10
Research Article DNA Methylation of Phosphatase and Actin Regulator 3 Detects Colorectal Cancer in Stool and Complements FIT Linda J.W. Bosch 1 , Frank A. Oort 2 , Maarten Neerincx 2 , Carolina A.J. Khalid-de Bakker 4 , Jochim S. Terhaar sive Droste 2 , Veerle Melotte 6 , Daisy M.A.E. Jonkers 4 , Ad A.M. Masclee 4 , Sandra Mongera 1 , Madeleine Grooteclaes 6 , Joost Louwagie 6 , Wim van Criekinge 6 , Veerle M.H. Coup e 3 , Chris J. Mulder 2 , Manon van Engeland 5 , Beatriz Carvalho 1 , and Gerrit A. Meijer 1 Abstract Using a bioinformatics-based strategy, we set out to identify hypermethylated genes that could serve as biomarkers for early detection of colorectal cancer (CRC) in stool. In addition, the complementary value to a Fecal Immunochemical Test (FIT) was evaluated. Candidate genes were selected by applying cluster alignment and computational analysis of promoter regions to microarray-expression data of colorectal adenomas and carcinomas. DNA methylation was measured by quantitative methylation-specific PCR on 34 normal colon mucosa, 71 advanced adenoma, and 64 CRC tissues. The performance as biomarker was tested in whole stool samples from in total 193 subjects, including 19 with advanced adenoma and 66 with CRC. For a large proportion of these series, methylation data for GATA4 and OSMR were available for comparison. The complementary value to FIT was measured in stool subsamples from 92 subjects including 44 with advanced adenoma or CRC. Phosphatase and Actin Regulator 3 (PHACTR3) was identified as a novel hypermethylated gene showing more than 70-fold increased DNA methylation levels in advanced neoplasia compared with normal colon mucosa. In a stool training set, PHACTR3 methylation showed a sensitivity of 55% (95% CI: 33–75) for CRC and a specificity of 95% (95% CI: 87–98). In a stool validation set, sensitivity reached 66% (95% CI: 50–79) for CRC and 32% (95% CI: 14–57) for advanced adenomas at a specificity of 100% (95% CI: 86–100). Adding PHACTR3 methylation to FIT increased sensitivity for CRC up to 15%. PHACTR3 is a new hypermethylated gene in CRC with a good performance in stool DNA testing and has complementary value to FIT. Cancer Prev Res; 5(3); 464–72. Ó2011 AACR. Introduction Screening for colorectal cancer (CRC) is the most efficient strategy for reducing death from this devastating disease. Colonoscopy is the gold standard for the detection and removal of early lesions and is highly sensitive, but also invasive and costly (1, 2). For population-wide screening simple and noninvasive procedures like stool testing are preferred (3). In follow-up to the guaiac-based Faecal Occult Blood Test (FOBT), the more sensitive immunochemical fecal occult blood test (Fecal Immunochemical Test or FIT; refs. 4–6) is now widely used in screening programs in Europe and Japan, and is expected to reduce CRC mortality by around 30% (7). This test performance though, still leaves room for improvement which could come from molecular stool tests like those testing for tumor DNA in stool. Multiple assays have been developed and evaluated for this purpose, but sensitivities still remain suboptimal (8–14). The recently introduced combination of mutation markers with DNA methylation markers has yielded sub- stantially improved test performance (13, 15, 16). Methyl- ation markers on their own, either alone or combined, have also yielded promising results, whereas the assays are tech- nically less demanding (17–23). Methylation markers are appealing for CRC screening even more because DNA methylation is an early event in colorectal development, preceding chromosomal abnormalities, and mutations (24). Yet, the ultimate marker, or combinations of markers, for stool DNA testing still remains to be determined. To this end, in this study, we aimed to identify new hypermethy- lated genes in CRC by applying dedicated bioinformatics to microarray expression data of colorectal adenomas and carcinomas and to explore their potential in whole stool Authors' Afliations: Departments of 1 Pathology, 2 Gastroenterology and Hepatology, and 3 Clinical Epidemiology and Biostatistics, VU University Medical Center, Amsterdam; 4 Department of Internal Medicine, Division of Gastroenterology & Hepatology; 5 Department of Pathology, GROWSchool for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands; and 6 MDxHealth, SA, Li ege, Belgium Note: Supplementary data for this article are available at Cancer Prevention Research Online (http://cancerprevres.aacrjournals.org/). Corresponding Author: Gerrit A. Meijer, Department of Pathology, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands. Phone: 312-044-44772; Fax: 312-044-42964; E-mail: [email protected] doi: 10.1158/1940-6207.CAPR-11-0315 Ó2011 American Association for Cancer Research. Cancer Prevention Research Cancer Prev Res; 5(3) March 2012 464 Cancer Research. on December 14, 2020. © 2012 American Association for cancerpreventionresearch.aacrjournals.org Downloaded from Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

Research Article

DNA Methylation of Phosphatase and Actin Regulator 3Detects Colorectal Cancer in Stool and Complements FIT

Linda J.W. Bosch1, Frank A. Oort2, Maarten Neerincx2, Carolina A.J. Khalid-de Bakker4,Jochim S. Terhaar sive Droste2, Veerle Melotte6, Daisy M.A.E. Jonkers4, Ad A.M. Masclee4,Sandra Mongera1, Madeleine Grooteclaes6, Joost Louwagie6, Wim van Criekinge6,Veerle M.H. Coup�e3, Chris J. Mulder2, Manon van Engeland5,Beatriz Carvalho1, and Gerrit A. Meijer1

AbstractUsing a bioinformatics-based strategy, we set out to identify hypermethylated genes that could serve as

biomarkers for early detection of colorectal cancer (CRC) in stool. In addition, the complementary value to a

Fecal Immunochemical Test (FIT) was evaluated. Candidate genes were selected by applying cluster

alignment and computational analysis of promoter regions to microarray-expression data of colorectal

adenomas and carcinomas. DNA methylation was measured by quantitative methylation-specific PCR on

34 normal colon mucosa, 71 advanced adenoma, and 64 CRC tissues. The performance as biomarker was

tested in whole stool samples from in total 193 subjects, including 19 with advanced adenoma and 66 with

CRC. For a large proportion of these series, methylation data for GATA4 and OSMR were available for

comparison. The complementary value to FITwasmeasured in stool subsamples from92 subjects including

44 with advanced adenoma or CRC. Phosphatase and Actin Regulator 3 (PHACTR3) was identified as a

novel hypermethylated gene showing more than 70-fold increased DNA methylation levels in advanced

neoplasia compared with normal colon mucosa. In a stool training set, PHACTR3 methylation showed a

sensitivity of 55% (95%CI: 33–75) for CRC and a specificity of 95% (95%CI: 87–98). In a stool validation

set, sensitivity reached 66% (95%CI: 50–79) for CRC and 32% (95%CI: 14–57) for advanced adenomas at

a specificity of 100% (95%CI: 86–100). Adding PHACTR3methylation to FIT increased sensitivity for CRC

up to 15%. PHACTR3 is a new hypermethylated gene in CRCwith a good performance in stool DNA testing

and has complementary value to FIT. Cancer Prev Res; 5(3); 464–72. �2011 AACR.

IntroductionScreening for colorectal cancer (CRC) is themost efficient

strategy for reducing death from this devastating disease.Colonoscopy is the gold standard for the detection andremoval of early lesions and is highly sensitive, but alsoinvasive and costly (1, 2). For population-wide screeningsimple and noninvasive procedures like stool testing arepreferred (3). In follow-up to the guaiac-based FaecalOccultBlood Test (FOBT), the more sensitive immunochemical

fecal occult blood test (Fecal Immunochemical Test or FIT;refs. 4–6) is now widely used in screening programs inEurope and Japan, and is expected to reduce CRCmortalityby around 30% (7). This test performance though, stillleaves room for improvement which could come frommolecular stool tests like those testing for tumor DNA instool. Multiple assays have been developed and evaluatedfor this purpose, but sensitivities still remain suboptimal(8–14). The recently introduced combination of mutationmarkers with DNA methylation markers has yielded sub-stantially improved test performance (13, 15, 16). Methyl-ationmarkers on their own, either alone or combined, havealso yielded promising results, whereas the assays are tech-nically less demanding (17–23). Methylation markers areappealing for CRC screening even more because DNAmethylation is an early event in colorectal development,preceding chromosomal abnormalities, and mutations(24).

Yet, the ultimatemarker, or combinations ofmarkers, forstool DNA testing still remains to be determined. To thisend, in this study, we aimed to identify new hypermethy-lated genes in CRC by applying dedicated bioinformatics tomicroarray expression data of colorectal adenomas andcarcinomas and to explore their potential in whole stool

Authors' Affiliations: Departments of 1Pathology, 2Gastroenterology andHepatology, and 3Clinical Epidemiology and Biostatistics, VU UniversityMedical Center, Amsterdam; 4Department of Internal Medicine, Division ofGastroenterology & Hepatology; 5Department of Pathology, GROW—

School for Oncology and Developmental Biology, Maastricht UniversityMedical Center, Maastricht, the Netherlands; and 6MDxHealth, SA, Li�ege,Belgium

Note:Supplementary data for this article are available atCancer PreventionResearch Online (http://cancerprevres.aacrjournals.org/).

Corresponding Author: Gerrit A. Meijer, Department of Pathology, VUUniversity Medical Center, PO Box 7057, 1007 MB, Amsterdam, theNetherlands. Phone: 312-044-44772; Fax: 312-044-42964; E-mail:[email protected]

doi: 10.1158/1940-6207.CAPR-11-0315

�2011 American Association for Cancer Research.

CancerPreventionResearch

Cancer Prev Res; 5(3) March 2012464

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 2: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

DNA testing for CRC (25, 26). Furthermore, we set out toevaluate complementary value of such markers to FIT in aseries of stool subsamples.

Materials and MethodsCell line authenticationThe CRC cell line HT29 was obtained from the American

Type Culture Collection (LGC Standards GmbH).COLO205, Colo320, HCT 116, and RKO cell lines werekindly provided by Dr. G.J. Peters, Department of Oncol-ogy, VU University Medical Center, Amsterdam, theNetherlands. The method for authentication was by arraycomparative genomic hybridization (aCGH, 244 k Agilentoligonucleotide platform), conducted at the VU UniversityMedicalCenter, Amsterdam, theNetherlands,most recentlyin October 2008. The patterns of chromosomal changesobserved were in concordance to the previously describedchromosomal changes in these cell lines (27). LS513 waskindly provided by Dr. F. Praz, Centre de Recherche Saint-Antoine, Paris, France. Array CGH confirmed the genomicprofile as described in literature (theWellcome Trust SangerInstitute Cancer Genome Project web site; ref. 28).

Strategy for methylation marker selectionThe strategy used for identification and validation of new

colon cancer-specific methylation markers included bothbioinformatics analysis of microarray-based mRNA expres-sion data and experimental validations of methylationlevels as outlined in Supplementary Fig. S1. First, genesdownregulated in CRC compared with adenomas weresubjected to a bioinformatics strategy for predicting can-cer-specific methylation (25). In the experimental valida-tion, the presence of DNAmethylation was tested by meth-ylation-specific PCR (MSP) using the BioTrove OpenArrayplatform (Ref. 29; BioTrove, Inc.), and high-throughputLightCycler assays. Full details are provided in the Supple-mentary Methods.

Strategy for evaluating PHACTR3 methylation as amarker in CRC tissue and stoolCell lines. For validating PHACTR3methylation inCRC

2 different regions around the Transcription Start Site (TSS)were investigated for the presence of methylation in 6 CRCcell lines; regions �149 to �63 bp and þ411 to þ526relative to the TSS. Next, HT29 and HCT116 cells weretreated with 5-aza-20-deoxycytidine (5-AZA) to evaluate theeffect of demethylation on mRNA expression.Tissue. PHACTR3methylation levels in the regionþ411

to þ526 relative to the TSS and mRNA expression weremeasured in cancer and matched normal tissue samplesfrom 9 CRC patients by quantitative MSP and quantitativeRT-PCR, respectively. Then, PHACTR3 methylation levelswere evaluated in an independent series of 34 normal colonmucosa tissue samples from cancer-free patients, 71advanced adenomas, and 64 carcinomas. Finally, toevaluate the specificity of PHACTR3 hypermethylation forCRC, methylation levels in other tumor types were ana-

lyzed, being tumor tissue samples from 44 breast, 20 cervix,20 lung, 11 esophagus (5 adenocarcinomas and 6 squa-mous cell carcinomas), 20 gliomas, 19 pancreas, and 15stomach.

Stool. To test the performance of PHACTR3 in a stool-based methylation test for CRC, a collection of 193 wholestool samples was split in a training set and a validationset of equal sizes. The training set consisted of a total of100 stool samples, of which 66 from patients withoutcolorectal neoplasia (58 healthy controls, 4 patients withcolonic diverticula and 4 patients with hemorrhoids), 9from patients with hyperplastic polyps, 3 from patientswith nonadvanced adenoma, and 22 from CRC patients.The training set was designed with more controls thencases, to better assess specificity. The validation set con-sisted of a total of 93 stool samples, 30 of which fromhealthy individuals, 19 from patients with advancedadenoma and 44 from CRC patients. The validation setcontained more advanced neoplasia, including advancedadenomas, to better assess sensitivity. (Patient character-istics are described in Supplementary Table S3). In addi-tion, methylation data for 2 other markers, that is, GATA4and OSMR were available for comparison in 95% of casesand 91% of controls from the training set and in all stoolsamples from the validation set tested for PHACTR3(18, 20).

Moreover, to examine the complementary value to FIT, anindependent series of 92 stool subsamples was analyzed forboth FIT and PHACTR3 methylation. This stool seriesoriginated from a retrospective collection from referralsubjects, and included 48 stool samples from subjectswithout colon neoplasia, 24 from patients with advancedadenomas and 20 from patients with carcinomas.

All details on tissue and stool collection, sample proces-sing, and methodologies used, are presented in the Sup-plementary Methods.

Statistical analysis of DNA methylation in tissues andstool

Mean differences in methylation or mRNA expressionlevels in tissue samples were analyzed with the Mann–Whitney test or ANOVA. The relation between methylationlevels in stool and the presence or absence of an advancedlesionwas studied by receiver operator characteristic (ROC)analysis. The Area under the curve (AUC) was used as ameasure of the test performance. CIs of proportions werecalculated using the Wilson score method. To test whetherage or gender were confounders in the relation betweenmethylation levels and the presence or absence of a lesion,linear regression was used. For the combination of FIT andPHACTR3 methylation, we used a distribution-free rank-based method (30) to calculate linear combination of the 2markers giving highest diagnostic accuracy. Sensitivitieswere compared with FIT or PHACTR3 methylation aloneat fixed specificities of 92%, 96%, and 98% using McNe-mar’s test. Calculations for the combination of FIT andPHACTR3 methylation was carried out in the R package(version 2.8.1.). All other analyses were carried out using

PHACTR3 Methylation as Stool DNA Test

www.aacrjournals.org Cancer Prev Res; 5(3) March 2012 465

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 3: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

SPSS software (version 15.0; SPSS Inc.). Values of P � 0.05were considered statistically significant.

ResultsBioinformatics for discovery of candidate genes

A total of 397 genes were downregulated in carcinomascompared with adenomas as determined by mRNA expres-sion microarray analysis (Wilcoxon rank test P < 1e–5 andThas P < 0.05; FDR < 0.05; ref. 26). In 284 of these genes, areported transcription start site (TSS) could be identified.These were subsequently subjected to a bioinformaticsapproach to predict cancer-specific methylation (25). Thisyielded 18 candidate genes, of which Phosphatase and actinregulator 3 (PHACTR3, NM_080672) was the only one thatpassed all further steps of experimental validation (seeSupplementary Fig. S1 and Supplementary Table S1).

PHACTR3 methylation and mRNA expression analysisin CRC cell lines and cancer and matched normaltissues

PHACTR3 harbors a dense CpG island located�678 andþ1,353 bp relative to the TSS (GC content: 65%,CpG(obs)/CpG(exp): 1.17). We designed conventional MSP primersin a region �149 to �63 bp and þ411 to þ526 relative tothe TSS (see Supplementary Methods and SupplementaryFig. S2). The region þ411 to þ526 relative to the TSS islocated in exon 1 and is the same region as analyzed at theinitial screen and validation of methylation status (seeSupplementary Fig. S1 and Supplementary Fig. S2A). Of6 cell lines tested, only HCT116 showedmethylation in theregion �149 to �63 bp relative to the TSS. For the regionþ411 toþ526 relative to the TSS,methylationwas found inall 6 cell lines tested (see Supplementary Fig. S2). At themRNA level these 6 CRC cell lines only showed marginalPHACTR3 expression levels compared with the positivecontrol (brain tissue; see Supplementary Fig. S2). Treatmentof HT29 and HCT116 with the demethylating agent 5-AZAresulted in reexpression of the gene (see Supplementary Fig.S2), consistent with PHACTR3 expression being downre-gulated by methylation in these cell lines.

Next, PHACTR3 methylation levels and mRNA expres-sion were measured in cancer tissue and matched normaltissue from 9 CRC patients. All 9 CRC tissue samplesshowed significantly increased methylation levels com-pared with their normal counterparts. Although mRNAexpression levels were low in both normal and tumortissues, still in 6 of 9 tumors PHACTR3 was significantlydownregulated compared with expression levels in theirnormal counterparts (see Fig. 1).

PHACTR3 methylation analysis in colorectal advancedadenoma and carcinoma tissues

To confirm the differential levels of PHACTR3 methyla-tion in CRC compared with normal mucosa, we tested anindependent series of tissue samples and included a set ofadvanced adenomas as well. PHACTR3 methylation levelswere 72-fold and 71-fold higher in advanced adenoma and

carcinoma samples, respectively, compared with normalmucosa samples (see Fig. 2A, P < 0.01). ROC analysisyielded an AUC of 0.93 (95% CI: 0.87–0.98) for CRC and0.95 (95% CI: 0.93–0.98, see Fig. 3A) for advanced neo-plasia (advanced adenomas and carcinomas; see Fig. 3).When fixing the cutoff for specificity at 100%, 72% of CRCand 81% of advanced neoplasia could be discriminatedfrom normal mucosa. No significant differences in meth-ylation levels were observed between advanced adenomaand carcinoma tissue samples or between carcinomas ofdifferent UICC stages (P ¼ 0.5 and P ¼ 0.07, respectively).Age or gender was no confounding factors (P¼ 0.1 and P¼0.9, respectively). Methylation levels in other tumor typesshowed high levels of methylation in tissue samples fromtumors of the intestinal tract (pancreatic, gastric, and esoph-ageal cancer) and in cervical cancer, whereas lower levels ofmethylation were seen in cancers of lung and bladder, andlittle or no methylation was seen in cancers of breast,prostate, and brain (glioma; Supplementary Fig. S3).

Sensitivity and specificity of PHACTR3 methylation instool for detecting colorectal cancer

To investigate the performance of PHACTR3methylationas a biomarker for CRC detection in stool, we measured

Figure 1. PHACTR3 methylation levels and mRNA expression in cancerandmatched normal tissues. A,PHACTR3methylation levels in tumor (T)and matched normal (N) tissues from 9 CRC patients. Quantificationsrepresent mean methylation levels (error bars correspond to SD) relativeto methylation levels in the normal tissues from 3 independentexperiments. Methylation levels are calculated as [relative quantity ofmethylated PHACTR3/relative quantity of unmethylated Beta-actin(ACTB)] ratio� 1,000. Asterisksmark significant differences (P < 0.05). B,mRNA expression analysis of PHACTR3 by RT-PCR in tumor (T) andmatched normal (N) tissues from 9 CRC patients. Quantificationsrepresent mean expression values (error bars correspond to SD) relativeto expression levels in the normal tissue from3 independent experiments.Asterisks mark significant differences (P < 0.05).

Bosch et al.

Cancer Prev Res; 5(3) March 2012 Cancer Prevention Research466

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 4: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

methylation levels in 2 independent series of stool-derivedDNA samples (see Fig. 2B and C). A training set was used todetermine the optimal cutoff to detect CRC patients com-pared with controls and nonadvanced adenoma patients.The validation set was used to validate the results from thetraining set and to test the detection rate of advancedadenomas.Training set. The training set consisted of 100 stool

samples from individuals who all had undergone completecolonoscopy. ROCanalysis of CRC (n¼ 22) comparedwithnonadvanced adenomas and control samples (n ¼ 78)yielded an AUC of 0.77 (95% CI: 0.64–0.90, see Fig. 3B).Maximum sensitivity at a fixed specificity of 95% (95% CI:87–98) was reached at a cutoff value of 82.5 relative copies.At that cutoff, sensitivity was 55% (95% CI:33–75) fordetecting CRC.Validation set. The validation set consisted of 93 stool

samples from individuals who all had undergone completecolonoscopy. ROC analysis with advanced neoplasia(44 CRC and 19 advanced adenoma) compared withhealthy controls (n ¼ 30) resulted in an AUC of 0.83(95% CI: 0.75–0.91, see Fig. 3C). Using a cutoff of valueof 82.5 relative copies as defined with the training setyielded a specificity of 100%(95%CI: 86–100), a sensitivity

of 66% (95% CI: 50–79) to detect CRC, and a sensitivity of32% (95%CI: 14–57) to detect advanced adenomas. Usinga cutoff of 28 relative copies, the highest possible sensitivityfor advanced adenomas was 53% (95% CI: 32–73), at aspecificity of 93% (95% CI: 79–98). Age or gender was noconfounding factor (P ¼ 1.0 and P ¼ 0.4, respectively).

Performance of PHACTR3 in comparison with otherstool methylation markers

To compare the test performance of PHACTR3 methyla-tion to other stool methylation markers, detection rateswere compared with those obtained with the previouslypublished markers, GATA4 (18) and OSMR (20), whichwere tested by QMSP in a large proportion of the currenttraining and validation series of stool samples. With thestool training set, the cutoffs for GATA4 and OSMRwere determined to detect CRC at equal specificities asPHACTR3. Table 1 shows the AUC, the cutoffs, sensitivitiesand specificities of these 3 markers. Compared withOSMR,in the training set, PHACTR3 showed a higher AUC andsensitivity (52% vs. 29%) for carcinomas, and in the val-idation set PHACTR3 showed a higher sensitivity for bothadvanced adenomas (32% vs. 21%) and carcinomas(66% vs. 43%) with higher specificity (100% vs. 90%).

Figure 2. PHACTR3 methylationlevels in tissue and stool. A,PHACTR3 methylation levels intissue samples of 34 normal colonmucosa, 71 advanced adenomasand 64 carcinomas (UICC stage I toIV). B, PHACTR3 methylation levelsin stool samples from 66 patientswithout colon neoplasia, 12 patientswith hyperplastic polyps (HP) ornonadvanced adenoma and 22 CRCpatients (training set). Methylationlevels are shown as relative quantityof methylated PHACTR3 � 1,000.C, PHACTR3 methylation levels instool samples from 30 patientswithout colon neoplasia, 19 patientswith advancedadenomaand44CRCpatients (validation set). Methylationlevels are shown as relative quantityof methylated PHACTR3 � 1,000.Box plots show first quartile, median,third quartile, and range ofmethylation levels. Dots representindividual data points, asterisksrepresent extremes.

PHACTR3 Methylation as Stool DNA Test

www.aacrjournals.org Cancer Prev Res; 5(3) March 2012 467

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 5: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

PHACTR3 andGATA4 showed equal AUCs and comparablesensitivities (52% vs. 57%) for detecting carcinomas in thetraining set. In the validation set, PHACTR3 showed ahigher sensitivity for both advanced adenomas (32% vs.16%) and carcinomas (66% vs. 39%) than GATA4, withhigher specificity (100% vs. 93%).

The combination of these 3 markers, calling positivewhen at least 1 of the 3markerswould be positive, increasedthe sensitivity to detect advanced adenoma from 32% to

42% and the sensitivity to detect carcinoma from 52% to62% (training set) and from 66% to 68% (validation set),however, at cost of specificity (decreased from 94% to 87%and from 100% to 83% for the training and validation set,respectively).

Combination of FIT and PHACTR3methylation in stoolTo investigate whether FIT and PHACTR3 methylation

would have complementary value for detecting colorectal

Figure 3. Receiver operatorcharacteristic analysis ofPHACTR3 methylation in tissueand stool. A, ROC curves of tissuesamples. Sensitivity and specificityat various cutoff values ofPHACTR3 methylation in tissuesamples from 64 carcinomasversus 34 normal colon mucosas(left) and 135 advanced neoplasiaversus 34 normal colon mucosas(right). The areas under the curvesare 0.93 and 0.95, respectively. B,ROC curves of stool samples(training set). Sensitivity andspecificity at various cutoff valuesof whole stool samples from 22CRC patients versus 66 controlpatients without colon neoplasia(left) and 22 CRC patients versus78 control patients (66 patientswithout colon neoplasia and 12patients with hyperplastic polyps(HP) or nonadvanced adenoma;right). The areas under the curvesare 0.78 and 0.77, respectively. C,ROC curves of stool samples(validation set). Sensitivity andspecificity at various cutoff valuesof whole stool samples from 44CRC patients versus 30 controlpatients without colon neoplasia(left) and 63patientswith advancedneoplasia versus 30 controlpatients without colon neoplasia(right). The areas under the curvesare 0.87and0.83, respectively. Thesolid line represents the ROCcurve, the dashed line shows thereference line of no discrimination.

Bosch et al.

Cancer Prev Res; 5(3) March 2012 Cancer Prevention Research468

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 6: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

cancer and advanced neoplasia, both tests were evaluated inan independent series of stool subsamples. Because forDNA methylation analysis, the stool subsamples were pro-cessed in a different way compared with whole stool sam-ples, different cutoffs were used, based on ROC analysis, todetermine sensitivity and specificity (see SupplementaryMethods). The sensitivities to detect advanced adenomaswere 21% (5/24, 95% CI: 9–40) for PHACTR3 and 21%(5/24, 95% CI: 9–40) for FIT. Combining PHACTR3 withFIT, meaning positive for at least 1 of the 2 measurements,increased the sensitivity to 33% (7/24, 95%CI: 18–53). Thesensitivities to detect CRC were 50% (10/20, 95% CI: 30–70) for PHACTR3 and 65% (13/20, 95%CI: 43–82) for FIT.Combining PHACTR3 with FIT increased the sensitivity to95% (19/20, 95% CI: 76–99). The specificity of the com-bination remained high [94% (45/48, 95% CI: 83–98)

compared with 96% (47/48, 95%CI: 86–99) for PHACTR3alone and 98% (47/48, 95% CI: 89–100) for FIT alone]. Inaddition, a positive test for both FIT and PHACTR3, occur-ring in 7 of the 44 advanced neoplasia cases (see Supple-mentary Fig. S4), revealed 100% specificity.

To evaluate whether the sensitivity of the combination ofFIT and PHACTR3 was significantly higher than the sensi-tivity of either FIT or PHACTR3 methylation alone, wecompared the sensitivities at equal specificity of 92%,96%, and98%(see SupplementaryMethods). ROCanalysisresulted in an AUC of 0.97 (95% CI: 0.93–1.0) for CRCand 0.79 (95% CI: 0.69–0.92) for advanced neoplasia(see Fig. 4), with sensitivity/specificity combinations of61%/92%, 55%/96%, and 48%/98% (see Table 2 foradvanced adenoma and cancer separately). At these specifi-cities, sensitivities for detecting CRC increased up to 15%

Table 1. Test performances of PHACTR3, GATA4, and OSMR

PHACTR3 GATA4 OSMR

Training setAUC (95% CI) 0.76 (0.62–0.89) 0.76 (0.62–0.90) 0.60 (0.45–0.75)Cutoff 82.5 16.5 3.6Sensitivity CRC (n ¼ 21) 52% 57% 29%Specificity (n ¼ 71) 94% 94% 94%

Validation setCutoff 82.5 16.5 3.6Sensitivity AA (n ¼ 19) 32% 16% 21%Sensitivity CRC (n ¼ 44) 66% 39% 43%Specificity (n ¼ 30) 100% 93% 90%

Abbreviations: AA, Advanced adenoma; GATA4, GATA-binding protein 4; OSMR, oncostatin M receptor.

Figure 4. Receiver operator characteristic analysis of FIT and PHACTR3 methylation in stool. A, sensitivity and specificity at various cutoff values of stoolsubsamples from20CRCpatients versus 48control patientswithout colonneoplasia for FIT,PHACTR3methylation and for the combination of both tests. Theareas under the curves are 0.92, 0.77, and 0.97, respectively. B, sensitivity and specificity at various cutoff values of partial stool samples from 44 patientswith advanced neoplasia versus 48 control patients without colon neoplasia for FIT, PHACTR3 methylation and for the combination of both tests. "A"represents the ROCcurve forPHACTR3methylation only, "B" represents the ROCcurve for FIT only, "C" represents the ROC curve for the combination of FITand PHACTR3 methylation. The areas under the curves are 0.76, 0.68, and 0.79, respectively.

PHACTR3 Methylation as Stool DNA Test

www.aacrjournals.org Cancer Prev Res; 5(3) March 2012 469

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 7: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

using the combination of FIT and PHACTR3 methylationcompared with FIT alone, although statistical significancewas not reached.

DiscussionStool-based DNA testing is an appealing approach for

noninvasive early detection of CRC. Both DNA mutationsand DNA promoter hypermethylation have been investi-gated as targets of potential screening assays, and especiallypanels of markers have shown promising performance ininitial studies. In this study, we applied a bioinformaticsapproach for identifying novel hypermethylated genes inCRC. This resulted in the identification of PHACTR3 as anew hypermethylated gene in CRC, which showed to havecomplementary diagnostic power to FIT in a pilot series.

PHACTR3 was first described in 2003 as a protein asso-ciated with the nuclear scaffold in human promyelocyticHL-60 leukemia cells. It was found to bind to actin and tothe catalytic subunit of Protein Phosphatase 1 (PP1), there-by inhibiting the function of PP1 (31), which in turn cantrigger apoptosis and can inhibit oncogenic signaling due toits interaction with pRb (32). Induced expression ofPHACTR3 in HeLa cervical cancer cells stimulated cellspreading and motility (33). Finally, in a small study ofnon–small cell lung cancer patients gene mutations werefound in 6/20 patients, which was associated with short-ened overall survival (34). Yet, looking at PHACTR3mRNAexpression, abundant expression has been found in adultnormal human brain and to a lesser extent in ovary, but notin other organs (31). Accordingly, in this study,weobservedhigh PHACTR3 mRNA expression in human brain tissue,but much lower expression levels in normal colon tissuesand again even lower expression in 6 of 9 matched tumortissues. Although PHACTR3 showed high methylationlevels in these 6 tumor tissue samples, methylation levelswere equally high in the 3 tumor tissues from patients in

which mRNA expression was not decreased compared withtheir normal counterparts. In addition, a direct correlationbetween the level ofmethylation and level ofmRNA expres-sion in all 9 tumor tissues could not be shown (Pearsoncorrelation of�0.3, P¼ 0.5, data not shown). Treatment ofHT29 andHCT116 cells with the demethylating agent 5-azadid result in reexpression of PHACTR3, however, consistentwith the recent observation that methylation in the regionof the first exon, which is the case for PHACTR3, is tightlylinked to transcriptional silencing (35). Yet, the possibilitythat the observed reexpression of PHACTR3 could be sec-ondary to demethylation of another gene or locus thenPHACTR3 itself cannot be excluded. Although a directrelationship between PHACTR3 hypermethylation andsilencing of expression in CRC remains to be established,the fact thatPHACTR3methylation is highly associatedwithcancer still makes it an interesting candidate biomarker.

Levels of PHACTR3 methylation in CRC tissues wereprominently high and could significantly discriminateadvanced adenoma and carcinoma from normal mucosaat the tissue level. Highmethylation levels are important forobtaining a good signal to noise ratio in a stool-based assay,especially when lesions to be detected are small. Interest-ingly, advanced adenoma tissue samples showed methyla-tion levels as high as carcinoma tissues, making PHACTR3methylation attractive as a biomarker, in compliance withrecent guidelines which have stated that the detection ofadvanced adenomas and not only early carcinoma shouldbe the goal of CRC screening (36).

When further exploring its potential as a biomarker, inwhole stool samples a sensitivity of 55% to 66% for detect-ing CRC and a sensitivity of 32% for detecting advancedadenoma was observed at a specificity of 95% to 100%.The test performance of PHACTR3 can be furtherimproved, which is illustrated by the higher AUC in tissuescompared with stool, in particular the sensitivity to detectadvanced adenomas. With the currently used method

Table 2. Cutoffs and sensitivities of FIT, PHACTR3 methylation, and their combination (Y) at equalspecificities

FIT PHACTR3 FITþPHACTR3 (Y)

Specificity Cutoff Sensitivity Cutoff Sensitivity P Cutoff Sensitivity P

AA 92% 1.0 33% 0.2 29% 1.0 16.0 33% 1.0CRC 92% 1.0 90% 0.2 60% 0.2 19.5 95% 1.0Advanced neoplasia 92% 1.0 59% 0.2 43% 0.2 16.0 61% 1.0AA 96% 49.5 25% 0.7 17% 0.7 55.5 25% 1.0CRC 96% 30.5 75% 0.7 50% 0.3 38.0 90% 0.3Advanced neoplasia 96% 30.5 48% 0.7 32% 0.2 38.0 55% 0.3AA 98% 99.5 21% 1.5 17% 1.0 81.5 25% 1.0CRC 98% 86.5 65% 1.1 50% 0.6 78.0 75% 0.5Advanced neoplasia 98% 86.5 41% 1.1 32% 0.5 78.0 48% 0.3

NOTE: P values are based on the comparison with FIT.Abbreviation: AA, Advanced adenoma.

Bosch et al.

Cancer Prev Res; 5(3) March 2012 Cancer Prevention Research470

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 8: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

higher sensitivities could also be reached, however, at costof specificity. Thehighest sensitivity that couldbe reached todetect advanced adenomas for example was 53%, resultingin a specificity of 93%. With these test performances,PHACTR3methylation is one of the best performing singlemethylation markers described so far (14). Also in com-parison to 2 previously published stool methylation mar-kers GATA4 and OSMR, PHACTR3 showed superior testperformance.A strategy to improve test performance is to combine

multiple markers in a single assay. Especially the highspecificity makes PHACTR3 attractive as a marker for sucha panel that would have increased sensitivity withoutmajoreffects on specificity. The combination of PHACTR3 withGATA4 and OSMR in this study indeed did increase sensi-tivity, but at cost of specificity, which mainly is due to thelower individual specificities of GATA4 and OSMR. Never-theless, it will be difficult to reach 100% sensitivity withmethylation markers only, because a portion of CRCs haveno or low frequencies of methylated genes, the so-calledCpG IslandMethylator Phenotype (CIMP)-negative tumors(37). Therefore, an attractive alternative could be to com-binemethylationmarkerswith a completely differentmark-er like FIT. Because FIT is already being used in severalscreeningprograms throughout theworld and logistics havealready been put in place, adding a DNA methylationmarker would be relatively easy. We therefore carried outa pilot study on the performance of PHACTR3methylationin combination with FIT. The combination of FIT andPHACTR3 methylation increased the sensitivity foradvanced neoplasia, that is, adenomas and CRC takentogether, while maintaining a high specificity. These experi-ments were done in a series of stool subsamples, whichwould be a good alternative for whole stool samples con-cerning logistics, stool processing, and storage of samples inlarge-scale screening programs. Because these stool sub-

samples were processed in a different manner comparedwith the whole stool samples described above, systematicdifferences existwhen comparing these results to those fromwhole stool samples (4). Still, the results obtained show thepower of combining FIT with a DNA methylation marker.This is in line with recent findings that showed an improvedtest performance of combining other molecular markers(i.e., APC, BAT26, and long-DNA) with FOBT (12, 38).

In conclusion, using a bioinformatics approach,PHACTR3 was identified as a new hypermethylated geneinCRC. Althoughwe could not unravel the functional effectof PHACTR3 hypermethylation in CRC, we clearly showedits highpotential as a biomarker in stool-basedDNA testing.Furthermore, this study suggests that combining PHACTR3methylation with FIT could be particularly promising. Thefull potential of this marker or its combination with FITawaits validation in a larger, well-controlled cohort study totest its performance in an asymptomatic population.

Disclosure of Potential Conflict of InterestJ. Louwagie andW. van Criekinge were employees of MDxHealth and are

holding stock options. G.A.Meijer is a consultant toMDxHealth and receivesresearch funding.

AcknowledgmentsThe authors thank Iedan Verly and Kim Wouters for their technical

assistance.

Grant SupportThis study was financially supported by SenterNovum (IS052034),

ZonMW (61200022), and performed within the framework of CTMM, theCenter for Translational Molecular Medicine, DeCoDe project (grant 03O-101).

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

Received June 20, 2011; revised November 15, 2011; accepted November17, 2011; published OnlineFirst December 1, 2011.

References1. Frazier AL, Colditz GA, Fuchs CS, Kuntz KM. Cost-effectiveness of

screening for colorectal cancer in the general population. JAMA2000;284:1954–61.

2. Rabeneck L, Paszat LF, Hilsden RJ, Saskin R, Leddin D, Grunfeld E,et al. Bleeding and perforation after outpatient colonoscopy and theirrisk factors in usual clinical practice. Gastroenterology 2008;135:1899–906,1906.

3. de Visser M, van Ballegooijen M, Bloemers SM, van Deventer SJ,Jansen JB, Jespersen J, et al. Report on the Dutch consensusdevelopment meeting for implementation and further development ofpopulation screening for colorectal cancer based on FOBT. Cell Oncol2005;27:17–29.

4. Oort FA, Droste JS, Van Der Hulst RW, Van Heukelem HA, Loffeld RJ,Wesdorp IC, et al. Colonoscopy-controlled intra-individual comparisonsto screen relevant neoplasia: faecal immunochemical test vs. guaiac-based faecal occult blood test. Aliment Pharmacol Ther 2010;31:432–9.

5. Hol L,Wilschut JA, vanBM, vanVuurenAJ, vander ValkHj, van der ZeeAG, et al. Screening for colorectal cancer: random comparison ofguaiac and immunochemical faecal occult blood testing at differentcut-off levels. Br J Cancer 2009;100:1103–10.

6. van Rossum LG, van Rijn AF, Laheij RJ, van Oijen MG, Fockens P, vanKriekenHH, et al. Randomcomparison of guaiac and immunochemical

fecal occult blood tests for colorectal cancer in a screening population.Gastroenterology 2008;135:82–90.

7. van Veen W, Mali WP. [Colorectal cancer screening: advice from theHealth Council of the Netherlands]. Ned Tijdschr Geneeskd 2009;153:A1441.

8. Ahlquist DA, Skoletsky JE, Boynton KA, Harrington JJ, Mahoney DW,Pierceall WE, et al. Colorectal cancer screening by detection of alteredhuman DNA in stool: feasibility of a multitarget assay panel. Gastro-enterology 2000;119:1219–27.

9. Tagore KS, Lawson MJ, Yucaitis JA, Gage R, Orr T, Shuber AP, et al.Sensitivity and specificity of a stoolDNAmultitarget assay panel for thedetection of advanced colorectal neoplasia. Clin Colorectal Cancer2003;3:47–53.

10. Imperiale TF, Ransohoff DF, Itzkowitz SH, Turnbull BA, RossME. FecalDNA versus fecal occult blood for colorectal-cancer screening in anaverage-risk population. N Engl J Med 2004;351:2704–14.

11. Whitney D, Skoletsky J, Moore K, Boynton K, Kann L, Brand R, et al.Enhanced retrieval of DNA from human fecal samples results inimprovedperformanceof colorectal cancer screening test. JMolDiagn2004;6:386–95.

12. Kutzner N, Hoffmann I, Linke C, Thienel T, Grzegorczyk M, Urfer W,et al. Non-invasive detection of colorectal tumours by the combined

PHACTR3 Methylation as Stool DNA Test

www.aacrjournals.org Cancer Prev Res; 5(3) March 2012 471

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 9: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

application of molecular diagnosis and the faecal occult blood test.Cancer Lett 2005;229:33–41.

13. Ahlquist DA, Sargent DJ, Loprinzi CL, Levin TR, Rex DK, Ahnen DJ,et al. Stool DNA and occult blood testing for screen detection ofcolorectal neoplasia. Ann Intern Med 2008;149:441–50, W81.

14. Bosch LJ, Carvalho B, Fijneman RJ, Jimenez CR, Pinedo HM, van EM,et al. Molecular tests for colorectal cancer screening. Clin ColorectalCancer 2011;10:8–23.

15. Itzkowitz S, Brand R, Jandorf L, Durkee K, Millholland J, Rabeneck L,et al. A simplified, noninvasive stool DNA test for colorectal cancerdetection. Am J Gastroenterol 2008;103:2862–70.

16. Itzkowitz SH, Jandorf L, BrandR, Rabeneck L, Schroy PC III, Sontag S,et al. Improved fecal DNA test for colorectal cancer screening. ClinGastroenterol Hepatol 2007;5:111–7.

17. Glockner SC, Dhir M, Yi JM, McGarvey KE, Van NL, Louwagie J, et al.Methylation of TFPI2 in stool DNA: a potential novel biomarker for thedetection of colorectal cancer. Cancer Res 2009;69:4691–9.

18. Hellebrekers DM, Lentjes MH, van den Bosch SM, Melotte V, WoutersKA, Daenen KL, et al. GATA4 and GATA5 are potential tumor sup-pressors and biomarkers in colorectal cancer. Clin Cancer Res2009;15:3990–7.

19. Huang ZH, Li LH, Yang F, Wang JF. Detection of aberrant meth-ylation in fecal DNA as a molecular screening tool for colorectalcancer and precancerous lesions. World J Gastroenterol 2007;13:950–4.

20. Kim MS, Louwagie J, Carvalho B, Terhaar Sive Droste JS, Park HL,Chae YK, et al. Promoter DNA methylation of oncostatin m receptor-beta as a novel diagnostic and therapeutic marker in colon cancer.PLoS One 2009;4:e6555.

21. LeungWK, ToKF,ManEP,ChanMW,Hui AJ,NgSS, et al. Detection ofhypermethylated DNA or cyclooxygenase-2 messenger RNA in fecalsamples of patients with colorectal cancer or polyps. Am J Gastro-enterol 2007;102:1070–6.

22. Melotte V, Lentjes MH, van den Bosch SM, Hellebrekers DM, de HoonJP,Wouters KA, et al. N-Myc downstream-regulated gene 4 (NDRG4):a candidate tumor suppressor gene and potential biomarker for colo-rectal cancer. J Natl Cancer Inst 2009;101:916–27.

23. WangDR, TangD.HypermethylatedSFRP2gene in fecal DNA is a highpotential biomarker for colorectal cancer noninvasive screening.WorldJ Gastroenterol 2008;14:524–31.

24. Derks S, Postma C, Moerkerk PT, van den Bosch SM, Carvalho B,HermsenMA,etal.Promotermethylationprecedeschromosomalaltera-tions in colorectal cancer development. Cell Oncol 2006;28:247–57.

25. Hoque MO, Kim MS, Ostrow KL, Liu J, Wisman GB, Park HL, et al.Genome-wide promoter analysis uncovers portions of the cancermethylome. Cancer Res 2008;68:2661–70.

26. Carvalho B, Postma C, Mongera S, Hopmans E, Diskin S, van de WielMA, et al. Multiple putative oncogenes at the chromosome 20qamplicon contribute to colorectal adenoma to carcinoma progression.Gut 2009;58:79–89.

27. Hermsen M, Snijders A, Guervos MA, Taenzer S, Koerner U, Baak J,et al. Centromeric chromosomal translocations show tissue-specificdifferences between squamous cell carcinomas and adenocarcino-mas. Oncogene 2005;24:1571–9.

28. TheWellcome Trust Sanger Institute Cancer Genome Project web site2011. Available from: http://www.sanger.ac.uk/genetics/CGP

29. Morrison T, Hurley J, Garcia J, Yoder K, Katz A, Roberts D, et al.Nanoliter high throughput quantitative PCR. Nucleic Acids Res2006;34:e123.

30. Pepe MS, Thompson ML. Combining diagnostic test results toincrease accuracy. Biostatistics 2000;1:123–40.

31. Sagara J, Higuchi T, Hattori Y, Moriya M, SarvothamH, Shima H, et al.Scapinin, a putative protein phosphatase-1 regulatory subunit asso-ciated with the nuclear nonchromatin structure. J Biol Chem2003;278:45611–9.

32. Wang RH, Liu CW, Avramis VI, Berndt N. Protein phosphatase1alpha-mediated stimulation of apoptosis is associated with dephos-phorylation of the retinoblastoma protein. Oncogene 2001;20:6111–22.

33. Sagara J, Arata T, Taniguchi S. Scapinin, the protein phosphatase 1binding protein, enhances cell spreading and motility by interactingwith the actin cytoskeleton. PLoS One 2009;4:e4247.

34. Bankovic J, Stojsic J, Jovanovic D, Andjelkovic T, Milinkovic V,Ruzdijic S, et al. Identification of genes associated with non-small-cell lung cancer promotion and progression. Lung Cancer 2010;67:151–9.

35. Brenet F, Moh M, Funk P, Feierstein E, Viale AJ, Socci ND, et al. DNAmethylation of the first exon is tightly linked to transcriptional silencing.PLoS One 2011;6:e14524.

36. Levin B, Lieberman DA, McFarland B, Andrews KS, Brooks D, Bond J,et al. Screening and surveillance for the early detection of colorectalcancer and adenomatous polyps, 2008: a joint guideline from theAmerican Cancer Society, the US Multi-Society Task Force on Colo-rectal Cancer, and the American College of Radiology. Gastroenter-ology 2008;134:1570–95.

37. Ostwald C, Linnebacher M, Weirich V, Prall F. Chromosomally andmicrosatellite stable colorectal carcinomas without the CpG islandmethylator phenotype in a molecular classification. Int J Oncol2009;35:321–7.

38. Kalimutho M, Del Vecchio BG, Cretella M, Mannisi E, Sileri P, FormosaA, et al. A simplified, non-invasive fecal-basedDNA integrity assay andiFOBT for colorectal cancer detection. Int J Colorectal Dis 2011;26:583–92.

Bosch et al.

Cancer Prev Res; 5(3) March 2012 Cancer Prevention Research472

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315

Page 10: DNA Methylation of Phosphatase and Actin …...Sandra Mongera1, Madeleine Grooteclaes 6, Joost Louwagie , Wim van Criekinge6, Veerle M.H. Coupe3, Chris J. Mulder2, Manon van Engeland5,

2012;5:464-472. Published OnlineFirst December 1, 2011.Cancer Prev Res   Linda J.W. Bosch, Frank A. Oort, Maarten Neerincx, et al.   Colorectal Cancer in Stool and Complements FITDNA Methylation of Phosphatase and Actin Regulator 3 Detects

  Updated version

  10.1158/1940-6207.CAPR-11-0315doi:

Access the most recent version of this article at:

  Material

Supplementary

  1

http://cancerpreventionresearch.aacrjournals.org/content/suppl/2011/11/30/1940-6207.CAPR-11-0315.DCAccess the most recent supplemental material at:

   

   

  Cited articles

  http://cancerpreventionresearch.aacrjournals.org/content/5/3/464.full#ref-list-1

This article cites 37 articles, 5 of which you can access for free at:

  Citing articles

  http://cancerpreventionresearch.aacrjournals.org/content/5/3/464.full#related-urls

This article has been cited by 4 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://cancerpreventionresearch.aacrjournals.org/content/5/3/464To request permission to re-use all or part of this article, use this link

Cancer Research. on December 14, 2020. © 2012 American Association forcancerpreventionresearch.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2011; DOI: 10.1158/1940-6207.CAPR-11-0315