Discriminant markers are required for accurate cancer screening. We evaluated genes frequently methylated in colorectal neoplasia to identify the most discriminant ones. Four genes specifically methylated in colorectal cancer [bone morphogenetic protein 3 (BMP3), EYA2, aristaless-like homeobox-4 (ALX4), and vimentin] were selected from 41 candidate genes and evaluated on 74 cancers, 62 adenomas, and 70 normal epithelia. Methylation status was analyzed qualitatively and quantitatively and confirmed by bisulfite genomic sequencing. Effect of methylation on gene expression was evaluated in five colon cancer cell lines. K-ras and BRAF mutations were detected by sequencing. Methylation of BMP3, EYA2, ALX4, or vimentin was detected respectively in 66%, 66%, 68%, and 72% of cancers; 74%, 48%, 89%, and 84% of adenomas; and 7%, 5%, 11%, and 11% of normal epithelia (P < 0.01, cancer or adenoma versus normal). Based on area under the curve analyses, discrimination was not significantly improved by combining markers. Comethylation was frequent (two genes or more in 72% of cancers and 84% of adenomas), associated with proximal neoplasm site (P < 0.001), and linked with both BRAF and K-ras mutations (P < 0.01). Cell line experiments supported silencing of expression by methylation in all four study genes. This study shows BMP3, EYA2, ALX4, and vimentin genes are methylated in most colorectal neoplasms but rarely in normal epithelia. Comethylation of these genes is common, and pursuit of complementary markers for methylation-negative neoplasms is a rational strategy to optimize screening sensitivity. (Cancer Epidemiol Biomarkers Prev 2007;16(12):2686–96)

Colorectal cancer is the second leading cause of cancer-related death in the United States, and currently, ∼40% of affected individuals die from this cancer (1).Colorectal cancer mortality can be reduced by screen detection of premalignant adenomas and early stage cancers (2-5). An emerging approach to cancer screening involves the assay of tumor-specific DNA alterations in bodily fluids from cancer patients, such as stool, serum, and urine (6-15). It is important to select markers with high accuracy if efficiency and effectiveness are to be achieved in a cancer screening application. Due to the molecular heterogeneity of colorectal neoplasia, high detection rates will likely require a panel of markers.

Several methylated genes have been detected in the stool and serum/plasma samples from colorectal cancer patients (8, 9, 11, 14, 16-20). Whereas some methylated genes have been found in a majority of colorectal cancers, the yield of bodily fluid–based assays remains suboptimal (8-11, 13-20). It is unclear as to what extent biological or technical factors account for such observations.

A subset of colorectal cancers exhibiting gene methylation and associated with proximal tumor site has been described as the CpG island methylator phenotype (CIMP; refs. 21, 22). Reported prevalences of CIMP in colorectal cancer vary (21-28). CIMP has been associated with BRAF mutations and microsatellite instability (26-30), but the relationship to other gene alterations is less studied. The degree to which CIMP may influence tumor detection is incompletely understood.

This study was designed to (a) evaluate high-yield methylated genes as candidate markers for screening colorectal neoplasia, (b) explore the effect of combining gene markers on detection sensitivity, and (c) examine the relationship of aberrant promoter methylation to the expression of bone morphogenetic protein 3 (BMP3), EYA2, aristaless-like homeobox-4 (ALX4), and vimentin genes.

Approval of this study was obtained from the Institutional Review Board of Mayo Foundation.

Subjects

Two hundred and six colon tissues, including 74 cancers, 62 adenomas, and 70 normal colon epithelia, were collected at the Mayo Clinic and evaluated in two studies. Tissue study I comprised 104 tissues, including 43 cancers, 32 adenomas, and 29 normal epithelia. Tissue study II comprised 102 tissues, including 31 cancers, 30 adenomas, and 41 normal epithelia. Samples in study I included 22 frozen and 82 paraffin-embedded tissues; methylation markers were assayed qualitatively. Samples in study II were all frozen, and markers were assayed quantitatively. The demographic and clinical characteristics of these subjects are shown in Table 1.

Table 1.

Clinical characteristics of subjects

StudyCancerAdenomaNormal
No. 43 32 29 
 II 31 30 41 
 Total 74 62 70 
Median age (range), y 66 (27-93) 67 (42-87) 67 (22-84) 
 II 66 (34-90) 65 (37-86) 65 (31-82) 
 Total 66 (27-93) 66 (37-87) 66 (22-84) 
Sex (M/F) 20/23 17/15 13/16 
 II 17/14 15/15 17/24 
 Total 37/37 32/30 30/40 
Location (proximal/distal) 21/22 19/13  
 II 12/19 18/12  
 Total 33/41 37/25  
Dukes stage (A/B/C/D) 2/21/19/1   
 II 6/9/15/1   
 Total 8/30/34/2   
Grade (1/2/3/4) or dysplasia (low/high) 1/10/28/4 25/7  
 II 0/4/26/1 21/9  
 Total 1/14/54/5 46/16  
StudyCancerAdenomaNormal
No. 43 32 29 
 II 31 30 41 
 Total 74 62 70 
Median age (range), y 66 (27-93) 67 (42-87) 67 (22-84) 
 II 66 (34-90) 65 (37-86) 65 (31-82) 
 Total 66 (27-93) 66 (37-87) 66 (22-84) 
Sex (M/F) 20/23 17/15 13/16 
 II 17/14 15/15 17/24 
 Total 37/37 32/30 30/40 
Location (proximal/distal) 21/22 19/13  
 II 12/19 18/12  
 Total 33/41 37/25  
Dukes stage (A/B/C/D) 2/21/19/1   
 II 6/9/15/1   
 Total 8/30/34/2   
Grade (1/2/3/4) or dysplasia (low/high) 1/10/28/4 25/7  
 II 0/4/26/1 21/9  
 Total 1/14/54/5 46/16  

Microdissection and DNA Extraction

Tissue sections were examined by a pathologist who circled out histologically distinct lesions to direct careful microdissection. Genomic DNA was extracted using Qiagen DNA minikit (Qiagen) or DNAzol (Invitrogen).

Conventional Methylation-Specific PCR

DNA was bisulfite treated using the EZ DNA methylation kit (Zymo Research) and eluted in 30 μL of elution buffer. One microliter of bisulfite-modified DNA was amplified in a total volume of 25 μL containing 1× PCR buffer (Applied Biosystem), 1.5 mmol/L MgCl2, 200 μmol/L of each deoxynucleotide triphosphate, 400 nmol/L of each primer, and 1.25 unit of AmpliTaq Gold polymerase (Applied Biosystem). Amplification included hot start at 95°C for 12 min, denaturing at 95°C for 30 s, annealing at certain temperatures for 30 s, extension at 72°C for 45 s for 35 cycles, and a final 10-min extension step at 72°C. Primer sequences and annealing temperatures were listed in Table 2, and primer locations were shown in Fig. 1. Bisulfite-treated human genomic DNA (Novagen) and CpGenomeTM universal methylated DNA (Chemicon) were used as positive controls for unmethylation and methylation, respectively.

Table 2.

Primers used in this study

GenePrimerPrimer sequence (5′→3′)Product size (bp)Annealing temperature (°C)Note*
BMP3 Unmethylated TTTAGTGTTGGAGTGGAGATGGTGTTTG 146 60  
  AAACACAACCAAATACAACAAAATAACAA    
 Methylated TTTAGCGTTGGAGTGGAGACGGCGTTC 143 68  
  CGCGACCGAATACAACGAAATAACGA    
 Real-time MSP AATATTCGGGTTATATACGTCGC 87 62  
  CCTCACCCGCGCAAAACG    
  6FAM-TAGCGTTGGAGTGGAGACGGCGTTCG-TAMRA    
 Bisulfite sequencing GAGGAGGGAAGGTATAGATAGA 256 60  
  AATTAAACTCCAAACCAACTAAAAC    
 RT-PCR CCCAAGTCCTTTGATGCCTA 147 62  
  TGGTACACAGCAAGGCTCAG    
EYA2 Unmethylated GGGAGGAGAAGGGGTTGGTTTTTTTG 209 60  
  CCTAAAATAAACACCACTAACAATACTCACCA    
 Methylated TTTCGGCGTAGGTAGTAGTCGC 190 66  
  GACCTAAAATAAACGCCGCTAACGA    
 Real-time MSP TTTTCGGCGTAGGTAGTAGTC 97 62  
  GACGAAACCGAACTAACTACGA    
  6FAM-CGGTAACGGTAGAGATAGTAACGTGTTC-TAMRA    
 Bisulfite sequencing GGTTTAGGGAGGAGAAGGGGT 370 60  
  CCTCTACCCTTATACCTTCCTAAC    
 RT-PCR GGACAATGAGATTGAGCGTGT 90 60 Ref. (57) 
  ATGTCCCCGTGAGTAAGGAGT    
ALX4 Unmethylated TGTGTTTTTTATTGTGAGTTGTTGGTT 295 60  
  ACAACAACAACTAAAACTACAAAATCAAC    
 Methylated TGCGTTTTTTATTGCGAGTCGTCGGTC 293 68  
  GACGACGACTAAAACTACGAAATCGACGA    
 Real-time MSP TTGTAGAGGTTTCGTTTTTCGTC 132 62  
  GCCTAAATTTCCCGTAAACTTTCGA    
  6FAM-CGTCGTCGTAGGTGAGAGCGTCGT-TAMRA    
 Bisulfite sequencing GGATAGTAGGATTGTAGAGGT 188 60  
  CTAAAACCCTAAAATCTCTAACTC    
 RT-PCR AGACCCACTACCCAGACGTG 222 63  
  GCCAGGACGGGTTCTGAAT    
Vimentin Unmethylated TTGGTGGATTTTTTGTTGGTTGATG 188 60  
  CACAACTTACCTTAACCCTTAAACTACTCA    
 Methylated TCGTTTCGAGGTTTTCGCGTTAGAGAC 216 68 Ref. (9) 
  CGACTAAAACTCGACCGACTCGCGA    
 Real-time MSP GTTTTAGTCGGAGTTACGTGATTAC 97 62  
  GAAAACGAAACGTAAAAACTACGA    
  6FAM-CGTATTTATAGTTTGGGCGACGCGTTGC-TAMRA    
 Bisulfite sequencing GTAGTTATGTTTATTAGGTT 342 55  
  CATTCAACTCCTACAACTC    
 RT-PCR GGACCAGCTAACCAACGACA 247 60  
  CTGGATTTCCTCTTCGTGGA    
β-actin Real-time bisulfite PCR TGGTGATGGAGGAGGTTTAGTAAGT Unknown 62 Ref. (31) 
  AACCAATAAAACCTACTCCTCCCTTAA    
  6FAM-ACCACCACCCAACACACAATAACAAACACA-TAMRA    
GAPDH RT-PCR CATCACCATCTTCCAGGAGCG 442 60 Ref. (50) 
  TGACCTTGCCCACAGCCTTG    
K-ras PCR AAGGCCTGCTGAAAATGACTGAAT 179 64  
  CTGTATCAAAGAATGGTCCTGCACC    
BRAF PCR CCACAAAATGGATCCAGACA 173 60  
  TGCTTGCTCTGATAGGAAAATG    
GenePrimerPrimer sequence (5′→3′)Product size (bp)Annealing temperature (°C)Note*
BMP3 Unmethylated TTTAGTGTTGGAGTGGAGATGGTGTTTG 146 60  
  AAACACAACCAAATACAACAAAATAACAA    
 Methylated TTTAGCGTTGGAGTGGAGACGGCGTTC 143 68  
  CGCGACCGAATACAACGAAATAACGA    
 Real-time MSP AATATTCGGGTTATATACGTCGC 87 62  
  CCTCACCCGCGCAAAACG    
  6FAM-TAGCGTTGGAGTGGAGACGGCGTTCG-TAMRA    
 Bisulfite sequencing GAGGAGGGAAGGTATAGATAGA 256 60  
  AATTAAACTCCAAACCAACTAAAAC    
 RT-PCR CCCAAGTCCTTTGATGCCTA 147 62  
  TGGTACACAGCAAGGCTCAG    
EYA2 Unmethylated GGGAGGAGAAGGGGTTGGTTTTTTTG 209 60  
  CCTAAAATAAACACCACTAACAATACTCACCA    
 Methylated TTTCGGCGTAGGTAGTAGTCGC 190 66  
  GACCTAAAATAAACGCCGCTAACGA    
 Real-time MSP TTTTCGGCGTAGGTAGTAGTC 97 62  
  GACGAAACCGAACTAACTACGA    
  6FAM-CGGTAACGGTAGAGATAGTAACGTGTTC-TAMRA    
 Bisulfite sequencing GGTTTAGGGAGGAGAAGGGGT 370 60  
  CCTCTACCCTTATACCTTCCTAAC    
 RT-PCR GGACAATGAGATTGAGCGTGT 90 60 Ref. (57) 
  ATGTCCCCGTGAGTAAGGAGT    
ALX4 Unmethylated TGTGTTTTTTATTGTGAGTTGTTGGTT 295 60  
  ACAACAACAACTAAAACTACAAAATCAAC    
 Methylated TGCGTTTTTTATTGCGAGTCGTCGGTC 293 68  
  GACGACGACTAAAACTACGAAATCGACGA    
 Real-time MSP TTGTAGAGGTTTCGTTTTTCGTC 132 62  
  GCCTAAATTTCCCGTAAACTTTCGA    
  6FAM-CGTCGTCGTAGGTGAGAGCGTCGT-TAMRA    
 Bisulfite sequencing GGATAGTAGGATTGTAGAGGT 188 60  
  CTAAAACCCTAAAATCTCTAACTC    
 RT-PCR AGACCCACTACCCAGACGTG 222 63  
  GCCAGGACGGGTTCTGAAT    
Vimentin Unmethylated TTGGTGGATTTTTTGTTGGTTGATG 188 60  
  CACAACTTACCTTAACCCTTAAACTACTCA    
 Methylated TCGTTTCGAGGTTTTCGCGTTAGAGAC 216 68 Ref. (9) 
  CGACTAAAACTCGACCGACTCGCGA    
 Real-time MSP GTTTTAGTCGGAGTTACGTGATTAC 97 62  
  GAAAACGAAACGTAAAAACTACGA    
  6FAM-CGTATTTATAGTTTGGGCGACGCGTTGC-TAMRA    
 Bisulfite sequencing GTAGTTATGTTTATTAGGTT 342 55  
  CATTCAACTCCTACAACTC    
 RT-PCR GGACCAGCTAACCAACGACA 247 60  
  CTGGATTTCCTCTTCGTGGA    
β-actin Real-time bisulfite PCR TGGTGATGGAGGAGGTTTAGTAAGT Unknown 62 Ref. (31) 
  AACCAATAAAACCTACTCCTCCCTTAA    
  6FAM-ACCACCACCCAACACACAATAACAAACACA-TAMRA    
GAPDH RT-PCR CATCACCATCTTCCAGGAGCG 442 60 Ref. (50) 
  TGACCTTGCCCACAGCCTTG    
K-ras PCR AAGGCCTGCTGAAAATGACTGAAT 179 64  
  CTGTATCAAAGAATGGTCCTGCACC    
BRAF PCR CCACAAAATGGATCCAGACA 173 60  
  TGCTTGCTCTGATAGGAAAATG    

Abbreviation: GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

*

Oligos were designed by us except those with references.

Figure 1.

Schematic graph of the 5′ regions of BMP3, EYA2, ALX4, and vimentin genes. Vertical bars, CpG sites. Regions analyzed by MSP, quantitative MSP (qMSP), and bisulfite genomic sequencing, and the start codons were indicated.

Figure 1.

Schematic graph of the 5′ regions of BMP3, EYA2, ALX4, and vimentin genes. Vertical bars, CpG sites. Regions analyzed by MSP, quantitative MSP (qMSP), and bisulfite genomic sequencing, and the start codons were indicated.

Close modal

Real-time Quantitative Methylation-Specific PCR

Bisulfite-treated DNA above was used as a template for methylation quantification with a fluorescence-based real-time PCR as described previously (31). Primers and probes were designed to target the bisulfite-modified methylated sequences of gene promoters (Fig. 1; Table 2). A region without CpG site in β-actin gene was also quantified with real-time PCR using primers and probe recognizing bisulfite-converted sequence as a reference of bisulfite treatment and DNA input (31). PCR reactions were done in a volume of 25 μL consisting of 600 nmol/L of each primer, 200 of nmol/L probe, 0.75 units of platinum Taq polymerase (Invitrogen), 200 μmol/L each of deoxynucleotide triphosphate, 16.6 mmol/L ammonium sulfate (Sigma), 67 mmol/L Trizma (Sigma), 6.7 mmol/L MgCl2, 10 mmol/L mercaptoethanol, and 0.1% DMSO. One microliter of bisulfite-treated DNA was used in each PCR reaction. The gene methylation level was defined as the ratio of the fluorescence emission intensity value of target gene PCR product to that of β-actin PCR product multiplied by 1,000 (31).

Amplifications were done in 96-well plates in a real-time iCycler (Bio-Rad) under the following conditions: 95°C for 2 min, followed by 45 cycles of 95°C for 10 s and 62°C for 60 s. Bisulfite-treated CpGenomeTM universal methylated DNA (Chemicon) was used as positive control and serially diluted to create standard curve for all plates. Each plate consisted of bisulfite-treated DNA samples, positive and negative controls, and water blanks.

Selection of Tumor-Specific Methylated Markers

Forty-one genes were analyzed with methylation-specific PCR (MSP). These genes consisted of seven candidates identified in colorectal cancer by our group, including EYA2, EYA3, BMP1, BMP2, BMP3, SIX2, and SIX6, and 16 commonly methylated genes, including p16, hMLH1, MGMT, CDH1, HIC1, GSTP1, RASSF1A, RUNX1, SLC5A8, SFRP1, vimentin, EYA4, BMP3b, TPEF, GATA4, and GATA5 (refs. 9, 32-46), as well as 18 methylated genes reported recently in the SW480 colon cancer cell line, including ALX4, FOXF1, SHH, ZNF677, RASL11A, PAX6, ADAM12, KIAA0789, TGFB2, ZNF566, CDCA2, RPS27L, FLJ25439, TAZ, LOC283514, DAP, GATA3, and a predicted gene (47). Methylated primers for the common methylated genes were from the literature, and the rest were designed by us with at least four CpGs and four Cs on each primer to discriminate methylated DNA sequence from unmethylated and wild-type ones.

The specificity of the primers to methylated sequence was first tested with bisulfite-treated universally methylated DNA, unmethylated human genomic DNA, and wild-type human genomic DNA. Primers that only amplified bisulfite-treated universally methylated DNA were further triaged in an age-matched independent set of colon tissues, including four cancers and four normal mucosa. Four genes, BMP3, EYA2, ALX4, and vimentin, were found to be methylated in three or more of the cancers but in none of the normal tissues (Fig. 2); thus, these four methylation markers were selected for more extensive evaluation in the present investigation as described above. Primers for BMP3, EYA2, ALX4, and vimentin were presented in Table 2, and primers for other genes are available upon request. The schematic graphs of the 5′ regions of BMP3, EYA2, ALX4, and vimentin were shown in Fig. 1.

Figure 2.

Tumor-specific methylated gene markers selected for study. Among 41 candidate genes, BMP3, EYA2, ALX4, and vimentin were methylated in at least three of four of the colorectal cancers, but in none of four normal colon tissues screened. HIC1, TPEF, and FOXF1 as representative of less specific or less sensitive markers for comparison. Universally methylated DNA and water were amplified as positive control and negative control, respectively.

Figure 2.

Tumor-specific methylated gene markers selected for study. Among 41 candidate genes, BMP3, EYA2, ALX4, and vimentin were methylated in at least three of four of the colorectal cancers, but in none of four normal colon tissues screened. HIC1, TPEF, and FOXF1 as representative of less specific or less sensitive markers for comparison. Universally methylated DNA and water were amplified as positive control and negative control, respectively.

Close modal

Bisulfite Genomic Sequencing

Methylation status of representative samples was confirmed by bisulfite genomic sequencing using primers (Table 2) that flank the MSP and/or real-time MSP regions of BMP3, EYA2, ALX4, and vimentin (Fig. 1). One microliter of bisulfite-modified DNA was amplified in a total volume of 25 μL containing 1× PCR buffer (Applied Biosystem), 3.0 mmol/L MgCl2, 200 μmol/L of each deoxynucleotide triphosphate, 400 nmol/L of each primer, and 1.25 unit of AmpliTaq Gold polymerase (Applied Biosystem). Amplification included 95°C for 10 min, denaturing at 95°C for 30 s, annealing at certain temperatures for 30 s, extension at 72°C for 45 s for 40 cycles, and a final 10-min extension step. PCR products were cut from gels, purified using QIAquick gel extraction kit (Qiagen), and then ligated into pCR 2.1-TOPO cloning vector using a TOPO TA cloning kit (Invitrogen). For each cloning, six colonies were grown, extracted with Wizard Plus Minipreps DNA purification system (Promega), and then sequenced with ABI Prism 377 DNA sequencer (Perkin-Elmer) to get detailed methylation status of each CpG site.

Mutation Detection

Mutations on K-ras at codons 12 and 13 and on BRAF (V600E) were assayed by Sanger sequencing. Genomic DNA (100 ng) was amplified with primers flanking the mutant loci (Table 2). Five microliters of PCR products were incubated with 2 μL ExoSAP-IT (U.S. Biochemical Corporation) at 37°C for 30 min to get rid of residual deoxynucleotide triphosphates, primers, and possible dimmers and then directly sequenced in an ABI Prism 377 DNA sequencer (Perkin-Elmer).

Cell Lines and 5-Aza-Deoxycytidine Treatment

Five colon cancer cell lines, including SW480, SNUC4, HCT15, SW620, and WIDR, were used in this study. SW480, SW620, and WIDR were grown in DMEM supplemented with 10% fetal bovine serum, and SNUC4 and HCT15 were grown in RPMI 1640 supplemented with 10% fetal bovine serum. These cells were split to low density in 4-mL flasks, grown for 12 to 24 h, and then treated using 5 μmol/L 5-aza-deoxycytidine or mock treated with PBS for 96 h. Medium containing 5-aza-deoxycytidine and with PBS was changed every 24 h. The dose and timing of 5-aza-deoxycytidine were based on prior tests showing optimal reactivation of gene expression and published studies (48, 49).

Real-time Reverse Transcription–PCR

The mRNA expression of BMP3, EYA2, ALX4, and vimentin in these colon cancer cell lines with or without 5-aza-deoxycytidine treatment was quantified with real-time reverse transcription–PCR (RT-PCR). Briefly, RNA was extracted with RNeasy minikit (Qiagen). Reverse transcription was done on 2 μg of total RNA using Omniscript RT kit (Qiagen). One microliter of cDNA was amplified in a real-time iCycler (Bio-Rad) using a reaction volume of 25 μL containing 1× iQ SYBR Green Supermix (Bio-Rad) and 200 nmol/L of each primer under the following conditions: 95°C for 3 min followed by 40 cycles of 95°C for 10 s and 60°C for 45 s. Primers for each gene were designed on different exons to guarantee specific amplification of cDNA (Table 2). Glyceraldehyde-3-phosphate dehydrogenase was used as an internal reference gene for normalizing the cDNA input (50). The mRNA expression ratios of the four genes were defined as the ratio fluorescence emission intensity value of target gene PCR products to that of β-actin PCR products multiplied by 1,000. Amplification was done in 96-well plates. Each plate consisted of cDNA samples and multiple water blanks, as well as positive and negative controls. Each assay was done in duplicate. Serial dilutions of positive controls were used to make standard curves for each plate. Melt curve was conducted for each reaction to guarantee that only one identical product was amplified, and the PCR products were further confirmed by agarose gel electrophoresis.

Statistical Analysis

χ2 test was used to compare gene methylation frequencies between each of the three different tissue groups in study I, and Fisher exact test was used to analyze the association of gene methylation frequencies with clinical characteristics of tumor patients in study I. Wilcoxon rank-sum test was used to compare the methylation levels between each of the three different tissue groups and evaluate the association of methylation levels with tumor location, gender, Dukes stage, and differentiation grade in study II. Correlation of methylation levels with tumor size and patient age in study II was calculated with logistic procedure. Receiver operating curve was constructed to compare methylation level in cancers or adenomas versus normal subjects for each of the four markers and their combinations in study II, and area under the curve (AUC) value was also calculated for each curve. The association of gene comethylation with clinical characteristics of tumor patients and K-ras or BRAF mutations were calculated with χ2 test. Statistical analysis was conducted with SAS software (SAS Institute).

Methylation of BMP3, EYA2, ALX4, and Vimentin Genes in Colorectal Tumors

From a total of 41 candidates, four genes (BMP3, EYA2, ALX4, and vimentin) were found to be methylated in at least three of four colon cancers but in none of four normal colon epithelia on prestudy triage. Methylation of these four selected genes was evaluated more comprehensively in this investigation in two tissue studies.

In tissue study I, using conventional MSP, methylation of BMP3, EYA2, ALX4, and vimentin was detected in 60%, 51%, 74%, and 77% of 43 cancers; 72%, 44%, 91%, and 91% of 32 adenomas; and 7%, 3%, 17%, and 17% of 29 normal mucosa samples, respectively. Methylation was more frequently detected in cancer or adenoma than in normal epithelia for each of the four genes (P < 0.01; Fig. 3). Methylation was significantly more frequent in cancer from proximal colon than from distal colon for BMP3, EYA2, ALX4, and vimentin (P < 0.05; Table 3), but not associated with age, sex, tumor size, Dukes stage, or grade for any of the four genes (P > 0.05). Methylation in adenomas was not associated with age, sex, tumor size, degree of dysplasia, or villous component (P > 0.05).

Figure 3.

Neoplasm-specific methylation of BMP3, EYA2, ALX4, and vimentin genes. Methylation status was determined by conventional MSP using methylation-specific primers. Representative tissues from normal colon epithelia, adenomas, and cancers. Universally methylated DNA and water were amplified as positive and negative controls, respectively.

Figure 3.

Neoplasm-specific methylation of BMP3, EYA2, ALX4, and vimentin genes. Methylation status was determined by conventional MSP using methylation-specific primers. Representative tissues from normal colon epithelia, adenomas, and cancers. Universally methylated DNA and water were amplified as positive and negative controls, respectively.

Close modal
Table 3.

Gene methylation associated with tumor location in cancer subjects

StudyGeneLocationMethylation rate or level (median; range)P
Study I BMP3 Proximal 90% (19 of 21) 0.0002 
  Distal 32% (7 of 22)  
 EYA2 Proximal 71% (15 of 21) 0.02 
  Distal 32% (7 of 22)  
 ALX4 Proximal 95% (20 of 21) 0.004 
  Distal 55% (12 of 22)  
 Vimentin Proximal 95% (20 of 21) 0.01 
  Distal 59% (13 of 22)  
Study II BMP3 Proximal 34 (0-628) 0.01 
  Distal 1 (0-302  
 EYA2 Proximal 155 (0-1082) 0.03 
  Distal 2 (0-360)  
 ALX4 Proximal 458 (17-1182) 0.002 
  Distal 25 (0-379)  
 Vimentin Proximal 418 (0-1055) 0.005 
  Distal 10 (0-276)  
StudyGeneLocationMethylation rate or level (median; range)P
Study I BMP3 Proximal 90% (19 of 21) 0.0002 
  Distal 32% (7 of 22)  
 EYA2 Proximal 71% (15 of 21) 0.02 
  Distal 32% (7 of 22)  
 ALX4 Proximal 95% (20 of 21) 0.004 
  Distal 55% (12 of 22)  
 Vimentin Proximal 95% (20 of 21) 0.01 
  Distal 59% (13 of 22)  
Study II BMP3 Proximal 34 (0-628) 0.01 
  Distal 1 (0-302  
 EYA2 Proximal 155 (0-1082) 0.03 
  Distal 2 (0-360)  
 ALX4 Proximal 458 (17-1182) 0.002 
  Distal 25 (0-379)  
 Vimentin Proximal 418 (0-1055) 0.005 
  Distal 10 (0-276)  

In tissue study II, methylation levels were quantified using quantitative MSP. Mean methylation levels in 31 cancers, 30 adenomas, and 41 normal colon epithelia were observed respectively as follows (Fig. 4): 116 (0-628), 189 (0-712), and 0.3 (0-8.2) for BMP3; 158 (0-1082), 167 (0-1066), and 1.5 (0-51) for EYA2; 230 (0-1182), 335 (0-868), and 10.1 (0-113) for ALX4; and 193 (0-1055), 258 (0-955), and 5.0 (0-144) for vimentin. Methylation levels were significantly higher in cancer or adenoma than in normal epithelium for each of the four genes (P < 0.01 for each gene) but were comparable between cancer and adenoma for each gene after stratification by tumor location (P > 0.05 for each gene). Methylation levels were significantly higher in cancers from the proximal colon than from the distal colon for all four genes (P < 0.05; Table 3) but higher in adenomas from proximal colon than from distal colon for ALX4 only (P = 0.02). Methylation levels in cancers correlated with larger size for ALX4 only (P = 0.004) but were not associated with age, sex, Dukes stage, and grade for any of the four genes (P > 0.05). Methylation levels of adenomas correlated with larger size for BMP3 only (P = 0.04) and with older age for vimentin only (P = 0.04) but not with other clinical characteristics, including sex, degree of dysplasia, and villous component, for any of the four genes (P > 0.05).

Figure 4.

Methylation levels of BMP3, EYA2, ALX4, and vimentin measured by quantitative real-time MSP in colorectal cancer, adenoma, and normal epithelia. Each dot reresents a sample.

Figure 4.

Methylation levels of BMP3, EYA2, ALX4, and vimentin measured by quantitative real-time MSP in colorectal cancer, adenoma, and normal epithelia. Each dot reresents a sample.

Close modal

For quantitative data obtained in study II, receiver operating curves were constructed for each of the four genes (Fig. 5). Comparing cancer to normal epithelia, AUC values were 0.85, 0.9, 0.89, and 0.88 for BMP3, EYA2, ALX4, and vimentin, respectively (Fig. 5A); comparing adenoma to normal epithelia, AUC values were 0.87, 0.79, 0.93, and 0.89 for BMP3, EYA2, ALX4, and vimentin, respectively (Fig. 5A). AUC value was not significantly improved by combining any or all markers compared with the best single marker (P > 0.05; Fig. 5B). At a specificity of 93%, methylation of BMP3, EYA2, ALX4, and vimentin detected 74%, 87%, 58%, and 65% of 31 cancers and 77%, 53%, 93%, and 77% of 30 adenomas.

Figure 5.

A, receiver operating curves for gene methylation levels in colorectal cancers or adenomas versus normal controls. For cancers versus normal controls, AUC values were 0.85, 0.9, 0.89, and 0.88 for BMP3, EYA2, ALX4, and vimentin, respectively; for adenomas versus normal controls, AUC values were 0.87, 0.79, 0.93, and 0.89 for BMP3, EYA2, ALX4, and vimentin, respectively. B, predicted receiver operating curves of best combinations of methylated markers in cancers or adenomas versus normal controls. AUC values were 0.92 for the predicted combination (BMP3, EYA2, and ALX4) in cancers and 0.94 for the predicted combination (ALX4, BMP3, and vimentin) in adenomas, which are not significantly higher than with single markers.

Figure 5.

A, receiver operating curves for gene methylation levels in colorectal cancers or adenomas versus normal controls. For cancers versus normal controls, AUC values were 0.85, 0.9, 0.89, and 0.88 for BMP3, EYA2, ALX4, and vimentin, respectively; for adenomas versus normal controls, AUC values were 0.87, 0.79, 0.93, and 0.89 for BMP3, EYA2, ALX4, and vimentin, respectively. B, predicted receiver operating curves of best combinations of methylated markers in cancers or adenomas versus normal controls. AUC values were 0.92 for the predicted combination (BMP3, EYA2, and ALX4) in cancers and 0.94 for the predicted combination (ALX4, BMP3, and vimentin) in adenomas, which are not significantly higher than with single markers.

Close modal

Combining studies I and II, methylation of BMP3, EYA2, ALX4, and vimentin was detected in 66%, 66%, 68%, and 72% of 74 cancers; 74%, 48%, 89%, and 84% of 62 adenomas; and 7%, 5%, 11%, and 11% of 70 normal epithelia, respectively (P < 0.01, cancer or adenoma versus normal for each gene).

Using representative samples, bisulfite genomic sequencing confirmed that these four genes are densely methylated in cancer and adenoma but rarely or not methylated in normal colon mucosa (Fig. 6).

Figure 6.

Methylation status of representative colon tissues confirmed by bisulfite genomic sequencing. The analyzed regions of the four CpG islands evaluated with the methylation status of each. Six clones were sequenced for each sample. Closed circles, methylated CpGs; open circles, unmethylated CpGs.

Figure 6.

Methylation status of representative colon tissues confirmed by bisulfite genomic sequencing. The analyzed regions of the four CpG islands evaluated with the methylation status of each. Six clones were sequenced for each sample. Closed circles, methylated CpGs; open circles, unmethylated CpGs.

Close modal

Comethylation in Colorectal Tumors

BMP3, EYA2, ALX4, and vimentin genes were commonly comethylated in colorectal neoplasms, and the subset of subjects with neoplasms showing comethylation shared certain characteristics. Methylation levels in study II were dichotomized to simplify panel assembly and to allow easier translation of quantitative to qualitative panels as obtained in study I (27). The dichotomization threshold at a methylation level of 10 was chosen as a point sufficiently above background levels measured with quantitative MSP but well below the much higher levels for the four markers in both colorectal cancers and adenomas (27). Methylation of one or more of four (at least one), two or more of four, three or more of four, or four of four genes was noted in 88%, 72%, 53%, and 41% of 74 cancers (Fig. 7; Table 4) and 98%, 84%, 60%, and 39% of 62 adenomas (Fig. 7; Table 5) compared with 24%, 7%, 3%, and 0% of 70 normal epithelia, respectively. Thus, comethylation is much more common in neoplasia than in normal epithelia, and comethylation is associated, progressively so with increasing specificity but decreasing sensitivity for colorectal neoplasia. Comethylation of two or more of four and three or more of four genes in cancer was significantly associated with older age (P < 0.05) and proximal colon location (P ≤ 0.001) but not with other clinical characteristics (Table 4); comethylation of four of four genes in cancer was associated with proximal location only (P = 0.0004; Table 4). Comethylation of two or more of four and three or more of four genes in adenoma was significantly associated with proximal location (P < 0.01; Table 5), and comethylation of four of four genes in adenoma was associated with older age (P = 0.008; Table 5).

Figure 7.

Heat maps demonstrating relationship of specific gene methylation, K-ras and BRAF mutations, and categorization as CIMP in colorectal cancer and adenoma. Red bars, methylated samples for the corresponding gene. The methylation levels across all cancer or adenoma samples are indicated from low to high using a long bar with increasing depth of red color. Blue bars, BRAF and K-ras mutations.

Figure 7.

Heat maps demonstrating relationship of specific gene methylation, K-ras and BRAF mutations, and categorization as CIMP in colorectal cancer and adenoma. Red bars, methylated samples for the corresponding gene. The methylation levels across all cancer or adenoma samples are indicated from low to high using a long bar with increasing depth of red color. Blue bars, BRAF and K-ras mutations.

Close modal
Table 4.

The association of gene comethylation with clinical variables and gene mutations in cancer subjects

Comethylated genes ≥2
Comethylated genes ≥3
Comethylated genes = 4
+P+P+P
Total  53 21  39 35  30 44  
Age ≤60 y 12 11 0.01 15 0.04 17 0.09 
 >60 y 41 10  31 20  24 27  
Sex Male 25 12 0.4 17 20 0.2 12 25 0.2 
 Female 28  22 15  18 19  
Location Proximal 31 0.0002 28 6 × 10−7 21 12 0.0004 
 Distal 22 19  11 30  32  
Dukes stage A/B 27 11 0.9 16 22 0.06 11 27 0.04 
 C/D 26 10  23 13  19 17  
Grade 1/2 12 0.5 1.0 10 0.5 
 3/4 41 18  31 28  25 34  
BRAF Mutant 15 0.006 15 1 × 10−5 15 9 × 10−8 
 Wild-type 38 21  24 35  15 44  
K-ras Mutant 19 0.007 15 0.02 12 1.0 
 Wild-type 34 20  24 30  22 32  
Comethylated genes ≥2
Comethylated genes ≥3
Comethylated genes = 4
+P+P+P
Total  53 21  39 35  30 44  
Age ≤60 y 12 11 0.01 15 0.04 17 0.09 
 >60 y 41 10  31 20  24 27  
Sex Male 25 12 0.4 17 20 0.2 12 25 0.2 
 Female 28  22 15  18 19  
Location Proximal 31 0.0002 28 6 × 10−7 21 12 0.0004 
 Distal 22 19  11 30  32  
Dukes stage A/B 27 11 0.9 16 22 0.06 11 27 0.04 
 C/D 26 10  23 13  19 17  
Grade 1/2 12 0.5 1.0 10 0.5 
 3/4 41 18  31 28  25 34  
BRAF Mutant 15 0.006 15 1 × 10−5 15 9 × 10−8 
 Wild-type 38 21  24 35  15 44  
K-ras Mutant 19 0.007 15 0.02 12 1.0 
 Wild-type 34 20  24 30  22 32  
Table 5.

The association of gene comethylation with clinical variables in adenoma subjects

Comethylated genes ≥2
Comethylated genes ≥3
Comethylated genes = 4
+P+P+P
Total  52 10  37 25  24 38  
Age ≤60 y 18 0.4 12 11 0.4 19 0.008 
 >60 y 34  25 14  20 19  
Sex Male 27 0.9 22 10 0.1 11 21 0.5 
 Female 25  15 15  13 17  
Location Proximal 35 0.005 27 10 0.01 14 23 0.9 
 Distal 17  10 15  10 15  
Dysplasia Low 37 0.2 25 21 0.1 14 23 0.9 
 High 15    10 15  
Comethylated genes ≥2
Comethylated genes ≥3
Comethylated genes = 4
+P+P+P
Total  52 10  37 25  24 38  
Age ≤60 y 18 0.4 12 11 0.4 19 0.008 
 >60 y 34  25 14  20 19  
Sex Male 27 0.9 22 10 0.1 11 21 0.5 
 Female 25  15 15  13 17  
Location Proximal 35 0.005 27 10 0.01 14 23 0.9 
 Distal 17  10 15  10 15  
Dysplasia Low 37 0.2 25 21 0.1 14 23 0.9 
 High 15    10 15  

Association of BRAF and K-ras Mutations with Tumor Methylation

BRAF V600E and K-ras codons 12 and 13 mutations were found in 20% (15 of 74) and 27% (20 of 74) of cancers and were mutually exclusive. All BRAF and K-ras mutations occurred in tumors exhibiting methylation in at least one of the four study genes, and addition of neither BRAF nor K-ras mutations improved sensitivity over the most informative methylation marker alone. BRAF was strongly associated with gene comethylation; each of the 15 cancers with BRAF mutations also showed methylation in all four study genes (odds ratio, ∞; P = 9 × 10−8; Fig. 7; Table 4). Most cancers (19 of 20) with mutant K-ras also showed methylation of two or more genes (odds ratio, 11.2; P = 0.007; Fig. 7; Table 4), but this association was not apparent when tumors were dichotomized into those with all four genes methylated and those with less than four genes (Fig. 7; Table 4).

Re-expression of Methylated Genes in Colon Cancer Cell Lines by Demethylation

In SNUC4, HCT15, and WIDR cell lines, all four genes were found to be methylated; in the SW620 cell line, methylation was found in BMP3 and ALX4 genes; and in the SW480 cell line, only the ALX4 gene was methylated (Fig. 8). Suppression of mRNA expression in these genes was generally observed in the methylated cell lines without 5-aza-deoxycytidine treatment. With the 5-aza-deoxycytidine treatment, BMP3 mRNA was re-expressed from an undetectable level in HCT15 and increased by 22-fold, 26-fold, or 3225-fold in SNUC4, SW620, and WIDR cell lines, respectively. No changes in mRNA expression of BMP3 were observed in the unmethylated cell SW480. EYA2 mRNA was increased or re-expressed by 5-aza-deoxycytidine in methylated cell lines SNUC4 and WIDR but also in an unmethylated cell line SW620. ALX4 mRNA was re-expressed from an undetectable level in four of five methylated cancer cells, SNUC4, HCT15, SW620, and WIDR; vimentin mRNA expression was increased by 8-fold, 147-fold, and 346-fold, respectively, in the methylated cells SNUC4, HCT15, and WIDR, but only slightly changed in the unmethylated cells SW480 and SW620 (Table 6).

Figure 8.

Methylation status of colon cancer cell lines checked with MSP. PCR products in lanes U or M indicates the presence of unmethylated or methylated genes, respectively. Universally methylated DNA and human genomic DNA were used as positive controls for methylation and unmethylation. Water was used as negative control.

Figure 8.

Methylation status of colon cancer cell lines checked with MSP. PCR products in lanes U or M indicates the presence of unmethylated or methylated genes, respectively. Universally methylated DNA and human genomic DNA were used as positive controls for methylation and unmethylation. Water was used as negative control.

Close modal
Table 6.

The effect of 5-aza-deoxycytidine treatment on the mRNA expression levels of genes in colon cancer cells

CellFold change after 5-aza-deoxycytidine treatment
BMP3EYA2ALX4Vimentin
SW480 −0.1 −0.1 +0.5 +0.3 
SNUC4 +22 +3 Re-expressed* +8 
HCT15 Re-expressed* Re-expressed* +147 
SW620 +26 Re-expressed* Re-expressed* +0.5 
WIDR +3225 Re-expressed* Re-expressed* +346 
CellFold change after 5-aza-deoxycytidine treatment
BMP3EYA2ALX4Vimentin
SW480 −0.1 −0.1 +0.5 +0.3 
SNUC4 +22 +3 Re-expressed* +8 
HCT15 Re-expressed* Re-expressed* +147 
SW620 +26 Re-expressed* Re-expressed* +0.5 
WIDR +3225 Re-expressed* Re-expressed* +346 
*

mRNA expression was only detected in these cells with 5-aza-deoxycytidine treatment and was not detectable without 5-aza-deoxycytidine treatment.

Methylated genes have been detected in the blood and stool of patients with colorectal cancer and proposed as candidate screening markers (8, 9, 11, 14-20). In this study, we found four genes, BMP3, EYA2, ALX4, and vimentin, to be methylated in the majority of both colorectal cancers and premalignant adenomas. As these methylated gene markers were rarely found in normal epithelia, their methylation seems to be neoplasm specific or cancer related (type C; ref. 51). Each of these candidate markers can be considered for further evaluation in screening or diagnostic applications for colorectal neoplasia because of their broad coverage and early onset in the tumorigenesis of colorectal cancer.

Of note, the four methylation markers evaluated in the current study were found in the same subset of neoplasms and were associated with certain clinical features and genetic alterations. Comethylation of these markers was particularly associated with BRAF mutations, proximal colon location, and older age, which is consistent with the previous reports of the so-called CIMP (22, 26-28, 52). K-ras gene mutation occurred almost exclusively (95%) in cancers with methylation of two or more genes, but the relationship was lost when comparing tumor subsets with all four genes methylated against those with fewer than four methylated genes; and this observation suggests the possibility of a mutant K-ras–related CIMP-low group (28).

From a clinical standpoint, this phenomenon of marker comethylation has potential relevance to the performance of methylation markers in colorectal cancer screening. Because comethylation disproportionately affects neoplastic tissue, comethylation could be incorporated into a stringent definition of test positivity to improve specificity if a panel of markers was assayed. For example, when the definition of test positivity in the present study is changed from “methylation of any of the four target genes” to “comethylation of at least two genes,” the false-positive rate drops by 71% (24% rate to 7% rate, respectively) but the true-positive rate for cancer decreases by only 18% (88% rate to 72% rate, respectively).

Due to the biology underlying CIMP, methylation events may be insufficient to provide a panel of completely informative tumor markers. Although methylation markers identified in this study covered most colorectal tumors, no methylation markers were found in an important minority subset. It is not known if the addition of any other tumor-specific methylation markers would have improved lesion detection in the methylation-negative subset because the panel of markers we evaluated was not exhaustive. Some have reported that methylated genes could yield near 100% coverage, such as with the combination of ER, MYOD1, and SFRP1 (32, 41, 53); however, as such genes are also frequently methylated in normal mucosa of older individuals and may be more age-related (type A; ref. 51), nonspecificity rates could be unacceptably high. Thus, to accurately detect both CIMP-positive and CIMP-negative tumors, it would seem logical to consider a screening panel that combines tumor-specific methylated markers with genetic markers mutant in CIMP-negative lesions, as others have suggested (15, 21, 30). In the present study, neither BRAF nor K-ras mutations proved to be complementary to the most informative single methylation marker for tumor detection sensitivity.

Recently, vimentin, ALX4, and BMP3 have been found to be methylated in colorectal cancer (9, 15, 19, 54), and the present study extends and corroborates these findings. Vimentin has been evaluated as a candidate stool marker, alone (9) and in combination with other markers (14), and has proved to be informative for colorectal neoplasia. This is the first report that EYA2 is frequently methylated and epigenetically silenced in cancer. Despite their frequent methylation in colorectal neoplasia, the carcinogenic roles of these genes are not well understood. As inactivation of tumor suppressor genes by aberrant promoter methylation is a mechanism of oncogenesis (55, 56), it is possible that these genes function as tumor suppressors. Evidence from the current study to support this hypothesis includes the findings that methylation is associated with markedly reduced or absent gene expression in colon cancer cell lines and that in vitro demethylation with 5-aza-deoxycytidine re-expressed these genes. Further basic investigation will be helpful to elucidate their cellular function and mechanisms of action.

In summary, BMP3, EYA2, ALX4, and vimentin genes are commonly methylated in colorectal cancers and adenomas but rarely in normal epithelia. Application studies on stool, serum, or other biological samples are indicated to explore the value of these methylated genes as markers for screening colorectal cancer. The comethylation of these four genes across a majority of colorectal cancers supports the existence of a subset that may be broader than the conventionally described CIMP. Such a broad panel of methylation markers has special relevance to neoplasm detection but may need to be distinguished from methylation markers that are mechanistically linked to CIMP-high associated with BRAF mutation and the serrated pathway to carcinogenesis (27, 29). An important minority of colorectal neoplasms does not seem to exhibit gene methylation and may be missed by tests that target methylated genes only. Complementary use of markers that detect nonmethylated or rarely methylated neoplasia is a biologically rational approach to optimize screening sensitivity.

Grant support: Charles Oswald Foundation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

The authors thank Ann Kolb and Mary Devens for collecting samples, Lisa Boardman for providing some samples, David Smith and William Taylor for useful discussion, and Jaci McCormick for clerical help.

1
Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006.
CA Cancer J Clin
2006
;
56
:
106
–30.
2
Mandel JS, Bond JH, Church TR, et al. Reducing mortality from colorectal cancer by screening for fecal occult blood. Minnesota Colon Cancer Control Study.
N Engl J Med
1993
;
328
:
1365
–71.
3
Kronborg O, Fenger C, Olsen J, Jorgensen OD, Sondergaard O. Randomised study of screening for colorectal cancer with faecal-occult-blood test.
Lancet
1996
;
348
:
1467
–71.
4
Hardcastle JD, Chamberlain JO, Robinson MH, et al. Randomised controlled trial of faecal-occult-blood screening for colorectal cancer.
Lancet
1996
;
348
:
1472
–7.
5
Winawer SJ, Flehinger BJ, Schottenfeld D, Miller DG. Screening for colorectal cancer with fecal occult blood testing and sigmoidoscopy.
J Natl Cancer Inst
1993
;
85
:
1311
–8.
6
Osborn NK, Ahlquist DA. Stool screening for colorectal cancer: molecular approaches.
Gastroenterology
2005
;
128
:
192
–206.
7
Ahlquist DA, Skoletsky JE, Boynton KA, et al. Colorectal cancer screening by detection of altered human DNA in stool: feasibility of a multi-target assay panel.
Gastroenterology
2000
;
119
:
1219
–27.
8
Ahlquist DA, Klatt KK, Harrington JJ, Cunningham JM. Novel Use of hypermethylated DNA markers in stool for detection of colorectal cancer: a feasibility study.
Gastroenterology
2002
;
122
:
Suppl A40
.
9
Chen WD, Han ZJ, Skoletsky J, et al. Detection in fecal DNA of colon cancer-specific methylation of the nonexpressed vimentin gene.
J Natl Cancer Inst
2005
;
97
:
1124
–32.
10
Zou H, Harrington JJ, Klatt KK, Ahlquist DA. A sensitive method to quantify human long DNA in stool: relevance to colorectal cancer screening.
Cancer Epidemiol Biomarkers Prev
2006
;
15
:
1115
–9.
11
Zou HZ, Yu BM, Wang ZW, et al. Detection of aberrant p16 methylation in the serum of colorectal cancer patients.
Clin Cancer Res
2002
;
8
:
188
–91.
12
Hoque MO, Topaloglu O, Begum S, et al. Quantitative methylation-specific polymerase chain reaction gene patterns in urine sediment distinguish prostate cancer patients from control subjects.
J Clin Oncol
2005
;
23
:
6569
–75.
13
Belinsky SA, Liechty KC, Gentry FD, et al. Promoter hypermethylation of multiple genes in sputum precedes lung cancer incidence in a high-risk cohort.
Cancer Res
2006
;
66
:
3338
–44.
14
Itzkowitz SH, Jandorf L, Brand R, et al. An improved fecal DNA test for colorectal cancer screening.
Clin Gastroenterol Hepatol
2007
;
5
:
111
–7.
15
Kann L, Han J, Ahlquist DA et al. Improved marker combination for detection of de novo genetic variation and aberrant DNA in colorectal neoplasia.
Clin Chem
2006
;
52
:
2299
–302.
16
Petko Z, Ghiassi M, Shuber A, et al. Aberrantly methylated CDKN2A, MGMT, and MLH1 in colon polyps and in fecal DNA from patients with colorectal polyps.
Clin Cancer Res
2005
;
11
:
1203
–9.
17
Muller HM, Oberwalder M, Fiegl H, et al. Methylation changes in faecal DNA: a marker for colorectal cancer screening?
Lancet
2004
;
363
:
1283
–5.
18
Leung WK, To KF, Man EP, et al. Detection of epigenetic changes in fecal DNA as a molecular screening test for colorectal cancer: a feasibility study.
Clin Chem
2004
;
50
:
2179
–82.
19
Ebert MP, Model F, Mooney S, et al. Aristaless-like homeobox-4 gene methylation is a potential marker for colorectal adenocarcinomas.
Gastroenterology
2006
;
131
:
1418
–30.
20
Grady WM, Rajput A, Lutterbaugh JD, Markowitz SD. Detection of aberrantly methylated hMLH1 promoter DNA in the serum of patients with microsatellite unstable colon cancer.
Cancer Res
2001
;
61
:
900
–2.
21
Issa JP, Shen L, Toyota M. CIMP, at last.
Gastroenterology
2005
;
129
:
1121
–4.
22
Toyota M, Ahuja N, Ohe-Toyota M, et al. CpG island methylator phenotype in colorectal cancer.
Proc Natl Acad Sci U S A
1999
;
96
:
8681
–6.
23
Eads CA, Lord RV, Wickramasinghe K, et al. Epigenetic patterns in the progression of esophageal adenocarcinoma.
Cancer Res
2001
;
61
:
3410
–8.
24
Esteller M, Tortola S, Toyota M, et al. Hypermethylation-associated inactivation of p14(ARF) is independent of p16(INK4a) methylation and p53 mutational status.
Cancer Res
2000
;
60
:
129
–33.
25
Yamashita K, Dai T, Dai Y, Yamamoto F, Perucho M. Genetics supersedes epigenetics in colon cancer phenotype.
Cancer Cell
2003
;
4
:
121
–31.
26
Samowitz WS, Albertsen H, Herrick J, et al. Evaluation of a large, population-based sample supports a CpG island methylator phenotype in colon cancer.
Gastroenterology
2005
;
129
:
837
–45.
27
Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer.
Nat Genet
2006
;
38
:
787
–93.
28
Ogino S, Kawasaki T, Kirkner GJ, et al. Molecular correlates with MGMT promoter methylation and silencing support CpG island methylator phenotype-low (CIMP-low) in colorectal cancer. Gut. Epub 2007 Mar 5.
29
O'Brien MJ, Yang S, Mack C, et al. Comparison of microsatellite instability, CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and traditional adenomas indicates separate pathways to distinct colorectal carcinoma end points.
Am J Surg Pathol
2006
;
30
:
1491
–501.
30
Toyota M, Ohe-Toyota M, Ahuja N, Issa JP. Distinct genetic profiles in colorectal tumors with or without the CpG island methylator phenotype.
Proc Natl Acad Sci U S A
2000
;
97
:
710
–5.
31
Hoque MO, Begum S, Topaloglu O, et al. Quantitative detection of promoter hypermethylation of multiple genes in the tumor, urine, and serum DNA of patients with renal cancer.
Cancer Res
2004
;
64
:
5511
–7.
32
Ahuja N, Li Q, Mohan AL, Baylin SB, Issa JP. Aging and DNA methylation in colorectal mucosa and cancer.
Cancer Res
1998
;
58
:
5489
–94.
33
Herman JG, Umar A, Polyak K, et al. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma.
Proc Natl Acad Sci U S A
1998
;
95
:
6870
–5.
34
Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia.
Cancer Res
1999
;
59
:
793
–7.
35
Wheeler JM, Kim HC, Efstathiou JA, et al. Hypermethylation of the promoter region of the E-cadherin gene (CDH1) in sporadic and ulcerative colitis associated colorectal cancer.
Gut
2001
;
48
:
367
–71.
36
Wales MM, Biel MA, el Deiry W, et al. p53 activates expression of HIC-1, a new candidate tumour suppressor gene on 17p13.3.
Nat Med
1995
;
1
:
570
–7.
37
Lee S, Hwang KS, Lee HJ, Kim JS, Kang GH. Aberrant CpG island hypermethylation of multiple genes in colorectal neoplasia.
Lab Invest
2004
;
84
:
884
–93.
38
Dammann R, Li C, Yoon JH, et al. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3.
Nat Genet
2000
;
25
:
315
–9.
39
Sakakura C, Hagiwara A, Miyagawa K, et al. Frequent downregulation of the runt domain transcription factors RUNX1, RUNX3 and their cofactor CBFB in gastric cancer.
Int J Cancer
2005
;
113
:
221
–8.
40
Li H, Myeroff L, Smiraglia D, et al. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers.
Proc Natl Acad Sci U S A
2003
;
100
:
8412
–7.
41
Zou H, Molina JR, Harrington JJ, et al. Aberrant methylation of secreted frizzled-related protein genes in esophageal adenocarcinoma and Barrett's esophagus.
Int J Cancer
2005
;
116
:
584
–91.
42
Zou H, Osborn NK, Harrington JJ, et al. Frequent methylation of eyes absent 4 gene in Barrett's esophagus and esophageal adenocarcinoma.
Cancer Epidemiol Biomarkers Prev
2005
;
14
:
830
–4.
43
Osborn NK, Zou H, Molina JR, et al. Aberrant methylation of the eyes absent 4 gene in ulcerative colitis-associated dysplasia.
Clin Gastroenterol Hepatol
2006
;
4
:
212
–8.
44
Dai Z, Lakshmanan RR, Zhu WG, et al. Global methylation profiling of lung cancer identifies novel methylated genes.
Neoplasia
2001
;
3
:
314
–23.
45
Liang G, Robertson KD, Talmadge C, Sumegi J, Jones PA. The gene for a novel transmembrane protein containing epidermal growth factor and follistatin domains is frequently hypermethylated in human tumor cells.
Cancer Res
2000
;
60
:
4907
–12.
46
Akiyama Y, Watkins N, Suzuki H, et al. GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer.
Mol Cell Biol
2003
;
23
:
8429
–39.
47
Weber M, Davies JJ, Wittig D, et al. Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells.
Nat Genet
2005
;
37
:
853
–62.
48
Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer.
Nat Genet
1999
;
21
:
103
–7.
49
Kondo Y, Shen L, Issa JP. Critical role of histone methylation in tumor suppressor gene silencing in colorectal cancer.
Mol Cell Biol
2003
;
23
:
206
–15.
50
Berggren P, Kumar R, Sakano S, et al. Detecting homozygous deletions in the CDKN2A (p16(INK4a))/ARF(p14(ARF)) gene in urinary bladder cancer using real-time quantitative PCR.
Clin Cancer Res
2003
;
9
:
235
–42.
51
Toyota M, Issa JP. CpG island methylator phenotypes in aging and cancer.
Semin Cancer Biol
1999
;
9
:
349
–57.
52
Goel A, Nagasaka T, Arnold CN, et al. The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer.
Gastroenterology
2007
;
132
:
127
–38.
53
Suzuki H, Gabrielson E, Chen W, et al. A genomic screen for genes upregulated by demethylation and histone deacetylase inhibition in human colorectal cancer.
Nat Genet
2002
;
31
:
141
–9.
54
Koinuma K, Kaneda R, Toyota M, et al. Screening for genomic fragments that are methylated specifically in colorectal carcinoma with a methylated MLH1 promoter.
Carcinogenesis
2005
;
26
:
2078
–85.
55
Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer.
Nat Rev Genet
2002
;
3
:
415
–28.
56
Jones PA, Laird PW. Cancer epigenetics comes of age.
Nat Genet
1999
;
21
:
163
–7.
57
Zhang L, Yang N, Huang J, et al. Transcriptional coactivator Drosophila eyes absent homologue 2 is up-regulated in epithelial ovarian cancer and promotes tumor growth.
Cancer Res
2005
;
65
:
925
–32.