Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers

10
Relation of a Hypoxia Metagene Derived from Head and Neck Cancer to Prognosis of Multiple Cancers Stuart C. Winter, 1 Francesca M. Buffa, 1,2 Priyamal Silva, 5,6 Crispin Miller, 7 Helen R. Valentine, 5 Helen Turley, 1 Ketan A. Shah, 3 Graham J. Cox, 4 Rogan J. Corbridge, 4 Jarrod J. Homer, 6 Brian Musgrove, 5 Nick Slevin, 5 Philip Sloan, 8 Pat Price, 5 Catharine M.L. West, 5 and Adrian L. Harris 1 1 Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital; 2 Gray Cancer Institute, University of Oxford; 3 Department of Cellular Pathology, John Radcliffe Hospital; 4 Department of Otorhinolaryngology, Head and Neck Surgery, Radcliffe Infirmary, Oxford, United Kingdom; 5 Academic Radiation Oncology, The University of Manchester and Department of Clinical Oncology, Christie Hospital NHS Trust; 6 Department of Head and Neck Surgery, Manchester Royal Infirmary; 7 Paterson Institute for Cancer Research; and 8 Department of Pathology, Dental Hospital, Manchester, United Kingdom Abstract Affymetrix U133plus2 GeneChips were used to profile 59 head and neck squamous cell cancers. A hypoxia metagene was obtained by analysis of genes whose in vivo expression clustered with the expression of 10 well-known hypoxia- regulated genes (e.g., CA9, GLUT1 , and VEGF ). To minimize random aggregation, strongly correlated up-regulated genes appearing in >50% of clusters defined a signature comprising 99 genes, of which 27% were previously known to be hypoxia associated. The median RNA expression of the 99 genes in the signature was an independent prognostic factor for recur- rence-free survival in a publicly available head and neck cancer data set, outdoing the original intrinsic classifier. In a published breast cancer series, the hypoxia signature was a significant prognostic factor for overall survival independent of clinicopathologic risk factors and a trained profile. The work highlights the validity and potential of using data from analysis of in vitro stress pathways for deriving a biological metagene/gene signature in vivo . [Cancer Res 2007;67(7):3441–9] Introduction Head and neck cancer represents the fifth most common cancer in men and the eighth in women worldwide, with f600,000 new cases each year (1). Surgery, radiotherapy, and chemotherapy play a role in the management of the disease, and 5-year survival rates for patients with advanced cancers are f50% (2, 3). Many factors contribute to this poor prognosis, including late presentation of disease, nodal metastases, and the failure of advanced cancers to respond to conventional treatments (4). Hypoxia, a key factor defining tumor behavior, is a characteristic of head and neck squamous cell carcinomas (HNSCC; refs. 5, 6). Although hypoxia results in cell death if it is severe or prolonged, cancer cells can adapt to this hostile environment, allowing them to survive and proliferate. It is, in part, this ability to adapt to a hostile environment that defines their malignant potential and characterizes a more aggressive phenotype (7). Up to 1.5% of the human genome is estimated to be transcriptionally responsive to hypoxia (8). One way cells respond to reduced oxygen levels is through the hypoxia-inducible factor (HIF)-1 pathway. The HIF DNA-binding complex is a heterodimer composed of a and h subunits. In normoxia, HIF-1a undergoes rapid hydroxylation and degradation. In hypoxia, hydroxylation is prevented, stabilized HIF-1a binds to HIF-1h, and the heterodimer binds to hypoxia response elements in genes involved in the regulation of cellular processes, such as cell proliferation, angiogenesis, cell metabolism, apoptosis, and migration (7). Tumor hypoxia is an independent adverse prognostic factor in many tumors, including HNSCC (6, 7). Evidence showing that hypoxia is important in tumor progression (9) and prognosis (7) has spurred research into developing therapies that target hypoxic cells. Therapeutic strategies include modification of the hypoxic environment or targeting components of the HIF-1 signaling pathway (10, 11). Although these approaches have shown some promising results, it remains difficult to identify hypoxic tumors and those patients most likely to benefit from hypoxia modification therapy. Currently, the level of tumor oxygenation is assessed by direct or indirect methods. The main direct approach is to measure intratumoral pO 2 with polarographic electrodes (6). Indirect techniques being explored include measuring the immunohisto- chemical expression of hypoxia-regulated proteins, such as carbonic anhydrase 9 (CA9) and HIF-1a (12, 13). The latter approach is attractive for routine clinical use. It is limited, however, by their variability of expression within a tumor, the lack of hypoxia specificity of individual proteins, and the complex interrelation- ships between molecular pathways in different types of tumors. Establishing tumor type–specific or tumor type–independent hypoxia gene signatures would be a major advance in this area of research. Therefore, we assessed the mRNA profile of HNSCC samples and defined an in vivo hypoxia metagene by clustering around the RNA expression of a set of known hypoxia-regulated genes. The validity of the method was confirmed by showing that the metagene contained many previously described in vitro –derived hypoxia response genes and was prognostic for treatment outcome in independent data sets. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). S.C. Winter and F.M. Buffa are joint first authors. C.M.L. West and A.L. Harris are joint senior authors. Supplemental and microarray data will be made available at: http:// bioinformatics.picr.man.ac.uk/experiments. Requests for reprints: Adrian L. Harris, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom. Phone: 44-1865-222-457; Fax: 44- 1865-222-431; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-3322 www.aacrjournals.org 3441 Cancer Res 2007; 67: (7). April 1, 2007 Research Article Research. on November 25, 2015. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers

Relation of a Hypoxia Metagene Derived from Head and

Neck Cancer to Prognosis of Multiple Cancers

Stuart C. Winter,1Francesca M. Buffa,

1,2Priyamal Silva,

5,6Crispin Miller,

7Helen R. Valentine,

5

Helen Turley,1Ketan A. Shah,

3Graham J. Cox,

4Rogan J. Corbridge,

4

Jarrod J. Homer,6Brian Musgrove,

5Nick Slevin,

5Philip Sloan,

8

Pat Price,5Catharine M.L. West,

5and Adrian L. Harris

1

1Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John RadcliffeHospital; 2Gray Cancer Institute, University of Oxford; 3Department of Cellular Pathology, John Radcliffe Hospital; 4Department ofOtorhinolaryngology, Head and Neck Surgery, Radcliffe Infirmary, Oxford, United Kingdom; 5Academic Radiation Oncology,The University of Manchester and Department of Clinical Oncology, Christie Hospital NHS Trust; 6Department of Head andNeck Surgery, Manchester Royal Infirmary; 7Paterson Institute for Cancer Research; and 8Department of Pathology,Dental Hospital, Manchester, United Kingdom

Abstract

Affymetrix U133plus2 GeneChips were used to profile 59 headand neck squamous cell cancers. A hypoxia metagene wasobtained by analysis of genes whose in vivo expressionclustered with the expression of 10 well-known hypoxia-regulated genes (e.g., CA9, GLUT1, and VEGF). To minimizerandom aggregation, strongly correlated up-regulated genesappearing in >50% of clusters defined a signature comprising99 genes, of which 27% were previously known to be hypoxiaassociated. The median RNA expression of the 99 genes in thesignature was an independent prognostic factor for recur-rence-free survival in a publicly available head and neckcancer data set, outdoing the original intrinsic classifier. In apublished breast cancer series, the hypoxia signature was asignificant prognostic factor for overall survival independentof clinicopathologic risk factors and a trained profile. Thework highlights the validity and potential of using data fromanalysis of in vitro stress pathways for deriving a biologicalmetagene/gene signature in vivo. [Cancer Res 2007;67(7):3441–9]

Introduction

Head and neck cancer represents the fifth most common cancerin men and the eighth in women worldwide, with f600,000 newcases each year (1). Surgery, radiotherapy, and chemotherapy play arole in the management of the disease, and 5-year survival rates forpatients with advanced cancers are f50% (2, 3). Many factorscontribute to this poor prognosis, including late presentation ofdisease, nodal metastases, and the failure of advanced cancers torespond to conventional treatments (4).Hypoxia, a key factor defining tumor behavior, is a characteristic

of head and neck squamous cell carcinomas (HNSCC; refs. 5, 6).Although hypoxia results in cell death if it is severe or prolonged,cancer cells can adapt to this hostile environment, allowing them

to survive and proliferate. It is, in part, this ability to adapt to ahostile environment that defines their malignant potential andcharacterizes a more aggressive phenotype (7). Up to 1.5% of thehuman genome is estimated to be transcriptionally responsive tohypoxia (8).One way cells respond to reduced oxygen levels is through the

hypoxia-inducible factor (HIF)-1 pathway. The HIF DNA-bindingcomplex is a heterodimer composed of a and h subunits. Innormoxia, HIF-1a undergoes rapid hydroxylation and degradation.In hypoxia, hydroxylation is prevented, stabilized HIF-1a binds toHIF-1h, and the heterodimer binds to hypoxia response elementsin genes involved in the regulation of cellular processes, such ascell proliferation, angiogenesis, cell metabolism, apoptosis, andmigration (7).Tumor hypoxia is an independent adverse prognostic factor in

many tumors, including HNSCC (6, 7). Evidence showing thathypoxia is important in tumor progression (9) and prognosis (7)has spurred research into developing therapies that target hypoxiccells. Therapeutic strategies include modification of the hypoxicenvironment or targeting components of the HIF-1 signalingpathway (10, 11). Although these approaches have shown somepromising results, it remains difficult to identify hypoxic tumorsand those patients most likely to benefit from hypoxia modificationtherapy.Currently, the level of tumor oxygenation is assessed by direct or

indirect methods. The main direct approach is to measureintratumoral pO2 with polarographic electrodes (6). Indirecttechniques being explored include measuring the immunohisto-chemical expression of hypoxia-regulated proteins, such ascarbonic anhydrase 9 (CA9) and HIF-1a (12, 13). The latterapproach is attractive for routine clinical use. It is limited, however,by their variability of expression within a tumor, the lack of hypoxiaspecificity of individual proteins, and the complex interrelation-ships between molecular pathways in different types of tumors.Establishing tumor type–specific or tumor type–independenthypoxia gene signatures would be a major advance in this areaof research.Therefore, we assessed the mRNA profile of HNSCC samples and

defined an in vivo hypoxia metagene by clustering around the RNAexpression of a set of known hypoxia-regulated genes. The validityof the method was confirmed by showing that the metagenecontained many previously described in vitro–derived hypoxiaresponse genes and was prognostic for treatment outcome inindependent data sets.

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).S.C. Winter and F.M. Buffa are joint first authors. C.M.L. West and A.L. Harris are

joint senior authors.Supplemental and microarray data will be made available at: http://

bioinformatics.picr.man.ac.uk/experiments.Requests for reprints: Adrian L. Harris, Cancer Research UK Molecular Oncology

Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, JohnRadcliffe Hospital, Oxford OX3 9DS, United Kingdom. Phone: 44-1865-222-457; Fax: 44-1865-222-431; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-3322

www.aacrjournals.org 3441 Cancer Res 2007; 67: (7). April 1, 2007

Research Article

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Materials and Methods

Sample procurement and RNA extraction. Oxford and South Man-chester Ethics Committees approved the work, and all patients gaveinformed consent. The study comprised patients with histologically proven,previously untreated HNSCC who were undergoing surgical resection withcurative intent. Tumor samples taken at operation were placed in RNAlater(Applied Biosystems, Warrington, United Kingdom) for up to 12 h beforecryopreservation in liquid nitrogen. Where possible, biopsies of healthymucosa were taken from the contralateral side and used as ‘‘normal’’samples. Subsequently, the samples were divided and half paraffinembedded for histologic analysis. RNA was extracted from the remainingtissue using Tri Reagent (Sigma-Aldrich, Poole, United Kingdom) andwashed using the Qiagen spin column (Qiagen, Crawley, United Kingdom).RNA quality and quantity were confirmed using the NanoDrop ND-1000spectrophotometer and the Agilent 2100 Bioanalyzer (Agilent Technologies,Santa Clara, CA).

Immunohistochemistry. Immunohistochemistry for CA9 and HIF-1awas done on sections from formalin-fixed, paraffin-embedded tumor biopsies(one section per protein). CA9 immunohistochemistry has been describedpreviously and involved a mouse monoclonal anti-human antibody (M75)raised to the external domain of CA9, a gift from Profs. S. Pastorekova andJ. Pastorek (14). The primary HIF-1a antibody was ESEE122, a mousemonoclonal IgG1 antibody to human HIF-1a (University of Oxford, Oxford,United Kingdom). Tumor membrane CA9 expression was estimated over thewhole section as a percentage. Tumor nuclear HIF-1a expression wasassessed in 10 separate fields under intermediate (�200) magnification. Thepercentage of tumor cells with nuclear reactivity in each field was recorded asa continuous variable, and the overall score for each tumor was the meanpercentage score across the 10 fields.

Microarray data. Affymetrix U133plus2 GeneChips were used. Biotiny-lated target RNA was prepared with minor modifications from the

manufacturer’s recommendations. A description of procedures is available

online.9 Target RNA generated from each sample was then processed as per

the manufacturer’s recommendation using an Affymetrix GeneChipInstrument System.10 Briefly, spike controls were added to 10 Ag of

fragmented cRNA before overnight hybridization to HGU133plus2 arrays.

Arrays were then washed and stained with streptavidin-phycoerythrin

before imaging on an Affymetrix GeneChip (3000) scanner.11 Followingscanning, data files were processed using Affymetrix GeneChip Operating

Software version 1.1.1.052. Subsequent data processing was done using affy

(15), simpleaffy (16), and GCRMA (17) packages from BioConductor and R.mRNA was estimated using GCRMA, and log2 values were used.

After scanning, array images were assessed visually to confirm scanner

alignment and the absence of significant bubbles or scratches. The 3¶/5¶ratios for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and h-actinfell within acceptable limits (0.9–1.45; 1.14–2.87), and BioB spike controls

were present on all arrays, with BioC, BioD, and CreX also present in

increasing intensity. When scaled to a target intensity of 100 (using the

MAS5 algorithm, as implemented in simpleaffy), scaling factors for all arrayswere within acceptable limits, as were background, Q values, and mean

intensities. Quality control data are provided as Supplementary Material.

The hypoxia metagene. A flow chart summarizing the method used toderive the hypoxia metagene is in the Supplementary Material. The in vivoexpression of genes that correlated strongly with 10 hypoxia-regulated

genes was investigated. Genes representing a wide range of known hypoxia-

regulated pathways were chosen, including those involved in angiogenesis(VEGF and ADM), glucose transport and glycolysis (GLUT1, PDK-1 , and

ENO1 ), linking mitochondrial metabolism and glycolysis (HK2), pH

regulation (CA9), reuse of ATP breakdown products (AK3), and inhibition

of cell cycle progression (CCNG2). PFKB3 was also used, which regulates theoverall rate of glycolysis and is the most active isozyme of the four that

regulate the concentration of fructose-2,6-bisphosphate, which activates the

rate-limiting enzyme 6-phosphofructo-1-kinase. Genes selected have been

widely investigated justifying their classification as hypoxia-responsivegenes, particularly in terms of promoter analysis, and have been frequently

reported in gene array analysis of hypoxia-induced cell lines. Pearson

correlation was used within the framework of significance analysis of

microarrays (SAM; refs. 17–19). To account for multiple testing, the localfalse discovery rate (FDR) was calculated for each probe set using SAM 2.21

within BioConductor. For each gene, a local FDR of <0.05 was used as cutoff

to determine cluster membership. This tells us which probe sets are

associated with the hypoxia proband genes in HNSCC in vivo . The probesets chosen comprised up-regulated and down-regulated genes; the up-

regulated genes made up the hypoxia metagene. However, the presence of a

probe set in a single cluster does not guarantee the involvement of its targetgene in hypoxia regulation in vivo . Some probe sets might have binding

artifacts or defective probes; the in vitro and in vivo behavior of the genes

could differ or probe sets might be aggregated from genes unrelated to

hypoxia pathways. To minimize random aggregation, the number of clustersin which a probe set appeared was calculated. Probe sets appearing in >50%

of the clusters were chosen. This conservative cutoff was selected after a

Monte-Carlo simulation was done computing cluster aggregation around

randomly selected sets of genes; this simulation showed that the probabilityof having an overlap >50% between 10 randomly selected genes was

P < 0.001.

Calculation of hypoxia scores. The pattern of expression of the up-regulated and down-regulated genes was investigated in the 59 HNSCC

using both a hierarchical clustering algorithm with Bayesian optimization

of the number of clusters and a continuous hypoxia score (HS), which

ranked patients according to expression of genes in the hypoxia metagene.The clustering method has been described previously (20) but used a two-

way approach by clustering on the genes first and then using the median

centroids of the gene clusters to cluster on the samples. HSs were calculated

from the distribution of the level of RNA expression of the up-regulated anddown-regulated genes (HS-up and HS-down) for each patient. Median

values were chosen to avoid effects of outliers and nonnormality of the

distribution. Furthermore, to make HS-up and HS-down general andtransportable to any data set, the fractional rank was computed, where

patients were ranked based on their score, and rank was normalized

between 0 and 1.

Statistical analyses. Two published data sets were analyzed: series of 60HNSCC (21) and 295 breast cancers (22–24). Details on the treatment and

follow-up are in the cited publications. Briefly, HNSCC patients underwent a

variety of treatments, including various combinations of surgery, radio-

therapy, and/or chemotherapy, and had a median follow-up of 16 months.Breast cancer patients underwent mastectomy or breast-conserving surgery

followed by radiotherapy if indicated, and the median duration of follow-up

was 7.8 years. HSs were calculated from the publicly available microarray

data set as described above. Univariate and multivariate Cox survivalanalyses were carried out. HSs were introduced together with relevant

clinical variables, and stepwise backward likelihood was used to select

covariates that correlated with outcome. A flow chart in the SupplementaryMaterial summarizes the approach used to analyze the published data sets.

Results

Samples and RNA extraction. Sixty-eight patients with primaryHNSCC were recruited, and samples from 9 were found to contain<10% tumor cells. Patient demographics for the remaining 59patients are presented in Table 1. Normal tissue samples wereobtained from 11 patients. All RNA was of high quality and met themanufacturer’s quality control criteria.Evaluation of HIF-1A and CA9 protein expression to obtain

a hypoxia metagene. There was a significant correlation betweenHIF-1a and CA9 protein expression in the 59 tumors (Spearman

9 http://bioinf.picr.man.ac.uk/mbcf/downloads/GeneChip_Target_Prep_Protocol-CR-UK_v4.pdf

10 http://www.affymetrix.com/support/technical/manual/expression_manual.affx11 http://bioinf.picr.man.ac.uk/mbcf/downloads/GeneChip_Hyb_Wash_Scan_

Protocol-CR-UK_v4.pdf

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3442 www.aacrjournals.org

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

q = 0.53; P < 0.001; Supplementary Fig. S1). Supervised clusteringon the percentage expression of the two proteins showed that CA9mRNA correlated strongest with CA9 protein expression (Spear-man q = 0.60), but the overall FDR was high for both lists and aheterogeneous pattern of gene expression was produced, noobvious groupings and only 1% overlap between the top 50 genesin each list. This was true both when using Pearson and Spearmancorrelation within SAM. Comparison of the genes with a list of 245published hypoxia-regulated genes taken from relevant reviews(Supplementary Table S1; refs. 7, 8, 11, 25, 26) showed that none ofthe top 20 HIF-1a–associated genes appeared in the literature listand only 3 of the top 20 CA9-associated genes. Therefore, clusteringon percentage of protein expression of a hypoxia-regulated genewas unreliable for deriving a hypoxia metagene.Generation of the hypoxia metagene by clustering HNSCC

around the RNA expression of known in vitro hypoxia-regulated genes. Clustering around the CA9 RNA expressionidentified several genes that correlated strongly with CA9 and wereknown to be hypoxia associated (10 of the top 20 were in theliterature list). This suggested that the approach might identifyother hypoxia-related genes in vivo . Furthermore, the cohort ofpatients were divided broadly into two groups (SupplementaryFig. S2), with one group showing high expression of some knownhypoxia-regulated genes.A single gene would not reflect the molecular diversity of the

response of a cell to hypoxia and was considered a high-riskapproach due to possible binding artifacts, differences betweenin vitro and in vivo expression for single probes or geneticvariability in response. Therefore, 10 widely investigated hypoxia-regulated genes were selected for the clustering process (VEGF,ADM, GLUT1, PDK-1, ENO1, HK2, PFKB3, CA9, AK3 , and CCNG2).

Genes clustering around the 10 feeder genes were compared with245 hypoxia-associated genes taken from the literature. Two genesshowed an unusual clustering pattern: CCNG2 clustered genescorrelated inversely with the literature genes and PFKFB3-associated genes correlated with none of the literature genes.The remaining eight clusters contained a varying percentage ofliterature genes; this percentage increased as the list of highestranked genes was shortened (14–30% when the 50 most correlatedgenes were considered). This improved when genes appearing inmore than one cluster were considered; 67% of the up-regulatedgenes found in >70% of clusters were in the literature list(Supplementary Fig. S3). Obviously, the number of genes selectedusing this method decreased as the amount of overlap increased. Afinal level of 50% overlap was chosen (i.e., transcripts appearing in>50% of clusters were considered). This was based on a Monte-Carlo simulation and offered a balance between identifying newgenes and including previously reported genes.There were 210 probe sets in the final gene list, 129 up-regulated

and 81 down-regulated, representing 99 (126 probe sets withknown gene identities) and 57 (68 probe sets with known geneidentities) genes, respectively. Of these, 27% (27) and 2% (1),respectively, were present in the literature list (i.e., they were knownto be hypoxia regulated). Table 2 lists the 99 up-regulated genesthat comprised the hypoxia metagene. Supplementary Tables S2and S3 list the up-regulated and down-regulated genes withcorrelation coefficients.Composition of the hypoxia metagene. The up-regulated gene

list (Table 2) contained several genes previously described (7, 11) as

Table 2. The 99 genes in the hypoxia metagene

MTX1 P4HA1 GAPD PSMA7ADORA2B PFKFB4 GMFB PSMB7

AK3 PGAM1 GSS PSMD2

ALDOA PVR HES2 PTGFRN

ANGPTL4 SLC16A1 HIG2 PYGLC20orf20 SLC2A1 IL8 RAN

MRPS17 TEAD4 KCTD11 RNF24

PGF TPBG KRT17 RNPS1

PGK1 TPI1 Kua RUVBL2AFARP1 TUBB2 LOC149464 S100A10

ANLN VEGF LOC56901 S100A3

B4GALT2 VEZT LRP2BP SIP1

BCAR1 AD-003 MGC14560 SLC6A10BMS1L ANKRD9 MGC17624 SLC6A8

BNIP3 C14orf156 MGC2408 SLCO1B3

HOMER1 C15orf25 MIF SMILEHSPC163 CA12 MRPL14 SNX24

IMP-2 CA9 NUDT15 SPTB

KIAA1393 CDCA4 PAWR TFAP2C

LDHA COL4A5 PDZK11 TIMM23LDLR CORO1C PLAU TMEM30B

MGC2654 CTEN PLEKHG3 TPD52L2

MNAT1 DKFZP564D166 PPARD VAPB

NDRG1 DPM2 PPP2CZ XPO5NME1 EIF2S1 PPP4R1

NOTE: Ordered by decreasing number of clusters in which the genes

were found from MTX1 (highest) to XPO5 (lowest). The abbreviationsare defined in Supplementary Table S2.

Table 1. Summary of HNSCC patient data

N 59

Age (y) Median (range) 60 (39�88)Gender Male 48 (81%)

Female 11 (19%)

pT 1 52 15

3 9

4 27

pN 0 211 9

2 12

3 1

Tumor site Larynx 9 (15%)Oropharynx 23 (39%)

Oral cavity 12 (20%)

Hypopharynx 12 (20%)

Other 3 (6%)Stage I 1 (2%)

II 9 (15%)

III 16 (27%)IVA 32 (54%)

IVB 1 (2%)

Differentiation Well 6 (10%)

Moderate 27 (46%)Poor 26 (44%)

% Tumor cells/biopsy Median (range) 60% (10�95%)CA9 staining Median (range) 20% (0�85%)HIF-1a staining Median (range) 31% (0�99%)

Hypoxia Gene Signature

www.aacrjournals.org 3443 Cancer Res 2007; 67: (7). April 1, 2007

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

induced by hypoxia, including those involved in glucose metabo-lism (e.g., ALDOA and GAPDH), angiogenesis (ANGPTL4 and PGF),cell migration (TPBG), and the regulation of apoptosis (BNIP3). Theknown genes represent a broad spectrum of molecular pathwaysinvolved in cellular response to hypoxia. Some of the genes havealso been described in RNA profiles of HNSCC [e.g., IL8 (27) andPLAU (28)]. Genes not previously reported to be up-regulatedunder hypoxia include MTX1, BCAR1, HES2, PSMA7 , and SLCO1B3 .Pattern of expression of the hypoxia metagene in HNSCC.

Investigation of the pattern of gene expression in the 59 HNSCCshowed that the genes were clustered into two main groups,whereas tumors were clustered into three groups. The two geneclusters are a consequence of how the genes were selected (i.e.,positively and negatively correlated with known hypoxia-regulatedgenes). The three tumor clusters are characterized by differingpatterns of expression, with one showing high expression of the 99up-regulated genes and low expression of the 57 down-regulatedgenes. However, when bootstrapping was applied to the tumorclusters (random sampling of patients with replacement), therewas a high level of variability in the classification of tumors intoclusters. This suggested that the hypoxia metagene was better

described as a continuous or graded characteristic rather than adefined binary status, and it might be difficult to assign reliablyfuture patients to a specific cluster. Using a continuous score alsoenables patient risk adjustment (based on hypoxia) on anindividual basis.Two HSs were calculated for each patient from the distribution

of the level of RNA expression of the list of up-regulated and down-regulated genes (HS-up and HS-down). The mean and SD of HS-upwere 8.76 F 0.69 (median, 8.80; range, 5.68–10.04). In Fig. 1A ,patients were ranked based on HS-up and hierarchical clusteringwas carried out on all the up-regulated and down-regulated genes.On the X axis, tumors are ordered by increasing HS-up so those onthe far right have high expression of hypoxia-associated genes.Guidelines indicate median and quartiles of HS-up for the 59patients. The Y axis shows the results of hierarchical clustering onthe whole gene list to illustrate which transcripts cluster together(e.g., different transcripts of the same gene cluster together and onecluster contains VEGF, CA9, and ANGPTL4). Using HS-down torank patients produced a different, but qualitatively similar,heatmap as HS-up and HS-down were inversely correlated (q =�0.75; P < 0.001).

Figure 1. A, dendogram of 11 normal tissue samples and 59 HNSCC. Two HSs were calculated for each patient by looking at the distribution of the level of RNAexpression of up-regulated and down-regulated hypoxia-associated gene (HS-up and HS-down). The median of this distribution was then calculated for each patient.X axis, patients are ordered by increasing HS-up; Y axis, results of hierarchical clustering on all the genes. The guidelines indicate the median and quartiles ofthe HS-up for the 59 patients. B, Kaplan-Meier plot of recurrence-free survival in 60 patients with HNSCC stratified according to HS-up (the hypoxia metagene). Thepatients are from a published study involving the molecular classification of HNSCC using patterns of gene expression (21). C, patients were first stratified according toquartiles, which suggested the separation into the two groups shown [i.e., the highest HS-up quartile (0.75�1) versus the remaining three quartiles (0�0.75)].The number of events/patients in the two arms was 10/45 and 8/15, respectively.

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3444 www.aacrjournals.org

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

The pattern of expression of the hypoxia metagene innormal tissue. Normal tissue samples showed a pattern ofexpression similar to the normoxic tumors (i.e., low median scoreof the up-regulated cluster when a continuous HS-up wasconsidered; Fig. 1A). For example, the median expression of VEGFwas similar in normal tissue (9.7) and ‘‘oxygenated’’ tumors (9.5)but significantly lower than in ‘‘hypoxic’’ tumors (11.0; P = 0.005).There was also no correlation between tumor and normal tissueexpression levels in the same patient. In a paired comparison usingSAM with a FDR of <0.05, 55 of the 210 transcripts (representing 44of the genes) in the gene list were expressed significantly higher intumor versus normal tissue.Hypoxia metagene as a prognostic factor in an indepen-

dent HNSCC data set. As no treatment outcome data wereavailable for our HNSCC data set due to a short follow-up, it wasnot possible to train our hypoxia metagene on outcome or test itssignificance on the present data set. Therefore, to explore whetherour signature could provide prognostic information, it wasapplied to a publicly available data set (21). The characteristicsof our 59 metagene-generating and the 60 literature HNSCC werecompared using m2. There were no significant differences in T, N,pT, and pN status, but our series contained more oropharynx andfewer larynx cancers (P = 0.010) and more poorly differentiatedtumors (P = 0.045) than the published data set. The data setdescribed a four-class expression profile, one of which wasassociated with a poor recurrence-free survival. Patients in theChung data set were stratified according to HS-up quartiles or

two groups (upper quartile versus rest). High HS-up expressionwas an adverse prognostic factor for recurrence-free survival(Fig. 1); the hazard ratio (HR) for the upper quartile versus therest was 3.64 [95% confidence interval (95% CI), 1.43–9.31]. Amultivariate Cox regression analysis was done, which includedHS-up entered as a continuous fractional rank, Chung’s intrinsicprofile, age, gender, smoking history, alcohol, clinical stage, lymphnode status, tumor grade, tumor subsite (hypopharyngeal versusothers), and array batch (Table 3). These factors were chosen asthey were used in the multivariate analysis in the original study.Significant adverse prognostic factors were Chung’s intrinsicprofile (P = 0.022), poor tumor differentiation (P = 0.003), and HS-up (P = 0.012; Fig. 2A). There was no significant difference insurvival when HS-down was considered. When the individualgenes were analyzed separately, VEGF, GLUT1 , and HK2 predicteda worse prognosis but were associated with large confidenceintervals (Fig. 2B). A HS for the whole literature list had noprognostic significance in either univariate or multivariateanalysis.Hypoxia metagene as a prognostic factor in another cancer

type. The ability of HS-up to predict outcome was investigated in abreast cancer series (22). Increasing HS-up was a significantadverse prognostic factor for metastasis-free (Fig. 3A and B) andoverall survival when patients were stratified by either quartiles ortwo groups. The HRs for metastasis-free survival were 1.62 (95% CI,1.33�1.98) for quartiles and 2.83 (95% CI, 0.78�3.38) for twogroups. In a multivariate analysis, including HS-up with the clinical

Table 3. Cox multivariate regression analyses of survival in published data sets

Cancer Survival Factor P HR 95% CI

Lower Upper

HNSCC* Recurrence-free Intrinsic classifier 0.022 1.84 1.09 3.10

HS-up (hypoxia metagene) 0.012 1.96 (14.83) 1.16 (1.80) 3.33 (122.35)Poor differentiation 0.003 4.15 1.62 10.64

Breastc

Metastasis-free HS-up (hypoxia metagene) 0.003 1.38 (3.58) 1.11 (1.53) 1.70 (8.39)

Age (decreasing decades) 0.021 1.49 1.06 2.09Increasing diameter 0.003 1.41 1.13 1.77

No. positive lymph nodes 0.009 1.12 1.03 1.22

Grade of differentiation 0.129

Moderate versus well 0.052 2.14 0.99 4.59Poor versus well 0.059 2.13 0.97 4.67

No adjuvant therapy 0.005 2.07 1.24 3.45

Breastc

Overall HS-up (hypoxia metagene) 0.002 1.43 (4.24) 1.14 (1.66) 1.81 (10.82)

Estrogen receptor negative 0.024 1.78 1.08 2.92Grade of differentiation 0.043

Moderate versus well 0.024 3.44 1.17 10.08

Poor versus well 0.012 4.04 1.36 12.04

Age (decreasing decades) 0.015 1.57 1.09 2.25

NOTE: Reduced models after backward likelihood selection are shown (entry P = 0.05, removal P = 0.10).

*Data from Chung et al. (21). Variables included in the model were intrinsic four-class classifier, age, gender, subsite (hypopharynx versus others),

smoking history (yes/no), alcohol use (yes/no), clinical stage (II to IV), nodal status (0/1), differentiation (poor/moderate/well), batch (as stratificationvariable), and HS-up. HS-up is the median RNA expression of up-regulated hypoxia-associated genes. HS-up was entered as a continuous variable

( fractional rank varying from 0 to 1), and the HR reported represents the risk for an increasing HS-up quartile. The numbers in parenthesis are the risks

at two ends of the spectrum.cData from Chang et al. (22). Variables included in the analysis were age (decreasing decade), increasing tumor diameter (cm), number of positive lymph

nodes, grade (poor/moderate/well), estrogen receptor status (negative versus positive), mastectomy (no versus yes), adjuvant therapy (no versus yes),

and HS-up.

Hypoxia Gene Signature

www.aacrjournals.org 3445 Cancer Res 2007; 67: (7). April 1, 2007

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

variables included in the original study (Table 3), HS-up retainedsignificance for metastasis-free (P = 0.003; Fig. 2C) and overall(P = 0.002) survival. In a multivariate analysis, including the 70-gene trained intrinsic signature, although HS-up was not significantfor metastasis-free survival, it retained significance for overallsurvival (P = 0.003; Supplementary Table S4). When a cell line–derived wound response signature (22) was included, HS-upretained independent prognostic significance for metastasis-free(HR, 2.80; 95% CI, 0.09�7.22; P = 0.033) and overall (HR, 3.01; 95%CI, 1.06�8.58; P = 0.038) survival. There was only 2.2% overlapbetween the wound response and our hypoxia signature (9 of 401known genes: LDLR, MNAT1, NME1, SLC16A1, CDCA4, CORO1C,NUDT15, PSMA7 , and PSMD2). When both the 70-gene trained andwound response signatures were included in the analysis, HS-upretained prognostic significance for overall survival but the woundsignature did not.Comparison with a published cell line–derived hypoxia

signature. A gene expression signature of cellular response tohypoxia in vitro was recently published (29). There was littleoverlap between the Chi and our signature, with only eight genes inboth lists. Whereas our signature contained 27% of the literaturegenes, the Chi profile contained 17%. To compare the differentapproaches used to derive hypoxia signatures, the above analyseson the Chang data set were repeated, including the Chi signature.In the Chi et al. article, mean RNA expression was used as ahypoxia signature metric rather than the median used here andpatients were stratified according to high versus low geneexpression. Analyses were carried out, therefore, using bothmethods, and in both cases, our approach proved superior. Inthe breast cancer series of van’t Veer, the Chi signature quantifiedas a median score was a significant prognostic factor formetastasis-free survival in univariate (HR for quartile increase,

1.39; 95% CI, 1.14�1.70; P = 0.004, log-rank test) and multivariate(HR, 2.74; 95% CI, 1.29�5.83; P = 0.0090) analyses. These datacompare less favorably with our signature in both univariate (HRfor quartile increase, 1.62; 95% CI, 1.33�1.98; P < 0.0001, log-ranktest; Fig. 3A) and multivariate (HR, 3.58; 95% CI, 1.53�8.39; P =0.0030) analyses. When both signatures were included in themultivariate analysis, the Chi signature lost prognostic significance.Similar results were obtained when considering overall survival.The analyses were repeated using mean RNA expression ofhypoxia-associated genes and stratifying by high versus lowhypoxia response groups as done by Chi et al. (29). Similar resultswere seen in univariate (Chi: HR, 2.15; 95% CI, 1.40�3.30; P < 0.001,log-rank test; ours: HR, 2.67; 95% CI, 1.76�4.06; P < 0.001, log-ranktest; Fig. 3C and D) and multivariate (Chi: HR, 1.91; 95% CI,1.22�3.00; P = 0.0047; ours: HR, 2.06; 95% CI, 1.29�3.31; P = 0.0019)analyses. Again, when both signatures were included, only oursretained prognostic significance. Likewise, repeating the analysesfor overall survival produced similar results.

Discussion

The limitations of using protein markers to reflect tumorhypoxia. High expression of HIF-1a and CA9 is associated withadverse prognosis in HNSCC (12, 30). Although high expression ofHIF-1a and CA9 was thought to reflect the hypoxic nature of atumor and activation of the HIF pathway, other studies reported noassociation with survival (31, 32) or association for only one factor(33). Some of these anomalous findings have been explained by thedifferent half-lives for CA9 (days) and HIF-1a (minutes) proteins(34). It is more probable that, because hypoxia influences manybiological pathways, a single factor is incapable of adequatelydescribing this complex response.

Figure 2. Cox multivariate analysis derived HRs with95% CIs. A, variables used in the original publication ofan independent HNSCC patient data set were enteredin the model along with HS-up. HS-up was entered as acontinuous variable (fractional rank varying from 0 to1), and the HR reported represents the risk for anincreasing HS-up quartile (the HR for the two ends ofthe spectrum was 14.83; 95% CI, 1.80�122.35). B, HRfor the RNA expression of the individual genes used toderive the hypoxia metagene for a Cox multivariateanalysis of recurrence-free survival in the independentHNSCC data set. No data were available for ENO1.C, variables used in the original publication in anindependent breast cancer data set were entered into aCox multivariate regression model along with HS-up.The HR for two ends of the HS-up spectrum was 3.58(95% CI, 1.53�8.39).

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3446 www.aacrjournals.org

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

In this study, a cluster analysis based on the protein expressionof HIF-1a or CA9 produced gene profiles with minimal overlap.The CA9 protein and RNA expression correlated, indicating thatmRNA is readily translated into the protein in HNSCC. In contrast,the post-translational modification of HIF-1a (11) explains the lackof correlation for HIF. A cluster analysis around the mRNAexpression of CA9 identified several genes recognized as hypoxiainducible involving a variety of molecular signaling pathways,suggesting the potential to identify novel hypoxia-regulated genes.Furthermore, the cluster analysis divided the cohort of patientsinto two groups, with one showing high expression of knownhypoxia up-regulated genes and thus possibly reflecting hypoxicversus oxygenated tumors.The hypoxia metagene based on clustering HNSCC around

the RNA expression of known in vitro hypoxia-regulated genes.Ten genes shown to be hypoxia regulated in vitro were used tocluster other genes in vivo . To test the validity of this approach inidentifying hypoxia-regulated genes, clustered genes were com-pared with a set of 245 genes reported in literature reviews ashypoxia regulated. The approach clustered other well-recognizedhypoxia-regulated genes, suggesting that the method might identify

novel hypoxia-associated genes. Our approach highlighted aninteresting finding about differences in hypoxia response in vivoversus in vitro . Two of our selected genes showed unexpectedclustering patterns. Genes clustering around CCNG2, involved incell cycle regulation, correlated inversely with genes in theliterature list. None of the genes clustering around PFKFB3,important in the metabolism of fructose, correlated with any ofthe genes in the literature list. Interestingly, both are up-regulatedin vitro in response to hypoxia. Tumor type–specific differences inhypoxia response might also be important, as CCNG2 was shownto be down-regulated in another gene profiling study in HNSCC(27).In addition, some studies advocate the use of laser microdis-

section to ensure that only the tumor RNA is extracted (35), butthis ignores the effects the tumor may have on the surroundingstroma. Therefore, in this study, we have used tumor andsurrounding stroma to extract RNA while quantitatively confirmingthe presence of tumor. Several genes were identified that clusteredwith the hypoxia-associated genes but have not previously beenshown to be hypoxia regulated [e.g., MTX1 , a component of atransport complex in the outer mitochondrial membrane (36);

Figure 3. Kaplan-Meier plots of metastases-free survival in 295 patients with breast cancer. The data were taken from Chang et al. (22). A, stratified according toHS-up quartiles. The numbers of events/patients for increasing quartiles were 12/73, 18/73, 20/74, and 38/73. B, stratified by highest HS-up quartile (0.75�1)versus the remaining three quartiles (0�0.75). The number of events/patients for the two arms was 50/220 and 38/73, respectively. C, stratified using the Chi geneexpression signature of cellular response to hypoxia (29). Two arms are for patients with high (mean score, >0; 35 events/77 patients) and low (mean score,<0; 53 events/218 patients) mean RNA expression of hypoxia-associated genes. D, stratified using the hypoxia gene signature derived in this article but using the Chiet al. (29) approach to compute the hypoxia score. The two arms are for patients with high (mean score, >0; 42 events/87 patients) and low (mean score, <0; 46 events/208 patients) mean RNA expression of hypoxia-associated genes.

Hypoxia Gene Signature

www.aacrjournals.org 3447 Cancer Res 2007; 67: (7). April 1, 2007

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

BCAR1 , which encodes a docking protein critical in controllingintegrin-dependent signaling (37); PSMA7 , involved in ubiquitin-dependent protein catabolism and plays a part in the regulation ofHIF-1a (38); and SLCO1B3 , a member of the organic aniontransporting polypeptide family of transporters (39)].Deriving a metric for the clinical application of the

hypoxia metagene. The hypoxia metagene clustered the 59HNSCCs studied into three tumor groups, which were not robustto bootstrapping. Thus, a continuous score was used to rank thetumors from low to high hypoxia metagene expression (Fig. 1).While this work was in progress, Chi et al. (29) published ahypoxia gene profile derived in a different way. They analyzed theexpression of genes induced in vitro by hypoxia in a panel ofnormal cell lines and showed a dependency on both cell typeand the intensity/duration of hypoxia (29). They used a set ofcommon genes expressed in the normal cell types studied fortheir profile, which predicted relapse in ovarian and breastcancers.In both studies, HSs were derived to provide a metric

applicable to other data sets. Chi et al. (29) used the mean geneexpression levels of the genes in their signature, whereas in ourstudy median values were calculated as the median is a morerobust statistic not affected by outliers. Furthermore, to makeHS general and transportable, the ranked score was used ratherthan raw values. It is of interest that, in our profile, 27% of thegenes were previously recognized as hypoxia regulated in theliterature, suggesting the identification of new pathways regulatedby hypoxia. There were nine transcripts/eight genes that appearedin both signatures: P4HA1, PGK1, CA9, ANGPTL4, NDRG1, LDHA,SLC2A1 , and SLC6A8 . These eight genes reflect a variety of path-ways (glycolysis, angiogenesis, extracellular matrix deposition, andpH regulation) and may represent a core set for future classi-fications. It is of interest that VEGF is not present in the Chihypoxia gene profile.The hypoxia metagene as a prognostic factor. In both

univariate and multivariate analyses, the hypoxia metagene was asignificant prognostic factor for recurrence-free survival inan independent HNSCC data set. In multivariate analysis, HS-up(P = 0.012) compared favorably with the original study four-classintrinsic classifier (P = 0.022). Interestingly, the down-regulatedgenes did not correlate with outcome, suggesting that they play aless significant part in tumor treatment response. Another findingof interest was that HS-up correlated with the intrinsic profile inthe Chung study (Kruskal-Wallis m2 = 22.3; P < 0.001); specifically,HS-up was higher in cluster 1 than in the other three Chungclusters. Cluster 1 correlated with a poor recurrence-free survival inthe Chung study and had a high expression of genes that wereassociated with activation of the epidermal growth factor receptorpathway. Hypoxia up-regulated genes in Chung cluster 1 wereSLC16A2, TGF-a , and KRT14 .

In the breast cancer data set, our hypoxia metagene was superiorto both the wound response and Chi signatures. When the trainedintrinsic classifier was included in the multivariate analysis, onlyour hypoxia metagene retained independent prognostic signifi-cance for overall survival. The difference in efficacy of the twohypoxia signatures may reflect the way they were developed: fromnormal cells in vitro (Chi signature) versus a combination of tumorcells in vitro plus cancers in vivo (our signature).It is of interest to note that both Chi (29) and our studies initially

investigated a specific tumor type, renal cancer or HNSCC,respectively, yet found applicability across tumor types. Ein-Doret al. (40) highlighted the lack of overlap between expressionprofiles, which predict cancer treatment outcome and showed thatmany equally predictive gene lists could be produced from thevan’t Veer breast cancer signature. It was suggested that this is duein part to the many genes that correlate with survival. However,Shen et al. (41) analyzed four independent microarray studies toderive an interstudy validated metasignature associated with breastcancer prognosis, which was comparable or better at providingprognostic information compared with the intrinsic signatures.With the publication of further hypoxia metagenes (42), therefore,our study and that of Chi et al. (29) are working toward thederivation of a future hypoxia metasignature with potentialapplication across a range of tumor types. A rationale behind thederivation of phenotype-specific profiles is that metagenes derivedfrom defined pathways may be more powerful markers ofprognosis but also particularly of value in selecting patients forspecific interventions.In summary, hypoxia results in molecular changes that promote

an aggressive phenotype and reduce the efficacy of conventionaltreatments, resulting in a significant therapeutic challenge. Usinga combined in vitro and in vivo approach, a hypoxia metagenewas derived that outperformed the original intrinsic profile of apublished HNSCC data set and was an independent prognosticfactor in a widely studied breast cancer microarray series. Ourcombined in vitro and in vivo strategy has potential for the futuredevelopment of metasignatures, which are not based on theextensive training of intrinsic profiles. In conclusion, genesignatures can be derived in vivo that reflect biological phenotypesrelevant in determining cancer patient prognosis.

Acknowledgments

Received 9/7/2006; revised 12/13/2006; accepted 1/25/2007.Grant support: Cancer Research UK, National Translational Cancer Research

Network of the United Kingdom, and EU Integrated Project ACGT (FP6-IST-026996).The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.We thank Dr. Jo Cresswell for administrative assistance in running the project,

Stuart Pepper for advice on the microarray work, and the Molecular Biology Corefacility of the Paterson Institute (Manchester, United Kingdom), which houses theCancer Research UK Affymetrix facility.

References1. Parkin DM, Bray F, Ferlay J, Pisani P. Estimating theworld cancer burden: Globocan 2000. Int J Cancer 2001;94:153–6.

2. Boring CC, Squires TS, Tong T, Montgomery S. Cancerstatistics, 1994. CA Cancer J Clin 1994;44:7–26.

3. Bernier J, Domenge C, Ozsahin M, et al. Postoperativeirradiation with or without concomitant chemotherapy

for locally advanced head and neck cancer. N Engl J Med2004;350:1945–52.

4. Sessions DG, Spector GJ, Lenox J, et al. Analysis oftreatment results for oral tongue cancer. Laryngoscope2002;112:616–25.

5. Adam MF, Gabalski EC, Bloch DA, et al. Tissue oxygendistribution in head and neck cancer patients. HeadNeck 1999;21:146–53.

6. Nordsmark M, Bentzen SM, Rudat V, et al. Prog-

nostic value of tumor oxygenation in 397 head andneck tumors after primary radiation therapy. Aninternational multi-center study. Radiother Oncol 2005;77:18–24.

7. Harris AL. Hypoxia—a key regulatory factor in tumourgrowth. Nat Rev Cancer 2002;2:38–47.

8. Denko NC, Fontana LA, Hudson KM, et al. Investigat-ing hypoxic tumor physiology through gene expressionpatterns. Oncogene 2003;22:5907–14.

Cancer Research

Cancer Res 2007; 67: (7). April 1, 2007 3448 www.aacrjournals.org

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

9. Giaccia AJ. Hypoxic stress proteins: survival of thefittest. Semin Radiat Oncol 1996;6:46–58.

10. Wouters BG, Weppler SA, Koritzinsky M, et al.Hypoxia as a target for combined modality treatments.Eur J Cancer 2002;38:240–57.

11. Semenza GL. Targeting HIF-1 for cancer therapy. NatRev Cancer 2003;3:721–32.

12. Koukourakis MI, Giatromanolaki A, Sivridis E, et al.Hypoxia-regulated carbonic anhydrase-9 (CA9) relatesto poor vascularization and resistance of squamous cellhead and neck cancer to chemoradiotherapy. ClinCancer Res 2001;7:3399–403.

13. Koukourakis MI, Giatromanolaki A, Sivridis E, et al.Hypoxia-inducible factor (HIF1A and HIF2A), angiogen-esis, and chemoradiotherapy outcome of squamous cellhead-and-neck cancer. Int J Radiat Oncol Biol Phys 2002;53:1192–202.

14. Loncaster JA, Harris AL, Davidson SE, et al. Carbonicanhydrase (CA IX) expression, a potential new intrinsicmarker of hypoxia: correlations with tumor oxygenmeasurements and prognosis in locally advancedcarcinoma of the cervix. Cancer Res 2001;61:6394–9.

15. Gautier L, Cope L, Bolstad BM, Irizarry RA. affy—analysis of Affymetrix GeneChip data at the probe level.Bioinformatics 2004;20:307–15.

16. Wilson CL, Miller CJ. Simpleaffy: a BioConductorpackage for Affymetrix Quality Control and dataanalysis. Bioinformatics 2005;21:3683–5.

17. Wu Z, Irizarry RA, Gentleman R, Martinez Murillo F,Spencer F. A model based background adjustment foroligonucleotide expression arrays. Available from:http://www.bepress.com/jhubiostat/paper1 2004.

18. Efron B, Tibshirani R. Empirical bayes methods andfalse discovery rates for microarrays. Genet Epidemiol2002;23:70–86.

19. Storey JD, Tibshirani R. Statistical methods foridentifying differentially expressed genes in DNA micro-arrays. Methods Mol Biol 2003;224:149–57.

20. Buffa FM, Bentzen SM, Daley FM, et al. Molecularmarker profiles predict locoregional control of head andneck squamous cell carcinoma in a randomized trial ofcontinuous hyperfractionated accelerated radiotherapy.Clin Cancer Res 2004;10:3745–54.

21. Chung CH, Parker JS, Karaca G, et al. Molecularclassification of head and neck squamous cell carcino-mas using patterns of gene expression. Cancer Cell 2004;5:489–500.

22. Chang HY, Nuyten DS, Sneddon JB, et al. Robustness,scalability, and integration of a wound-response geneexpression signature in predicting breast cancer surviv-al. Proc Natl Acad Sci U S A 2005;102:3738–43.

23. van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene-expression signature as a predictor of survival in breastcancer. N Engl J Med 2002;347:1999–2009.

24. van ’t Veer LJ, Dai H, van de Vijver MJ, et al. Geneexpression profiling predicts clinical outcome of breastcancer. Nature 2002;415:530–6.

25. Le QT, Denko NC, Giaccia AJ. Hypoxic geneexpression and metastasis. Cancer Metastasis Rev2004;23:293–310.

26. Papandreou I, Powell A, Lim AL, Denko N. Cellularreaction to hypoxia: sensing and responding to anadverse environment. Mutat Res 2005;569:87–100.

27. Choi P, Chen C. Genetic expression profiles andbiologic pathway alterations in head and neck squa-mous cell carcinoma. Cancer 2005;104:1113–28.

28. Roepman P, Wessels LF, Kettelarij N, et al. Anexpression profile for diagnosis of lymph node metas-tases from primary head and neck squamous cellcarcinomas. Nat Genet 2005;37:182–6.

29. Chi JT, Wang Z, Nuyten DS, et al. Gene expressionprograms in response to hypoxia: cell type specificityand prognostic significance in human cancers. PLoSMed 2006;3:e47.

30. Aebersold DM, Burri P, Beer KT, et al. Expression ofhypoxia-inducible factor-1a: a novel predictive andprognostic parameter in the radiotherapy of oropha-ryngeal cancer. Cancer Res 2001;61:2911–6.

31. Swinson DE, Jones JL, Richardson D, et al. Carbonicanhydrase IX expression, a novel surrogate markerof tumor hypoxia, is associated with a poor prognosisin non-small-cell lung cancer. J Clin Oncol 2003;21:473–82.

32. Giatromanolaki A, Koukourakis MI, Sivridis E, et al.Relation of hypoxia inducible factor 1a and 2a inoperable non-small cell lung cancer to angiogenic/

molecular profile of tumours and survival. Br J Cancer2001;85:881–90.

33. Hui EP, Chan AT, Pezzella F, et al. Coexpression ofhypoxia-inducible factors 1a and 2a, carbonic anhy-drase IX, and vascular endothelial growth factor innasopharyngeal carcinoma and relationship to survival.Clin Cancer Res 2002;8:2595–604.

34. Turner KJ, Crew JP, Wykoff CC, et al. The hypoxia-inducible genes VEGF and CA9 are differentiallyregulated in superficial vs invasive bladder cancer. Br JCancer 2002;86:1276–82.

35. Leethanakul C, Patel V, Gillespie J, et al. Distinctpattern of expression of differentiation and growth-related genes in squamous cell carcinomas of the headand neck revealed by the use of laser capturemicrodissection and cDNA arrays. Oncogene 2000;19:3220–4.

36. Armstrong LC, Komiya T, Bergman BE, Mihara K,Bornstein P. Metaxin is a component of a preproteinimport complex in the outer membrane of the mamma-lian mitochondrion. J Biol Chem 1997;272:6510–8.

37. Holcomb M, Rufini A, Barila D, Klemke RL.Deregulation of proteasome function induces Abl-mediated cell death by uncoupling p130CAS and c-CrkII. J Biol Chem 2006;281:2430–40.

38. Cho S, Choi YJ, Kim JM, et al. Binding and regulationof HIF-1a by a subunit of the proteasome complex,PSMA7. FEBS Lett 2001;498:62–6.

39. Konig J, Cui Y, Nies AT, Keppler D. Localization andgenomic organization of a new hepatocellular organicanion transporting polypeptide. J Biol Chem 2000;275:23161–8.

40. Ein-Dor L, Kela I, Getz G, Givol D, Domany E.Outcome signature genes in breast cancer: is there aunique set? Bioinformatics 2005;21:171–8.

41. Shen R, Ghosh D, Chinnaiyan AM. Prognostic meta-signature of breast cancer developed by two-stagemixture modeling of microarray data. BMC Genomics2004;5:94.

42. Dressman HK, Hans C, Bild A, et al. Gene expressionprofiles of multiple breast cancer phenotypes andresponse to neoadjuvant chemotherapy. Clin CancerRes 2006;12:819–26.

Hypoxia Gene Signature

www.aacrjournals.org 3449 Cancer Res 2007; 67: (7). April 1, 2007

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:3441-3449. Cancer Res   Stuart C. Winter, Francesca M. Buffa, Priyamal Silva, et al.   Cancer to Prognosis of Multiple CancersRelation of a Hypoxia Metagene Derived from Head and Neck

  Updated version

  http://cancerres.aacrjournals.org/content/67/7/3441

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2007/03/27/67.7.3441.DC1.html

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/7/3441.full.html#ref-list-1

This article cites 41 articles, 14 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/7/3441.full.html#related-urls

This article has been cited by 21 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

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

Research. on November 25, 2015. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from