clinical human cancer biology cancer research€¦ · human cancer biology characterization of...

11
Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru Qin 1 , Hong Tang 1 , Fajun Xie 2 , Haibo Liu 2 , Yinghui Zhu 2 , Jiaoyu Ai 1 , Leilei Chen 3 , Yan Li 2 , Dora L. Kwong 3 , Li Fu 2,3 , and Xin-Yuan Guan 2,3 Abstract Purpose: By using cDNA microarray analysis, we identified a transcriptional factor, SOX6, was frequently downregulated in esophageal squamous cell carcinoma (ESCC). The aim of this study is to investigate the role of SOX6 in human esophageal cancer development, and to examine the prevalence and clinical significance of SOX6 downregulation in ESCC. Experimental Design: Expressions of SOX6 mRNA in 50 ESCCs and SOX6 protein in 300 ESCCs were investigated by semiquantitative RT-PCR and immunohistochemistry, respectively. The tumor-suppressive function of SOX6 was characterized by cell growth, foci formation, wound-healing and cell invasive assays, and tumor xenograft experiment. Western blot analysis was applied to detect protein expression levels. Results: SOX6 was frequently downregulated in primary ESCCs in both mRNA level (29/50, 58%) and protein level (149/219, 68.0%), which was significantly associated with the poor differentiation (P ¼ 0.029), lymph node metastases (P ¼ 0.014), advanced TNM stage (P ¼ 0.000), and disease-specific survival (P < 0.001). Multivariate analysis indicated that the downregulation of SOX6 (P ¼ 0.000) was a significant independent prognostic factors for ESCC. Functional studies showed that SOX6 was able to suppress both in vitro and in vivo tumorigenic ability of ESCC cells. The tumor-suppressive mechanism of SOX6 was associated with its role in G1/S cell-cycle arrest by upregulating expressions of p53 and p21 WAF1/CIP1 and downregulating expressions of cyclin D1/CDK4, cyclin A, and b-catenin. Conclusions: We provided the first evidence that SOX6 is a novel tumor-suppressor gene in ESCC development and is a potential prognostic marker in ESCC. Clin Cancer Res; 17(1); 46–55. Ó2010 AACR. Introduction Esophageal squamous cell carcinoma (ESCC) is one of the most common fatal malignances of the digestive tract (1). Despite advances in diagnosis and multimodality therapies, ESCC continues to carry a grim prognosis (2). The major reason for this poor survival is the fact that ESCC frequently metastasizes to regional and distant lymph nodes, even at initial diagnosis. Recently, the treatment of cancer using molecular targets has brought promising results and attracted more and more attention. However, most of the molecular targets, which were commonly found in other epithelial neoplasias, have failed to show prognostic value for ESCC (3). Hence, characterization of genes involved in the development and progression of ESCC may lead to the identification of new prognostic markers and therapeutic targets (4, 5). Systematic analysis of expression levels of thousands of genes by cDNA microarray is an effective approach to identify novel genes and pathways with importance in cancer development and progression, or as targets for new therapies. Recently, genome-wide expression profiles between pooled cDNA from 10 primary ESCCs and their corresponding nontumorous tissues were compared by cDNA microarray containing 47,000 genes. Using an arbi- trary cutoff line at signal log ratio 2.0 or 2.0, 185 upregulated genes and 225 downregulated genes were detected in ESCC tumors as compared with normal con- trols (unpublished data). One frequently downregulated gene in ESCC specimen, SOX6, was further characterized. SOX6, a member of the D subfamily of sex determining region y-related transcription factors, encodes a protein that binds to DNA through a high-mobility-group (HMG) domain and plays critical roles in cell fate deter- mination, differentiation, and proliferati on (6–9). Some Sox factors, such as, SOX2, SOX4, and SOX9, have been Authors' Affiliations: 1 Department of Clinical Oncology, the First affiliated hospital, Zhengzhou University, Zhengzhou, China; 2 State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-Sen University, Guangzhou, China; and 3 Department of Clinical Oncology, University of Hong Kong, Hong Kong, China Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Xin-Yuan Guan, State Key Laboratory of Oncol- ogy in Southern China, Cancer Center, Sun Yat-Sen University, Room 605, 651 Dongfeng Road East, Guangzhou 510060, China. Phone: 86-20- 87343166; E-mail: [email protected]; or Li Fu, Department of Clinical Oncology, L10-56, Laboratory Block, 21 Sassoon Road, Hong Kong, China. Phone: 852-28199782; E-mail: [email protected]. doi: 10.1158/1078-0432.CCR-10-1155 Ó2010 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 17(1) January 1, 2011 46 on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Upload: others

Post on 14-Jun-2020

14 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

Human Cancer Biology

Characterization of Tumor-Suppressive Function of SOX6 in HumanEsophageal Squamous Cell Carcinoma

Yan-Ru Qin1, Hong Tang1, Fajun Xie2, Haibo Liu2, Yinghui Zhu2, Jiaoyu Ai1, Leilei Chen3, Yan Li2,Dora L. Kwong3, Li Fu2,3, and Xin-Yuan Guan2,3

AbstractPurpose: By using cDNA microarray analysis, we identified a transcriptional factor, SOX6, was

frequently downregulated in esophageal squamous cell carcinoma (ESCC). The aim of this study is to

investigate the role of SOX6 in human esophageal cancer development, and to examine the prevalence and

clinical significance of SOX6 downregulation in ESCC.

Experimental Design: Expressions of SOX6 mRNA in 50 ESCCs and SOX6 protein in 300 ESCCs were

investigated by semiquantitative RT-PCR and immunohistochemistry, respectively. The tumor-suppressive

function of SOX6was characterized by cell growth, foci formation, wound-healing and cell invasive assays,

and tumor xenograft experiment. Western blot analysis was applied to detect protein expression levels.

Results: SOX6 was frequently downregulated in primary ESCCs in both mRNA level (29/50, 58%) and

protein level (149/219, 68.0%), which was significantly associated with the poor differentiation (P ¼0.029), lymph nodemetastases (P¼ 0.014), advanced TNM stage (P¼ 0.000), and disease-specific survival

(P < 0.001). Multivariate analysis indicated that the downregulation of SOX6 (P¼ 0.000) was a significant

independent prognostic factors for ESCC. Functional studies showed that SOX6 was able to suppress both

in vitro and in vivo tumorigenic ability of ESCC cells. The tumor-suppressive mechanism of SOX6 was

associated with its role in G1/S cell-cycle arrest by upregulating expressions of p53 and p21WAF1/CIP1 and

downregulating expressions of cyclin D1/CDK4, cyclin A, and b-catenin.Conclusions: We provided the first evidence that SOX6 is a novel tumor-suppressor gene in ESCC

development and is a potential prognostic marker in ESCC. Clin Cancer Res; 17(1); 46–55. �2010 AACR.

Introduction

Esophageal squamous cell carcinoma (ESCC) is one ofthe most common fatal malignances of the digestive tract(1). Despite advances in diagnosis and multimodalitytherapies, ESCC continues to carry a grim prognosis (2).Themajor reason for this poor survival is the fact that ESCCfrequently metastasizes to regional and distant lymphnodes, even at initial diagnosis. Recently, the treatmentof cancer using molecular targets has brought promisingresults and attracted more and more attention. However,

most of the molecular targets, which were commonlyfound in other epithelial neoplasias, have failed to showprognostic value for ESCC (3). Hence, characterization ofgenes involved in the development and progression ofESCC may lead to the identification of new prognosticmarkers and therapeutic targets (4, 5).

Systematic analysis of expression levels of thousands ofgenes by cDNA microarray is an effective approach toidentify novel genes and pathways with importance incancer development and progression, or as targets fornew therapies. Recently, genome-wide expression profilesbetween pooled cDNA from 10 primary ESCCs and theircorresponding nontumorous tissues were compared bycDNA microarray containing 47,000 genes. Using an arbi-trary cutoff line at signal log ratio �2.0 or ��2.0, 185upregulated genes and 225 downregulated genes weredetected in ESCC tumors as compared with normal con-trols (unpublished data). One frequently downregulatedgene in ESCC specimen, SOX6, was further characterized.SOX6, a member of the D subfamily of sex determiningregion y-related transcription factors, encodes a proteinthat binds to DNA through a high-mobility-group(HMG) domain and plays critical roles in cell fate deter-mination, differentiation, and proliferati on (6–9). SomeSox factors, such as, SOX2, SOX4, and SOX9, have been

Authors' Affiliations: 1Department of Clinical Oncology, the First affiliatedhospital, Zhengzhou University, Zhengzhou, China; 2State Key Laboratoryof Oncology in Southern China, Cancer Center, Sun Yat-Sen University,Guangzhou, China; and 3Department of Clinical Oncology, University ofHong Kong, Hong Kong, China

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

Corresponding Author: Xin-Yuan Guan, State Key Laboratory of Oncol-ogy in Southern China, Cancer Center, Sun Yat-Sen University, Room 605,651 Dongfeng Road East, Guangzhou 510060, China. Phone: 86-20-87343166; E-mail: [email protected]; or Li Fu, Department of ClinicalOncology, L10-56, Laboratory Block, 21 Sassoon Road, Hong Kong,China. Phone: 852-28199782; E-mail: [email protected].

doi: 10.1158/1078-0432.CCR-10-1155

�2010 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 17(1) January 1, 201146

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 2: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

identified as tumor suppressors that were frequently down-regulated in various cancers (10–15). However, to ourknowledge, the role of SOX6 in esophageal cancer devel-opment and progression has not been explored.In this study, the expression pattern of SOX6 in clinical

ESCC samples and ESCC cell lines was studied by RT-PCR.Both in vitro and in vivo assays were used to study the tumor-suppressive function of SOX6. In addition, the tumor-suppressive mechanism of SOX6 and its clinical signifi-cance in ESCC was also investigated.

Materials and Methods

Cell lines and primary tumor specimensChinese ESCC cell line HKESC1 was kindly provided by

Professor Srivastava (Department of Pathology, The Uni-versity of Hong Kong), and 2 Chinese ESCC cell lines (EC18and EC109) were kindly provided by Professor Tsao(Department of Anatomy, The University of Hong Kong).Six Japanese ESCC cell lines (KYSE30, KYSE140, KYSE180,KYSE410, KYSE510, and KYSE520; ref. 16) were obtainedfrom DSMZ, the German Resource Centre for BiologicalMaterial. Fifty pairs of primary ESCC tumors and theiradjacent nontumorous tissues from the proximal resectionmargins were collected immediately after surgical resectionat Linzhou Cancer Hospital. A total of 300 formalin-fixedand paraffin-embedded ESCCs and their correspondingnontumorous tissue samples were also kindly provided byLinzhou Cancer Hospital. Clinical data of patients includedin this study are detailed in Supplementary Table S1 and S2.No patients recruited in this study have received any pre-operative treatment. Samples used in this study wereapproved by the individual institutional Committees forEthical Review of Research involving Human Subjects.

Semiquantitative reverse transcription-PCRTotal RNA was extracted from cell lines and frozen ESCC

tissues by the TRIzol reagent (Invitrogen). Reverse tran-scription of total RNA (2 mg) was done using an Advantage

RT for PCR kit (Clontech), and cDNAwas subjected to PCRfor 28 cycles of amplification with the following pair ofprimers: SOX6Fw, 5’-GGCGTCCCCCTACCCTGTCATCCand SOX6Rv, 5’-TGCTGCACACGGCTCCTCAC- TG.GAPDH gene was used as a control.

Tissue microarray and immunohistochemistryTissue microarrays (TMA) containing 300 pairs of pri-

mary ESCC tissue samples and their corresponding non-tumorous tissues were constructed as described previously(17). Standard streptavidin–biotin–peroxidase complexmethod was used for immunohistochemical staining(17). Briefly, TMA section was deparaffinized, blocked with10% normal rabbit serum for 10 minutes, and incubatedwith anti-SOX6 polyclonal antibody (Novous; 1:75 dilu-tion) overnight at 4�C. The TMA sectionwas then incubatedwith biotinylated goat anti-rabbit immunoglobulin at aconcentration of 1:75 at 37�C for 30 minutes. The statusof nuclear expression of SOX6 was assessed by 3 indepen-dent investigatorswithout prior knowledge of clinicopatho-logic data. Positive expression of SOX6 in normal andmalignant ESCC tissues was primarily a nuclear pattern.Because the intensity of staining within each tumor tissuecore wasmostly homogeneous, the intensity of SOX6 stain-ing was semiquantitatively evaluated on the basis of criteriaused by our previous publication (18): strong positive(scored as 2þ), dark brown staining in greater than 50%of normal or malignant esophageal squamous cells com-pletely obscuring nucleus; weak positive(scored as 1þ), anylesser degree of brown staining appreciable in cell nucleus;absent (scored as 0), no appreciable staining in normal ormalignant esophageal squamous cells.

Tumor-suppressive function of SOX6To test tumor-suppressive function of SOX6 gene, it was

PCR amplified, sequence-verified, cloned into pcDNA3.1(þ) vector (Invitrogen), and transfected into ESCC cell lineKYSE30 and KYSE510 cells. Stable SOX6-expressing clones(SOX6-30/SOX6-510) were pooled and the SOX6 cDNAwas resequenced. Blank vector-transfected KYSE30 andKYSE510 cells (Vec-30/Vec-510) were used as control.For foci formation assay, 1 � 103SOX6-30/SOX6510 cellsor Vec-30/ Vec-510 cells were plated in wells of a 6-wellplate. After 7 days culture, surviving colonies (>50 cells percolony) were counted with Giemsa staining. Triplicateindependent experiments were carried out.

Cell growth rates of SOX6-30/SOX6-510 and Vec-30/Vec-510 cells were detected byMTT assay. Cells were seededin 96-well plate at a density of 1 � 103 per well. The cellgrowth rate was detected using cell proliferation MTT kit(Sigma) according to the manufacturer’s instruction. Tri-plicate independent experiments were carried out.

Tumor formation in nude miceThe in vivo tumor-suppressive ability of SOX6 was inves-

tigated by tumor xenograft experiment. About 2 �106SOX6-30 cells or Vec-30 cells were injected subcuta-neously into the right and left hind legs of 4-week-old nude

Translational Interference

Increasing knowledge of genes and their biologicalpathways associated with cancer development and pro-gression has provided opportunities to develop targetedtherapeutics for cancer treatment with higher specificity,more efficiency, and safety. Here, we identify SOX6 as atumor suppressor in esophageal squamous cell carci-noma (ESCC). Loss of SOX6 expression is significantlycorrelated with poor prognosis in ESCC patients. Thetumor-suppressive mechanism of SOX6 is associatedwith its role in G1/S cell-cycle arrest via the upregulationof p53 and p21WAF1/CIP1, along with the downregula-tion of cyclin D1/CDK4, cyclin A, and b-catenin. Thesefindings suggest that SOX6 could be used as a prognosticmarker and a potential therapeutic target in ESCC.

SOX6 in ESCC

www.aacrjournals.org Clin Cancer Res; 17(1) January 1, 2011 47

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 3: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

mice (10 mice per group). Tumor formation in nude micewas monitored over a 4-week period. The tumor volumewas calculated by the formula V ¼ 0.5 � L � W2 (19).

Migration and invasion assaysFor cell migration assay, SOX6-30/SOX6-510 or Vec-

30/Vec-510 cells were cultured in a 6-well plate untilconfluent. The cell layer was wounded using a steriletip. After incubation for 24 or 36 hours, the cells werephotographed under a phase-contrast microscope. Theexperiment was carried out in triplicate. For invasionassay, SOX6-30 cells or Vec-30 cells were starved withserum-free medium for 24 hours before the assay. Cells (5� 104) were suspended in 0.5-mL serum-free mediumand loaded on the upper compartment of invasion cham-ber coated with Matrigel (BD Biosciences). The lowercompartment was filled with complete medium as che-moattractant. After 24 hours, invasive cells were fixed,stained, and counted under a microscope. Triplicateindependent experiments were carried out.

Cell-cycle analysisSOX6-30/SOX6-510 or Vec-30/Vec-510 cells (1 � 106 to

2 � 106) were cultured in RPMI medium containing 10%

fetal bovine serum (FBS). Serum was withdrawn fromculture medium when cells were 70% confluent. After 72hours, 10% FBSwas added in themedium for an additional12 hours. Cells were fixed in 70% ethanol, stained withpropidium iodide, and DNA content was analyzed byCytomics FC (Beckman Coulter).

Western blot analysisWestern blotting was done according to the standard

protocol with antibodies for SOX6 (Novous), p53,p21WAF1/CIP1, cyclin A, cyclin D1, CDK4 (Cell SignalingTechnology), and b-catenin (Santa Cruz Biotechnology).

Statistical analysisStatistical analysis was done with the SPSS standard

version 13.0 (SPSS Inc). The correlation between SOX6expression and clinicopathologic characteristics was ana-lyzed using the chi-square test. Disease-specific survival(DSS) was calculated from the date of diagnosis to thedate of cancer-related death or last follow-up. Survivalcurves were assessed by the Kaplan–Meier method andcompared by the log-rank test. Relative risks of cancer-related death associated with SOX6 expression status andother predictor variables were estimated by univariate

A

B

C

Figure 1.Downregulation of SOX6in ESCC. SOX6 expression wasfrequently downregulated inprimary ESCCs (A) and ESCC celllines (B) detected by RT-PCR. Forprimary ESCCs, expression ofSOX6 in tumor tissues (T) wascompared with their pairednontumorous tissues (N). GAPDHwas used as a loading control. C,representative of SOX6expression in a pair of ESCC (right)and adjacent normal tissue (left)detected by immunostaining withanti-SOX6 antibody (brown). Theslide was counterstained withhematoxylin. Originalmagnification � 200.

Qin et al.

Clin Cancer Res; 17(1) January 1, 2011 Clinical Cancer Research48

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 4: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

analyses. Multivariate survival analysis was done on allparameters that were found to be significant on univariatelevel using the Cox regression model. Differences wereconsidered significant when P value was less than 0.05.

Results

Downregulation of SOX6 is frequently detected inESCCs

The mRNA expression of SOX6 in 9 ESCC cell lines and50 primary ESCC tumors and their paired nontumoroustissues were studied by semiquantitative RT-PCR. Expres-sion of SOX6 was observed in all 50 tested nontumoroustissues. However, downregulation of SOX6 was detectedin 29 of 50 (58%) of primary ESCCs (Fig. 1A). Similarly,downregulation of SOX6 was detected in 5 of 9 (EC109,HKESC1, KYSE30, 510, and 520) ESCC cell lines(Fig. 1B).

Downregulation of SOX6 correlates with poorsurvival outcome in ESCC

To investigate the clinical significance of downregulationof SOX6 in esophageal carcinogenesis, SOX6 expression inprotein level was also studied using ESCC TMA containing300 primary ESCC cases. The clinicopathologic features ofthese cases were summarized in Table 1. Positive staining ofSOX6 was mainly observed at the nucleus (Fig. 1C) and theexpression of SOX6 was classified into absent (scored as 0),weak-positive (scored as 1þ) and strong-positive (scored as2þ) staining. Informative expression of SOX6 was detectedin 219 ESCC cases. Noninformative samples included lost

Table 1. Association between SOX6 expression and clinicopathologic characteristics of patients withESCC (n ¼ 219)

Clinicopathologic characteristics N SOX6 expression, n (%) c2 P

Downregulation Normal

Age, y�60 115 78 (67.8) 37 (32.2) 0.005 1.000>60 104 71 (68.3) 33 (31.7)

SexMale 97 67 (69.1) 30 (30.9) 0.086 0.773Female 122 82 (66.4) 40 (33.6)

Tumor cell differentiationWell 25 13 (52.0) 12 (48.0) 7.416 0.029a

Moderate 141 93 (66.0) 48 (34.0)Poor 53 43 (79.2) 10 (20.8)

Lymph nodes metastasis (N)N0 114 69 (60.5) 45 (39.5) 6.167 0.014a

N1 105 80 (76.2) 25 (23.8)TNM stage

I 5 3 (60.0) 2 (40.0) 17.277 0.000a

II 131 76 (58.0) 55 (42.0)III 83 70(84.3) 13 (15.7)

General classificationMedullary type 119 82 (68.9) 37 (31.1) 1.950 0.597Ulcerative type 61 43 (70.5) 18 (29.5)Sclerotic type 14 10 (71.4) 4 (28.6)Mushroom type 25 14 (56.0) 11 (44.0)

aP < 0.05.

Figure 2. Kaplan–Meier analysis of survival in patients with ESCC. Black,patients with normal SOX6 expression (n ¼ 70, median survival 18months); gray, patients with downregulation of SOX6 (n ¼ 149, mediansurvival 37 months; P < 0.001, log-rank test).

SOX6 in ESCC

www.aacrjournals.org Clin Cancer Res; 17(1) January 1, 2011 49

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 5: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

samples, unrepresentative samples, samples with too fewtumor cells, and samples with inappropriate staining; suchwere not used in data complication. Normal expression ofSOX6 (strong-positive staining) was observed in 205 of 219(93.6%) of normal esophageal epithelial cells. Downregu-lation of SOX6 (absent- and weak-positive staining) wasdetected in 149 of 219 (68.0%) of informative ESCC cases(Table 1).

The correlation of SOX6 expression with various clin-icopathologic features was investigated and the resultshowed that downregulation of SOX6 was significantassociated with poor differentiation (P ¼ 0.029), lymphnode metastasis (P ¼ 0.014), and advanced stage (P ¼0.000, Table 1). Furthermore, log-rank test showed thatESCC patients with SOX6 downregulation (median sur-vival time, 18 months) experienced a shorter DSS thanpatients with normal SOX6 expression (median survivaltime, 37 months; P < 0.001; Fig. 2). By univariableanalysis, downregulation of SOX6 (P < 0.0001), poordifferentiation (P ¼ 0.012), presence of lymph nodemetastasis (P < 0.0001), and advanced stage (P <0.0001) were significant negative prognostic factors forDSS in ESCC patients (Table 2). Nevertheless, multivari-ate analysis showed that downregulation of SOX6 andpresence of lymph node metastasis were 2 independentprognostic predictors for ESCC patients enrolled in thisstudy (P < 0.001, Table 3).

SOX6 has strong tumor-suppressive abilityTo determine if SOX6 has tumor-suppressive ability,

SOX6 gene was stably transfected into KYSE30 andKYSE510 cells (SOX6-30/SOX6-510). Empty vector-trans-

fected KYSE30 and KYSE510 cells (Vec-30/Vec-510) wereused as control. Expression of SOX6 in SOX6-30 and SOX6-510 was confirmed by RT-PCR or Western blot analysis,respectively (Fig. 3A). Tumor-suppressive function of SOX6was studied by foci formation assay, cell growth assay, andtumor xenograft experiment. Foci formation assay showedthat the efficiency of foci formation was significantlyinhibited (P < 0.001) in SOX6-transfectants compared withcontrol cells (Fig. 3B). Cell growth assay also revealed thatthe cell growth rates in SOX6-30 cells were significantlyinhibited by SOX6 (P < 0.01) compared with Vec-30 cells(Fig. 3C). A similar result was shown in SOX6-510 cells(Fig. 3D).

SOX6 inhibits cell migration and invasionAs the TMA result showed that downregulation of

SOX6 was closely associated with ESCC metastasis, theeffects of SOX6 on cell migration and invasion werestudied by wound-healing and cell invasion assays,respectively. Wound-healing assay showed that the ecto-pic expression of SOX6 could dramatically inhibit cellmigration ability in SOX6-transfectants compared withcontrol cells (Fig. 4A). Matrigel invasion assay also foundthat SOX6 could inhibit the invasiveness of ESCC cells, asshowed by a significant decrease in the number ofinvaded cells in SOX6-30 cells compared with Vec-30control (P < 0.001, Fig. 4B).

SOX6 inhibits tumor formation in vivoTo further explore the in vivo tumor-suppressive ability

of SOX6, tumor formation in nude mouse was carried outby injection of SOX6-30 cells (n ¼ 10), whereas Vec-30

Table 2. Univariate Cox regression analysis of factors possibly influencing disease-specific survival inpatients with ESCC

Variables Hazard ratio 95% CI P

SOX6 0.338 0.242–0.471 0.000a

Age, y 1.206 0.912–1.595 0.188Gender 1.052 0.793–1.395 0.726Differentiation 1.349 1.067–1.706 0.012a

pN factor 1.859 1.401–2.467 0.000a

TNM stage 2.148 1.622–2.843 0.000a

aP < 0.05.

Table 3. Multivariate Cox regression analysis for factors possibly influencing disease-specific survival inpatients with ESCC

Variables Hazard ratio 95% CI P

SOX6 0.360 0.257–0.506 0.000a

Differentiation 1.220 0.963–1.548 0.100pN factor 1.733 1.301–2.309 0.000a

aP < 0.05.

Qin et al.

Clin Cancer Res; 17(1) January 1, 2011 Clinical Cancer Research50

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 6: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

cells were used as controls. Within 4 weeks, solid tumorswere readily visible in left hind legs of all 10 mice(injected with Vec-30 cells), but only observed in 4 of10 of mice injected with SOX6-30 cells. In addition, thesize of tumors caused by SOX6-30 cells (tumor volume:385 � 50 mm3) was significantly smaller than tumors(750 � 37 mm3) induced by Vec-30 cells (P < 0.001;

Fig. 4C). These results suggested that SOX6 had a strongtumor-suppressive ability in vivo.

SOX6 arrests cell cycle at G1/S transitionTo elucidate the mechanism underlying growth inhibi-

tion by SOX6, flow-cytometry was used to compare celldistributions in cell cycle between SOX6-transfectants and

Figure 3. Tumor-suppressivefunction of SOX6 in ESCC cells. A,expression of SOX6 in SOX6-transfected ESCC cells (SOX6-30/SOX6-510) was confirmed by RT-PCR and Western blot analysis.Empty vector-transfected ESCCcells (Vec-30/Vec-510) were usedas controls. B, representativeinhibition of foci formation inmonolayer culture by SOX6 (left).Quantitative analyses of focinumbers (right). Columns, mean ofat least 3 independentexperiments; bars, SD. **,P < 0.001 versus controls by usingthe Student's t test. Growthcurves of SOX6-30 cells (C) andSOX6-510 cells (D) werecompared with control cells byMTT assay. Points, mean of atleast 3 independent experiments;bars, SD. **, P < 0.01.

A

B

C D

SOX6 in ESCC

www.aacrjournals.org Clin Cancer Res; 17(1) January 1, 2011 51

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 7: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

control cells. The percentage of SOX6-transfectants in G0/G1 phases and in S-phase were significantly increased anddecreased, respectively, compared with that in control cells,suggesting that SOX6 was able to inhibit DNA synthesisand G1/S phase transition (Fig. 5A and B). To further revealthe potential molecular mechanism of SOX6 in cell-cyclearrest, the effects of SOX6 on several key cell-cycle regula-tors, including p53, p21WAF1/CIP1, cyclin A, cyclin D1,CDK4, and b-catenin were investigated by Western blotanalysis. Increased expression of p53 and p21WAF1/CIP1,downregulation of cyclin D1/CDK4 complex, cyclin A, andb-catenin were detected in SOX6-30 and SOX6-510 cells,compared with those in control cells (Fig. 5C).

Discussion

The known functions of the SOX protein in developmentand disease are both diverse and complex (20–22). In thisstudy, we found that SOX6 was frequently downregulatedin ESCC cell lines and primary ESCC tumors. TMA studyshowed that the downregulation of SOX6 was detected in68% primary ESCCs, which was significantly associatedwith advanced stage, increased lymph nodemetastasis, andpoor survival of patients with ESCC. Multivariate analysisshowed that downregulation of SOX6 could be used as anindependent prognostic predictors for ESCC patients. Incontrast, it has been reported that high SOX6 expressionmay be a potential diagnostic marker for some types ofbrain tumors (23, 24). However, the biological role of high

SOX6 expression in brain tumor is still unknown. Thecontradictory results indicated that SOX6 expression mightthus be tissue specific. In this study, the mechanismsunderlying silencing of SOX6 in ESCC cells have also beeninvestigated. However, neither methylation nor mutationof SOX6 was detected (data not shown), suggesting thatother mechanisms such as, micro-RNA regulation or his-tone modification might be involved in the inactivation ofSOX6.

The tumor-suppressive function of SOX6 showed byboth in vitro and in vivo assays further supports its roleas a TSG in the development and progression of ESCC. Theresults showed that SOX6 could suppress cell growth,decrease foci formation and cell motility, and inhibittumor formation in nude mice. Further study revealed thatSOX6 was able to inhibit G1/S-phase transition throughthe upregulation of p53 and p21WAF1/CIP1, and downregu-lation of cyclin D1/CDK4 complex and cyclin A in ESCCcancer cells. G1/S phase transition is known to be a majorcheckpoint for cell-cycle progression. The cyclin-dependentkinase inhibitor (CKI), p21WAF1/CIP1, serves as a criticalnegative mediator during this transition through CDKinhibition and regulate the activity of cyclin D-CDK com-plex, which are essential for S phase entry (25–27). Becausean inhibition of CKI activity is one of the factors causinguncontrolled tumor cell growth (27), one possible strategyto control cancer cell proliferation is to induce CKI expres-sion, which would lead to G1 arrest and inhibit tumorgrowth. Although the tumor suppressor p53 itself is not a

A

B C

Figure 4. SOX6 inhibitsmetastasis of ESCC cells. A, theeffect of SOX6 on cell migrationwas determined by wound-healing assay. During a period ofincubation, the spreading speedof SOX6-expressing cells alongthe wound edge was slower thanthat in control cells. B,representative images showed theSOX6-30 cells and Vec-30 cellsthat invaded through the matrigel.Number of invaded tumor cellswas quantified in the right panel.Columns, mean of triplicateexperiments. **, P < 0.001. C,tumor growth curves of SOX6-30cells in nude mice were comparedwith Vec-30 cells by tumorxenograft experiment. Theaverage tumor volume of SOX6-30 cells versus Vec-30 cells wasexpressed as mean � SD in 10inoculated sites for each group. **,P < 0.001, Student's t test.

Qin et al.

Clin Cancer Res; 17(1) January 1, 2011 Clinical Cancer Research52

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 8: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

CKI, it has been implicated in various cellular processes(28, 29), including induction of G1 arrest by transactivat-ing its downstream target gene, p21WAF1/CIP1 (30). Takentogether, it is reasonable to speculate that SOX6 is able toinduce p53 expression, in turn, activate the expression ofp21WAF1/CIP1, resulting in its inhibition of cyclin D1/CDK4activity in human esophageal cancer cells. However, theinteraction between the transcription regulator SOX6 andthe tumor suppressor p53 need to be further investigated.In addition to the suppression of cyclin D1/CDK4 com-

plex, our data also showed that ectopic expression of SOX6resulted in the inhibition of the late G1-phase regulatorcyclin A. Expression of cyclin A has been strongly impli-cated in the control of cell proliferation and prognoses inhuman cancers (31–33). These findings are complemen-tary to ours and suggest that downregulation of SOX6would favor tumor progression.

Recent study by Iguchi et al. has shown that SOX6suppresses cyclin D1 promoter activity by interacting withb-catenin, and its downregulation induces pancreatic b-cellproliferation (9). Because the Wnt/b-catenin signalingpathway regulates a variety of cellular processes, includingcell proliferation, differentiation, development, and apop-tosis (34, 35), the suppression of b-catenin signaling bySOX6 may be of biological significance. Previous studieshave also shown that some other Sox proteins, includingSOX1, SOX3, SOX9, and SOX17, could bind b-catenin andinhibit b-catenin signaling (36–39). In this study, we didfound that SOX6 could downregulate b-catenin expressionin ESCC cells, suggesting that the tumor-suppressive func-tion of SOX6 might also be associated with the inhibitionof b-catenin signal transduction pathway. Interestingly, theoncogenic role of b-catenin in enhancing proliferationcould also be inhibited by p53 in response to growth arrest

Figure 5. SOX6 induces G1–Scell-cycle arrest. Representativeand summary of DNA contentdetected by flow cytometryshowed that the percentage ofcells in S phase was much lower inSOX6-30 cells (A) and SOX6-510cells (B) than that in control cells.Values are the mean � SD of 3independent experiments. **, P <0.05. C, protein expression of p21,p53, b-catenin, cyclin A, cyclin D1,and CDK4 were determined byWestern blot analysis. GAPDHwas used as an internal control.

A

B

C

SOX6 in ESCC

www.aacrjournals.org Clin Cancer Res; 17(1) January 1, 2011 53

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 9: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

(40–42). Thus, it will be important to know whether SOX6could exert the tumor suppressor effect in esophagealcancer through interaction with p53 directly.

In summary, we provided evidences that SOX6 is a tumorsuppressor in ESCC, based on the following facts: (i) SOX6is frequently downregulated in ESCC tissues; (ii) down-regulation of SOX6 was significantly associated with poorprognosis; (iii) the introduction of SOX6 into ESCC cellscould inhibit cell proliferation and tumor formation innude mice; (iv) the ectopic expression of SOX6 is able toarrest cell cycle at G1/S checkpoint by upregulating p53 andp21, downregulating cyclin D1/CDK4, cyclin A and b-cate-nin. A better understanding of the tumor-suppressive roleof SOX6 will significantly improve our knowledge in thedevelopment of ESCC, and may lead to a more effectivemanagement of ESCC with the inactivation of SOX6.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Grant Support

Doctoral Program of Henan Province (200703042), National Natural ScienceFoundation of China (30772475, 30700820, and 30971606), Sun Yat-Sen Uni-versity "Hundred Talents Program" (85000-3171311), Major State Basic ResearchProgram of China (2006CB910104), Research Fund for the Doctoral Program ofHigher Education of China (20070558272), and Research Grant Council CentralAllocation (HKUST 2/06C).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received May 5, 2010; revised October 6, 2010; accepted October 22,2010; published OnlineFirst November 17, 2010.

References1. Shimada H, Nabeya Y, Ochiai T, Matsubara H, Shiratori T, Gunji Y,

et al. Prediction of survival with squamous cell carcinoma antigen inpatients with resectable esophageal squamous cell carcinoma. Sur-gery 2003;133:486–94.

2. Jemal A, Siegel R, Ward E, Murray T, Xu J, ThunMJ. Cancer statistics,2007. CA Cancer J Clin 2007;57:43–66.

3. Metzger R, Schneider PM, Warnecke-Eberz U, Brabender J,Holscher AH. Molecular biology of esophageal cancer. Onkologie2004;27:200–6.

4. He LR, Liu MZ, Li BK, Rao HL, Liao YJ, Zhang LJ, et al. Overexpres-sion of AIB1 predicts resistance to chemoradiotherapy and poorprognosis in patients with primary esophageal squamous cell carci-noma. Cancer Sci. 2009;100:1591–6.

5. Yuen HF, Chan YP, Law S, Srivastava G, El-Tanani M, Mak TW, et al.DJ-1 could predict worse prognosis in esophageal squamous cellcarcinoma. Cancer Epidemiol Biomarkers Prev 2008;17:3593–602.

6. Connor F, Wright E, Denny P, Koopman P, Ashworth A. The Sry-related HMG box-containing gene Sox6 is expressed in the adulttestis and developing nervous system of the mouse. Nucleic AcidsRes 1995;23:3365–72.

7. Lefebvre V, Li P, de Crombrugghe B. A new long form of Sox5 (L-Sox5), Sox6 and Sox9 are coexpressed in chondrogenesis andcooperatively activate the type II collagen gene. EMBO J1998;17:5718–33.

8. Hamada-Kanazawa M, Ishikawa K, Ogawa D. Suppression of Sox6 inP19 cells leads to failure of neuronal differentiation by retinoic acid andinduces retinoic acid-dependent apoptosis. FEBS Lett 2004;577:60–6.

9. Iguchi H, Urashima Y, Inagaki Y, Okamura M, Tanaka T, Uchida A,et al. SOX6 suppresses cyclin D1 promoter activity by interacting withbeta-catenin and histone deacetylase 1, and its down-regulationinduces pancreatic beta-cell proliferation. J Biol Chem2007;282:19052–61.

10. Otsubo T, Akiyama Y, Yanagihara K, Yuasa Y. SOX2 is frequentlydownregulated in gastric cancers and inhibits cell growth through cell-cycle arrest and apoptosis. Br J Cancer 2008;98:824–31.

11. AaboeM, Birkenkamp-Demtroder K,Wiuf C, Sorensen FB, Thykjaer T,Sauter G, et al. SOX4 expression in bladder carcinoma: clinicalaspects and in vitro functional characterization. Cancer Res2006;66:3434–42.

12. Afonja O, Raaka BM, Huang A, Das S, Zhao X, Helmer E, et al. RARagonists stimulate SOX9 gene expression in breast cancer cell lines:evidence for a role in retinoid-mediated growth inhibition. Oncogene2002;21:7850–60.

13. Drivdahl R, Haugk KH, Sprenger CC, Nelson PS, Tennant MK, Ply-mate SR. Suppression of growth and tumorigenicity in the prostate

tumor cell line M12 by overexpression of the transcription factorSOX9. Oncogene 2004;23:4584–93.

14. Aleman A, Adrien L, Lopez-Serra L, Cordon-Cardo C, Esteller M,Belbin TJ, et al. Identification of DNA hypermethylation of SOX9 inassociation with bladder cancer progression using CpG microarrays.Br J Cancer 2008;98:466–73.

15. Passeron T, Valencia JC, Namiki T, Vieira WD, Passeron H, MiyamuraY, et al.Upregulation of SOX9 inhibits the growth of human andmousemelanomas and restores their sensitivity to retinoic acid. J Clin Invest2009;119:954–63.

16. Shimada Y, Imamura M, Wagata T, Yamaguchi N, Tobe T. Character-ization of 21 newly established esophageal cancer cell lines. Cancer1992;69:277–84.

17. Xie D, Sham JS, Zeng WF, Lin HL, Che LH, Wu HX, et al.Heterogeneous expression and association of beta-catenin,p16 and c-myc in multistage colorectal tumorigenesis and pro-gression detected by tissue microarray. Int J Cancer 2003;107:896–902.

18. Fu L, Qin YR, Xie D, ChowHY, Ngai SM, Kwong DL, et al. Identificationof alpha-actinin 4 and 67 kDa laminin receptor as stage-specificmarkers in esophageal cancer via proteomic approaches. Cancer2007;110:2672–81.

19. Cao ZA, Daniel D, Hanahan D. Sub-lethal radiation enhances anti-tumor immunotherapy in a transgenic mouse model of pancreaticcancer. BMC Cancer 2002;2:11.

20. Wegner M. From head to toes: the multiple facets of Sox proteins.Nucleic Acids Res 1999;27:1409–20.

21. Schepers GE, Teasdale RD, Koopman P. Twenty pairs of sox: extent,homology, and nomenclature of the mouse and human sox transcrip-tion factor gene families. Dev Cell 2002;3:167–70.

22. Chew LJ, Gallo V. The Yin and Yang of Sox proteins: activation andrepression in development and disease. J Neurosci Res2009;87:3277–87.

23. Ueda R, Yoshida K, Kawakami Y, Kawase T, Toda M. Immunohisto-chemical analysis of SOX6 expression in human brain tumors. BrainTumor Pathol 2004;21:117–20.

24. Ueda R, Yoshida K, Kawakami Y, Kawase T, Toda M. Expression of atranscriptional factor, SOX6, in human gliomas. Brain Tumor Pathol2004;21:35–8.

25. Carnero A, Hannon GJ. The INK4 family of CDK inhibitors. Curr TopMicrobiol Immunol 1998;227:43–55.

26. Parry D, Mahony D, Wills K, Lees E. Cyclin D-CDK subunit arrange-ment is dependent on the availability of competing INK4 and p21 classinhibitors. Mol Cell Biol 1999;19:1775–83.

27. Malumbres M, Barbacid M. To cycle or not to cycle: a critical decisionin cancer. Nat Rev Cancer 2001;1:222–31.

Qin et al.

Clin Cancer Res; 17(1) January 1, 2011 Clinical Cancer Research54

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 10: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

28. Kohn KW. Molecular interaction map of the mammalian cell cyclecontrol and DNA repair systems. Mol Biol Cell 1999;10:2703–34.

29. el-Deiry WS, Harper JW, O’Connor PM, Velculescu VE, Canman CE,Jackman J, et al. WAF1/CIP1 is induced in p53-mediated G1 arrestand apoptosis. Cancer Res 1994;54:1169–74.

30. Waldman T, Kinzler KW, Vogelstein B. p21 is necessary for thep53-mediated G1 arrest in human cancer cells. Cancer Res1995;55:5187–90.

31. Aaltomaa S, Eskelinen M, Lipponen P. Expression of cyclin A and Dproteins in prostate cancer and their relation to clinopathologicalvariables and patient survival. Prostate 1999;38:175–82.

32. Mrena J, Wiksten JP, Kokkola A, Nordling S, Haglund C, Ristimaki A.Prognostic significance of cyclin A in gastric cancer. Int J Cancer2006;119:1897–901.

33. Ahlin C, Zhou W, Holmqvist M, Holmberg L, Nilsson C, Jirstrom K, etal. Cyclin A is a proliferative marker with good prognostic value innode-negative breast cancer. Cancer Epidemiol Biomarkers Prev2009;18:2501–6.

34. Huelsken J, Birchmeier W. New aspects of Wnt signaling pathways inhigher vertebrates. Curr Opin Genet Dev 2001;11: 547–53.

35. Logan CY, Nusse R. The Wnt signaling pathway in development anddisease. Annu Rev Cell Dev Biol 2004;20:781–810.

36. Zhang C, Basta T, Jensen ED, Klymkowsky MW. The beta-catenin/VegT-regulated early zygotic gene Xnr5 is a direct target of SOX3regulation. Development 2003;130:5609–24.

37. Kan L, Israsena N, Zhang Z, Holmberg L, Nilsson C, Jirstrom K, et al.Sox1 acts through multiple independent pathways to promote neu-rogenesis. Dev Biol 2004;269:580–94.

38. Akiyama H, Lyons JP, Mori-Akiyama Y, Yang X, Zhang R, Zhang Z,et al. Interactions between Sox9 and beta-catenin control chondro-cyte differentiation. Genes Dev 2004;18:1072–87.

39. Sinner D, Kordich JJ, Spence JR, Opoka R, Rankin S, Lin SC, et al.Sox17 and Sox4 differentially regulate beta-catenin/T-Cell factoractivity and proliferation of colon carcinoma cells. Mol cell Biol2007;27:7802–15.

40. Orford K, Orford CC, Byers SW. Exogenous expression of the b-cate-nin regulates contact inhibition, anchorage-independent growth,anoikis and radiation-induced cell cycle arrest. J Cell Biol1999;146:855–67

41. Polakis P. More than one way to skin a catenin. Cell 2001;105:563–6.42. Sadot E, Geiger B, Oren M, Ben-Ze’ev A. Down-regulation of beta-

catenin by activated p53. Mol Cell Biol 2001;21:6768–81.

SOX6 in ESCC

www.aacrjournals.org Clin Cancer Res; 17(1) January 1, 2011 55

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155

Page 11: Clinical Human Cancer Biology Cancer Research€¦ · Human Cancer Biology Characterization of Tumor-Suppressive Function of SOX6 in Human Esophageal Squamous Cell Carcinoma Yan-Ru

2011;17:46-55. Published OnlineFirst November 17, 2010.Clin Cancer Res   Yan-Ru Qin, Hong Tang, Fajun Xie, et al.   Esophageal Squamous Cell Carcinoma

in HumanSOX6Characterization of Tumor-Suppressive Function of

  Updated version

  10.1158/1078-0432.CCR-10-1155doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2011/01/03/1078-0432.CCR-10-1155.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/1/46.full#ref-list-1

This article cites 42 articles, 14 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/1/46.full#related-urls

This article has been cited by 7 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/17/1/46To request permission to re-use all or part of this article, use this link

on June 21, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 17, 2010; DOI: 10.1158/1078-0432.CCR-10-1155