.

16. Ren W, Korchin B, Lahat G, Wei C, Bolshakov S, Nguyen T, et al.Combined vascular endothelial growth factor receptor/epidermalgrowth factor receptor blockade with chemotherapy for treatmentof local, uterine, and metastatic soft tissue sarcoma. Clin Cancer Res2008;14:5466–75.

17. Chou TC. Preclinical versus clinical drug combination studies. LeukLymphoma 2008;49:2059–80.

18. Chou TC, Talalay P. Generalized equations for the analysis of inhibi-tions of Michaelis-Menten and higher-order kinetic systems with twoormoremutually exclusive and nonexclusive inhibitors. Eur J Biochem1981;115:207–16.

19. Reynolds CP, Maurer BJ. Evaluating response to antineoplastic drugcombinations in tissue culture models. Methods Mol Med 2005;110:173–83.

20. Rich JN, Reardon DA, Peery T, Dowell JM, Quinn JA, Penne KL, et al.Phase II trial of gefitinib in recurrent glioblastoma. J Clin Oncol2004;22:133–42.

21. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al.Erlotinib plus gemcitabine compared with gemcitabine alone inpatients with advanced pancreatic cancer: a phase III trial of theNational Cancer Institute of Canada Clinical Trials Group. J Clin Oncol2007;25:1960–6.

22. Zhu QS, Ren W, Korchin B, Lahat G, Dicker A, Lu Y, et al. Soft tissuesarcoma cells are highly sensitive to AKT blockade: a role for p53-independent up-regulation of GADD45 alpha. Cancer Res2008;68:2895–903.

23. Gridelli C, Bareschino MA, Schettino C, Rossi A, Maione P, CiardielloF. Erlotinib in non-small cell lung cancer treatment: current status andfuture development. Oncologist 2007;12:840–9.

24. Galizia G, Lieto E, De Vita F, Orditura M, Castellano P, Troiani T, et al.Cetuximab, a chimeric human mouse anti-epidermal growth factorreceptor monoclonal antibody, in the treatment of human colorectalcancer. Oncogene 2007;26:3654–60.

25. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA,Brannigan BW, et al. Activating mutations in the epidermal growthfactor receptor underlying responsiveness of non-small-cell lungcancer to gefitinib. N Engl J Med 2004;350:2129–39.

26. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al.Acquired resistance of lung adenocarcinomas to gefitinib or erlotinibis associated with a second mutation in the EGFR kinase domain.PLoS Med 2005;2:e73.

27. Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancertherapy. Nat Rev Cancer 2004;4:335–48.

28. Schmelzle T, Hall MN. TOR, a central controller of cell growth. Cell2000;103:253–62.

29. Chapuis N, Tamburini J, Green AS, Willems L, Bardet V, Park S, et al.Perspectives on inhibiting mTOR as a future treatment strategy forhematological malignancies. Leukemia 2010;24:1686–99.

30. Fouladi M, Laningham F, Wu J, O'Shaughnessy MA, Molina K,Broniscer A, et al. Phase I study of everolimus in pediatric patientswith refractory solid tumors. J Clin Oncol 2007;25:4806–12.

31. Soria JC, Lee HY, Lee JI, Wang L, Issa JP, Kemp BL, et al. Lack ofPTEN expression in non-small cell lung cancer could be related topromoter methylation. Clin Cancer Res 2002;8:1178–84.

32. Carlson M, Laurent BC. The SNF/SWI family of global transcriptionalactivators. Curr Opin Cell Biol 1994;6:396–402.

33. She QB, Solit D, Basso A, Moasser MM. Resistance to gefitinib inPTEN-null HER-overexpressing tumor cells can be overcome throughrestoration of PTEN function or pharmacologic modulation of consti-tutive phosphatidylinositol 30-kinase/Akt pathway signaling. Clin Can-cer Res 2003;9:4340–6.

34. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ,et al. Molecular determinants of the response of glioblastomas toEGFR kinase inhibitors. N Engl J Med 2005;353:2012–24.

35. Sos ML, Koker M, Weir BA, Heynck S, Rabinovsky R, Zander T, et al.PTEN loss contributes to erlotinib resistance in EGFR-mutantlung cancer by activation of Akt and EGFR. Cancer Res 2009;69:3256–61.

36. Jhawer M, Goel S, Wilson AJ, Montagna C, Ling YH, Byun DS, et al.PIK3CA mutation/PTEN expression status predicts response of coloncancer cells to the epidermal growth factor receptor inhibitor cetux-imab. Cancer Res 2008;68:1953–61.

37. Bianco R, Garofalo S, Rosa R, Damiano V, Gelardi T, Daniele G, et al.Inhibition of mTOR pathway by everolimus cooperates with EGFRinhibitors in human tumours sensitive and resistant to anti-EGFRdrugs. Br J Cancer 2008;98:923–30.

38. Chiu CW, Nozawa H, Hanahan D. Survival benefit with proapoptoticmolecular and pathologic responses from dual targeting of mamma-lian target of rapamycin and epidermal growth factor receptor in apreclinical model of pancreatic neuroendocrine carcinogenesis. J ClinOncol 2010;28:4425–33.

39. Wang MY, Lu KV, Zhu S, Dia EQ, Vivanco I, Shackleford GM, et al.Mammalian target of rapamycin inhibition promotes response toepidermal growth factor receptor kinase inhibitors in PTEN-defi-cient and PTEN-intact glioblastoma cells. Cancer Res 2006;66:7864–9.

Xie et al.

Clin Cancer Res; 17(18) September 15, 2011 Clinical Cancer Research5912

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660

Page 13: Combining EGFR and mTOR Blockade for the Treatment of Epithelioid Sarcoma

2011;17:5901-5912. Published OnlineFirst August 5, 2011.Clin Cancer Res   Xianbiao Xie, Markus P.H. Ghadimi, Eric D. Young, et al.   Epithelioid SarcomaCombining EGFR and mTOR Blockade for the Treatment of

  Updated version

  10.1158/1078-0432.CCR-11-0660doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2011/08/05/1078-0432.CCR-11-0660.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/18/5901.full#ref-list-1

This article cites 39 articles, 14 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/18/5901.full#related-urls

This article has been cited by 5 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]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/17/18/5901To request permission to re-use all or part of this article, use this link

on April 9, 2019. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 5, 2011; DOI: 10.1158/1078-0432.CCR-11-0660