phosphoproteinpathwaymapping:akt/mammaliantarget...

11
Phosphoprotein Pathway Mapping: Akt/Mammalian Target of Rapamycin Activation Is Negatively Associated with Childhood Rhabdomyosarcoma Survival Emanuel F. Petricoin III, 1 Virginia Espina, 2 Robyn P. Araujo, 2 Brieanne Midura, 3 Choh Yeung, 3 Xiaolin Wan, 3 Gabriel S. Eichler, 4 Donald J. Johann, Jr., 2 Stephen Qualman, 5 Maria Tsokos, 2 Kartik Krishnan, 3 Lee J. Helman, 3 and Lance A. Liotta 2 1 Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Cellular and Gene Therapy; 2 Laboratory of Pathology, 3 Department of Pediatric Oncology, and 4 Genomics and Bioinformatics Group, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland; and 5 Department of Pathology, Columbus Children’s Hospital, Columbus, Ohio Abstract Mapping of protein signaling networks within tumors can identify new targets for therapy and provide a means to stratify patients for individualized therapy. Despite advances in combination chemotherapy, the overall survival for childhood rhabdomyosarcoma remains f60%. A critical goal is to identify functionally important protein signaling defects associated with treatment failure for the 40% nonresponder cohort. Here, we show, by phosphoproteomic network analysis of microdissected tumor cells, that interlinked components of the Akt/mammalian target of rapamycin (mTOR) pathway exhibited increased levels of phosphorylation for tumors of patients with short-term survival. Specimens (n = 59) were obtained from the Children’s Oncology Group Intergroup Rhabdomyosarcoma Study (IRS) IV, D9502 and D9803, with 12- year follow-up. High phosphorylation levels were associated with poor overall and poor disease-free survival: Akt Ser 473 (overall survival P < 0.001, recurrence-free survival P < 0.0009), 4EBP1 Thr 37/46 (overall survival P < 0.0110, recur- rence-free survival P < 0.0106), eIF4G Ser 1108 (overall survival P < 0.0017, recurrence-free survival P < 0.0072), and p70S6 Thr 389 (overall survival P < 0.0085, recurrence-free survival P < 0.0296). Moreover, the findings support an altered interrela- tionship between the insulin receptor substrate (IRS-1) and Akt/mTOR pathway proteins (P < 0.0027) for tumors from patients with poor survival. The functional significance of this pathway was tested using CCI-779 in a mouse xenograft model. CCI-779 suppressed phosphorylation of mTOR downstream proteins and greatly reduced the growth of two different rhabdomyosarcoma (RD embryonal P = 0.00008; Rh30 alveolar P = 0.0002) cell lines compared with controls. These results suggest that phosphoprotein mapping of the Akt/mTOR pathway should be studied further as a means to select patients to receive mTOR/IRS pathway inhibitors before administration of chemotherapy. [Cancer Res 2007;67(7):3431–40] Introduction Rhabdomyosarcoma arises from undifferentiated mesenchymal cells bearing skeletal muscle features (1, 2). Rhabdomyosarcoma is the most common soft tissue sarcoma in children, consisting of three histologic subtypes—alveolar, embryonal, and botyroid. Despite the recent advances in combination chemotherapy, and the molecular knowledge of the translocations t(2;13)(q35;q14) and t(1;13)(p36;q14) in alveolar rhabdomyosarcoma, the overall survival of all patients with childhood rhabdomyosarcoma has remained in the range of 60% to 70% (3, 4). The Children’s Oncology Group (COG) completed a series of treatment protocols evaluating the activity of a combination regimen using vincristine, actinomycin D and cyclophosphamide (VAC), VA plus ifosfamide (VAI), or VI and etoposide (VIE) in newly diagnosed children who presented with nonmetastatic rhabdomyosarcoma (3, 4). IRS-IV accrued patients between 1991 and 1997 with patients randomized to chemotherapy regimens (except for those with group I paratesticular and group I/II orbital primary tumors and patients with preexisting renal dysfunction). Additionally, patients were randomized to conventional or hyper- fractionated radiotherapy for patients with group III disease. No difference in outcome was seen among the chemotherapy regimens or between the radiotherapy regimens for group III patients (4). D9502 was a pilot study to assess the feasibility of cyclophos- phamide dose intensification during the first 12 weeks of VAC therapy for patients with intermediate risk rhabdomyosarcoma. Dose intensification therapy did not result in outcome differences compared with VAC with lower cyclophosphamide cycle doses (3). D9803 is an on-going study comparing VAC to VAC alternating with vincristine, topotecan, and cyclophosphamide for patients with intermediate risk rhabdomyosarcoma. The overall disease-free survival rate in these studies was 67%, but the newer VA plus ifosfamide or VI and etoposide combina- tions did not improve overall outcome and survival compared with the standard VAC regimen (3, 4). Unfortunately, there is no way to identify the 33% of children destined to fail initial therapy, regardless of disease stage or histologic subtype. On the other hand, the 60% to 70% of children that respond to standard therapy do so exceedingly well, with a vast majority of these patients currently living disease-free. Consequently, an urgent clinical goal is to identify functionally important molecular networks associated with the 30% to 40% nonresponder rhabdomyosarcoma subjects to develop new treatment strategies for this group. Because kinases are important drug targets, kinase network information could Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Current address for E.F. Petricoin III, V. Espina, R.P. Araujo, and L.A. Liotta: George Mason University, Center for Applied Proteomics and Molecular Medicine, Manassas, VA 20110. Requests for reprints: Virginia Espina, George Mason University, Center for Applied Proteomics and Molecular Medicine, 10900 University Boulevard MS 1A9, Manassas, VA 20110. Phone: 703-993-8062; Fax: 703-993-8606; E-mail: vespina@ gmu.edu. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-1344 www.aacrjournals.org 3431 Cancer Res 2007; 67: (7). April 1, 2007 Research Article Research. on January 2, 2020. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 17-Sep-2019

0 views

Category:

Documents


0 download

TRANSCRIPT

Phosphoprotein Pathway Mapping: Akt/Mammalian Target

of Rapamycin Activation Is Negatively Associated with

Childhood Rhabdomyosarcoma Survival

Emanuel F. Petricoin III,1Virginia Espina,

2Robyn P. Araujo,

2Brieanne Midura,

3Choh Yeung,

3

Xiaolin Wan,3Gabriel S. Eichler,

4Donald J. Johann, Jr.,

2Stephen Qualman,

5Maria Tsokos,

2

Kartik Krishnan,3Lee J. Helman,

3and Lance A. Liotta

2

1Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Cellular and Gene Therapy; 2Laboratory ofPathology, 3Department of Pediatric Oncology, and 4Genomics and Bioinformatics Group, Laboratory of MolecularPharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland; and5Department of Pathology, Columbus Children’s Hospital, Columbus, Ohio

Abstract

Mapping of protein signaling networks within tumors canidentify new targets for therapy and provide a means tostratify patients for individualized therapy. Despite advancesin combination chemotherapy, the overall survival forchildhood rhabdomyosarcoma remains f60%. A critical goalis to identify functionally important protein signaling defectsassociated with treatment failure for the 40% nonrespondercohort. Here, we show, by phosphoproteomic network analysisof microdissected tumor cells, that interlinked components ofthe Akt/mammalian target of rapamycin (mTOR) pathwayexhibited increased levels of phosphorylation for tumors ofpatients with short-term survival. Specimens (n = 59) wereobtained from the Children’s Oncology Group IntergroupRhabdomyosarcoma Study (IRS) IV, D9502 and D9803, with 12-year follow-up. High phosphorylation levels were associatedwith poor overall and poor disease-free survival: Akt Ser473

(overall survival P < 0.001, recurrence-free survival P <0.0009), 4EBP1 Thr37/46 (overall survival P < 0.0110, recur-rence-free survival P < 0.0106), eIF4G Ser1108 (overall survivalP < 0.0017, recurrence-free survival P < 0.0072), and p70S6Thr389 (overall survival P < 0.0085, recurrence-free survival P <0.0296). Moreover, the findings support an altered interrela-tionship between the insulin receptor substrate (IRS-1) andAkt/mTOR pathway proteins (P < 0.0027) for tumors frompatients with poor survival. The functional significance of thispathway was tested using CCI-779 in a mouse xenograft model.CCI-779 suppressed phosphorylation of mTOR downstreamproteins and greatly reduced the growth of two differentrhabdomyosarcoma (RD embryonal P = 0.00008; Rh30 alveolarP = 0.0002) cell lines compared with controls. These resultssuggest that phosphoprotein mapping of the Akt/mTORpathway should be studied further as a means to selectpatients to receive mTOR/IRS pathway inhibitors beforeadministration of chemotherapy. [Cancer Res 2007;67(7):3431–40]

Introduction

Rhabdomyosarcoma arises from undifferentiated mesenchymalcells bearing skeletal muscle features (1, 2). Rhabdomyosarcoma isthe most common soft tissue sarcoma in children, consisting ofthree histologic subtypes—alveolar, embryonal, and botyroid.Despite the recent advances in combination chemotherapy, andthe molecular knowledge of the translocations t(2;13)(q35;q14) andt(1;13)(p36;q14) in alveolar rhabdomyosarcoma, the overall survivalof all patients with childhood rhabdomyosarcoma has remainedin the range of 60% to 70% (3, 4).

The Children’s Oncology Group (COG) completed a series oftreatment protocols evaluating the activity of a combinationregimen using vincristine, actinomycin D and cyclophosphamide(VAC), VA plus ifosfamide (VAI), or VI and etoposide (VIE) innewly diagnosed children who presented with nonmetastaticrhabdomyosarcoma (3, 4). IRS-IV accrued patients between 1991and 1997 with patients randomized to chemotherapy regimens(except for those with group I paratesticular and group I/II orbitalprimary tumors and patients with preexisting renal dysfunction).Additionally, patients were randomized to conventional or hyper-fractionated radiotherapy for patients with group III disease.No difference in outcome was seen among the chemotherapyregimens or between the radiotherapy regimens for group IIIpatients (4).

D9502 was a pilot study to assess the feasibility of cyclophos-phamide dose intensification during the first 12 weeks of VACtherapy for patients with intermediate risk rhabdomyosarcoma.Dose intensification therapy did not result in outcome differencescompared with VAC with lower cyclophosphamide cycle doses (3).D9803 is an on-going study comparing VAC to VAC alternatingwith vincristine, topotecan, and cyclophosphamide for patientswith intermediate risk rhabdomyosarcoma.

The overall disease-free survival rate in these studies was 67%,but the newer VA plus ifosfamide or VI and etoposide combina-tions did not improve overall outcome and survival compared withthe standard VAC regimen (3, 4). Unfortunately, there is no way toidentify the 33% of children destined to fail initial therapy,regardless of disease stage or histologic subtype. On the otherhand, the 60% to 70% of children that respond to standard therapydo so exceedingly well, with a vast majority of these patientscurrently living disease-free. Consequently, an urgent clinical goal isto identify functionally important molecular networks associatedwith the 30% to 40% nonresponder rhabdomyosarcoma subjects todevelop new treatment strategies for this group. Because kinasesare important drug targets, kinase network information could

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

Current address for E.F. Petricoin III, V. Espina, R.P. Araujo, and L.A. Liotta: GeorgeMason University, Center for Applied Proteomics and Molecular Medicine, Manassas,VA 20110.

Requests for reprints: Virginia Espina, George Mason University, Center forApplied Proteomics and Molecular Medicine, 10900 University Boulevard MS 1A9,Manassas, VA 20110. Phone: 703-993-8062; Fax: 703-993-8606; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-1344

www.aacrjournals.org 3431 Cancer Res 2007; 67: (7). April 1, 2007

Research Article

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

become the basis of therapeutic strategies for improving treatmentoutcome (5).

Although gene microarrays can provide important informationabout somatic genetic taxonomy, they are unable to provide aneffective recapitulation of the posttranslational, fluctuating signal-ing events that occur at the proteomic level. The phosphorylation,or activation state, of kinase-driven signaling networks containsimportant information concerning both the disease pathogenesisas well as potential for therapeutic target selection (5, 6). It is forthis reason that modulation of ongoing cellular kinase activityrepresents one of the most rapidly growing arenas in new drugdevelopment (7). The phosphorylation status of proteins can bedetected and measured using specific antiphosphoprotein anti-bodies. Antibodies have been developed to specifically recognizethe phosphorylated isoform of kinase substrates. Through the useof these phosphospecific antibodies, it is now possible to evaluate

the state of entire portions of a signaling pathway or cascade,although the cell is lysed, by looking at dozens of kinase substratesat once through multiplexed phosphospecific antibody analysis(5, 8, 9). Applying these antibodies to reverse-phase protein arraysprovides the opportunity to profile the state of the ongoing cellularsignaling events within small numbers of human tumor cellsobtained by biopsy (5, 9, 10). Sandwich two-site antibody pairs arenot available for most phosphorylated epitopes. The advantage ofthe reverse phase arrays is that this format requires only oneantibody to measure each phosphorylated epitope.

We applied the reverse phase array phosphoprotein pathwaymapping to evaluate the hypothesis that prosurvival pathways,including Akt/mammalian target of rapamycin (mTOR), aredifferentially activated for rhabdomyosarcoma tumors associatedwith a poor versus favorable outcome. A secondary goal was tofunctionally test the effect of treatment on the pathway we found to

Figure 1. Characteristics of the rhabdomyosarcoma sample sets. A, two independent study sets, sets 1A and 1B, were evaluated by reverse phase protein microarrayto profile the state of cellular signaling proteins. B, survival analysis of rhabdomyosarcoma study sets 1A and 1B. Overall survival and recurrence-free survival forboth heterogeneous study sets was not significantly different by a Kaplan-Meier survival estimate (set 1A, red line ; set 1B, blue line ; overall survival log-rank P = 0.2111and recurrence-free survival P = 0.5824). C, histologic subtype did not show a significant difference between the study sets (embryonal, blue line ; alveolar,red line ; Kaplan-Meier overall survival log-rank P = 0.4103 and recurrence-free survival P = 0.4312).

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3432 www.aacrjournals.org

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

be activated in a xenograft model of human rhabdomyosarcoma.This study shows the importance of phosphoprotein cell signalingevents and how they may constitute a new and functionally relevantanalyte for deriving therapeutic insights for rhabdomyosarcoma.

Materials and Methods

Specimens and patient data. All specimens (n = 59) and relevantclinical data were obtained from the Intergroup Rhabdomyosarcoma Study(IRS) IV, D9502 and D9803, studies from the COG with appropriateinstitutional review board approval (3, 4).

All specimens were snap frozen in liquid nitrogen and procured beforetherapy. The sample set was analyzed in two groups, 1A and 1B (Fig. 1A).Figure 1B to C shows the survival characteristics for the two study sets.Samples were anonymized and blinded as to clinical survival outcome beforefinal data analysis. The samples representing study set 1A (Fig. 1A) consistedof nine snap-frozen surgical specimens and 290 frozen section slides for 33different patients with a pathologic diagnosis of rhabdomyosarcoma. Allpatients used here had stage III (tumors <5 cm or regional lymph nodeinvolvement) disease and group III tumors (gross residual disease remainingafter treatment) before study entry. An additional set of 26 frozen sectionsamples and clinical data were provided by the COG for patients from thesame protocols (Fig. 1A). Pathologic diagnosis was rendered before therapy.An independent board-certified pathologist verified the diagnosis beforelaser capture microdissection. The histologic subtypes represented alveolar,

embryonal, botryoid, and mixed morphologic types. Pure tumor cell popu-lations were microdissected from the tissue sections with a PixCell II lasercapture microdissection instrument (Molecular Devices, Sunnyvale, CA).

Reverse phase protein microarrays. The microdissected cells (5, 9, 10)were subjected to lysis and reverse phase protein microarrays were printed

in duplicate with the whole-cell protein lysates as described by Sheehan

et al. (10). Briefly, the lysates were printed on glass backed nitrocellulose

array slides (FAST Slides Whatman, Florham Park, NJ) using a GMS 417

arrayer (Affymetrix, Santa Clara, CA) equipped with 500 Am pins. Each

lysate was printed in a dilution curve representing neat, 1:2, 1:4, 1:8, 1:16,

and negative control dilutions. The slides were stored with desiccant

(Drierite, W.A. Hammond, Xenia, OH) at �20jC before immunostaining.

Protein microarray immunostaining. Immunostaining was done on an

automated slide stainer per manufacturer’s instructions (Autostainer CSA

kit, DAKO, Carpinteria, CA). Each slide was incubated with a single primary

antibody at room temperature for 30 min. Polyclonal primary antibodies

were as follows: glycogen synthase kinase-3 (GSK3) a/h Tyr279/216

(Invitrogen-Biosource, Carlsbad, CA), BCL-2, HIF-1a (BD, Franklin Lakes,

NJ), 4EBP1, FKHR ser256, eIF4E, eIF4E ser209, eIF4G, eIF4G Ser1108, IGFR-h,

IRS-1, IRS-2, IRS-1 Ser612, SGK, Bak, Bax, BAD, BAD Ser112, BAD Ser136, BAD

Ser155, B-Raf, mTOR, mTOR Ser2448, p70S6 Thr389, p70S6 kinase, p70S6 Ser371,

S6 kinase Ser240/244, Akt, Akt Ser473, Akt Thr308, 4EBP1 Ser65, 4EBP1 Ser70, and

4EBP1 Thr37/46 (Cell Signaling Technology, Danvers, MA). The negative

control slide was incubated with antibody diluent. Secondary antibody was

goat anti-rabbit IgG H+L (1:5,000; Vector Labs, Burlingame, CA).

Figure 2. Exploratory data analysis of rhabdomyosarcoma study set 1A. A, unsupervised Bayesian clustering of normalized protein end points (columns ) indicatedtwo major clusters of tumors (rows ). These clusters seem unrelated to clinical parameters in (B). The two clusters were compared by Fisher’s exact test, P < 0.05.Five samples were excluded from analysis due to values below the limit of detection, precluding sample ranking.

Akt/mTOR Pathway and Rhabdomyosarcoma Survival

www.aacrjournals.org 3433 Cancer Res 2007; 67: (7). April 1, 2007

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Bioinformatics method for microarray analysis. Each array was

scanned; spot intensity was analyzed; data were normalized; and a

standardized, single data value was generated for each sample on the

array (Image Quant v5.2, GE Healthcare, Piscataway, NJ). Spot intensity was

integrated over a fixed area. Local area background intensity was calculated

for each spot with the unprinted adjacent slide background. This resulted in

a single data point for each sample, for comparison with every other spot on

the array. The Ward method for two-way hierarchical clustering was done

using JMP v5.0 (SAS Institute, Cary, NC). Wilcoxon two-sample rank sum

test was used to compare values between two groups. P values <0.05 were

considered significant. When we could not assume a normal distribution of

the variables, we used nonparametric methods. We used Kaplan-Meier (log-

rank) survival estimates for univariate survival analysis.

In vivo xenograft tumor model. Animal studies were done in

accordance with guidelines of the NIH Animal Care and Use Committee.

Female 4- to 6-week-old beige-severe combined immunodeficient (SCID)mice were purchased from Charles River Laboratories (Wilmington, MA).

Two million viable cells harvested from mid confluent cultures of either

Rh30 alveolar or RD embryonal cells in 0.2 mL diluent (5% Tween 80, 5%polyethylene glycol 400; Sigma, St. Louis, MO) were injected orthotopically

into the gastrocnemius muscle in the left hind leg, and after 1 week mice

were randomly assigned to control (n = 8) or CCI-779 treatment groups

(n = 8). Mice were treated i.p. every 3 days for 30 consecutive days with20 mg/kg/i.p. of CCI-779 (Developmental Therapeutics Program, National

Cancer Institute and Wyeth, Madison, NJ) or vehicle alone. Tumor growth

was measured every 3 days with calipers, and tumor volume was calculated

by the formula V (mm3) = a � b2, where a is the longest tumor axis and b isthe shortest tumor axis. All mice were sacrificed by asphyxiation with CO2

and underwent necropsy for confirmation of tumor growth. Tumors were

excised and snap frozen at �80jC until analysis.

Results

Exploratory data analysis of rhabdomyosarcoma tumor set1A. Enrichment of tumor cells by laser capture microdissectionwas done before analysis to ensure that the cells for analysis camefrom within the cancer cell population, without contamination bynoncancer cells (5, 11). For study set 1A (n = 33), 15 specificsignaling proteins (Fig. 2A) were initially chosen for reverse phaseprotein microarray analysis because they constituted a broad surveyof multiple prosurvival related events related to the Akt/mTORpathways that have been shown to play a role in rhabdomyosar-coma (12, 13). Unsupervised hierarchical clustering analysis of the15 protein end points revealed two major classes of tumors: onecluster with Akt/mTOR activation/phosphorylation and the otherwith a comparatively low level of signaling (Fig. 2A). After clinicaloutcome data was obtained from the COG, these two clusters werecompared by Fisher’s exact test based on patient characteristics ofage, sex, primary site, histology, invasion, and lymph nodeinvolvement (Fig. 2B ; ref. 14). Although none of the characteristicsreached P < 0.05 statistical significance, patients with para-meningeal head and neck primary site tumors comprised 62% ofcluster 1, whereas cluster 2 had 27% of patients with parameningealprimary site tumors (Fisher’s exact test, P = 0.06). Additionally,cluster 2 contained 73% alveolar tumors, whereas cluster 1 had 62%embryonal tumors (Fisher’s exact test, P = 0.06). Typically, patientswith embryonal rhabdomyosarcoma tumors from orbital or non-parameningeal sites have the best prognosis (15). These two clusterswere not statistically different for commonly accepted prognostic/clinical factors associated with rhabdomyosarcoma.

We proceeded to correlate the protein analyte values withdisease-free and overall survival clinical outcome data provided bythe COG for study set 1A. A clear partitioning of the tumorsemerged after clinical outcome data was obtained from the COG. Adecision tree analysis of three proteins—4EBP1, phosphorylated4EBP1 Thr37/46, and eIF4E—all components of the Akt/mTORpathway, partitioned patients who were in continuous completeremission from those who recurred and died after being treated

Figure 3. Reverse phase protein microarray kinase pathway profiling results forrhabdomyosarcoma sample set 1A. A, 4EBP1 and 4EBP1 Thr37/46 showed astatistically significant correlation for segregation of nonsurvivor and survivorstatus in study set 1A (4EBP1 Wilcoxon one-way m2, 0.0064; degree of freedom,1; n = 32; nonsurvivor mean 4EBP1, 82.4; SE, 11.48; survivor mean 4EBP1,145.61; SE, = 14.89; 4EBP1 Thr37/46 m2, 0.0135, degree of freedom, 1; n = 33).B, decision tree analysis indicated 4EBPI was a discriminator for survival in studyset 1A; therefore, Kaplan-Meier survival estimates for 4EBP1 were calculated.The Kaplan-Meier plots indicated relatively high levels of 4EBP1 (red line ) had asignificant statistical correlation with overall survival [log-rank P < 0.0177, n = 32(data not usable for one sample)] and recurrence-free survival (P = 0.0370)compared with samples with relatively low 4EBP1 levels (blue line ) for patientsat time of last contact.

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3434 www.aacrjournals.org

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

with standard therapy (data not shown). Among these end points,4EBP1 and 4EBP1 Thr37/46 individually were found to besignificantly correlated with survival by Wilcoxon one-way analysis,4EBP1 (P < 0.0064) and 4EBP1 Thr37/46 (P < 0.0135; Fig. 3A). A log-rank univariate survival analysis (Kaplan-Meier) supported theassociation of 4EBP1 with outcome in overall and recurrence-freesurvival in study set 1A (Fig. 3B ; overall survival P = 0.0177,recurrence-free survival P = 0.0370; ref. 16).

For recurrence-free survival in study set 1A, 4EBP1 level (P2 =0.0074; hazard ratio, 7.44; 95% confidence interval, 1.71–32.36)emerged as significant prognostic factor (17, 18). Thus, for study set1A (Figs. 1–3), individual components within the Akt/mTORpathway seemed to correlate with survival.

Disease-free and overall survival in rhabdomyosarcomapatients is associated with phosphorylated components ofthe Akt and mTOR pathways. Based on the findings of study set1A, an independent set of samples (set 1B, Fig. 1A) were obtainedfrom COG (n = 26) for analysis of an expanded set of proteinsassociated with the Akt/mTOR pathway. Univariate log-rankanalysis of the two heterogeneous sample sets (set 1A and 1B)revealed no significant difference in overall or recurrence-freesurvival by sample set (overall survival P = 0.2111, recurrence-freesurvival P = 0.5824; Fig. 1B) or histology (overall survival P = 0.4103,recurrence-free survival P = 0.4312; Fig. 1C). We analyzed set 1B bylaser capture microdissection and reverse phase protein microarrayas in set 1A. We expanded the number of end points to 27 to include

Figure 4. Reverse phase protein microarray kinase pathway profiling results for rhabdomyosarcoma sample set 1B. A, Kaplan-Meier survival analysis showedstatistically significant correlation in both overall and recurrence-free survival by log-rank analysis in set 1B for Akt Ser473 (overall survival P < 0.001, recurrence-freesurvival P < 0.0009), eIF4G Ser1108 (overall survival P < 0.0017, recurrence-free survival P < 0.0072), 4EBP1 Thr37/46 (overall survival P < 0.0110,recurrence-free survival P < 0.0106), and p70S6 Thr389 (overall survival P < 0.0085, recurrence-free survival P < 0.0296) for patients at time of last contact. Red line,relatively high levels of the indicated protein end point; blue line, relative low levels. B, protein end points evaluated by reverse phase protein microarray forrhabdomyosarcoma sample set 1B (open columns, survivor status; filled columns, nonsurvivor status). 4EBP1 Thr37/46 (P < 0.0348), GSK3a/h Tyr279/216 (P < 0.0348),eIF4G Ser1108 (P < 0.0196), Akt Ser473 (P < 0.0227), Bak (P < 0.0321), and p70S6 Thr389 (P < 0.0373) were found to be statistically significantly associated withoverall survival by Wilcoxon one-way analysis. Columns, mean; bars, SE. Total and phosphorylated forms were independently evaluated. Although the phosphorylatedform of the seven end points reached the level of statistical significance as indicated, the total protein for the same end points did not reach statistical significance,except in the case of total Akt (P < 0.0455). Data for all 27 end points is indicated in Supplementary Data.

Akt/mTOR Pathway and Rhabdomyosarcoma Survival

www.aacrjournals.org 3435 Cancer Res 2007; 67: (7). April 1, 2007

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

additional signaling proteins upstream and downstream of Akt andmTOR for an independent evaluation of pathway activation.

Following unblinding of the data, the results for study set 1B(Fig. 4) showed a significant association of disease-free and overallsurvival with phosphorylated components of the Akt-mTORpathway. High levels of Akt Ser473, 4EBP1 Thr37/46, eIF4G Ser1108,and p70S6 Thr389 were all significantly associated with poor overalland poor disease-free survival: Akt Ser473 (overall survival P < 0.001,recurrence-free survival P < 0.0009), 4EBP1 Thr37/46 (overall survivalP < 0.0110, recurrence-free survival P < 0.0106), eIF4G Ser1108

(overall survival P < 0.0017, recurrence-free survival P < 0.0072), andp70S6 Thr389 (overall survival P < 0.0085, recurrence-free survivalP < 0.0296; Fig. 4A). Each of the 27 components was also evaluatedindividually for statistical correlation with survivor versus non-survivor status. Six end points—again, all components of the Akt/

mTOR network (4EBP1 Thr37, Akt Ser473, eIF4G Ser1108, p70S6Thr389, Bak, and GSK3a/h Tyr279/216)—correlated independentlywith survival [Wilcoxon one-way analysis 4EBP1 Thr37/46 (P <0.0348), GSK3a/h Tyr279/216 (P < 0.0348), eIF4G Ser1108 (P < 0.0196),Akt Ser473 (P < 0.0227), Bak (P < 0.0321), and p70S6 Thr389 (P <0.0373); Fig. 4B].IRS-1/Akt/mTOR feedback loop is dysregulated in non-

survivor cohort. Although tyrosine phosphorylated insulin recep-tor substrate-1 (IRS-1) activates Akt/mTOR signaling throughphosphatidylinositol 3-kinase (PI3K), serine phosphorylation ofIRS-1 at serine612 by mTOR and p70S6 down-regulates IRS-1 tyrosineactivation (19–21). Thus, IRS-1 is subject to negative feedbackregulation in response to Akt/mTOR activation (Fig. 5A). Weexamined levels of phosphorylated members of the IRS-1/Akt/mTOR feedback loop by reverse phase protein microarray for

Figure 5. Proposed IRS-1 cell signaling pathway linkages in rhabdomyosarcoma study set 1B. A, IRS-1 feedback loop diagram. IRS-1 is regulated by both apositive feedback loop through Akt and a negative feedback loop through mTOR and p70S6 via IRS-1 Ser612. B, nonparametric and survival analysis of IRS-1/Akt/mTOR pathway proteins in sample set 1B (Fig. 1A). Spearman’s Rho table of selected prosurvival and apoptotic signaling proteins evaluated for sample set 1Bsupports the hypothesis of an altered negative feedback loop between IRS-1 Ser612 and the Akt/mTOR pathway in patients with poor survival outcome. Comparisonsare made between the total and phosphorylated form of the indicated end points. The phosphorylated form reaches a level of statistical significance (P < 0.05)compared with the total form of the protein, or the ratio of the phosphorylated to total protein. (Wilcoxon one-way analysis of overall survival outcome and KaplanMeier log-rank survival analysis). C, Spearman’s Rho nonparametric analysis showed a correlation between IRS-1 Ser612 and mTOR Ser2448 for tumors from patientswith survivor status (P = 0.0027) compared with tumors from patients with nonsurvivor status (P = 0.7358). D, similar correlations were noted between IRS-1Ser612 and p70S6 Thr389 for tumor samples from patients with survivor status (P = 0.00004) versus tumor samples from patients with nonsurvivor status (P = 0.1827).

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3436 www.aacrjournals.org

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

the tumors in study set 1B (n = 26). Although levels of IRS-1 Ser612

were no different between the survivors and nonsurvivors,phosphorylation of IRS-1 Ser612 correlated strongly with phosphor-ylation of mTOR at Ser2448 in the survivor cohort (Spearman’s Rhononparametric P < 0.0027), suggesting a linkage between these twosignaling events (Fig. 5B). By contrast, the phosphorylation of thesesame two signaling proteins was not correlated in the nonsurvivorcohort (Spearman’s Rho nonparametric P = 0.7358; Fig. 5B–C). Thislack of correlation with IRS-1 Ser612 phosphorylation also prevailedfor the mTOR downstream components eIF4E Ser209 (survivor P =0.0006, nonsurvivor P = 0.102) and p70S6 Thr389 (survivor P =0.00004, nonsurvivor P = 0.1827; Fig. 5B and D). Thus, the inter-relationship between IRS-1 activity and the mTOR pathway proteinsmay be altered in the tumors of patients, which subsequently arefound to have poor survival after chemotherapy compared with thetumors of patients who have long-term survival (Fig. 5A–D).Interrogation of the phosphorylated versus nonphosphory-

lated state of proteins. Phosphorylation is an important

posttranslational modification that has potential significance as areadout for the activation state of pathways and kinase inhibitortargets. To further investigate potential significant cell signalingproteins within the IRS-1/Akt/mTOR pathway, we extended ouranalysis to include the following additional end points: BAD, eIF4G,IRS-1, IRS-2, IGFR-h, and S6 Ser240/244. We conducted Wilcoxon one-way analysis and Kaplan-Meier survival analysis for the phosphor-ylated protein, the total protein form, and the ratio of thephosphorylated to total forms of key protein end points (Fig. 5B ;Supplementary Data). The results clearly show that the specificphosphorylated forms of the protein end points within the Akt-mTOR and associated pathways are independently associated withsurvival (P < 0.05) compared with the nonphosphorylated total formof the analyte protein (4EBP1 Thr37/46 P < 0.035, p70S6 Thr389

P < 0.037, Akt Ser473 P < 0.023, eIF4G Ser1108 P < 0.02). This is animportant distinction because it is likely that the population of thetotal protein in a signal pathway node is in excess compared withthe phosphorylated form. The phosphorylated form, actively

Figure 6. CCI-779 suppression of human rhabdomyosarcoma tumor growth in a mouse xenograft model. A, time-dependent CCI-779 inhibition of phosphorylationof mTOR pathway downstream substrates within tumor tissue in a xenograft treatment model. CCI-779 inhibited phosphorylation of mTOR pathway substrates,pS6 Ser235/236, and 4EBP1 Thr70 in both noninvolved muscle and tumor tissue compared with actin. B, CCI-779 inhibited tumor growth in Rh30 and RD mouse xenograftmodels. Tumor volume, as measured with calipers, was less in the CCI-779 treatment group compared with the vehicle alone for both the Rh30 and RD xenografts.E, RD control; n, Rh30 plus CCI-779; y, Rh30 control; �, RD plus CCI-779 (RD P = 0.00008; Rh30 P = 0.0002, Student’s t test). C, CCI-779 was administeredat 20 mg/kg/i.p. every 3 d for 30 d. Protein extracts from Rh30 and RD mouse xenograft tumors or uninvolved muscle were treated with CCI-779 or vehicle for 30 dand analyzed by Western blotting for S6 and 4EBP1 phosphorylation. D, Western blotting for total Akt and phosphorylated Akt Ser473 for the indicted duration oftreatment from RD and Rh30 tumor xenografts.

Akt/mTOR Pathway and Rhabdomyosarcoma Survival

www.aacrjournals.org 3437 Cancer Res 2007; 67: (7). April 1, 2007

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

engaged in signaling, constitutes a subpopulation of the totalprotein. Thus, the phosphorylated form constitutes a variable that isindependently correlated with survival compared with the totalprotein (Fig. 5; Supplementary Data).Suppression of the mTOR pathway in a mouse xenograft

model reduces tumor growth. To validate the functionalsignificance of our IRS-1/Akt/mTOR network analysis, we usedrapamycin analogues, which are well-characterized inhibitors ofthe mTOR protein kinase pathway, using a mouse xenografttreatment model. Either RD embryonal cells or Rh30 alveolar cellswere injected orthotopically into the hind leg of beige SCID mice.These two different cell lines were used to determine the effects ofmTOR inhibition in different histologic tumor categories. Therapamycin analogue CCI-779 (Wyeth, Madison, NJ) dosage was 20mg/kg, which corresponds to dosages currently administered tohumans in phase I and II clinical trials (22, 23). Administration ofCCI-779 at doses that were verified to suppress the phosphoryla-tion of mTOR downstream targets profoundly reduced the growthof rhabdomyosarcoma xenografts as measured in the beige SCIDmurine model (Rh30 xenograft group P = 0.0002; RD xenograftgroup P = 0.00008, n = 8 for both groups; Fig. 6A–D). Suppression ofthe mTOR pathway was monitored by measuring the phosphor-ylation of 4EBP1 and S6 ribosomal protein, which are well-established downstream targets of mTOR (12). CCI-779 inhibitedthe phosphorylation of these downstream targets commensuratewith a blockade in mTOR signaling in both the Rh30 alveolar– andRD embryonal xenograft–derived tumors. In addition, there was aslight but definite increase in phosphorylated Akt at Ser473 in boththe RD and Rh30 xenograft tumors over the course of treatmentwith CCI-779 (Fig. 6D).

Discussion

Recent real-time reverse transcription-PCR and fluorescencein situ hybridization assays for PAX3-FKHR and PAX7-FKHR fusiontranscripts have been developed and applied to rhabdomyosarco-ma. Nevertheless, genomic and transcriptomic assays do notprovide an effective recapitulation of the posttranslational,fluctuating signaling events that occur at the proteomic level(24–26). Therefore, we used reverse phase protein microarrays as ameans of monitoring the in vivo state of selected kinase pathways.

In the present investigation, analysis of protein signalingpathways was conducted blinded to treatment or survival usingtwo independent rhabdomyosarcoma tumor study sets for which12-year follow-up data was available. Two independent study sets(Fig. 1A) were procured randomly from the pool of frozenspecimens. Each study set represented a variety of treatmentmodalities, histologic subtypes, and tumor sites. The two setsdiffered in the proportion of samples with alveolar versusembryonal histology (Fig. 1A and C ; refs. 3, 4). Although thesample sets were heterogeneous, there was no statisticallysignificant difference in either overall survival or recurrence-freesurvival between the two sample sets (overall survival P = 0.2111,recurrence-free survival P = 0.5824; Fig. 1B).

Current prognostic indicators for patients diagnosed withrhabdomyosarcoma are age, stage, group, histology, and primarysite, with patients in the 1- to 8-year age group with embryonalrhabdomyosarcoma from orbital or nonparameningeal head andneck sites having the best prognosis (15). Using unsupervisedclustering analysis, we sought to determine if any protein signalingsignature correlated with histologic subtype. For the first study set,

15 specific signaling proteins (Figs. 2A and 3A) were initiallychosen because they constituted a broad survey of multipleprosurvival-related events. A multiplexed measurement of thechosen phosphorylation states provided an averaged portrait of theongoing kinase activity events within selected networks that drivecellular growth or survival.

The initial unsupervised clustering analysis was not significantlyassociated with histology but there was clear partitioning of thesamples into two clusters, with one cluster exhibiting activation ofAkt/mTOR proteins (Fig. 2A). Therefore, clinical outcome data wasobtained from the COG for further exploratory associationsbetween the protein end points and clinical data. The results ofset 1A revealed a statistically significant association betweensurvival and the activation/suppression of proteins linked to theAkt/mTOR signaling pathway (Fig. 3A–B).

Based on the results of set 1A, we expanded this exploratoryanalysis to 27 end points applied to a second independent set ofsamples (Fig. 1A ; and Supplementary Data). Proteins that seemedto correlate with survival or failure in the second study set werelinked together in the Akt/mTOR kinase pathway (12, 27, 28).Phosphorylated components of the Akt/mTOR pathway, specif-ically Akt, eIF4G, 4EBP1 (elongation binding factor), GSK3a/h,and p70S6 were found to be associated with outcome (Fig. 4B).IRS-1, Akt, and GSK3h are associated with cell growth, survival,insulin response, and glucose metabolism. mTOR, 4EBP1, andp70S6 are essential components of protein translation, in whichphosphorylation of 4EBP1 releases 4EBP1 from eIF4E, activatingcap-dependent translation (29). These pathways are involvedin the regulation of prosurvival and translation for a group ofproteins that are important for cell cycle and apoptosis, includingseveral known oncogenes such as cyclin D, c-myc , and Hif-1a (30).Although Akt Ser473 correlated with survival (P < 0.02) for studyset 1B, it did not correlate with survival in set 1A (P = 0.2460).This may have been due to differences in the relative composi-tion of tumor histologies and sites of origin between the twogroups (Fig. 1A).

A variety of autocrine and paracrine stimuli, including hormones,growth factors, mitogens, cytokines, and G-protein–coupledreceptor agonists, elicit 4EBP1 hyperphosphorylation and concom-itant loss of eIF4E-binding activity in the mTOR pathway (12, 27, 28,30). Activation of PI3K or the downstream effector kinase Akt leadsto 4EBP1 hyperphosphorylation, affecting its release from eIF4E.Phosphorylation of 4EBP1 on multiple loci is associated withlinkage to the insulin receptor pathway and the PI3K pathway. Sixphosphorylation sites have been identified on 4EBP1. Thr37, Thr46,Ser65, and Thr70 become phosphorylated after insulin stimulation,and such phosphorylation can be blocked by rapamycin (inhibitorof mTOR) and wortmannin (inhibitor of PI3K; refs. 31, 32). It hasbeen shown that mTOR itself, as well as an mTOR-associatedkinase, directly phosphorylates sites on 4EBP1. Gingras et al. (31)established that phosphate groups are first added to Thr37 andThr46. This priming phosphorylation is required for the phosphor-ylation of other sites necessary for binding. Thus, multiplephosphorylation events triggered from multiple kinases, primedby Thr37/46, are involved in the release of 4E-BP1 from eIF4E.

Tyrosine-phosphorylated IRS-1 activates Akt/mTOR signalingthrough PI3K, serine phosphorylates IRS-1 (at Ser612) by mTOR,and p70S6 down-regulates IRS-1 tyrosine activation (19–21). Thus,it is hypothesized that IRS-1 is subject to negative feedbackregulation in response to Akt/mTOR activation through p70S6(Fig. 5A ; ref. 33). Based on this negative feedback between mTOR

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3438 www.aacrjournals.org

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

and IRS-1, O’Reilly et al. (33) recently hypothesized that inhibitionof mTOR may cause augmented phosphorylation of Akt throughdisruption of the negative feedback loop between p70S6 and IRS-1.We examined the IRS-1 feedback loop interrelationship withcomponents of the Akt and mTOR pathway by nonparametriccorrelations (Fig. 5B–D). Interrogation of IRS-1 Ser612 and variouspotential interacting proteins provided a means to assess theprotein interactions with the actual phosphorylation site involvedin the negative feedback regulation of IRS-1 (20, 21). The averagelevel of IRS-1 Ser12 was not statistically different (P < 0.098)between tumors from patients with favorable outcome comparedwith those with poor outcome (Fig. 4B), suggesting that the levelof IRS-1 upstream activity was similar. Although the average levelof IRS-1 Ser612 phosphorylation was similar in the favorableversus poor outcome cases, the correlation of individual IRS-1phosphorylation levels in each tumor with phosphorylation levelsof Akt and mTOR pathway proteins was highly dissimilar in thesetwo phenotypes. In addition Bak, FKHR Ser256, and IGFR-h weresignificantly correlated for both groups (Supplementary Data). Asshown in Fig. 5, there was a strong positive correlation (P =0.00269) of IRS-1 Ser612 with mTOR Ser2448 and with p70S6 Thr289

(P = 0.00004) in tumors with favorable outcome (Fig. 5B and C).This suggests a linkage or correlation consistent with a feedbackloop between mTOR and IRS-1 in these tumors with favorableoutcomes. These data support a selective alteration in the activeinterrelationship between IRS-Ser612 and downstream compo-nents of the mTOR pathway in tumors with poor outcome.

The implications of these differences in the IRS-Akt-mTORinterconnectivity of survivors and nonsurvivors are 2-fold. First,the apparent lack of interconnection between IRS-1 and mTORcould disrupt the normal negative feedback regulation asdescribed (20, 33). This could result in increased phosphorylationof Akt as we noted in the tumors from patients with pooroutcomes and illustrated in Fig. 5A . Baseline levels of phosphor-ylated Akt and mTOR may be elevated in aggressive tumors inwhich the negative feedback regulation of mTOR through IRS-1 isdisrupted, or phosphorylation of Akt is supplemented by IRS-1–independent mechanisms.

The identified 4E-BP1 phosphorylation sites are specificallyinhibited by rapamycin treatment (29, 31). To validate thefunctional significance of our network analysis, revealing mTORpathway suppression observed in patients who had a favorabletreatment outcome, we exploited the existence of rapamycinanalogues, which are well-characterized inhibitors of the mTORprotein kinase pathway. Some of these analogues are currently inphase I and II clinical trials of adults with cancer (22, 23).Suppression of the mTOR pathway was monitored by measuringthe state of phosphorylation of 4EBP1 and S6 kinase, which arewell-established downstream substrates of mTOR (12, 27, 28, 30,31). CCI-779 inhibited the expected phosphorylation of thedownstream targets commensurate with a blockade in mTORsignaling in xenograft tumors derived from Rh30 alveolar or RDembryonal cells (Fig. 6). Phosphorylation of Akt was present inboth of these cell lines grown in vivo (Fig. 6D) and there was aslight increase in the RH30 (alveolar) xenograft and a perceptibleaugmentation in the RD (embryonal) xenograft of phosphorylatedAkt on Ser473 over time during CCI-779 treatment (Fig. 6D). This isconsistent with the negative feedback loop as described by O’Reillyet al. (33). The growth of both histologic tumor types was inhibitedby CCI-779 treatment. In Fig. 4, both embryonal and alveolarrhabdomyosarcoma subtypes were represented in the cohort with

poor outcomes, which had higher levels of mTOR pathwayactivation.

The findings in the present study for both the patient’s tumortissue and the xenograft model are supported and complementedby small interfering RNA S6K1 knockdown experiments in Rh30and RD cell lines (34). Both the patient tumor and mouse xeno-graft findings (Figs. 5B–D and 6) support a key regulation ofAkt signaling through mTOR and IGF-IRS-1 as shown bythe correlation between IRS-1 Ser612 and other members of thesepathways. Moreover, a recent report from the COG usingoligonucleotide gene expression analysis revealed an associationof elevated platelet-derived growth factor receptor and insulin-likegrowth factor gene transcripts with decreased survival for patientsfrom the IRS-IV study set (35). Our finding of an increased amountof total IGFR-h associated with poor survival (Fig. 5B) is in keepingwith this finding. Thus, the present data support the conclusion ofWan et al. (34) that an optimal therapy strategy would be tocombine an mTOR inhibitor with an IGF-IR antibody (or smallmolecule) inhibitor.

The IGF-I/Akt/mTOR pathway found herein to be associatedwith rhabdomyosarcoma has previously been associated withregulation of hypertrophy and atrophy of muscle, the cellularprogenitor of rhabdomyosarcoma Song et al. (36) have reportedthat IGF-I, via the mTOR pathway, regulates angiotensin II–induced skeletal muscle wasting. Rat skeletal muscle atrophy isassociated with inactivation of the mTOR pathway. Activation ofthe mTOR pathway is also associated with active remodeling ofthe heart in vivo and protection against atrophy, whereasrapamycin treatment of rat hearts augmented atrophy (37).Cardiac myocyte reperfusion after transient ischemia induced byreversible occlusion was associated with activation of the PI3K/Akt/mTOR pathway (38). Reperfusion was associated withactivation of Akt and phosphorylation of GSK3h, and associatedstimulation of signaling through mTOR as evidenced byphosphorylation of eIF4E and S6 kinase. Activation of the mTORpathway and phosphorylation of S6K1 is also associated withhypertrophy and pressure overload hypertrophy. Post ischemia,surviving myocytes undergo 10% to 15% hypertrophy (39).Phosphorylation of S6K1 was unaltered during ischemia butincreased during reperfusion (38).

These results in nonneoplastic muscle indicate that a physiologiccycle of ischemia followed by reperfusion activates the Akt/mTORpathway, is associated with myocyte hypertrophy, and protectsagainst atrophy. This may be relevant during the precursor statebefore neoplastic growth, but they may also apply to cycles ofischemia, necrosis, and angiogenesis associated with aggressiveneoplastic growth. Because rhabdomyosarcoma is thought to arisefrom a myocyte lineage, signaling pathways involved in normalmuscle cell regrowth/remodeling could be similar to those involvedin tumorigenesis.

Mirk, a protein recently identified to possess antiapoptoticfunctions in rhabdomyosarcoma cell lines and tumors, exhibitsboth prosurvival and growth arrest functions in differentiatingskeletal myoblasts (40, 41). Mirk has been shown to be activated byMKK3, a stress-activated mitogen-activated kinase kinase (42).RNA interference (RNAi) depletion of endogenous Mirk reducedthe clonogenicity of RD embryonal and Rh30 alveolar rhabdomyo-sarcoma cell lines in colony formation experiments, indicating aprosurvival role in rhabdomyosarcoma (41). In addition, RNAidepletion of Mirk has been shown to block myoblast differentiationin skeletal muscle primary cell culture (40). These findings further

Akt/mTOR Pathway and Rhabdomyosarcoma Survival

www.aacrjournals.org 3439 Cancer Res 2007; 67: (7). April 1, 2007

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

strengthen the role of cell survival mechanisms in rhabdomyosar-coma and muscle development.

In summary, protein pathway analysis of microdissected humanrhabdomyosarcoma clinical specimens, procured before treat-ment, revealed a strong association between activation (phos-phorylation) of multiple interconnected Akt/mTOR pathwaycomponents and a poor disease-free or overall survival in thisinitial, exploratory analysis. This observation was found to beconsistent between two independently analyzed clinical study sets.Moreover, the functional significance of IRS-1/Akt/mTOR pathwayactivation in rhabdomyosarcoma was verified using the specifictargeted inhibitor CCI-779 to suppress tumor growth in a beigeSCID rhabdomyosarcoma xenograft model. These data provideimpetus for testing rapamycin analogues in this tumor type as apotential way to modulate poor prognosis patients into moredurable outcomes. In support of this combination therapyconcept, MacKiegan et al. (43) used RNAi libraries to search for

kinases whose inhibition resulted in increased rates of apoptosis.mTOR knockdown was found to sensitize cells to Taxol-inducedapoptosis (43). Based on the interrelationship of the Akt/mTORpathway and the IGF-IRS pathway (Fig. 5), future combinationtherapy strategies could be aimed at blocking both upstream IRS-1–mediated signaling factor activation, as well as downstreammTOR signaling, as a means of augmenting standard cytotoxicrhabdomyosarcoma therapy.

Acknowledgments

Received 4/12/2006; revised 12/8/2006; accepted 1/24/2007.Grant support: Intramural Research Program of the NIH, National Cancer

Institute, and George Mason University.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

The authors thank James Anderson for critical reading of the manuscript, andWilliam Mayer for assistance in providing samples.

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3440 www.aacrjournals.org

References1. Dagher R, Helman L. Rhabdomyosarcoma: an over-

view. Oncologist 1999;4:34–44.2. Merlino G, Helman LJ. Rhabdomyosarcoma—working

out the pathways. Oncogene 1999;18:5340–8.3. Spunt SL, Smith LM, Ruymann FB, et al. Cyclophos-

phamide dose intensification during induction therapyfor intermediate-risk pediatric rhabdomyosarcoma isfeasible but does not improve outcome: a report fromthe soft tissue sarcoma committee of the children’soncology group. Clin Cancer Res 2004;10:6072–9.

4. Crist WM, Anderson JR, Meza JL, et al. Intergrouprhabdomyosarcoma study-IV: results for patients withnonmetastatic disease. J Clin Oncol 2001;19:3091–102.

5. Petricoin EF III, Bichsel VE, Calvert VS, et al. Mappingmolecular networks using proteomics: a vision forpatient-tailored combination therapy. J Clin Oncol2005;23:3614–21.

6. Chan SM, Ermann J, Su L, Fathman CG, Utz PJ. Proteinmicroarrays for multiplex analysis of signal transductionpathways. Nat Med 2004;10:1390–6.

7. Meric-Bernstam F, Mills GB. Mammalian target ofrapamycin. Semin Oncol 2004;31:10–7; discussion 33.

8. Sachs K, Perez O, Pe’er D, Lauffenburger DA, NolanGP. Causal protein-signaling networks derived frommultiparameter single-cell data. Science 2005;308:523–9.

9. Liotta LA, Espina V, Mehta AI, et al. Protein micro-arrays: meeting analytical challenges for clinical appli-cations. Cancer Cell 2003;3:317–25.

10. Sheehan KM, Calvert VS, Kay EW, et al. Use of reversephase protein microarrays and reference standarddevelopment for molecular network analysis of meta-static ovarian carcinoma. Mol Cell Proteomics 2005;4:346–55.

11. Emmert-Buck MR, Bonner RF, Smith PD, et al. Lasercapture microdissection. Science 1996;274:998–1001.

12. Lynch M, Fitzgerald C, Johnston KA, Wang S, SchmidtEV. Activated eIF4E-binding protein slows G1 progres-sion and blocks transformation by c-myc withoutinhibiting cell growth. J Biol Chem 2004;279:3327–39.

13. Sun SY, Rosenberg LM, Wang X, et al. Activation ofAkt and eIF4E survival pathways by rapamycin-mediat-ed mammalian target of rapamycin inhibition. CancerRes 2005;65:7052–8.

14. Fisher R. On the interpretation of m2 from contin-gency tables, and the calculation of P . J R Stat Soc 1922;85:87–94.

15. Mazzoleni S, Bisogno G, Garaventa A, et al. Outcomesand prognostic factors after recurrence in children and

adolescents with nonmetastatic rhabdomyosarcoma.Cancer 2005;104:183–90.

16. Kaplan E, Meier P. Non-parametric estimationfrom incomplete observations. J Am Stat Assoc 1958;53:457–81.

17. Cox D. Regression models and life tables. J Royal StatSoc (B) 1972;34:187–202.

18. Simon R, Altman DG. Statistical aspects ofprognostic factor studies in oncology. Br J Cancer1994;69:979–85.

19. Harrington LS, Findlay GM, Lamb RF. RestrainingPI3K: mTOR signalling goes back to the membrane.Trends Biochem Sci 2005;30:35–42.

20. Gual P, Marchand-Brustel Y, Tanti J-F. Positive andnegative regulation of insulin signaling through IRS-1phosphorylation. Biochimie 2005;87:99–109.

21. Gallicchio MA, van Sinderen M, Bach LA. Insulin-likegrowth factor binding protein-6 and CCI-779, an esteranalogue of rapamycin, additively inhibit rhabdomyo-sarcoma growth. Horm Metab Res 2003;35:822–7.

22. Raymond E, Alexandre J, Faivre S, et al. Safety andpharmacokinetics of escalated doses of weekly intrave-nous infusion of CCI-779, a novel mTOR inhibitor, inpatients with cancer. J Clin Oncol 2004;22:2336–47.

23. Smolewski P. Recent developments in targeting themammalian target of rapamycin (mTOR) kinase path-way. Anticancer Drugs 2006;17:487–94.

24. Barr FG, Smith LM, Lynch JC, et al. Examination ofgene fusion status in archival samples of alveolarrhabdomyosarcoma entered on the Intergroup Rhabdo-myosarcoma Study-III trial: a report from the Children’sOncology Group. J Mol Diagn 2006;8:202–8.

25. Carey KA, Segal D, Klein R, et al. Identification ofnovel genes expressed during rhabdomyosarcomadifferentiation using cDNA microarrays. Pathol Int2006;56:246–55.

26. Nishio J, Althof PA, Bailey JM, et al. Use of a novelFISH assay on paraffin-embedded tissues as an adjunctto diagnosis of alveolar rhabdomyosarcoma. Lab Invest2006;86:547–56.

27. Houghton PJ, Huang S. mTOR as a target for cancertherapy. Curr Top Microbiol Immunol 2004;279:339–59.

28. Huang S, Shu L, Easton J, et al. Inhibition ofmammalian target of rapamycin activates apoptosissignal-regulating kinase 1 signaling by suppressingprotein phosphatase 5 activity. J Biol Chem 2004;279:36490–6.

29. Bjornsti MA, Houghton PJ. Lost in translation:dysregulation of cap-dependent translation and cancer.Cancer Cell 2004;5:519–23.

30. Dutcher JP. Mammalian target of rapamycin inhibi-tion. Clin Cancer Res 2004;10:6382–7S.

31. Gingras AC, Raught B, Gygi SP, et al. Hierarchicalphosphorylation of the translation inhibitor 4E-BP1.Genes Dev 2001;15:2852–64.

32. Takeuchi H, Kondo Y, Fujiwara K, et al. Synergisticaugmentation of rapamycin-induced autophagy inmalignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res 2005;65:3336–46.

33. O’Reilly KE, Rojo F, She QB, et al. mTOR inhibitioninduces upstream receptor tyrosine kinase signaling andactivates Akt. Cancer Res 2006;66:1500–8.

34. Wan X, Harkavy B, Shen N, Grohar P, Helman LJ.Rapamycin induces feedback activation of Akt signalingthrough an IGF-IR-dependent mechanism. Oncogene2006 Sep 25; [Epub ahead of print].

35. Blandford MC, Barr FG, Lynch JC, et al. Rhabdomyo-sarcomas utilize developmental, myogenic growthfactors for disease advantage: a report from theChildren’s Oncology Group. Pediatr Blood Cancer 2006;46:329–38.

36. Song YH, Li Y, Du J, et al. Muscle-specific expressionof IGF-1 blocks angiotensin II-induced skeletal musclewasting. J Clin Invest 2005;115:451–8.

37. Razeghi P, Sharma S, Ying J, et al. Atrophicremodeling of the heart in vivo simultaneously activatespathways of protein synthesis and degradation. Circu-lation 2003;108:2536–41.

38. Crozier SJ, Zhang X, Wang J, et al. Activation ofsignaling pathways and regulatory mechanisms ofmRNA translation following myocardial ischemia/reper-fusion. J Appl Physiol 2006;101:576–82.

39. Zhang XQ, Moore RL, Tillotson DL, Cheung JY.Calcium currents in postinfarction rat cardiac myocytes.Am J Physiol 1995;269:C1464–73.

40. Deng X, Ewton DZ, Pawlikowski B, Maimone M,Friedman E. Mirk/dyrk1B is a Rho-induced kinase activein skeletal muscle differentiation. J Biol Chem 2003;278:41347–54.

41. Mercer SE, Ewton DZ, Shah S, Naqvi A, Friedman E.Mirk/Dyrk1b mediates cell survival in rhabdomyosar-comas. Cancer Res 2006;66:5143–50.

42. Lim S, Jin K, Friedman E. Mirk protein kinase isactivated by MKK3 and functions as a transcriptionalactivator of HNF1a. J Biol Chem 2002;277:25040–6.

43. MacKeigan JP, Murphy LO, Blenis J. Sensitized RNAiscreen of human kinases and phosphatases identifiesnew regulators of apoptosis and chemoresistance. NatCell Biol 2005;7:591–600.

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:3431-3440. Cancer Res   Emanuel F. Petricoin III, Virginia Espina, Robyn P. Araujo, et al.   Childhood Rhabdomyosarcoma SurvivalRapamycin Activation Is Negatively Associated with Phosphoprotein Pathway Mapping: Akt/Mammalian Target of

  Updated version

  http://cancerres.aacrjournals.org/content/67/7/3431

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2007/03/26/67.7.3431.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/7/3431.full#ref-list-1

This article cites 42 articles, 18 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/7/3431.full#related-urls

This article has been cited by 33 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/67/7/3431To request permission to re-use all or part of this article, use this link

Research. on January 2, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from