cancer research epidermal growth factor receptor ... · molecular and cellular pathobiology...

15
Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct Tumor-Initiating Cells in Human Glioblastoma Multiforme and Is Required for Gliomagenesis Stefania Mazzoleni 1,5 , Letterio S. Politi 2 , Mauro Pala 6 , Manuela Cominelli 7 , Alberto Franzin 3 , Lucia Sergi Sergi 4 , Andrea Falini 2 , Michele De Palma 4 , Alessandro Bulfone 6 , Pietro L. Poliani 7 , and Rossella Galli 1 Abstract Epidermal growth factor receptor (EGFR) is a known diagnostic and, although controversial, prognostic marker of human glioblastoma multiforme (GBM). However, its functional role and biological significance in GBM remain elusive. Here, we show that multiple GBM cell subpopulations could be purified from the specimens of patients with GBM and from cancer stem cell (CSC) lines based on the expression of EGFR and of other putative CSC markers. All these subpopulations are molecularly and functionally distinct, are tumorigenic, and need to express EGFR to promote experimental tumorigenesis. Among them, EGFR- expressing tumor-initiating cells (TIC) display the most malignant functional and molecular phenotype. Accordingly, modulation of EGFR expression by gain-of-function and loss-of-function strategies in GBM CSC lines enhances and reduces their tumorigenic ability, respectively, suggesting that EGFR plays a fun- damental role in gliomagenesis. These findings open up the possibility of new therapeutically relevant scenarios, as the presence of functionally heterogeneous EGFR pos and EGFR neg TIC subpopulations within the same tumor might affect clinical response to treatment. Cancer Res; 70(19); 750013. ©2010 AACR. Introduction In the last 10 years, the identification of putative cancer stem cells (CSC) in different types of tumors has raised many expectations. CSCs represent a rare fraction of the tumor bulk, uniquely responsible for tumor initiation and progres- sion and, as such, are envisioned as elective targets of ther- apy (1, 2). The CSC model of tumorigenesis has been convincingly validated in many hemopoietic malignancies (3) and in solid tumors such as breast and colon cancers (47). However, in other tumors, CSCs seem to make up the majority of cells within the tumor bulk and all exhibit tumor-initiating ability, thus being more appropriately called tumor-initiating cells (TIC; refs. 811). This last model might also apply to glioblastoma multi- forme (GBM), as soundly shown by recent studies in cultured CSC lines (12, 13). To test whether this concept might hold true in specimens from patients with primary GBM, we se- lected the epidermal growth factor receptor (EGFR) as a pro- spective GBM CSC marker (14, 15). EGFR expression in GBM has both diagnostic and prognostic significance (1619) and it is associated with tumor progression (20). Likewise, EGFR gene amplification is positively correlated with molecularly defined GBM subclasses, characterized by active proliferation and poor prognosis (21). Valuable preclinical information has also been obtained by modulating EGFR expression in stan- dard serum-cultured glioma cell lines (2225) and by using these in vitro cultures for testing different therapeutic approaches (26). Here, we investigated the biological significance of EGFR expression in human primary GBM by assessing its functional role in tumor cell fractions purified from several GBM patientderived specimens and by validating it in GBM CSC lines, which reproduce the genotypic and phenotypic charac- teristics of GBM more faithfully than standard glioma cell lines (27, 28). By this experimental strategy, we identified mul- tiple subpopulations of GBM TICs, each characterized by a distinct gene signature and tumorigenic potential, and thus being able to reproduce the diverse pathologic and functional features of GBM, such as invasive and angiogenic behavior. Authors' Affiliations: 1 Neural Stem Cell Biology Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, 2 Neuroradiology Unit and CERMAC, 3 Neurosurgery Unit, and 4 Angiogenesis and Tumor Targeting Research Unit, Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute; 5 Vita-Salute San Raffaele University, Milan, Italy; 6 Bio))flag Ltd., Pula, Cagliari, Italy; and 7 Department of Pathology, University of Brescia-Spedali Civili of Brescia, Brescia, Italy Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Rossella Galli, Neural Stem Cell Biology Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy. Phone: 39-02-2643-4626; Fax: 39-02-2643-4621; E-mail: [email protected]. doi: 10.1158/0008-5472.CAN-10-2353 ©2010 American Association for Cancer Research. Cancer Research Cancer Res; 70(19) October 1, 2010 7500 Research. on April 22, 2021. © 2010 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

Upload: others

Post on 06-Nov-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

Mole

EpidFunin Hfor

StefanLuciaPietro

Abst

Intro

In tstem cexpecbulk, usion aapy (convin(3) an(4–7).the m

AuthorRegeneUnit anTargetiof RegeScienti6Bio))flUnivers

Note:Resear

CorresDivisionRaffael39-02-2

doi: 10

©2010

Cance7500

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

Canceresearch

cular and Cellular Pathobiology

ermal Growth Factor Receptor Expression Identifiesctionally and Molecularly Distinct Tumor-Initiating Cellsuman Glioblastoma Multiforme and Is Required

R

Gliomagenesis

ia Mazzoleni1,5, Letterio S. Politi2, Mauro Pala6, Manuela Cominelli7, Alberto Franzin3,

Sergi Sergi4, Andrea Falini2, Michele De Palma4, Alessandro Bulfone6, L. Poliani7, and Rossella Galli1

ractEpid

markein GBspecimand otumorexpresAccordCSC li

Howeverajority of

s' Affiliatirative Medid CERMACng Researcnerative Mfic Instituteag Ltd., Puity of Bresc

Supplemench Online (

ponding Aof Regene

e Scientific643-4626;

.1158/0008-

American A

r Res; 70(

Downlo

ermal growth factor receptor (EGFR) is a known diagnostic and, although controversial, prognosticr of human glioblastoma multiforme (GBM). However, its functional role and biological significanceM remain elusive. Here, we show that multiple GBM cell subpopulations could be purified from theens of patients with GBM and from cancer stem cell (CSC) lines based on the expression of EGFRf other putative CSC markers. All these subpopulations are molecularly and functionally distinct, areigenic, and need to express EGFR to promote experimental tumorigenesis. Among them, EGFR-sing tumor-initiating cells (TIC) display the most malignant functional and molecular phenotype.ingly, modulation of EGFR expression by gain-of-function and loss-of-function strategies in GBMnes enhances and reduces their tumorigenic ability, respectively, suggesting that EGFR plays a fun-tal role in gliomagenesis. These findings open up the possibility of new therapeutically relevant

damen

scenarios, as the presence of functionally heterogeneous EGFRpos and EGFRneg TIC subpopulations withinthe same tumor might affect clinical response to treatment. Cancer Res; 70(19); 7500–13. ©2010 AACR.

tumortumorThi

formeCSC litrue inlectedspectihas boit is a

duction

he last 10 years, the identification of putative cancerells (CSC) in different types of tumors has raised manytations. CSCs represent a rare fraction of the tumorniquely responsible for tumor initiation and progres-nd, as such, are envisioned as elective targets of ther-1, 2). The CSC model of tumorigenesis has beencingly validated in many hemopoietic malignanciesd in solid tumors such as breast and colon cancers

, in other tumors, CSCs seem to make upcells within the tumor bulk and all exhibit

gene adefineand poalso bdard stheseapproHer

expresrole ipatienlines,teristilines (tiple sdistincbeingfeatur

ons: 1Neural Stem Cell Biology Unit, Division ofcine, Stem Cells and Gene Therapy, 2Neuroradiology, 3Neurosurgery Unit, and 4Angiogenesis and Tumorh Unit, Telethon Institute for Gene Therapy, Divisionedicine, Stem Cells and Gene Therapy, San Raffaele; 5Vita-Salute San Raffaele University, Milan, Italy;la, Cagliari, Italy; and 7Department of Pathology,ia-Spedali Civili of Brescia, Brescia, Italy

tary data for this article are available at Cancerhttp://cancerres.aacrjournals.org/).

uthor: Rossella Galli, Neural Stem Cell Biology Unit,rative Medicine, Stem Cells and Gene Therapy, SanInstitute, Via Olgettina 58, 20132 Milan, Italy. Phone:Fax: 39-02-2643-4621; E-mail: [email protected].

5472.CAN-10-2353

ssociation for Cancer Research.

19) October 1, 2010

Research. on April 22, 202cancerres.aacrjournals.org aded from

-initiating ability, thus being more appropriately called-initiating cells (TIC; refs. 8–11).s last model might also apply to glioblastoma multi-(GBM), as soundly shown by recent studies in culturednes (12, 13). To test whether this concept might holdspecimens from patients with primary GBM, we se-the epidermal growth factor receptor (EGFR) as a pro-ve GBM CSC marker (14, 15). EGFR expression in GBMth diagnostic and prognostic significance (16–19) andssociated with tumor progression (20). Likewise, EGFRmplification is positively correlated with molecularlyd GBM subclasses, characterized by active proliferationor prognosis (21). Valuable preclinical information haseen obtained by modulating EGFR expression in stan-erum-cultured glioma cell lines (22–25) and by usingin vitro cultures for testing different therapeuticaches (26).e, we investigated the biological significance of EGFRsion in human primary GBM by assessing its functionaln tumor cell fractions purified from several GBMt–derived specimens and by validating it in GBM CSCwhich reproduce the genotypic and phenotypic charac-cs of GBM more faithfully than standard glioma cell27, 28). By this experimental strategy, we identified mul-ubpopulations of GBM TICs, each characterized by at gene signature and tumorigenic potential, and thus

able to reproduce the diverse pathologic and functionales of GBM, such as invasive and angiogenic behavior.

1. © 2010 American Association for Cancer

Page 2: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

Ourwith stinct GclinicaTIC su

Mate

CollecGBM

logic dcordareviewTumoent panot sawith ostudyproces

ImmuFou

and wanti-CmouseSectio(Chem

MolecTot

lines w(Qiagereversized toStandref. 29

FluorFoll

from pwereAbD SDickin(clone(BectoFACSVrangedsortinabove

Cultuand cEst

descri

MicroSee

EvaluimplaTum

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

findings unequivocally associate EGFR expressionpecific biological functions and molecular traits in dis-BM cell subpopulations, thus providing relevant pre-

l information to be exploited for the development of tinct

Magneas desImm

sectioCalbio

WesteLys

accordwere(Tyr10

rabbitphospSignalnalingwas u

Lentivcell inHum

into tcPPT.transdLen

humaInfectturer's

ImmuhybriImm

ref. 27bit poin situDako,tous aloidin

PharmAG1

centrasamewas m

InvasInva

poreEGFRside omigraAG147were

bpopulation–targeted effective therapies.

rials and Methods

tion and processing of patients' specimensspecimens were collected from patients with histo-

iagnosis of primary GBM (WHO grade 4 glioma) in ac-nce with the protocol approved by the institutionalboard of San Raffaele Scientific Institute (01-CSC07).

r diagnosis and grading were blind-reviewed by differ-thologists at two distinct institutions. Samples that didtisfy the inclusion criteria of primary GBMs (e.g., GBMligodendroglioma features) were excluded from the. See Supplementary Experimental Procedure forsing methods.

nohistochemical stainingr-micrometer sections were cut from paraffin blocksere stained with mouse antihuman EGFR, mouseD31 (all DakoCytomation), rabbit anti-SOX2 (Abcam),anti-Bmi1, and mouse anti-CD15 (Becton Dickinson).ns were then incubated with a secondary antibodyMATE Envision Rabbit/Mouse, DakoCytomation).

ular analysisal RNA from GBM tumor specimens and from CSCas extracted using the RNeasy Micro and Mini Kitsn). cDNA was obtained by using Superscript RNaseH−

e transcriptase (Invitrogen). All cDNAs were normal-the β-actin reverse transcription-PCR (RT-PCR) product.ard RT-PCR for EGFRvIII was done as described in.

escence-activated cell sortingowing enzymatic and mechanical dissociation, cellsrimary GBM tissues and from experimental xenograftsstained with rabbit antihuman EGFR (clone EGFR.1,erotec), rat antihuman CD34/CD45/CD11b (Bectonson), antihuman AC133 (Miltenyi), mouse anti-CD15MMA, Becton Dickinson), and antihuman pan-HLAn Dickinson). Cells were sorted on a Becton Dickinsonantage SE FACSDiVa. The purity of each cell fractionbetween 90% and 99%. Fluorescence-activated cell

g (FACS) analysis of CSC lines was done as described, without enzymatic digestion.

re propagation, population analysis,loningablished GBM CSCs were cultured as previouslybed (27).

array-based gene expression profilingSupplementary Experimental Procedure for details.

the lo(Dade

acrjournals.org

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

ation of CSC tumorigenicity by orthotopicntationorigenicity was determined by injecting all the dis-

cell preparations orthotopically in nu/nu mice (27).tic resonance imaging (MRI) analysis was performedcribed in Supplementary Experimental Procedure.unofluorescence analysis was performed on cryostat

ns using mouse monoclonal antihuman EGFR (1:100;chem). Details can be found in Supplementary Data.

rn blottingates from GBM tissues and CSCs were immunoblotteding to standard protocols. The primary antibodies usedrabbit anti-EGFR, rabbit anti–phosphorylated EGFR68; Cell Signaling), rabbit anti-ErbB2 (Calbiochem) andanti-Erbb3 (Santa Cruz Biotechnology), rabbit anti–horylatedmitogen-activated protein kinase (MAPK; Celling), and rabbit anti–phosphorylated Akt (Ser473; Cell Sig-). As a loading control, a mouse anti–β-tubulin antibodysed (Sigma-Aldrich).

iral vector generation, production, andfectionan wild-type (wt) EGFR cDNA (Upstate) was cloned

he monocistronic transfer lentiviral vector pCCL.sin.PGK.GFP.WPRE11. CSCs were transduced with 1 × 107

uction units/mL of lentiviral vectors for 16 hours.tiviral vectors coding for shRNAs targeted against then EGFR were purchased from Sigma (Mission RNAi).ion of CSCs was performed according to the manufac-instructions.

nofluorescence and fluorescence in situdizationunofluorescence was performed as described in

, using mouse antihuman EGFR (Calbiochem) and rab-lyclonal anti-GFP (Molecular Probes). Fluorescencehybridization (FISH) was performed using the kit fromaccording to the manufacturer's instructions. Filamen-ctin (F-actin) was stained by TRITC-conjugated phal-(Chemicon).

acologic EGFR inhibition in CSCs478 (Calbiochem) was used at the indicated final con-tions in complete medium. DMSO was used at theconcentration as the vehicle control. Cell proliferationeasured by an MTT incorporation assay.

ion assayssion assays were performed in Matrigel-coated 8-μm-Transwell chambers (Corning Costar). EGFRpos andneg CSCs were seeded in sister cultures on the upperf the chambers in complete medium and allowed tote for 5 and 7 days, respectively, with or without8. Noninvaded cells on the upper side of the filtersor were not scraped off, and those that migrated to

wer side were fixed and stained by using DiffQuickBehring). The extent of cell migration was normalized

Cancer Res; 70(19) October 1, 2010 7501

1. © 2010 American Association for Cancer

Page 3: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

to theunscra

Resu

EGFRwithinSev

mRNART-PCHowevembedature,EGFRroundmentanegligtumorples; Fthe improteiof humBy

EGFRthat, i(Fig. 1contawhereprolife214.1 arespecimensproteilary acInte

that Ecytomtrievedin thiscolabetopoiethat t(30) wstromwas dwas amentSuppleThu

GBM,from 0note,AC133not (FTo

biologpreviomenta

wheretein (Tof cellsubpopressiInte

contaitary Tfour oand C

EGFRdistinTo

pulatitumorCSC liEGF

contaiAC133of themcells (were eImm

withofractiowholepulatidistingnote,betwewhenthat Eleculato EGlateded toprogrConvewere mthe mcan (GBMstumorWh

turedlutionhighercells (Cel

were amediaEGFR(Fig. 2dicatefrom

Mazzoleni et al.

Cance7502

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

total number of viable cells in the correspondingped culture.

lts

expression identifies distinct cell subpopulationshuman primary GBM and GBM CSC lines

eral GBM surgical specimens were tested for EGFRexpression by both microarrays and semiquantitativeR and all were shown to express the transcript (Table 1).er, immunohistochemistry on corresponding paraffin-ded sections showed that, in agreement with the liter-only 50% of specimens were characterized by diffusestaining, which was particularly intense in small-sizedtumor cells (EGFRpos tumor samples; Fig. 1A, Supple-ry Fig. S1). Accordingly, the other specimens displayedible EGFR expression that was restricted mostly to-infiltrating inflammatory cells (EGFRneg tumor sam-ig. 1A). Western blotting (WB) analysis confirmedmunohistochemistry findings (Fig. 1B). Hence, EGFRn distribution identifies two main distinct subtypesan GBM (16).

FISH, a subset of EGFRpos tumor specimens showedgene amplification and expression of mutant EGFRvIIIn some patients, correlated with EGFR protein levelsC and D; Table 1). Interestingly, EGFRpos tumorsined small and thin CD31-positive vessels (Fig. 1E),as EGFRneg tumors were characterized by an activelyrating vasculature [mean vessel area (±SEM), 853.1 ±nd 2,500.2 ± 615.1 μm2 in EGFRpos and EGFRneg GBMs,tively; P > 0.05, Student's t test; n = 7; Fig. 1E]. All spec-expressed similar levels of stem cell–associated

ns, such as Sox2 and BMI1 (Fig. 1F), and of glial fibril-idic protein (GFAP; data not shown).restingly, whereas immunohistochemistry indicatedGFR was expressed in the totality of GBM cells, flowetry suggested that EGFR expression could be re-only in a smaller proportion of cells. Of note, with-EGFRpos cell fraction, a conspicuous subset of cells

led with the endothelial marker CD34 and the hema-tic markers CD45 and CD11b (Fig. 1G), indicatinghey were stromal cells. The CSC marker CD133as more expressed in proper tumor cells than inal cells in EGFRpos tumors, whereas the oppositeetected in EGFRneg tumors. On the contrary, CD15lways more expressed in the stromal cell compart-in both EGFRpos and EGFRneg tumors (refs. 31, 32;mentary Table S1).s, the frequency of EGFRpos proper tumor cells incalculated after the exclusion of stromal cells, ranged% to 35% of the total tumor cell number (Table 1). Ofsome EGFRpos proper tumor cells also labeled withand/or CD15 markers, whereas EGFRneg cells did

ig. 1F and H; Supplementary Table S2).formally show that EGFR might also play a role in they of GBM CSCs in vitro, we tested 13 GBM CSC lines

usly validated for being bona fide CSC lines (Supple-ry Fig. S2). All CSC lines expressed EGFR mRNA,

genesfractio

r Res; 70(19) October 1, 2010

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

as only 5 of 13 CSC lines also expressed the EGFR pro-able 2). Among the EGFRpos CSC lines, only 6% to 70%s expressed the receptor, suggesting that multiple GBMpulations could also be identified based on EGFR ex-on in CSC lines.restingly, many EGFRpos and EGFRneg CSC linesned AC133pos and CD15pos cells (Table 2; Supplemen-able S3). However, one of five EGFRpos CSC lines andf eight EGFRneg CSC lines did not contain any AC133pos

D15pos cells at all.

expression identifies GBM subpopulations with active gene signaturemolecularly analyze the above-described GBM subpo-ons, we purified them from (a) EGFRpos single patients'specimens, (b) EGFRneg specimens, and (c) EGFRpos

nes.Rpos patient samples T1224, T0131, T0222, and T0321ned (a) EGFRpos/AC133neg cells (region R1), (b) EGFRpos/pos cells (R2), and (c) EGFRneg/AC133neg (R3) cells. Twoalso contained a small proportion of EGFRpos/CD15pos

Fig. 2A and B; Supplementary Table S2). Stromal cellsxcluded from sorting.ediately after cell sorting and purity assessment,

ut any in vitro manipulation, the different GBMns were subjected to transcriptional profiling by-transcript microarrays (Fig. 2C). EGFRpos cell subpo-ons displayed a specific molecular signature clearlyuishable from that of double-negative cells (R3). Ofa subset of 163 genes was differentially expresseden EGFRpos and EGFRneg cell subpopulations, evenisolated from distinct patient specimens, suggestingGFR expression in GBM might affect common mo-r determinants (Supplementary List S1). In additionFR, the differentially expressed genes (DEG) upregu-in EGFRpos versus EGFRneg GBM fractions were relat-biological processes such as tumor invasion and

ession [e.g., periostin (POSTN), LMO3, and AQP4].rsely, genes overexpressed in EGFRneg GBM fractionsostly involved in the interaction of tumor cells with

icroenvironment [e.g., PDGFRB, Notch3, PTPRK, lumi-LUM), and endothelin receptor A (EDNRA)]. Thus,contain molecularly distinct subpopulations ofcells.

en the same freshly isolated cell fractions were cul-in vitro by the NeuroSphere Assay under limiting di-conditions (33), EGFRpos cell fractions gave rise to afrequency of secondary neurospheres than EGFRneg

Fig. 2D).l subpopulations expressing different levels of EGFRlso purified from EGFRpos CSC lines (Fig. 2E–G). Im-tely after sorting, EGFRhigh (R4), EGFRlow (R5), andneg (R6) CSC subpopulations were validated by WBH) and subjected to transcriptional profiling, which in-d that each subpopulation was molecularly distinctthe others (Fig. 2I; Supplementary List S2). Of note,

upregulated in EGFRhigh and EGFRlow versus EGFRneg

ns were also highly expressed in the most malignant

Cancer Research

1. © 2010 American Association for Cancer

Page 4: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

molecand mSupplEGFRp

lignanAs s

dilutioEGFRprolife

showefew supresorsamewas noWh

EGFRl

Table R s n s

Patientcode

WHO Age/sex EGFR wt EGFR vIII EGFR gene EGFR protein EGFR protein

EGFRp

T01 M + aT02 /F + + aT12 M + aT12 M + + aT04T06T03 M + + a

EGFRn

T01 M + mT01 M + mT01 M + mT01 /F + mT03 M + mT0512 IV 75/F 1+ 0 No amplification 5–10 1+T1003 IV 65/M 1+ 0 No amplification 5–10 1+

Patient code Total EGFRpos cells (%) EGFRpos tumor cells (%) EGFRpos stromal cells (%)

EGFRpositive GBMsT0131 20.8 4.0 16.8T0222 76.8 35.2 41.6T1210 13.7 10.7 3.0T1224 78.1 20.3 57.8T0418 36.6 12.4 24.2T0625 22.4 8.9 13.5T0321 39.1 21.7 17.4

EGFRnegative GBMsT0104 4.9 0.9 4.0T0109 17.3 0.0 17.3T0125 19.0 1.6 17.4T0130 25.3 3.3 22.0T0325 0.0 0.0 0.0T0512 4.4 0.0 4.4T1003 7.5 0.0 7.5

NOTE: FISH scores: high gene amplification, >10 gene copies in >10% of the cells; low gene amplification, 6 to 10 gene copies in>10% of the cells; high polysomy, >4 gene copies in >40% of the cells; low polysomy, >4 gene copies in >10% but <40% of thecells; no amplification, 1 to 4 gene copies. Immunohistochemistry scores: 3+, very intense membrane staining; 2+, intensemembrane staining; 1+, moderate staining; 0, no staining.

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

ular subclasses of primary GBM (i.e., the proliferativeesenchymal subtypes, as identified in refs. 21 and 34;ementary Fig. S3). Likewise, genes downregulated inos fractions were also downregulated in the same ma-t molecular subtypes.hown by clonogenic assays performed under limitingn conditions and by long-term population analysis,

low and EGFRneg cell fractions displayed the highestrative capacity, whereas EGFRhigh cell subpopulations

EGFRn

Howe

acrjournals.org

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

d a transiently reduced growth ability that required abculturing passages to be completely restored to theting levels (Fig. 2J). All subpopulations contained thepercentage of Bmi1- and Sox2-expressing cells, whicht affected by cell differentiation (Supplementary Fig. S4).en tested immediately after sorting, EGFRhigh andow fractions contained 100% of EGFRpos cells, whereas

1. EGF

expre sion in G BM patie ts' tumor pecimens

eg fractionsver, after s

1. © 2010 A

were devoid of any EGeveral subculturing p

Cancer Res; 70

merican Association

tumorstage

mRNA mRNA amplification expression(% of positive cells)

expression (membranestaining intensity)

os GBMs

31 IV 58/ 2 0 High mplification 70–90 3+ 22 IV 57 2 1 High mplification 80–100 3+ 10 IV 56/ 2 0 High mplification 80–100 3+ 24 IV 59/ 2 1 High mplification 70–90 3+ 18 IV 63/ 1 0 High olysomy 70–90 2+ M + p25 IV 74/F 1+ 0 Low amplification 70–90 2+ 21 IV 59/ 2 1 High mplification 70–90 3+ eg GBMs 04 IV 49/ 1 0 No a plification 10–20 1+ 09 IV 47/ 1 0 No a plification 10–20 1+ 25 IV 49/ 1 0 No a plification 10–20 1+ 30 IV 72 1 0 No a plification 15–25 1+ 25 IV 51/ 1 0 No a plification 0 0

FRpos cells (Fig. 2K).assages, each cell

(19) October 1, 2010 7503

for Cancer

Page 5: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

Figurefor EGF(black athat thetumorsCD31 scells caEGFRpo

EGFRpo

(top rig

Mazzoleni et al.

Cance7504

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

1. EGFR expression identifies distinct cell subpopulations within primary human GBM. A, EGFRpos GBM tumors show diffuse and intense reactivityR in small and round neoplastic cells (T1224). EGFRneg GBM specimens show EGFR staining restricted to tumor-infiltrating inflammatory cellsrrow; T0109). Magnification, ×400 and ×800. B, WB confirms EGFR protein levels in GBM patients' specimens. C, FISH analysis indicatesEGFR gene is amplified only in EGFRpos tumors. D, the mutant EGFRvIII can be detected in a subset of EGFRpos tumors by RT-PCR. E, EGFRpos

contain modestly proliferating vessels (arrows), whereas EGFRneg tumors show an enlarged and proliferating vasculature (arrows). H&E andtaining; magnification, ×200 and ×400. F, EGFRpos and EGFRneg tumors contain similar numbers of Sox2 and Bmi1pos cells. CD15pos proper tumorn be detected only in EGFRpos (arrow at bottom right, CD15pos monocytes within a vessel). Magnification, ×400. G, in EGFRpos tumors, manys cells are proper tumor cells, whereas in EGFRneg tumors the few EGFRpos cells are stromal/inflammatory cells. H, the EGFRpos fractions froms pos pos neg

specimens contain AC133 and CD15 cells (top middle), which are either tumor cells or stromal cells. On the contrary, EGFR fractionsht) and EGFRneg GBM tumors (bottom) contain only stromal AC133pos and CD15pos cells.

r Res; 70(19) October 1, 2010 Cancer Research

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

fractioEGFRn

ulatin

EGFRcapacTo

morigpurifieinto thnipulavolumterizedever, Etumorthan tanalysNot

graftsfromxenogor CDcentlyEGFRgiventationlates w

Althsameshownarray-in EGin angxenogIn a

ular d(Fig. 2highlygray mand GtumorboundmatteS6). Acharaca lowcellshigherand SThu

genic,Not

Table iz E n S n atCSC a 1 D

CSC l

EGFRp

L06 + 0 a ± ± ±L08 + 0 p ± ± ±L03L02L06 + 0 p ± 0. .0

EGFRn

L01 + 0 m ± ± ±L02 + 0 m .0 0. ±L01 + 0 m .0 0. .0L12 + 0 m ± 0. .0L03 + 0 m .0 0. ±LLL

NOTmeacopof tthrosubculturing passages.

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

n regenerated the same frequency of EGFRpos andeg cells found in the parental bulk population, recapit-g the original cellular heterogeneity (Fig. 2K).

expression confers increased tumorigenicity to GBM cellsassess whether EGFR expression might affect the tu-enic ability of GBM cells, R1, R2, and R3 fractions,d from EGFRpos GBM specimens, were transplantede brain of nu/nu mice, without any prior in vitro ma-tion (Fig. 3A; Supplementary Fig. S1). MRI-basedetric analysis showed that EGFRpos cells were charac-by enhanced tumorigenic capacity (Fig. 3A). How-GFRneg/AC133neg cells were also capable of initiatings, although these developed significantly more slowlyhose generated by EGFRpos GBM cells (Kaplan-Meieris; Fig. 3B).ably, EGFR expression was retrieved not only in xeno-derived from EGFRpos cells but also in those derivedEGFRneg fractions (Fig. 3C–E). Importantly, the latterrafts never contained either AC133pos cells (Fig. 3E)15pos cells (Fig. 3F, Supplementary Fig. S1). Thus, as re-shown for other stem cell markers in melanoma (35),expression in GBM TICs is dynamically regulated,that EGFRneg cells can become EGFRpos on transplan-

. Most importantly, EGFR expression positively corre-ith gliomagenesis.

xenogfrom

acrjournals.org

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

ough R1-, R2-, and R3-derived xenografts showed theexpression of SOX2 and Bmi1 (Fig. 3G and data not), they were molecularly different (Fig. 3H). By micro-based analysis, proinvasive genes were overexpressedFRpos cell–derived xenografts, whereas genes involvediogenesis were upregulated in EGFRneg cell–derivedrafts (Supplementary Fig. S5).greement with these latter findings and with molec-ata obtained on EGFRpos and EGFRneg subpopulationsC), xenografts from EGFRpos cells were shown to beinvasive, with cells spreading preferentially along theatter while avoiding white matter fiber tracts (Fig. 3C; Supplementary Figs. S1 and S6). On the contrary,s formed by EGFRneg cells showed well-demarcatedaries, with cells migrating preferentially along whiter tracts (Fig. 3C and G; Supplementary Figs. S1 andccordingly, EGFRpos cell–induced xenografts wereterized by thin, regularly shaped blood vessels andproliferative index, whereas tumors from EGFRneg

contained enlarged and proliferating vessels and afrequency of mitotic cells (Supplementary Figs. S1

6).s, EGFR expression confers different invasive, angio-and tumorigenic abilities to GBM cells in vivo.ably, the same functional differences observed between

2. Characterenrichment m

ation ofrkers AC

GFRpos a33 and C

d EGFRneg GBM C15

rG

1

C lines for their

afts generated by EGBM specimens coul

C

. © 2010 American

expressio

FRpos and EGd also be de

ancer Res; 70

Association

of the put

FRneg TICs ptected in xeno

(19) October 1

for Cancer

ive

ine code EGFR wtmRNA

EGFR vIIImRNA

EGFR geneamplification (FISH)

PTEN expression(WB)

EGFRpos

cells (%)AC133pos

cells (%)CD15pos

cells (%)

ositive CSC lines

27 2 High mplification 2+ 25.8 3.5 42.9 15.2 22.3 2.9 04 2 High olysomy 0 7.7 1.2 5.9 0.9 2.7 1.0 06 2 Low mplification 3+ 21.1 3.0 0 28.1 2.9 + 0 a ± 0. ±01 2+ 0 High polysomy 1+ 79.1 ± 3.9 3.1 ± 1.5 13.7 ± 0.9 05 2 Low olysomy 0 8.5 2.8 0 0 egative CSC lines 04 1 No a plification 0 2.0 0.8 30.1 13.8 50.8 3.2 23 1 No a plification 3+ 0 0 10.7 1.3 25 1 No a plification 1+ 0 0 0 10 1 No a plification 0 2.4 1.2 0 0 14 1 No a plification 3+ 0 0 5.9 1.7 25 1 No a plification 0 0 0 0 03 + 0 m .0 0. .0

0512 1+ 0 No amplification 3+ 1.7 ± 0.6 2.1 ± 1.4 11.7 ± 2.90418 1+ 0 No amplification 0 0.0 18.2 ± 4.5 0.0

E: EGFR wt and EGFRvIII mRNA levels were measured by semiquantitative RT-PCR. EGFR, AC133, and CD15 expression wassured by flow cytometry. High gene amplification, >10 gene copies in >10% of the cells; low gene amplification, 6 to 10 geneies in >10% of the cells; high polysomy, >4 gene copies in >40% of the cells; low polysomy, >4 gene copies in >10% but <40%he cells; no amplification, 1 to 4 gene copies. The frequency of EGFRpos cells within EGFRpos CSC lines remained stableughout extensive subculturing. Likewise, EGFRneg CSC lines were never shown to comprise EGFRpos cells even at very high

urifiedgrafts

, 2010 7505

Page 7: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

Figure(R1), EGexpress(right). Dcells. Mfrom EGEGFR isEGFRpo

and afte

Mazzoleni et al.

Cance7506

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

2. EGFR expression identifies GBMsubpopulationswith distinctive gene signatures and different growth characteristics in vitro. A andB, EGFRpos/AC133neg

FRpos/AC133pos (R2), and EGFRneg/AC133neg (R3) cells were isolated from EGFRpos tumor specimens by FACS (IgGs, isotype controls). C, global geneiondata in R1, R2, and R3 fractions purified from patients' specimens (left). The expression of a selection of coregulated genes is highlighted in the heatmaps, limiting dilution clonogenic assay of R1, R2, and R3 fractions immediately after sorting. E, different CSC lines comprise variable amounts of EGFRpos

agnification, ×200 (insets, ×600). F, some EGFRpos CSCs display EGFR gene amplification. G, different CSC subpopulations could be isolated by FACSFRpos CSC lines based on their EGFR expression (region R4, EGFRhigh CSCs; R5, EGFRlow CSCs; R6, EGFRneg CSCs; L0627). H, WB analysis shows thatactivated in EGFRhigh and EGFRlow CSCs but is absent in EGFRneg CSCs. I, global gene expression data of R4, R5, and R6 GBM fractions purified from

s

CSC lines.SemiquantitativeRT-PCRvalidationofDEGs. J, clonogenic assayand long-termgrowthcurvesofR4,R5, andR6CSCs immediately after sortingr in vitro culture. K, EGFR expression in CSC fractions immediately after sorting (top) and on in vitro culture (bottom; EGFR, green; magnification, ×200).

r Res; 70(19) October 1, 2010 Cancer Research

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

FigureAC133,experimEGFRne

dispersthe injeE, the scytomecould bconfersexperimonly in

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

3. EGFR expression identifies TICs with different tumorigenic capacities. A, EGFRpos cell–derived tumors, independently of the coexpression ofwere significantly larger than tumors induced from EGFRnegAC133neg cells. P < 0.05, one-way ANOVA with Bonferroni test (n = 5, six independentents). B, Kaplan-Meier survival curves (P < 0.01). C, EGFR expression detected by an antihuman EGFR is retrieved in all xenografts, includinggAC133neg cell–derived tumors (EGFR, green). EGFRpos cell–derived tumors are characterized by an infiltrative pattern of growth. Tumor cellse within the gray matter, avoiding the corpus callosum (white arrow). EGFRnegAC133neg cells give rise to expanding tumors, mostly confined withinction site, which migrate and invade along white matter tracts (white arrowhead). D, WB confirms EGFR protein expression in all xenografts.ame distinct EGFRpos and EGFRneg subpopulations as detected in the parental tumor were retrieved in the corresponding xenografts by flowtry. CD15pos cells (F) were not retrieved in any xenografts by immunohistochemistry. G, a similar frequency of Sox2 and Bmi1 immunoreactive cellse retrieved in R1- and R3-derived xenografts. H, global gene expression data of R1- and R3-derived xenografts. I to J, EGFR expressionenhanced tumorigenic potential to EGFRpos fractions from EGFRpos CSC lines (one-way ANOVA with Bonferroni test, n = 5, three independent

ents). P < 0.01, Kaplan-Meier survival curves. K, EGFR expression was retrieved in all xenografts, whereas ErbB2 was highly expressedEGFRhigh CSC–derived tumors (WB).

Cancer Res; 70(19) October 1, 2010acrjournals.org 7507

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

generalines (Bec

graftswe puthat wpropeTablexenogAC133play aFig. S7serialidentifbonaThe

EGFRn

(Figs.L0627testedspecimrivedthosedisplaEGFRconfirperimwas dgestin(Fig. 3To

EGFRtermsEGFRCD15n

EGFRn

L0627EGFRp

of ACmuchEGFRn

lines/sgenertwo mtary FThu

tionatEGFRthe Eand SexpressizedscriptEGFRIndeedadultefficieFig. S2

to staEGFRgressiogens i

Enforthe mLen

distincly funEGFR-develoknownenhanConsisEGFR-Gen

overexDEGssuggesameCSC lITGA3in FAmentaICAM1(Fig. 4mesentary FSPARCdownrregulaNot

mentacorres

GenetmaligTo

L0201in 70%shRNAdown(Fig. 5ysis coinhibisilencEGF

in vitrcells (cells dboth ptionsOf

develothose

Mazzoleni et al.

Cance7508

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

ted by in vitro expanded EGFRpos and EGFRneg CSCSupplementary Fig. S2; ref. 36).ause EGFR expression in EGFRneg cell–derived xeno-might be due to contaminating EGFRpos tumor cells,rified EGFRneg tumor cells from EGFRneg specimensere totally devoid of EGFRpos, AC133pos, and CD15pos

r tumor cells (Fig. 1A, G, and H; SupplementaryS1). FACS-purified EGFRneg/AC133neg cells generatedrafts that expressed EGFR but were negative forand CD15, thus confirming that EGFR might indeedprominent role in glioma formation (Supplementary). Notably, both in vivo limiting dilution analysis andtransplantation proved that all GBM subpopulationsied based on EGFR expression could be consideredfide TICs (Supplementary Fig. S8).tumorigenic potential of EGFRhigh, EGFRlow, and

eg cells purified from different EGFRpos CSC lines2G and 3I and J and Supplementary Fig. S9 forand Supplementary Fig. S9 for L0605) was also. As observed for xenografts obtained from GBMen–derived subpopulations (Fig. 3A–E), tumors de-from EGFRhigh and EGFRlow CSCs were larger thangenerated from EGFRneg cell fractions, which, again,yed tumor-initiating ability (Fig. 3I and J). Again,neg CSC fraction–derived tumors expressed EGFR,ming that EGFR upregulation occurred during ex-ental tumorigenesis (Fig. 3K). ErbB2 expressionetected only in EGFRhigh cell–derived tumors, sug-g that the xenografts were molecularly heterogeneousK).understand the relationship between the expression ofand that of the CSC markers CD15 and AC133 inof tumor-initiating potential, EGFRposCD15neg (R4),

posCD15pos (R5), EGFRnegCD15pos (R6), and EGFRneg

eg (R7) as well as EGFRposCD15negAC133neg (R1) andegCD15negAC133neg (R3) fractions were purified fromand L0605 (Supplementary Fig. S9). Interestingly,os subpopulations, independent of the coexpression133 and/or CD15, generated tumors that developedearlier and grew larger than those generated byeg fractions. Consistent with previous findings, CSCubpopulations negative for EGFR, AC133, and CD15ated xenografts that did not express either of thearkers in vivo but did express the EGFR (Supplemen-igs. S2 and S9).s, all of our GBM CSC lines, taken as a whole or frac-ed into subpopulations and independent of their basalexpression, gave rise to xenografts that upregulatedGFR significantly (Fig. 3K; Supplementary Figs. S29). To understand the mechanism underlying EGFRsion in EGFRneg cell–derived xenografts, we hypothe-that because EGFRneg GBM cells retain EGFR tran-expression (Tables 1 and 2; Fig. 2C and I), in vivoexpression might be regulated posttranscriptionally., when we simulated in vitro the mitogen-reducedbrain microenvironment by starvation, EGFRneg CSCs

ntly reexpressed the EGFR protein (Supplementary). Accordingly, when the same CSCs were reexposed

increamice

r Res; 70(19) October 1, 2010

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

ndard mitogen concentrations, they downregulatedexpression, as may occur in vivo during tumor pro-n when tumor-infiltrating stromal cells secrete mito-n a paracrine fashion (37).

ced EGFR expression by gene transfer increasesalignant behavior of CSCs in vitro and in vivotiviral vector–mediated EGFR expression in threet EGFRneg CSC lines allowed the expression of a ful-ctional receptor (Fig. 4A). Of note, the majority oftransduced cells acquired a fibroblastoid shape andped stress fibers and actin-driven lamellipodia, allmarkers of epithelial-mesenchymal transition and

ced malignancy (Fig. 4B; Supplementary Table S4).tently, the average cell size was increased in mosttransduced cells (Fig. 4C; Supplementary Table S4).e expression profiling of naïve, mock, and EGFR-pressing CSC lines identified a common subset ofthat was altered in both L0104 and L0125 CSC lines,sting that enforced EGFR expression affected themolecular determinants in different patient-derivedines (Fig. 4D and E). In addition to EGFR, LIF, and, genes previously shown to be also highly expressedCS-purified EGFRpos CSC fractions (Fig. 2I; Supple-ry List S2), proinvasive genes such as CXCL10 and(38, 39) were upregulated in EGFR-transduced CSCsD–E) and also overexpressed in the proliferative andchymal subtypes of human GBM (ref. 2I; Supplemen-ig. S3). Accordingly, tumor suppressors such asL1 (Fig. 2I), ERBB4, ASCL1, and EDNRB, which wereegulated in EGFR-transduced CSCs, were also down-ted in GBM malignant subtypes.ably, all EGFR-transduced CSC lines formed experi-l tumors that were larger than those induced by theponding mock lines (Fig. 4F).

ic loss of function of EGFR reduces thenancy of CSCs in vitro and in vivosilence EGFR expression stably, we exploited lineand line L0627, which express high levels of EGFRand 30% of the cells, respectively (Table 2). Theclones 204 and 206 induced EGFR protein knock-with an efficiency of 60% and 70% in lines L0201A) and L0627 (data not shown), respectively. WB anal-nfirmed the flow cytometry results and indicated antion of downstream signaling pathways on EGFRing (Fig. 5B).R-knockdown CSCs displayed morphologic changeso, with neurospheres appearing as well-differentiatedFig. 5C). Consistently, the frequency of differentiatedetected in shRNA-transduced CSCs increased underroliferative (Fig. 5D) and differentiative culture condi-(data not shown).note, tumors generated from EGFR-knockdown CSCsped more slowly and were significantly smaller thangenerated by the CTRL subclone, thus resulting in the

sed survival of EGFR-knockdown CSC–transplanted(Fig. 5E).

Cancer Research

1. © 2010 American Association for Cancer

Page 10: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

Figurea functin EGFR(phalloisize. FSare corvalidatit test, n

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

4. Overexpression of EGFR enhances the tumorigenic potential of CSCs. A, lentiviral-mediated EGFR gene transfer resulted in the expression ofional EGFR, as indicated by phosphorylation at residue Tyr1068 and by the hyperactivation of downstream pathways. B, enforced EGFR expressionneg CSCs (EGFR, red) correlates with changes in cell morphology, which are absent in GFP-transduced CSCs (GFP, green). F-actin staining

din, red). Magnifications: top, ×400; insets, ×1,000; bottom, ×1,000. C, EGFR overexpression in EGFRneg CSCs induces a significant increase in theirC-A and SSC-A. D, global gene expression data in naïve, mock-transduced, and EGFR-transduced CSC lines identify a subset of DEGs thategulated in different patient-derived CSC lines and modulated by enforced EGFR expression (EGFR gene signature). E, semiquantitative RT-PCR

neg

on of DEGs. F, ectopic expression of EGFR in EGFR CSC lines increases their tumorigenic potential and reduces mouse survival (Student's= 5, three independent experiments).

Cancer Res; 70(19) October 1, 2010acrjournals.org 7509

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

FigureshRNAC and DE, EGFP < 0.0responinhibitioPTEN e

Mazzoleni et al.

Cance7510

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

5. Inhibition of EGFR expression and activity negatively affects the malignant behavior of CSCs. A and B, flow cytometry and WB of L0201204 and shRNA 206 CSC subclones indicate efficient knockdown of EGFR protein and downregulation of MAPK and Akt downstream pathways., EGFR knockdown induces CSC differentiation. Tuj1, red; GFAP, green. Magnifications: top, ×200; bottom, ×400. *, P > 0.05, Student's t test.

R silencing in CSCs significantly decreases their tumorigenicity (one-way ANOVA with Bonferroni test, n = 4, two independent experiments).1, Kaplan-Meier survival curves. F, AG1478 treatment inhibits the phosphorylation status of EGFR and Akt in CSCs (WB). G, EGFRpos CSC linesd to treatment by decreasing their proliferation/survival, whereas EGFRneg CSC do not. *, P > 0.05; **, P > 0.01, Student's t test. H and I, EGFR

n by AG1478 negatively affects the invasive ability of CSCs. **, P > 0.01, Student's t test. J, CSC responsiveness to TKI is independent ofxpression (WB).

r Res; 70(19) October 1, 2010 Cancer Research

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 12: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

EGFRto phTo

the EGsis ofAG147of EGAKT pPha

AG147decreathe sunot aftransdgestinbecomsure osive cAgain,fectedInte

macolPTEN,ness iThu

bition

Discu

EGFRGBM

whichHowevonly tobut altinct cIn l

tionspoietimelancate thpend opropo(43). Ocontexdifferemorigular fsubpoThey smentathe tuon serBas

mightTICs,lines (

tumorexpresThu

jorityhierarto GB

MechGBMTum

progre(45). IcontriTIC pgenerin GBneedsgandstumortivatioand emal gtion ofollow

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

pos and EGFRneg CSC lines differentially respondarmacologic inhibition of EGFRpharmacologically inhibit EGFR activity, we exploitedFR tyrosine kinase inhibitor (TKI) AG1478. WB analy-two different EGFRpos CSCs that were exposed to8 for 2, 5, and 16 hours showed effective inhibitionFR autophosphorylation and a significant decrease inhosphorylation, a hallmark of drug sensitivity (Fig. 5F).rmacologic treatment of EGFRpos CSC lines with8 for up to 72 hours in vitro resulted in a significantse in CSC survival/proliferation (Fig. 5G). Relevantly,rvival and proliferation of EGFRneg CSC lines werefected by AG1478, even when the same lines wereuced with EGFR-coding vectors before treatment, sug-g that EGFR-overexpressing EGFRneg CSC lines did note addicted to EGFR (data not shown). Likewise, expo-f EGFRpos CSCs to AG1478 strongly reduced their inva-apacity as compared with controls (Fig. 5H and I).the invasive ability of EGFRneg CSC lines was not af-by AG1478 exposure.restingly, all the EGFRpos CSC lines responded to phar-ogic treatment independently of the expression ofa putative molecular determinant of TKI responsive-

n patients with GBM (Fig. 5J; ref. 40).

s, EGFRpos CSCs are responsive to pharmacologic inhi- In

the EmightEGFRcessivwe simtion. IGBMadditileculaTICs.sine kfractiosynergperimtion hthat dthe laPDGFEGF

mentatherefHowehancetheyConveefficieigenicwhichdownrate stIn lin

by TKIs, whereas EGFRneg CSCs are not.

ssion

expression contributes to GBM heterogeneityis characterized by a high degree of heterogeneity,

is efficaciously reflected in the definition “multiforme.”er, the intrinsic heterogeneity of GBM might refer notthe well-known molecular diversity of GBM subtypes

so to the existence of genetically and functionally dis-ohorts of TICs within the same tumor.ine with recent reports that provide new interpreta-of the process of tumorigenesis in mouse hemato-c malignancies (8), mouse colon cancer (41), humanoma (10, 11), and brain tumors (42), our findings indi-at the initiation and progression of GBM may not de-n the presence of a single and rare fraction of CSCs, assed by the hierarchical model of tumor developmentn the contrary, our data propose a different biologicalt, in which all GBM cells can be considered TICs withnt degrees of stemness as proven by their different tu-enic ability and by their distinct phenotypic and molec-eatures (44). In fact, all of our GBM-derived TICpulations satisfy the requirements for bona fide CSCs:elf-renew in vitro and in vivo and give rise to experi-l tumors that recapitulate the phenotypic traits ofmor of origin under limiting dilution conditions andial transplantation.ed on these observations, GBM tissue heterogeneitybe explained by the presence of a hierarchy of distinct

as recently suggested for in vitro generated GBM CSC13). These distinct TICs distribute along a gradient of

GBMson rec

acrjournals.org

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

igenic potential that is strictly associated with EGFRsion.s, our study provides conclusive evidence that the ma-of GBM cells have tumorigenic capacity and that thechical model of tumorigenesis might not fully applyM.

anisms underlying EGFR-dependentheterogeneityor cells might modify their phenotype during tumorssion, with multiple cohorts of TICs being generatedn this study, we have shown that EGFR expressionbutes to GBM cell diversity by generating differentopulations. As to the mechanisms involved in theation of EGFR-dependent phenotypic heterogeneityM, the contribution of tumor microenvironmentto be taken into consideration. Paracrine EGFR li-, such as epidermal growth factor, are released bystromal cells that, in this way, contribute to the ac-n of the EGFR signaling in both proper tumor cellsndothelial cells (37). When stroma-produced epider-rowth factor is secreted, EGFR protein downregula-ccurs in target cells by receptor internalization,ed by lysosomal degradation.line with this view, it is tempting to speculate thatGFRneg cell component detected in EGFRpos GBMbe generated from GBM cells that were originally

pos through receptor downregulation induced by ex-e ligand stimulation during tumor progression, asulated in vitro by modulation of mitogen concentra-n addition, termination of EGFR signaling in EGFRneg

cells might lead to the acquisition of compensatoryonal genetic events, which might account for the mo-r differences detected between EGFRpos and EGFRneg

For instance, the expression of other receptor tyro-inases, such as PDGFRs, observed in EGFRneg TICns might contribute to their tumorigenic ability byizing with the EGFR re-upregulation required for ex-ental gliomagenesis. Indeed, PDGFR pathway activa-as been recently shown to identify a GBM subtypeoes not express the EGFR (46), thus suggesting thattter GBM subtype might be sustained by EGFRneg

Rpos TICs.Rpos and EGFRneg TICs can both give rise to experi-l tumors that phenocopy the tumor of origin, andore, they both can be considered bona fide TICs.ver, because EGFRpos TICs are characterized by en-d tumorigenic potential and highly invasive behavior,might be envisioned as the “actual” GBM CSCs.rsely, EGFRneg TICs, which form tumors with lowncy and need to re-upregulate the EGFR to be tumor-, might be better defined as “potential” GBM CSCs,might be kept in a dormancy-like state by EGFRegulation, being reactivated on exposure to appropri-imuli when exposed to the in vivo microenvironment.e with this notion, it has been recently shown that

, which were EGFRneg in origin, expressed the EGFRurrence (47).

Cancer Res; 70(19) October 1, 2010 7511

1. © 2010 American Association for Cancer

Page 13: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

TheraGBMThe

and itDespiexpresmeansstrategTo

achievascribdepencoactirosinehibitioBy

gest ththe satargetbitionindepresponical pecompoEGFRn

cells tThereconsidthis ennot ontions bbe nec

Discl

No p

Ackn

We tAntoneediting

Grant

ComResearc(R. Gall

Theof pageaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Refe1. Bo

hie199

2. VeCa

3. Dic479

4. Al-ProNaAc

5. Peheco

6. Ricexp11

7. O'BcelNa

8. Kenee

9. Adste

10. QuMoce

11. HeBopro

12. Beglich

13. Chren20

Mazzoleni et al.

Cance7512

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

peutic implications of EGFR-dependentheterogeneityrole of EGFR in the malignant progression of GBMs effect on patient survival have been highly debated.te the controversial prognostic significance of EGFRsion in GBM, pharmacologic targeting of EGFR byof TKIs has been proposed as a possible therapeuticy (40).date, single-agent EGFR inhibition in GBM did note complete therapeutic efficacy. This failure has beened primarily to the maintenance of a high level of Akt-dent signaling in PTEN-mutated tumors (48) and tovation in the same tumor cell of multiple receptor ty-kinases, which might compensate for the effective in-n of EGFR-dependent pathways (49).complementing these explanations, our findings sug-at the simultaneous presence of distinct TICs withinme tumor might also influence the outcome of EGFR-ed therapies in GBM. Indeed, whereas effective inhi-of EGFR in our EGFRpos CSC lines can be achievedendently of PTEN expression, EGFRneg CSCs do notd to treatment. By translating these results into a clin-rspective, in spite of the inhibition of the EGFRpos cell

nent in EGFRpos GBM, the nonresponsiveness of the

en R, Nishimura MC, Bumbaca SM, et al. A hierarchy of self-ewing tumor-initiating cell types in glioblastoma. Cancer Cell06;17:362–75.

14. Nigrome

15. Bewitclin

16. Fuma26

17. Ohvivglio

18. Hetor20

19. Lagrogliooth14

20. Hogliotic

21. Phhigpro20

22. Suma

23. Arwof67

24. Lugepa

25. Yin

r Res; 70(19) October 1, 2010

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

o survive treatment and to support tumor relapse.fore, an effective therapeutic regimen should take intoeration the role of residual, nonresponder TICs. Tod, the development of a combination therapy targetedly against the most aggressive “actual” TIC popula-ut also against the less malignant “potential” TICs willessary to improve the management of GBM.

osure of Potential Conflicts of Interest

otential conflicts of interest were disclosed.

owledgments

hank Alessio Palini and Chiara Villa for technical assistance with FACS.lla Iadanza for technical assistance with MRI, and Carol Stayton forof the manuscript.

Support

pagnia S. Paolo grants CSP-2002.0735 and CSP-2005.0153, the Oncologyh Programme-Istituto Superiore di Sanità, and the Fondazione Cariploi).costs of publication of this article were defrayed in part by the paymentcharges. This article must therefore be hereby marked advertisement in

eg TIC fraction to EGFR in

hibitors might allow these Received06/28/2010; accepted07/18/2010; publishedOnlineFirst 09/21/2010.

rencesnnet D, Dick JE. Human acute myeloid leukemia is organized as ararchy that originates from a primitive hematopoietic cell. Nat Med7;3:730–7.

scovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nat Revncer 2006;6:425–36.k JE. Stem cell concepts renew cancer research. Blood 2008;112:3–807.Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF.spective identification of tumorigenic breast cancer cells. Proctl Acad Sci U S A 2003;100:3983–8, [erratum appears in Proc Natlad Sci U S A. 2003 May 27;100(11):6890].ce S, Tosoni D, Confalonieri S, et al. Biological and molecularterogeneity of breast cancers correlates with their cancer stem cellntent. Cell 2007;140:62–73.ci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification andansion of human colon-cancer-initiating cells. Nature 2007;445:

1–5.rien CA, Pollett A, Gallinger S, Dick JE. A human colon cancerl capable of initiating tumour growth in immunodeficient mice.ture 2007;445:106–10.lly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor growthd not be driven by rare cancer stem cells. Science 2007;317:337.ams JM, Strasser A. Is tumor growth sustained by rare cancerm cells or dominant clones? Cancer Res 2008;68:4018–21.intana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM,rrison SJ. Efficient tumour formation by single human melanomalls. Nature 2008;456:593–8.ld MA, Curley DP, Dankort D, McMahon M, Muthusamy V,senberg MW. Characterization of melanoma cells capable ofpagating tumors from a single cell. Cancer Res 2007;70:388–97.ier D, Hau P, Proescholdt M, et al. CD133+ and CD133−

oblastoma-derived cancer stem cells show differential growtharacteristics and molecular profiles. Cancer Res 2007;67:4010–5.

cholas MK, Lukas RV, Jafri NF, Faoro L, Salgia R. Epidermalwth factor receptor-mediated signal transduction in the develop-nt and therapy of gliomas. Clin Cancer Res 2006;12:7261–70.nito R, Gil-Benso R, Quilis V, et al. Primary glioblastomas with andhout EGFR amplification: relationship to genetic alterations andicopathological features. Neuropathology 2010;30:392–400.rnari FB, Fenton T, Bachoo RM, et al. Malignant astrocytic glio-: genetics, biology, and paths to treatment. Genes Dev 2007;21:83–710.gaki H, Kleihues P. Population-based studies on incidence, sur-al rates, and genetic alterations in astrocytic and oligodendroglialmas. J Neuropathol Exp Neurol 2005;64:479–89.imberger AB, Suki D, Yang D, Shi W, Aldape K. The natural his-y of EGFR and EGFRvIII in glioblastoma patients. J Transl Med05;3:38.yfield LJ, Willmore C, Tripp S, Jones C, Jensen RL. Epidermalwth factor receptor gene amplification and protein expression inblastoma multiforme: prognostic significance and relationship toer prognostic factors. Appl Immunohistochem Mol Morphol 2006;:91–6.elzinger DB, Mariani L, Weis J, et al. Gene expression profile ofblastoma multiforme invasive phenotype points to new therapeu-targets. Neoplasia 2005;7:7–16.illips HS, Kharbanda S, Chen R, et al. Molecular subclasses ofh-grade glioma predict prognosis, delineate a pattern of diseasegression, and resemble stages in neurogenesis. Cancer Cell06;9:157–73.gawa N, Yamamoto K, Ueda S, et al. Function of aberrant EGFR inlignant gliomas. Brain Tumor Pathol 1998;15:53–7.ert E, Hingtgen S, Figueiredo JL, et al. Visualizing the dynamics

EGFR activity and antiglioma therapies in vivo. Cancer Res 2007;:7335–42.KV, Zhu S, Cvrljevic A, et al. Fyn and SRC are effectors of onco-

nic epidermal growth factor receptor signaling in glioblastomatients. Cancer Res 2009;69:6889–98.g H, Zheng H, Scott K, et al. Mig-6 controls EGFR trafficking and

Cancer Research

1. © 2010 American Association for Cancer

Page 14: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

sup691

26. Yacomo

27. GatumCa

28. Leegliophcel

29. Sofacgro12

30. Sintum

31. SomeSte

32. Remabla

33. Rere-

34. Leeysithe200

35. Rotinccon

36. SaindNa

37. Hytar

38. MachCX

39. Chmoast

40. MetheMe

41. Shreslon

42. OgCD20

43. Rean

44. Viscu20

45. Hil20

46. Breclawa

47. Maglio20

48. HarecJ N

EGFR Identifies Malignant Glioma-Initiating Cells

www.a

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353

presses gliomagenesis. Proc Natl Acad Sci U S A 2008;107:2–7.ng W, Wu G, Barth RF, et al. Molecular targeting and treatment ofmposite EGFR and EGFRvIII-positive gliomas using boronatednoclonal antibodies. Clin Cancer Res 2008;14:883–91.lli R, Binda E, Orfanelli U, et al. Isolation and characterization oforigenic, stem-like neural precursors from human glioblastoma.ncer Res 2004;64:7011–21.J, Kotliarova S, Kotliarov Y, et al. Tumor stem cells derived fromblastomas cultured in bFGF and EGF more closely mirror the

enotype and genotype of primary tumors than do serum-culturedl lines. Cancer Cell 2006;9:391–403.k JC, Coppelli FM, Thomas SM, et al. Mutant epidermal growthtor receptor (EGFRvIII) contributes to head and neck cancerwth and resistance to EGFR targeting. Clin Cancer Res 2006;:5064–73.gh SK, Hawkins C, Clarke ID, et al. Identification of human brainour initiating cells. Nature 2004;432:396–401.n MJ, Woolard K, Nam DH, Lee J, Fine HA. SSEA-1 is an enrich-nt marker for tumor-initiating cells in human glioblastoma. Cellm Cell 2009;4:440–52.ad TA, Fogarty MP, Markant SL, et al. Identification of CD15 as arker for tumor-propagating cells in a mouse model of medullo-stoma. Cancer Cell 2009;15:135–47.ynolds BA, Rietze RL. Neural stem cells and neurospheres—evaluating the relationship. Nat Methods 2005;2:333–6.Y, Scheck AC, Cloughesy TF, et al. Gene expression anal-

s of glioblastomas identifies the major molecular basis forprognostic benefit of younger age. BMC Med Genomics8;1:52.esch A, Fukunaga-Kalabis M, Schmidt EC, et al. A temporarily dis-t subpopulation of slow-cycling melanoma cells is required fortinuous tumor growth. Cell 2006;141:583–94.

kariassen PO, Prestegarden L, Wang J, et al. Angiogenesis-ependent tumor growth mediated by stem-like cancer cells. Proctl Acad Sci U S A 2006;103:16466–71.

49. Stotorthe

acrjournals.org

Research. on April 22, 202cancerres.aacrjournals.org Downloaded from

nes NE, Lane HA. ERBB receptors and cancer: the complexity ofgeted inhibitors. Nat Rev Cancer 2005;5:341–54.ru SV, Holloway KA, Flynn G, et al. Chemokine production andemokine receptor expression by human glioma cells: role ofCL10 in tumour cell proliferation. J Neuroimmunol 2008;199:35–45.oi K, Benveniste EN, Choi C. Induction of intercellular adhesionlecule-1 by Fas ligation: proinflammatory roles of Fas in humanroglioma cells. Neurosci Lett 2003;352:21–4.llinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants ofresponse of glioblastomas to EGFR kinase inhibitors. N Engl Jd 2005;353:2012–24.melkov SV, Butler JM, Hooper AT, et al. CD133 expression is nottricted to stem cells, and both CD133 and CD133 metastatic co-cancer cells initiate tumors. J Clin Invest 2008;118:2111–20.den AT, Waziri AE, Lochhead RA, et al. Identification of A2B5+

133− tumor-initiating cells in adult human gliomas. Neurosurgery08;62:505–14, discussion 514–505.ya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer,d cancer stem cells. Nature 2001;414:105–11.vader JE, Lindeman GJ. Cancer stem cells in solid tumours: ac-mulating evidence and unresolved questions. Nat Rev Cancer08;8:755–68.l RP, Perris R. “Destemming” cancer stem cells. J Natl Cancer Inst07;99:1435–40.nnan C, Momota H, Hambardzumyan D, et al. Glioblastoma sub-sses can be defined by activity among signal transduction path-ys and associated genomic alterations. PLoS One 2009;4:e7752.rtinez R, Rohde V, Schackert G. Different molecular patterns inblastoma multiforme subtypes upon recurrence. J Neurooncol

05;96:321–9.as-Kogan DA, Prados MD, Tihan T, et al. Epidermal growth factoreptor, protein kinase B/Akt, and glioma response to erlotinib.atl Cancer Inst 2005;97:880–7.mmel JM, Kimmelman AC, Ying H, et al. Coactivation of recep-

tyrosine kinases affects the response of tumor cells to targetedrapies. Science 2007;318:287–90.

Cancer Res; 70(19) October 1, 2010 7513

1. © 2010 American Association for Cancer

Page 15: Cancer Research Epidermal Growth Factor Receptor ... · Molecular and Cellular Pathobiology Epidermal Growth Factor Receptor Expression Identifies Functionally and Molecularly Distinct

2010;70:7500-7513. Published OnlineFirst September 21, 2010.Cancer Res   Stefania Mazzoleni, Letterio S. Politi, Mauro Pala, et al.   GliomagenesisHuman Glioblastoma Multiforme and Is Required forFunctionally and Molecularly Distinct Tumor-Initiating Cells in Epidermal Growth Factor Receptor Expression Identifies

  Updated version

  10.1158/0008-5472.CAN-10-2353doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2010/09/20/0008-5472.CAN-10-2353.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/70/19/7500.full#ref-list-1

This article cites 49 articles, 14 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/70/19/7500.full#related-urls

This article has been cited by 19 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. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/70/19/7500To request permission to re-use all or part of this article, use this link

Research. on April 22, 2021. © 2010 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 21, 2010; DOI: 10.1158/0008-5472.CAN-10-2353