01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

13
Gene Expression Signature to Improve Prognosis Prediction of Stage II and III Colorectal Cancer Ramon Salazar, Paul Roepman, Gabriel Capella, Victor Moreno, Iris Simon, Christa Dreezen, Adriana Lopez-Doriga, Cristina Santos, Corrie Marijnen, Johan Westerga, Sjoerd Bruin, David Kerr, Peter Kuppen, Cornelis van de Velde, Hans Morreau, Loes Van Velthuysen, Annuska M. Glas, Laura J. Van’t Veer, and Rob Tollenaar From the Institut Catala ` d’Oncologia– Biomedical Research Institute of Bellvitge, L’Hospitalet de Llobregat; University of Barcelona, Barcelona, Spain; Agendia; Netherlands Cancer Institute; Slotervaart Hospital, Amsterdam; Leiden University Medical Center, Leiden, the Netherlands; and University of Oxford, Radcliffe Infir- mary, Oxford, United Kingdom. Submitted May 5, 2010; accepted Septem- ber 7, 2010; published online ahead of print at www.jco.org on November 22, 2010. RNA isolation and hybridization of the samples and part of the analysis were performed and funded at Agendia. The training set of the study was partly supported by the Leiden Medical Centre Institutional Grant and by the Dutch Genomics Initiative Cancer Genomics Center in the Netherlands Cancer Institute. The validation of the study was partly supported by the Catalan Institute of Oncology and the Private Foundation of the Biomedical Research Institute of Bellvitge, the Spanish Ministry of Science (Grants No. SAF 06-6084 and SAF 2009-07319), the Instituto de Salud Carlos III (Grants No. PI08-1635 and PI09-01037), Spanish Networks Red Tema ´tica de Investigacio ´n Cooperativa en Ca ´ncer (Grant No. RD06/ 0020/1050), Centro de Investigacio ´n Biome ´ dica en Red de Epidemiología y Salud Pu ´ blica G55 and the Accion Transver- sal del Cancer, the Catalan Government Departament d’Universitats, Recerca i Soci- etat de la Informacio ´ (Grants No. 2009SGR1489 and 2009SGR290), the European Commission (Grant No. FP7-COOP-Health-2007-B), HiperDart, and Fundacio ´ Gastroenterologia Dr Francisco Vilardell. Authors’ disclosures of potential con- flicts of interest and author contribu- tions are found at the end of this article. Corresponding author: Ramon Salazar, MD, Institut Catala ` d’Oncologia-IDIBELL, L’Hospitalet de Llobregat, Av Gran Via 199-203, Barcelona, Spain 08907; e-mail: [email protected]. © 2010 by American Society of Clinical Oncology 0732-183X/10/2899-1/$20.00 DOI: 10.1200/JCO.2010.30.1077 A B S T R A C T Purpose This study aims to develop a robust gene expression classifier that can predict disease relapse in patients with early-stage colorectal cancer (CRC). Patients and Methods Fresh frozen tumor tissue from 188 patients with stage I to IV CRC undergoing surgery was analyzed using Agilent 44K oligonucleotide arrays. Median follow-up time was 65.1 months, and the majority of patients (83.6%) did not receive adjuvant chemotherapy. A nearest mean classifier was developed using a cross-validation procedure to score all genes for their association with 5-year distant metastasis–free survival. Results An optimal set of 18 genes was identified and used to construct a prognostic classifier (ColoPrint). The signature was validated on an independent set of 206 samples from patients with stage I, II, and III CRC. The signature classified 60% of patients as low risk and 40% as high risk. Five-year relapse-free survival rates were 87.6% (95% CI, 81.5% to 93.7%) and 67.2% (95% CI, 55.4% to 79.0%) for low- and high-risk patients, respectively, with a hazard ratio (HR) of 2.5 (95% CI, 1.33 to 4.73; P .005). In multivariate analysis, the signature remained one of the most significant prognostic factors, with an HR of 2.69 (95% CI, 1.41 to 5.14; P .003). In patients with stage II CRC, the signature had an HR of 3.34 (P .017) and was superior to American Society of Clinical Oncology criteria in assessing the risk of cancer recurrence without prescreening for microsatellite instability (MSI). Conclusion ColoPrint significantly improves the prognostic accuracy of pathologic factors and MSI in patients with stage II and III CRC and facilitates the identification of patients with stage II disease who may be safely managed without chemotherapy. J Clin Oncol 28. © 2010 by American Society of Clinical Oncology INTRODUCTION The American Joint Committee on Cancer TNM staging system is the current standard for determin- ing the prognosis of patients with colorectal cancer (CRC). Patients with stage I CRC have a 5-year sur- vival rate of approximately 93%, which decreases to approximately 80% for patients with stage II disease and to 60% for patients with stage III disease. 1 De- spite numerous clinical trials, the benefit of adjuvant chemotherapy for patients with stage II CRC is still debatable. 2-4 In Western countries, official guide- lines give suggestions for risk stratification but no clear recommendations on the administration of adjuvant chemotherapy. 5 In contrast, adjuvant treatment is universally recommended for all pa- tients with stage III disease. 6 However, patients with T1-2N1M0 tumors (stage IIIA) have significantly higher survival rates than patients with stage IIB tumors, 1 suggesting that adjuvant chemotherapy se- lection needs optimization. To date, substantial effort has been put into the identification of clinicopathologic parameters that predict prognosis of patients with stage II disease. The most important factors for predicting the risk of systemic recurrence (ie, distant metastases) are emergency presentation, poorly differentiated tu- mor, depth of tumor invasion, and adjacent organ involvement (T4). 5,7 Inadequate sampling of lymph nodes is an additional risk factor. 8 Among the mo- lecular factors investigated as prognostic candidates in early CRCs, microsatellite instability (MSI) is the JOURNAL OF CLINICAL ONCOLOGY O R I G I N A L R E P O R T © 2010 by American Society of Clinical Oncology 1 http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2010.30.1077 The latest version is at Published Ahead of Print on November 22, 2010 as 10.1200/JCO.2010.30.1077 Copyright 2010 by American Society of Clinical Oncology from 196.3.50.254 Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010 Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

description

Copyright 2010 by American Society of Clinical Oncology JClinOncol28.©2010byAmericanSocietyofClinicalOncology INTRODUCTION Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010 Copyright © 2010 American Society of Clinical Oncology. All rights reserved. ©2010byAmericanSocietyofClinical Oncology Authors’disclosuresofpotentialcon- flictsofinterestandauthorcontribu- tionsarefoundattheendofthis article. from 196.3.50.254 1

Transcript of 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Page 1: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Gene Expression Signature to Improve Prognosis Predictionof Stage II and III Colorectal CancerRamon Salazar, Paul Roepman, Gabriel Capella, Victor Moreno, Iris Simon, Christa Dreezen,Adriana Lopez-Doriga, Cristina Santos, Corrie Marijnen, Johan Westerga, Sjoerd Bruin, David Kerr,Peter Kuppen, Cornelis van de Velde, Hans Morreau, Loes Van Velthuysen, Annuska M. Glas,Laura J. Van’t Veer, and Rob Tollenaar

From the Institut Catala d’Oncologia–Biomedical Research Institute of Bellvitge,L’Hospitalet de Llobregat; University ofBarcelona, Barcelona, Spain; Agendia;Netherlands Cancer Institute; SlotervaartHospital, Amsterdam; Leiden UniversityMedical Center, Leiden, the Netherlands;and University of Oxford, Radcliffe Infir-mary, Oxford, United Kingdom.

Submitted May 5, 2010; accepted Septem-ber 7, 2010; published online ahead of printat www.jco.org on November 22, 2010.

RNA isolation and hybridization of thesamples and part of the analysis wereperformed and funded at Agendia. Thetraining set of the study was partlysupported by the Leiden Medical CentreInstitutional Grant and by the DutchGenomics Initiative Cancer GenomicsCenter in the Netherlands Cancer Institute.The validation of the study was partlysupported by the Catalan Institute ofOncology and the Private Foundation of theBiomedical Research Institute of Bellvitge,the Spanish Ministry of Science (GrantsNo. SAF 06-6084 and SAF 2009-07319),the Instituto de Salud Carlos III (Grants No.PI08-1635 and PI09-01037), SpanishNetworks Red Tematica de InvestigacionCooperativa en Cancer (Grant No. RD06/0020/1050), Centro de InvestigacionBiomedica en Red de Epidemiología ySalud Publica G55 and the Accion Transver-sal del Cancer, the Catalan GovernmentDepartament d’Universitats, Recerca i Soci-etat de la Informacio (Grants No.2009SGR1489 and 2009SGR290), theEuropean Commission (Grant No.FP7-COOP-Health-2007-B), HiperDart, andFundacio Gastroenterologia Dr FranciscoVilardell.

Authors’ disclosures of potential con-flicts of interest and author contribu-tions are found at the end of thisarticle.

Corresponding author: Ramon Salazar, MD,Institut Catala d’Oncologia-IDIBELL,L’Hospitalet de Llobregat, Av Gran Via199-203, Barcelona, Spain 08907; e-mail:[email protected].

© 2010 by American Society of ClinicalOncology

0732-183X/10/2899-1/$20.00

DOI: 10.1200/JCO.2010.30.1077

A B S T R A C T

PurposeThis study aims to develop a robust gene expression classifier that can predict disease relapse inpatients with early-stage colorectal cancer (CRC).

Patients and MethodsFresh frozen tumor tissue from 188 patients with stage I to IV CRC undergoing surgery wasanalyzed using Agilent 44K oligonucleotide arrays. Median follow-up time was 65.1 months, andthe majority of patients (83.6%) did not receive adjuvant chemotherapy. A nearest mean classifierwas developed using a cross-validation procedure to score all genes for their association with5-year distant metastasis–free survival.

ResultsAn optimal set of 18 genes was identified and used to construct a prognostic classifier (ColoPrint).The signature was validated on an independent set of 206 samples from patients with stage I, II,and III CRC. The signature classified 60% of patients as low risk and 40% as high risk. Five-yearrelapse-free survival rates were 87.6% (95% CI, 81.5% to 93.7%) and 67.2% (95% CI, 55.4% to79.0%) for low- and high-risk patients, respectively, with a hazard ratio (HR) of 2.5 (95% CI, 1.33to 4.73; P � .005). In multivariate analysis, the signature remained one of the most significantprognostic factors, with an HR of 2.69 (95% CI, 1.41 to 5.14; P � .003). In patients with stage IICRC, the signature had an HR of 3.34 (P � .017) and was superior to American Society of ClinicalOncology criteria in assessing the risk of cancer recurrence without prescreening for microsatelliteinstability (MSI).

ConclusionColoPrint significantly improves the prognostic accuracy of pathologic factors and MSI in patientswith stage II and III CRC and facilitates the identification of patients with stage II disease who maybe safely managed without chemotherapy.

J Clin Oncol 28. © 2010 by American Society of Clinical Oncology

INTRODUCTION

The American Joint Committee on Cancer TNMstaging system is the current standard for determin-ing the prognosis of patients with colorectal cancer(CRC). Patients with stage I CRC have a 5-year sur-vival rate of approximately 93%, which decreases toapproximately 80% for patients with stage II diseaseand to 60% for patients with stage III disease.1 De-spite numerous clinical trials, the benefit of adjuvantchemotherapy for patients with stage II CRC is stilldebatable.2-4 In Western countries, official guide-lines give suggestions for risk stratification but noclear recommendations on the administration ofadjuvant chemotherapy.5 In contrast, adjuvanttreatment is universally recommended for all pa-

tients with stage III disease.6 However, patients withT1-2N1M0 tumors (stage IIIA) have significantlyhigher survival rates than patients with stage IIBtumors,1 suggesting that adjuvant chemotherapy se-lection needs optimization.

To date, substantial effort has been put into theidentification of clinicopathologic parameters thatpredict prognosis of patients with stage II disease.The most important factors for predicting the risk ofsystemic recurrence (ie, distant metastases) areemergency presentation, poorly differentiated tu-mor, depth of tumor invasion, and adjacent organinvolvement (T4).5,7 Inadequate sampling of lymphnodes is an additional risk factor.8 Among the mo-lecular factors investigated as prognostic candidatesin early CRCs, microsatellite instability (MSI) is the

JOURNAL OF CLINICAL ONCOLOGY O R I G I N A L R E P O R T

© 2010 by American Society of Clinical Oncology 1

http://jco.ascopubs.org/cgi/doi/10.1200/JCO.2010.30.1077The latest version is at Published Ahead of Print on November 22, 2010 as 10.1200/JCO.2010.30.1077

Copyright 2010 by American Society of Clinical Oncologyfrom 196.3.50.254

Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
W.Wifler
Highlight
W.Wifler
Highlight
Page 2: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

only one that has remained significant both in a meta-analysis andprospective trials.9-11

During the last decade, gene expression profiling has shown greatpromise in predicting the long-term outcome of an individual pa-tient.12 The power of applying customized microarray technology topredict the prognosis of patients with breast cancer has led to thesuccessful development of a US Food and Drug Administration–approved breast cancer prognostic test (MammaPrint; Agendia, Am-sterdam, the Netherlands).13,14 Several studies have already describedprognostic gene expression profiles for patients with CRC from tumorsamples10,15-21 and even from adjacent normal mucosa.22 However,few studies compared the genomic prognosis prediction with tradi-tional risk factors except for stage.20 In this study, we demonstrate thedevelopment and validation of a new prognosis signature to distin-guish low- and high-risk patients using gene expression analysis.

PATIENTS AND METHODS

Patients and Tumor Samples

Samples used for classifier training (n � 188) were prospectively col-lected between 1983 and 2002 at the Netherlands Cancer Institute (Amster-dam), the Leiden University Medical Center (Leiden), and the SlotervaartGeneral Hospital (Amsterdam) in the Netherlands. Samples for the validationset of patients (n � 206) were prospectively collected at the Institut Catalad’Oncologia in Barcelona, Spain, between 1996 and 2004. Clinical and patho-logic data were extracted from the medical records and centrally reviewed forthe purpose of this study. Patients with rectal cancer underwent total mesorec-tal excision controlled surgery. Patients were staged according to the AmericanJoint Committee on Cancer TNM staging system and monitored for relapse(development of distant metastases or locoregional recurrence) and overallsurvival (median follow-up time: training set, 65.1 months; validation set, 54.8months). Detailed patient information is listed in Table 1 and Appendix TableA1 (online only). The study was approved by the medical ethical boards of theparticipating medical centers. In all, 71.6% of the patients did not receiveadjuvant chemotherapy (83.6% and 61.9% of patients in the training andvalidation sets, respectively).

Mutational and MSI Analysis

Mutations in BRAF V600; KRAS codons 12, 13, and 61; and PI3KCAexons 9 and 20 were assessed in cDNA by means of direct sequencing ofpolymerase chain reaction products using M13 primers after reverse transcrip-tase polymerase chain reaction. Primers used and experimental conditions areavailable on request. In the training set, 5-�m slides were immunohistochemi-cally stained for the markers MLH1 and PMS2 using standard protocols toidentify MSI-high (MSI-H) patients. In the validation set, the MSI statusanalysis was performed as previously described.23

Gene Expression Analysis

RNA isolation, labeling, and hybridization to Agendia customizedwhole-genome oligonucleotide high-density microarrays followed proceduresas previously described.14 Samples were hybridized against a colon cancerreference pool, consisting of primary tumor tissue from 44 patients with CRC.Raw fluorescence intensities were quantified and normalized using AgilentFeature Extraction software (Agilent, Santa Clara, CA) according to the man-ufacturer’s protocols and imported into R/Bioconductor (http://www.bioconductor.org/) for further analysis.

A supervised training approach was performed to identify a prognosticCRC gene signature. Using a cross-validation procedure, all 33,834 geneprobes that showed variation across the 188 training samples were scored fortheir association (t test) with 5-year distant metastasis–free survival (DMFS).During each of the leave-one-out cross-validation iterations, the set of geneswith a significant DMFS association [abs(T) � 3.5] was marked. From thecomprehensive pool of genes, an optimal set of 18 nonredundant probesshowed robust DMFS association in more than 50% of all iterations, a selec-

tion criterion suggested by Michiels et al.24 These 18 probes corresponded to18 unique genes (Appendix Table A2, online only) and were used to constructa nearest centroid–based classifier (called ColoPrint). This type of classifier hasbeen proven to be useful for clinical use14 and scores a sample as either low riskor high risk for development of distant metastasis. The optimal threshold forthe classifier index score was selected to reach optimal sensitivity and specific-ity in the training set. If a sample’s index exceeded the set threshold, it wasclassified as a high-risk sample; if the index of the sample was below thethreshold, the sample was classified as a low-risk sample.

Statistical Analysis

For the analysis of the training set, we defined the probability that pa-tients remain free of distant metastases as the first event. For the analysis of thevalidation set, the primary end point was relapse-free survival (RFS), whichwas defined as the probability that patients remain free of recurrence (locore-gional or metastatic) as the first event; data on all other patients were censoredon the date of the last follow-up visit or date of death. Deaths of no specificcause were censored to evaluate true prognostic prediction. Data were ana-lyzed from the date of surgery to the time of the first event or the date on whichdata were censored, according to the Kaplan-Meier method, and the curveswere compared with use of the log-rank test. To increase the number of events,the training set and validation set were combined to analyze the prognostic

Table 1. Patient Demographics and Clinical Characteristics for theTraining and Validation Sets

Demographic or ClinicalCharacteristic

Training Set(n � 188)

Validation Set(n � 206)

No. ofPatients %

No. ofPatients %

HospitalLUMC 76 40.4NKI 52 27.7Slotervaart 48 25.5Other 12 6.4ICO Barcelona 206 100.0

Median age, years 67.9 69Median follow-up, months 65.1 54.1Sex

Male 84 44.7 132 64.1Female 104 55.3 74 35.9

LocalizationLeft 92 50.3 115 55.8Right 74 40.4 67 32.5Rectum 17 9.3 24 11.7

StageI (T2 only) 24 12.8 30 14.6II 100 53.2 114 55.3III 56 29.8 62 30.1IV 8 4.2 — —

Grade1 11 5.8 90 43.72 141 75.0 100 48.53 30 16.0 16 7.8NA 6 3.2 — —

Distant metastasisNo 137 72.9 173 84.0Yes 51 27.1 33 16.0

ChemotherapyNo 148 78.7 125 60.7Yes 36 19.1 77 37.4Unknown 4 2.1 4 1.9

Abbreviations: LUMC, Leiden University Medical Center; NKI, NetherlandsCancer Institute; ICO, Institut Catala d’Oncologia; NA, not available.

Salazar et al

2 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
Page 3: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

information of KRAS, BRAF, and PI3KCA mutation in univariate analysis.MSI status was analyzed in a subset of patients from the training set (n � 90)and in all patients from the validation set. To determine the independence ofour classifier to clinicopathologic variables in predicting an individual’s risk ofexperiencing relapse, we analyzed the validation set using univariate analysisfollowed by multivariate analysis. Sex; localization of the tumor; T stage; Nstage; number of lymph nodes assessed; histologic grade; lymphatic, vascular,and perineural invasion; adjuvant chemotherapy administration; MSI status;and gene expression profile were included as variables in this analysis. Log-rank tests were used in the univariate analysis, and a multivariate Cox modelwas built including the significant variables from univariate analysis on allpatients in the validation set and on subsets of patients with stage II and IIIdisease only. In addition, multivariate analysis with American Society of Clin-ical Oncology (ASCO) clinical risk criteria (defined as T stage of 4, poor gradetumor, � 13 assessed lymph nodes, or emergency presentation with obstruc-tion or perforation5) and ColoPrint as independent variables was performedin patients with stage II disease. All calculations were performed with SPSSstatistical package version 16.0 (SPSS, Chicago, IL).

RESULTS

Profile Development

Unsupervised hierarchical clustering of the 188 tumor tissues ofthe training set revealed three main molecular subtypes. Patients withsubtype A showed a good outcome, whereas patients with subtype Chad a relative poor outcome (84% and 58% 5-year disease-free sur-vival, respectively; hazard ratio [HR], 1.8; P � .015). Most patients(110 of 188 patients) fell into the intermediate prognosis cluster,subtype B (Fig 1). Further investigation of these subtypes indicatedthat both survival-associated subtypes, A and C, were enriched forpatients with an activating BRAF V600E mutation. In the subtype Agroup, 52% of patients had BRAF mutations, and in the poor-outcome subtype C group, 22% of patients had BRAF mutations;whereas in the subtype B group, 4% of patients were mutation carriers.Subtype A was enriched for patients with MSI (MSI-H). Fifteen of 90patients in the training set with known MSI status were MSI-H. Thir-teen of these 15 patients belonged to subtype A. The molecular sub-types had no correlation with stage (data not shown).

Only subtype B (n � 110) was used to develop a prognosticsignature to avoid building a classifier that was mainly based on theextreme expression patterns of subtypes A and C. An optimal set of 18genes was identified and used to construct the ColoPrint prognostic

classifier (see Patients and Methods). The classifier was applied to allsamples in the training set (n � 188) using a leave-one-out cross-validation procedure (Appendix Fig A1, online only). Five-year DMFSrates were 82% (95% CI, 76% to 89%) and 50% (95% CI, 38% to66%) for patients with a low-risk and high-risk signature, respectively(HR, 3.41; 95% CI, 1.95 to 5.91; P � .001). Among the 29 patients whoreceived adjuvant chemotherapy, 14 (48.3%) were classified as Colo-Print low risk, and 15 (51.7%) were classified as high risk.

Independent Validation

An independent patient cohort of 206 patients was used to eval-uate the performance of the colorectal prognosis classifier (Table 2).Most patients in the validation set were patients with stage II disease(n � 114). In the validation set of all patients, 60% of patients wereidentified as low risk, whereas 40% of patients were high risk (Fig 2).Low-risk patients had a 5-year RFS rate of 87.6% (95% CI, 81.5% to93.7%), whereas high-risk patients had a 5-year RFS rate of only 67.2%(95% CI, 55.4% to 79%). Male patients and patients with colon canceron the left side were more often classified as high risk than femalepatients and patients with cancer in the right colon. High risk was alsopositively associated with relapse, time to relapse, and time to death.Classification as ColoPrint low or high risk was not associated withgrade, age, stage, or number of assessed lymph nodes (Table 2).

When the classifier was applied to patients with stage II and stageIII disease separately, it correctly classified low- and high-risk patientsin both groups (Fig 2). In the analysis of patients with stage II disease,63.2% were classified as low risk, and 36.8% were classified as highrisk, with 5-year RFS rates of 90.9% (95% CI, 84% to 97.8%) and73.9% (95% CI, 59.2% to 88.6%), respectively (P � .017).

Comparison to Clinical Factors, Mutational Analysis,

and MSI

In the combined training and validation set, in patients withknown mutation status (n � 381), KRAS mutations were detected in115 patients (30.2%), PI3KCA mutations were detected in 45 patients(11.8%), and BRAF mutations were detected in 42 patients (11%). Inour data set, the mutations, either alone or in combination, were notpredictive for relapse or overall survival (data not shown). MSI statuswas known for 276 patients (90 patients in the training set, 186 patientsin the validation set), of whom 29 were classified as MSI-H. Patients

1.0

0.8

0.6

0.4

0.2

0

52% BRAFmut 4% BRAFmut 22% BRAFmut

20 40 60 80 100 120Dist

ant M

etas

tasi

s–Fr

ee S

urvi

val

(pro

porti

on)

Time (months)

BRAF

HR 1.8P = .015

ABC

A BC

Fig 1. Unsupervised clustering of 188 tumor samples (training set) revealed three molecular subtypes (A, B, and C). BRAF mutation status was known for 179 patientsand is associated with the subtypes.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 3

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 4: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Table 2. Association of Clinicopathologic Variables With Assessment of Colon Low- and High-Risk Signature in the Validation Set

Variable

Total (N � 206)

ColoPrint

P

Low Risk (n � 123) High Risk (n � 83)

No. of Patients % No. of Patients % No. of Patients %

Age, yearsMedian 69 69 69.45 .676� 70 113 54.9 68 55.3 45 54.2 .880� 70 93 45.1 55 44.7 38 45.8

Localization .000Left 115 55.8 57 46.3 58 69.9Right 67 32.5 54 43.9 13 15.7Rectum 24 11.7 12 9.8 12 14.5

Grade .3861 90 43.7 51 41.5 39 47.02 100 48.5 60 48.8 40 48.23 16 7.8 12 9.8 4 4.8

Sex .085Male 132 64.1 73 59.3 59 71.1Female 74 35.9 50 40.7 24 28.9

No. of LNs assessedMedian 17.5 19.0 15.0 .093� 12 55 26.7 29 23.6 26 31.3 .218� 12 151 73.3 94 76.4 57 68.7

Stage .405I 30 14.6 15 12.2 15 18.1II 114 55.3 72 58.5 42 50.6III 62 30.1 36 29.3 26 31.3

pT .4712 33 16.0 17 13.8 16 19.33 157 76.2 95 77.2 62 74.74 16 7.8 11 8.9 5 6.0

pN .3340 144 69.9 87 70.7 57 68.71 42 20.4 27 22.0 15 18.12 20 9.7 9 7.3 11 13.3

DM .027No 173 84.0 109 88.6 64 77.1Yes 33 16.0 14 11.4 19 22.9

Median time to DM, months 51.8 60 38.7 .001Relapse (local, regional, distant) .013

No 166 80.6 106 86.2 60 72.3Yes 40 19.4 17 13.8 23 27.7

Median time to relapse, months 51.8 60 38.7 .001Death .846

No 175 85.0 104 84.6 71 85.5Yes 31 15.0 19 15.4 12 14.5

Median survival time, months 54.1 60.2 49.5 .029Chemotherapy .971

No 125 61.9 75 62 50 61.7Yes 77 38.1 46 38 31 38.3

MSI-high .039No 172 83.5 99 80.0 73 88.0Yes 14 6.8 12 9.9 2 2.4NA 20 9.7 12 9.9 8 9.6

Lymphatic invasion .846No 175 85.0 104 84.6 71 85.5Yes 31 15.0 19 15.4 12 14.5

Venous invasion .718No 192 93.2 114 92.7 78 94.0Yes 14 6.8 9 7.3 5 6.0

(continued on following page)

Salazar et al

4 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 5: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

with MSI-H were mainly patients with stage II disease (21 of 29patients; 72%) and had a high frequency of BRAF mutation (15 of 29patients; 52%). These patients were also mostly classified as ColoPrintlow risk (26 of 29 patients; 90%), indicating that the good prognosis ofthe MSI-H patients is identified by the gene classifier. This is alsoverified in analysis of the validation set only (Table 2).

For comparison of performance of ColoPrint and clinical factors,only results from the validation set were used (Table 2). ColoPrint wasthe strongest predictor of RFS in the univariate analysis, where onlystage, T stage, and lymph node status showed a similar magnitude ofstatistical significance. Among the 77 patients who received adjuvantchemotherapy, 46 (59.7%) were classified as ColoPrint low risk, and31 (40.3%) were classified as ColoPrint high risk; chemotherapy ad-ministration was not a significant prognostic factor for RFS in thisseries (Table 3 and Appendix Table A3, online only). In the multivar-iate analysis of all samples and of samples from patients with stage IIIdisease only, ColoPrint remained a strong independent prognosticfactor (Table 4 and Appendix Table A4, online only). Analysis ofDMFS yielded similar results (Appendix Tables A5 and A6, on-line only).

In the subset of patients with stage II disease (n� 114), ColoPrintwas the strongest predictor for RFS in the univariate analysis (HR,3.34; 95% CI, 1.24 to 9.00; P � .017) and multivariate analysis (Tables3 and 4). The analysis of the relative performance of the gene classifierwith conventional clinicopathologic factors revealed that T stage wasalso associated with prognosis (HR, 3.15; 95% CI, 1.02 to 9.69;P � .045). In addition, the classifier performed independently fromthe ASCO risk criteria when they were analyzed either individually(Table 3) or combined (HR, 3.66; 95% CI, 1.24 to 9.08; P � .017;Appendix Table A7, online only). Interestingly, a high degree of dis-cordance (48.2%) in risk stratification between ColoPrint and ASCOcriteria was observed (Appendix Table A8, online only). Finally, in thesubgroup of patients with stage II disease, among 40 patients (36%)who received adjuvant chemotherapy, 28 patients (68%) were classi-fied as ColoPrint low risk, and 12 patients (32%) were classified as highrisk, and chemotherapy administration was not a significant prognos-tic factor for RFS (P � .34) or overall survival.

Additional In Silico Validation and Functional Analysis

To further explore the clinical and biologic relevance of Colo-Print, an additional in silico validation and functional analysis of theset of genes included was performed. Gene expression data of 322 stage

I to III colorectal tumor samples from three previously publishedstudies18,20,25 were available for in silico validation of the gene classi-fier. In the first data set of 100 patients (Gene Expression Omnibusaccession GSE5206),25 ColoPrint risk scores were significantly associ-ated with development of disease recurrence (Wilcoxon P � .0092),with an area under the receiver operating curve of 0.68 (data notshown). This data set was combined with two additional data sets(GSE10402 and ArrayExpress accession MEXP-1245),18,20 yielding atotal of 322 colorectal tumors. ColoPrint risk outcome was signifi-cantly associated with RFS (P � .001, McNemar test), with an oddsratio of 2.8 (95% CI, 1.6 to 4.7). In the analysis of all stages, ColoPrintlow-risk samples (n � 177, 55%) showed a 5-year RFS rate of 83.8%(95% CI, 79.3% to 87.5%) compared with a 5-year RFS of 64.8% (95%CI, 60.1% to 70.0%) for ColoPrint high-risk samples (n � 145).

Genes in the classifier were selected in an agnostic, data-drivenway. Nevertheless, some of the selected genes have been shown to playa role in colon cancer biology (Appendix Table A2), coding for serine/threonine protein kinases, transcription factors, proteases, and mem-brane components. The gene ontology analysis (Babelomics software;http://babelomics.bioinfo.cipf.es/) revealed that the selected genes areinvolved in cell proliferation, transforming growth factor � pathway,immune response, and metabolism. One of the genes is LAM3(laminin-322), whose abnormal expression, in addition to its integrinreceptors, is believed to promote invasion of colon, breast, and skincancer cells. Moreover, LAM3 and its protease degradation productsmay induce and/or promote tumor cell migration.26 Another gene,CTSC (cathepsin C), has also been shown to be involved in invasion.

DISCUSSION

In this study, we present the development and validation of agene expression signature that is associated with the risk of re-lapse in patients with stage II or III CRC. ColoPrint identifiestwo thirds of patients with stage II colon cancer who are at suffi-ciently low risk of recurrence who may be safely managed withoutadjuvant chemotherapy.

The unsupervised hierarchical clustering in three prognostic sub-types supports the underlying hypothesis that the transcripts of theprimary tumors yield prognostic information. Of note, the molecularcharacteristics and percentage of patients in these three subtypes are

Table 2. Association of Clinicopathologic Variables With Assessment of Colon Low- and High-Risk Signature in the Validation Set (continued)

Variable

Total (N � 206)

ColoPrint

P

Low Risk (n � 123) High Risk (n � 83)

No. of Patients % No. of Patients % No. of Patients %

Perineural invasion .566No 203 98.5 122 99.2 81 97.6Yes 3 1.5 1 0.8 2 2.4

Lymphatic, venous, or perineural invasion .925No 162 78.6 97 78.9 65 78.3Yes 44 21.4 26 21.1 18 21.7

NOTE. No patient had obstruction/perforation.Abbreviations: LN, lymph node; DM, distant metastasis; MSI, microsatellite instability; NA, not available.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 5

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
W.Wifler
Highlight
W.Wifler
Highlight
W.Wifler
Highlight
Page 6: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

reminiscent of the molecular CpG island methylation phenotype sub-types that are characterized by MSI, BRAF mutation, and methyl-ation status.27

On the basis of gene expression information in the primarytumor, ColoPrint can assist in more accurately identifying the 25% to35% of patients diagnosed with stage II disease who will experience arecurrence within 5 years after surgery. Our prognostic classifier iden-tified 36.8% of the validation stage II subset as high-risk patients with

A

B

C

0

Log -rank P = .003

Prob

abili

ty o

f Rel

apse

-Fre

e Su

rviv

al

Time to Relapse (months)

All stages1.0

0.8

0.6

0.4

0.2

25 50 75 100 125

Low riskHigh risk

0

Log -rank P = .012

Prob

abili

ty o

f Rel

apse

-Fre

e Su

rviv

al

Time to Relapse (months)

Stage II1.0

0.8

0.6

0.4

0.2

25 50 75 100 125

Low riskHigh risk

0

Log -rank P = .099

Prob

abili

ty o

f Rel

apse

-Fre

e Su

rviv

al

Time to Relapse (months)

Stage III1.0

0.8

0.6

0.4

0.2

25 50 75 100 125

Low riskHigh risk

Fig 2. Kaplan-Meier analysis of relapse-free survival (RFS) in the validationset. (A) All stages, n � 206; 5-year RFS rate for low-risk patients was 87.6%(95% CI, 81.5% to 93.7%) and for high-risk patients was 67.2% (95% CI,55.4% to 79.0%). (B) Stage II, n � 114; 5-year RFS rate for low-risk patientswas 90.9% (95% CI, 84.0% to 97.8%) and for high-risk patients was 73.9%(95% CI, 59.2% to 88.6%). (C) Stage III, n � 62; 5-year RFS rate for low-riskpatients was 78.2% (95% CI, 49.9% to 90.7%) and for high-risk patients was47.2% (95% CI, 25.8% to 68.6%).

Table 3. Univariate Analysis for Relapse-Free Survival in Validation Set

Variable P HR 95% CI

All stages, N � 206ColoPrint, high v low risk .005 2.51 1.33 to 4.73Age, � v � 70 years .071 1.78 0.95 to 3.33Localization, right v left .576 0.82 0.43 to 0.16Grade

Baseline .149 1Moderate v low 0.89 0.46 to 1.76High v low 2 0.82 to 5.72

Sex, male v female .739 1.12 0.58 to 2.14No. of LNs assessed, continuous .036 0.50 0.26 to 0.96� 12 LNs assessed, binary .036 0.50 0.26 to 0.96Stage

I v II .004 0.21 0.03 to 1.59Baseline � II 1III v II 2.36 1.25 to 4.47

pTBaseline � T2 .006 1T3 v T2 2.08 0.64 to 0.68T4 v T2 6.74 1.74 to 26.11

pT, continuous .003 2.8 1.41 to 5.54pN

Baseline .000 11-3 positive LNs v no positive LNs 1.88 0.88 to 4.01� 3 positive LNs v no positive LNs 5.73 2.69 to 12.21

Chemotherapy, yes v no .414 0.77 0.40 to 1.46MSI-H, yes v no .830 1.07 0.59 to 1.92Lymphatic invasion, yes v no .100 1.87 0.89 to 3.93Venous invasion, yes v no .101 2.20 0.86 to 5.62Perineural invasion, yes v no .651 1.58 0.22 to 11.54Any invasion, yes v no .051 1.93 1.00 to 3.76

Stage II only, n � 114ColoPrint, high v low risk .017 3.34 1.24 to 9.00Age, � v � 70 years .187 0.47 0.15 to 1.44Localization, right v left .73 0.82 0.15 to 2.46Grade

Baseline .515Moderate v low 0.66 0.23 to 1.91High v low 2.15 0.27 to 16.87

Sex, male v female .175 2.17 0.71 to 6.67No. of LNs assessed, continuous .553 0.98 0.94 to 1.04� 12 LNs assessed, binary .776 0.86 0.30 to 2.44pT, T4 v T3 .045 3.15 1.02 to 9.69ASCO risk, high v low .200 1.67 0.22 to 12.59Chemotherapy, yes v no .339 0.60 0.21 to 1.71MSI-H, yes v no .619 0.77 0.28 to 2.13Lymphatic invasion, yes v no .689 1.51 0.20 to 11.50Venous invasion, yes v no .496 2.02 0.27 to 15.41Perineural invasion, yes v no .237 3.41 0.45 to 26.03Any invasion, yes v no .209 2.23 0.64 to 7.80

Abbreviations: HR, hazard ratio; LN, lymph node; MSI-H, microsatelliteinstability-high; ASCO, American Society of Clinical Oncology.

Salazar et al

6 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
Page 7: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

higher accuracy than the recommended clinical risk factors, irrespec-tive of chemotherapy administration. Approximately two thirds of allpatients analyzed received a low-risk classification, 91% of whom didnot experience relapse. Chemotherapy was administered to 36% ofthese patients, where it was evenly distributed between ColoPrinthigh- and low-risk groups and had no influence in the global prognos-tic statistical analysis.

The suitability of gene expression profiles to identify high-riskpatients with CRC has been proven in several independentstudies.15-21 Similar to what has been observed in the breast cancerfield, these studies led to the construction of different gene signaturesthat may be secondary to differences in patient cohorts, technologicplatforms, and data mining strategies.12,28 Although signatures canoften be validated in silico, the signature presented in this study is thefirst prognostic CRC profile, to our knowledge, that has been validatedin independent patient series, using the same technology, gene set, andanalytic approach.

The ColoPrint signature adds value to more conventional prog-nostic clinicopathologic factors. Routine standardization of genomicassessment, which includes tissue handling and processing, RNA ex-traction techniques, and the hybridization process, is a critical issue ifit is to be used in the clinical setting.29 Much progress has been made toestablish high-quality standards for this new technology.14 Of note, itis often overlooked that more conventional parameters, such as vas-cular invasion or a precise cutoff number of analyzed lymph nodes,may not be consistently recorded or evaluated in a significant propor-tion of tumors,30 and a comparable standardization for clinical factorsshould be pursued.

Single molecular markers, such as loss of heterozygosity in 18q,MSI, thymidylate synthase expression, p53 or p21 expression, orKRAS or BRAF mutations, provide an additional means of character-izing individual tumors but are not routinely recommended for prog-nostic characterization.31 MSI is the most extensively investigated andvalidated of these markers. A published meta-analysis showed thatMSI is an independent prognostic predictor of improved survival and

time to recurrence.9 Data coming from the translational studies of theQuick and Simple and Reliable (QUASAR) trial and Pan-EuropeanTrials in Alimentary Tract Cancers (PETACC-3) have confirmed thisobservation in stage II disease.10,11 However, this prognostic factoridentifies only a small subgroup of low-risk patients.23 In our valida-tion data set, 8% of patients with known MSI status were MSI-H, andmost of the patients (86%) were also identified as low risk by theColoPrint classifier.

In this study, the prognostic classifier was validated in anindependent patient set collected from a different country, withfurther international validation studies currently underway. Addi-tionally, for use in routine clinical practice, the prognostic classifierwas translated into a robust and standardized assay with stringentquality controls following guidelines of the National Committeeon Clinical Laboratory Standards. Using the classifier in a clinicalsetting will provide more accurate information on the risk ofrecurrence compared with the use of conventional clinicopatho-logic criteria alone and can facilitate the selection of low-risk pa-tients who can be spared chemotherapy.

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTSOF INTEREST

Although all authors completed the disclosure declaration, the followingauthor(s) indicated a financial or other interest that is relevant to the subjectmatter under consideration in this article. Certain relationships markedwith a “U” are those for which no compensation was received; thoserelationships marked with a “C” were compensated. For a detaileddescription of the disclosure categories, or for more information aboutASCO’s conflict of interest policy, please refer to the Author DisclosureDeclaration and the Disclosures of Potential Conflicts of Interest section inInformation for Contributors.Employment or Leadership Position: Paul Roepman, Agendia (C); IrisSimon, Agendia (C); Christa Dreezen, Agendia (C); Annuska M. Glas,Agendia (C); Laura J. Van’t Veer, Agendia (C) Consultant or AdvisoryRole: David Kerr, Agendia (C) Stock Ownership: Paul Roepman,Agendia; Iris Simon, Agendia; Annuska M. Glas, Agendia; Laura J. Van’tVeer, Agendia Honoraria: None Research Funding: None ExpertTestimony: None Other Remuneration: None

AUTHOR CONTRIBUTIONS

Conception and design: Ramon Salazar, Paul Roepman, Gabriel Capella,Victor Moreno, Iris Simon, David Kerr, Laura J. Van’t Veer,Rob TollenaarFinancial support: Gabriel Capella, Victor Moreno, Laura J. Van’t Veer,Rob TollenaarAdministrative support: Iris Simon, Christa Dreezen, AdrianaLopez-Doriga, Sjoerd BruinProvision of study materials or patients: Ramon Salazar, GabrielCapella, Victor Moreno, Cristina Santos, Corrie Marijnen, JohanWesterga, Sjoerd Bruin, Peter Kuppen, Cornelis van de Velde, HansMorreau, Loes Van Velthuysen, Rob TollenaarCollection and assembly of data: Ramon Salazar, Paul Roepman,Gabriel Capella, Iris Simon, Adriana Lopez-Doriga, Cristina Santos,Corrie Marijnen, Johan Westerga, Sjoerd Bruin, Peter Kuppen, Cornelisvan de Velde, Hans Morreau, Loes Van VelthuysenData analysis and interpretation: Ramon Salazar, Paul Roepman, VictorMoreno, Iris Simon, Christa Dreezen, Corrie Marijnen, Annuska M.Glas, Laura J. Van’t Veer, Rob Tollenaar

Table 4. Multivariate Analysis for Relapse-Free Survival in Validation Set

Variable P HR 95% CI

All stages, N � 206ColoPrint, high v low .003 2.69 1.41 to 5.14pT

T2 .000T3 v T2 .038 0.19 0.04 to 0.91T4 v T2 .960 1.05 0.19 to 5.88

Stage, continuous .021 0.05 0.00 to 0.063pN

No positive LNs .0001-3 positive LNs v no positive LNs .327 1.52 0.66 to 3.52� 3 positive LNs v no positive LNs .000 5.97 2.62 to 13.63

No. of LNs assessed, continuous .059Lymphatic, venous, or perineural invasion, any .491

Stage II only, n � 114ColoPrint, high v low .018 3.29 1.24 to 8.83pT, T4 v T3 .051 3.06 0.99 to 9.44

NOTE. Multivariate analysis includes only variables that were significant(P � .05) in the univariate analysis.

Abbreviations: HR, hazard ratio; LN, lymph node.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 7

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
W.Wifler
Highlight
W.Wifler
Highlight
Page 8: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Manuscript writing: Ramon Salazar, Iris Simon, Laura J. Van’t Veer,Rob TollenaarFinal approval of manuscript: Ramon Salazar, Paul Roepman, GabrielCapella, Victor Moreno, Iris Simon, Christa Dreezen, Adriana

Lopez-Doriga, Cristina Santos, Corrie Marijnen, Johan Westerga, SjoerdBruin, David Kerr, Peter Kuppen, Cornelis van de Velde, Hans Morreau,Loes Van Velthuysen, Annuska M. Glas, Laura J. Van’t Veer,Rob Tollenaar

REFERENCES

1. O’Connell JB, Maggard MA, Ko CY: Coloncancer survival rates with the new American JointCommittee on Cancer sixth edition staging. J NatlCancer Inst 96:1420-1425, 2004

2. Quasar Collaborative Group, Gray R, BarnwellJ, et al: Adjuvant chemotherapy versus observationin patients with colorectal cancer: A randomisedstudy. Lancet 370:2020-2029, 2007

3. Sobrero A: Should adjuvant chemotherapybecome standard treatment for patients with stageII colon cancer? For the proposal. Lancet Oncol7:515-516, 2006

4. Kohne CH: Should adjuvant chemotherapybecome standard treatment for patients with stageII colon cancer? Against the proposal. Lancet Oncol7:516-517, 2006

5. Benson AB 3rd, Schrag D, Somerfield MR, etal: American Society of Clinical Oncology recom-mendations on adjuvant chemotherapy for stage IIcolon cancer. J Clin Oncol 22:3408-3419, 2004

6. Van Cutsem EJ, Oliveira J, Kataja VV: ESMOMinimum Clinical Recommendations for diagnosis,treatment and follow-up of advanced colorectal can-cer. Ann Oncol 16:i18-i19, 2005 (suppl 1)

7. Gill S, Loprinzi CL, Sargent DJ, et al: Pooledanalysis of fluorouracil-based adjuvant therapy forstage II and III colon cancer: Who benefits and byhow much? J Clin Oncol 22:1797-1806, 2004

8. Le Voyer TE, Sigurdson ER, Hanlon AL, et al:Colon cancer survival is associated with increasingnumber of lymph nodes analyzed: A secondarysurvey of intergroup trial INT-0089. J Clin Oncol21:2912-2919, 2003

9. Popat S, Hubner R, Houlston RS: Systematicreview of microsatellite instability and colorectalcancer prognosis. J Clin Oncol 23:609-618, 2005

10. Kerr D, Gray R, Quirke P, et al: A quantitativemultigene RT-PCR assay for prediction of recur-rence in stage II colon cancer: Selection of thegenes in four large studies and results of the inde-pendent, prospectively designed QUASAR valida-

tion study. J Clin Oncol 27:169s, 2009 (suppl; abstr4000)

11. Roth AD, Tejpar S, Yan P, et al: Stage-specificprognostic value of molecular markers in colon can-cer: Results of the translational study on thePETACC 3-EORTC 40993-SAKK 60-00 trial. J ClinOncol 27:169s, 2009 (suppl; abstr 4002)

12. Quackenbush J: Microarray analysis and tu-mor classification. N Engl J Med 354:2463-2472,2006

13. van’t Veer LJ, Dai H, van de Vijver MJ, et al:Gene expression profiling predicts clinical outcomeof breast cancer. Nature 415:530-536, 2002

14. Glas AM, Floore A, Delahaye LJ, et al: Con-verting a breast cancer microarray signature into ahigh-throughput diagnostic test. BMC Genomics7:278, 2006

15. Barrier A, Boelle PY, Roser F, et al: Stage IIcolon cancer prognosis prediction by tumor geneexpression profiling. J Clin Oncol 24:4685-4691,2006

16. Bertucci F, Salas S, Eysteries S, et al: Geneexpression profiling of colon cancer by DNA microar-rays and correlation with histoclinical parameters.Oncogene 23:1377-1391, 2004

17. Eschrich S, Yang I, Bloom G, et al: Molecularstaging for survival prediction of colorectal cancerpatients. J Clin Oncol 23:3526-3535, 2005

18. Garman KS, Acharya CR, Edelman E, et al: Agenomic approach to colon cancer risk stratificationyields biologic insights into therapeutic opportuni-ties. Proc Natl Acad Sci U S A 105:19432-19437,2008

19. Jiang Y, Casey G, Lavery IC, et al: Develop-ment of a clinically feasible molecular assay topredict recurrence of stage II colon cancer. J MolDiagn 10:346-354, 2008

20. Lin YH, Friederichs J, Black MA, et al: Multiplegene expression classifiers from different array plat-forms predict poor prognosis of colorectal cancer.Clin Cancer Res 13:498-507, 2007

21. Wang Y, Jatkoe T, Zhang Y, et al: Geneexpression profiles and molecular markers to predict

recurrence of Dukes’ B colon cancer. J Clin Oncol22:1564-1571, 2004

22. Barrier A, Roser F, Boelle PY, et al: Prognosisof stage II colon cancer by non-neoplastic mucosagene expression profiling. Oncogene 26:2642-2648,2007

23. Gonzalez-García I, Moreno V, Navarro M, et al:Standardized approach for microsatellite instabilitydetection in colorectal carcinomas. J Natl CancerInst 92:544-549, 2000

24. Michiels S, Koscielny S, Hill C: Prediction ofcancer outcome with microarrays: a multiple ran-dom validation strategy. Lancet 365:488-492, 2005

25. Kaiser S, Park YK, Franklin JL, et al: Transcrip-tional recapitulation and subversion of embryoniccolon development by mouse colon tumor modelsand human colon cancer. Genome Biol 8:R131,2007

26. Tsuruta D, Kobayashi H, Imanishi H, et al:Laminin-332-integrin interaction: A target for cancertherapy? Curr Med Chem 15:1968-1975, 2008

27. Poynter JN, Siegmund KD, Weisenberger DJ,et al: Molecular characterization of MSI-H colorectalcancer by MLHI promoter methylation, immunohis-tochemistry, and mismatch repair germline muta-tion screening. Cancer Epidemiol Biomarkers Prev17:3208-3215, 2008

28. Ein-Dor L, Kela I, Getz G, et al: Outcomesignature genes in breast cancer: Is there a uniqueset? Bioinformatics 21:171-178, 2005

29. Cascinu S, Zaniboni A, Scartozzi M, et al:Molecular biology for stage II colorectal cancer: Thejury is still out. J Clin Oncol 25:2861; author reply2862-2863, 2007

30. Morris EJ, Maughan NJ, Forman D, et al: Whoto treat with adjuvant therapy in Dukes B/stage IIcolorectal cancer? The need for high quality pathol-ogy. Gut 56:1419-1425, 2007

31. Locker GY, Hamilton S, Harris J, et al: ASCO2006 update of recommendations for the use oftumor markers in gastrointestinal cancer. J ClinOncol 24:5313-5327, 2006

■ ■ ■

Salazar et al

8 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

W.Wifler
Highlight
W.Wifler
Highlight
Page 9: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Appendix

Table A1. Patient Demographics and Clinical Characteristics of Training Set

Demographic or Clinical Characteristic

Training Set (n � 188) Training Set, Group B (n � 110)

No. of Patients % No. of Patients %

HospitalLUMC 40.4 30.9NKI 27.7 36.4Slotervaart 25.5 24.5Other 6.4 8.2

Median age, years 67.9 68Median follow-up, months 65.1 63.3Sex

Male 84 44.7 55 50Female 104 55.3 55 50

Localization 92 48.9 64 58.2Left 74 39.4 33 30.0Right 17 9.0 9 8.2Rectum 5 2.7 4 3.6

StageI (T2 only) 24 12.8 16 14.5II 100 53.2 57 51.8III 56 29.8 31 28.2IV 8 4.2 6 5.5

Grade1 11 5.8 7 6.42 141 75.0 90 81.83 30 16.0 10 9.1NA 6 3.2 3 2.7

Distant metastasisNo 137 72.9 79 71.8Yes 51 27.1 31 28.2

ChemotherapyNo 148 78.7 86 78.2Yes 36 19.1 21 19.1Unknown 4 2.1 3 2.7

Abbreviations: LUMC, Leiden University Medical Center; NKI, Netherlands Cancer Institute; NA, not available.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 9

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 10: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Table A2. Gene Function of 18-Gene Classifier

Gene Full Gene Name Function

MCTP1 Multiple C2 domains, transmembrane 1 Ca2�-binding transmembrane proteinLAMA3 Laminin, alpha 3 (laminin 5 � now laminin 332) Heterotrimeric epithelial-derived basement membrane component;

abnormal expression of laminin-332 and its integrin receptors isbelieved to promote invasion of colon, breast, and skin cancercells and likely induce and/or promote tumor cell migration

CTSC Cathepsin C A lysosomal cysteine proteinase that appears to be a centralcoordinator for activation of many serine proteinases in immune/inflammatory cells and plays a role in invasion

PYROX D1 Pyridine nucleotide-disulphide oxidoreductase domain 1 UnknownEDEM1 ER degradation enhancer, mannosidase alpha-like 1 Endoplasmatic reticulum protein involved in folding and maturationIL2RB Interleukin-2 receptor, beta Involved in T-cell–mediated immune responses; IL-2 enhances

susceptibility of colon cancer cells to FasR-mediated apoptosis byupregulating Fas receptor levels and by downregulating FAP-1expression

ZNF697 Zinc finger protein 697 UnknownSLC6A11 Solute carrier family 6 (neurotransmitter transporter,

GABA), member 11GABA is a major inhibitory neurotransmitter; GABAergic

neurotransmission is terminated by the uptake of GABA into thepresynaptic terminal and the surrounding astroglial cells bysodium-dependent transporters, such as SLC6A11

IL2RA Interleukin-2 receptor, alpha The IL-2 receptor � (IL2RA) and � (IL2RB) chains, together with thecommon � chain (IL2RG), constitute the high-affinity IL-2 receptor

CYFIP2 Cytoplasmic FMR1 interacting protein 2 The CYFIP2 promoter contains a p53-responsive element thatconfers p53 binding as well as transcriptional activation of aheterologous reporter

PIM3 Pim-3 oncogene PIM3 belongs to a family of proto-oncogenes that encodeserine/threonine protein kinases

LIF Leukemia inhibitory factor(cholinergic differentiation factor)

The protein encoded by this gene is a pleiotropic cytokine involved inthe induction of hematopoietic differentiation in normal andmyeloid leukemia cells, induction of neuronal cell differentiation,and regulation of mesenchymal to epithelial conversion duringkidney development

PLIN3 Mannose-6-phosphate receptor binding protein 1 Mannose-6-phophate receptors deliver lysosomal hydrolase from theGolgi complex to endosomes and then return to the Golgicomplex; this protein also binds directly to the GTPase RAB9(RAB9A), a member of the RAS oncogene family; also called TIP47

HSD3B1 Hydroxy-delta-5-steroid dehydrogenase, 3 beta- andsteroid delta-isomerase 1

Steroid metabolism: 3-�-hydroxysteroid dehydrogenase 1 (HSD3B1)is an enzyme that inactivates dihydrotestosterone in the prostateand has been shown to be involved in prostate cancer

ZBED4 Zinc finger, BED-type containing 4 ZBED4 has four predicted DNA binding domains, a dimerizationdomain, and two LXXLL motifs characteristic of co-activators/co-repressors of nuclear hormone receptors

PPARA Peroxisome proliferator-activated receptor alpha Peroxisome proliferators include hypolipidemic drugs, herbicides,leukotriene antagonists, and plasticizers and induce an increase inthe size and number of peroxisomes. The action of peroxisomeproliferators is thought to be mediated via specific receptors,called PPARs, which belong to the steroid hormone receptorsuperfamily. PPARs affect the expression of target genes involvedin cell proliferation, cell differentiation, and immune andinflammation responses. This gene encodes the subtype PPAR-�,which is a nuclear transcription factor

THNSL2 Threonine synthase-like 2 (Saccharomyces cerevisiae) Metabolic processCA4388O2 Unknown

Abbreviations: ER, endoplasmic reticulum; IL-2, interleukin-2; FAP-1, fas-associated phosphatase-1; GABA, �-aminobutyric acid; PPAR, peroxisome proliferator-activated receptor.

Salazar et al

10 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 11: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Table A3. Univariate Analysis for Relapse-Free Survival in Patients With Stage III Disease Only (n � 62)

Variable P HR 95% CI

ColoPrint, high v low .106 2.09 0.86 to 4.78Age, continuous .002 1.08 1.03 to 1.12Age � 70 years .002 4.79 1.81 to 12.71Localization, right v left .952 0.97 0.40 to 2.39Grade

Baseline � moderate .863 1Moderate v low 0.08 0.38 to 3.11High v low 1.38 0.39 to 4.83

Sex, female v male .481 0.74 0.37 to 1.72No. of LNs assessed, continuous .185 0.96 0.91 to 1.02pT, continuous .290 0.23 0.51 to 9.82pT

T2 .015 1T3 v T2 0.38 0.09 to 1.67T4 v T2 2.21 0.36 to 13.53

pN, continuous .013 2.90 1.25 to 6.75Chemotherapy, yes v no .015 0.35 0.15 to 0.82MSI-H, yes v no .361 1.41 0.68 to 2.92Lymphatic invasion .943 1.03 0.43 to 2.47Venous invasion .615 1.32 0.45 to 3.91Perineural invasion� — — —Lymphatic or venous invasion .959 0.98 0.42 to 2.27

Abbreviations: HR, hazard ratio; LN, lymph node; MSI-H, microsatellite instability-high.�No events occurred.

Table A4. Multivariate Analysis for Relapse-Free Survival in Patients With Stage III Disease Only (n � 62)

Variable P HR 95% CI

ColoPrint, high v low .014 3.36 1.27 to 8.88Age, continuous .003 1.10 1.03 to 1.16pT

T2 .001 1.00T3 v T2 0.12 0.02 to 0.63T4 v T2 1.91 0.28 to 13.00

pN, continuous .006 4.20 1.52 to 11.62Chemotherapy, yes v no .577

NOTE. Multivariate analysis included only variables that were significant (P � .1) in the univariate analysis.Abbreviation: HR, hazard ratio.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 11

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 12: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Table A5. Univariate Analysis of Distant Metastasis–Free Survival for All Stages in the Validation Set (N � 206)

Variable P HR 95% CI

ColoPrint, high v low .010 2.49 1.24 to 5.00Age

Continuous .538 1.01 0.98 to 1.04� v � 70 years .422 1.33 0.66 to 2.65

Localization, right v left .576 0.82 0.41 to 1.64Grade

Baseline .497 1Moderate v low 0.85 0.41 to 1.76High v low 1.65 0.54 to 5.06

Sex, female v male .388 1.39 0.66 to 2.92No. of LNs assessed .147 0.97 0.93 to 1.01

Continuous� 12 LNs, binary .074 0.52 0.26 to 1.07

StageI v II .025 0.24 0.03 to 1.82Baseline � II 1III v II 2.08 1.03 to 4.20

pTBaseline � T2 .466 1T3 v T2 1.89 0.57 to 6.23T4 v T2 2.65 0.53 to 13.12

pT, continuous .206 1.63 0.77 to 3.46pN

Baseline � no positive LNs .000 11-3 positive LNs v no positive LNs 1.34 0.55 to 3.29� 3 positive LNs v no positive LNs 6.08 2.74 to 13.50

pN, continuous .000 2.37 1.54 to 3.64Therapy, yes v no .825 1.08 0.54 to 2.15MSI-H, yes v no .391 0.42 0.06 to 3.07Lymphatic invasion .066 2.11 0.95 to 4.70Venous invasion .160 2.12 0.74 to 6.05Perineural invasion� — — —Lymphatic, perineural, or venous invasion .060 2.01 0.97 to 4.15

Abbreviations: HR, hazard ratio; LN, lymph node; MSI-H, microsatellite instability-high.�Low No. of events and extremely high borders.

Table A6. Multivariate Analysis for Distant Metastasis–Free Survival for All Stages in the Validation Set (N � 206)

Variable P HR 95% CI

ColoPrint, high v low .016 2.39 1.18 to 4.83pN�

Baseline � no positive LNs .000 1.001-3 positive LNs v no positive LNs 1.30 0.53 to 3.19� 3 positive LNs v no positive LNs 5.79 2.60 to 12.88

NOTE. Multivariate analysis included only variables that were significant (P � .05) from univariate analysis.Abbreviations: HR, hazard ratio; LN, lymph node.�Stage and pN are linearly dependent; therefore, only one variable was included.

Salazar et al

12 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.

Page 13: 01_Salazar_Gene_Expression_Signature_to_Improve_Prognosis_JCO_2011

Table A7. ColoPrint and ASCO Clinical Risk Index: Multivariate Analysis for Relapse-Free Survival in the Validation Set for Patients With Stage IIDisease Only (n � 115)

Variable P HR 95% CI

ColoPrint .017 3.660 1.24 to 9.08ASCO risk� .197 1.874 0.72 to 4.87

Abbreviations: ASCO, American Society of Clinical Oncology; HR, hazard ratio.�As per published recommendations, patients are considered high risk if they have any of the following: No. of assessed lymph nodes � 12; T4; histologic grade

3; and/or emergency presentation or obstruction.

Table A8. ColoPrint and ASCO Clinical Risk Index: Descriptive Distribution in Validation Set, Stage II Disease Only

Index

No. of Patients

ColoPrint Low Risk ColoPrint High Risk

ASCO low risk 43 29ASCO high risk 26 16

Abbreviation: ASCO, American Society of Clinical Oncology.

0

P = .1.7e-05

Time (months)

1.0

0.8

0.6

0.4

0.2

20 40 60 80 100 120

ColoPrint low-riskColoPrint high-risk

Prob

abili

ty o

f Dis

tant

M

etas

tasi

s–Fr

ee S

urvi

val

Fig A1. Kaplan-Meier analysis of the cross-validation in the training set (70% of patients were classified as low risk, and 30% were classified as high risk). Thehazard ratio is 3.41 (P � .001) with 5-year distant metastasis–free survival rates of 82% (95% CI, 76% to 89%) for low-risk patients and 50% (95% CI, 38%to 66%) for high-risk patients.

Colorectal Cancer Prognosis

www.jco.org © 2010 by American Society of Clinical Oncology 13

from 196.3.50.254Information downloaded from jco.ascopubs.org and provided by at HOFFMANN-LA ROCHE AG on November 23, 2010

Copyright © 2010 American Society of Clinical Oncology. All rights reserved.