Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2007 Dec;9(12):1091–1098. doi: 10.1593/neo.07760

IGFBP3 Promoter Methylation in Colorectal Cancer: Relationship with Microsatellite Instability, CpG Island Methylator Phenotype, and p531

Takako Kawasaki *,2, Katsuhiko Nosho *,2, Mutsuko Ohnishi *,2, Yuko Suemoto *, Gregory J Kirkner , Charles S Fuchs *,, Shuji Ogino *,
PMCID: PMC2134905  PMID: 18084616

Abstract

Insulin-like growth factor binding protein 3 (IGFBP3), which is induced by wild-type p53, regulates IGF and interacts with the TGF-β pathway. IGFBP3 promoter methylation may occur in colorectal cancer with or without the CpG island methylator phenotype (CIMP), which is associated with microsatellite instability (MSI) and TGFBR2 mutation. We examined the relationship between IGFBP3 methylation, p53 expression, CIMP and MSI in 902 population-based colorectal cancers. Utilizing real-time PCR (MethyLight), we quantified promoter methylation in IGFBP3 and eight other CIMP-high-specific promoters (CACNA1G, CDKN2A, CRABP1, IGF2, MLH1, NEUROG1, RUNX3, and SOCS1). IGFBP3 methylation was far more frequent in non-MSI-high CIMP-high tumors (85% = 35/41) than in MSI-high CIMP-high (49% = 44/90, P < .0001), MSI-high non-CIMP-high (17% = 6/36, P < .0001), and non-MSI-high non-CIMP-high tumors (22% = 152/680, P < .0001). Among CIMP-high tumors, the inverse relationship between MSI and IGFBP3 methylation persisted in p53-negative tumors (P < .0001), but not in p53-positive tumors. IGFBP3 methylation was associated inversely with TGFBR2 mutation in MSI-high non-CIMP-high tumors (P = .02). In conclusion, IGFBP3 methylation is inversely associated with MSI in CIMP-high colorectal cancers, and this relationship is limited to p53-negative tumors. Our data suggest complex relationship between global genomic/epigenomic phenomena (such as MSI/CIMP), single molecular events (e.g., IGFBP3 methylation, TP53 mutation, and TGFBR2 mutation), and the related pathways.

Keywords: Colon cancer, IGFBP3, methylation, CIMP, MSI

Introduction

Insulin-like growth factor binding protein 3 (IGFBP3) is the main carrier of insulin-like growth factors (IGFs) in the circulation, where this complex regulates biologic function of IGFs [1]. IGFBP3 has been shown to regulate both cell growth and death, independently of its interaction with IGFs [2,3]. IGFBP3 promoter methylation and gene silencing are observed in human cancers including colorectal cancer [4,5], and have been associated with poor clinical outcome in lung and ovarian cancers [6,7]. IGFBP3 is induced by wild-type p53 [8], and promoter methylation at the p53 regulatory element causes gene silencing resistant to p53 [9]. IGFBP3 enhances the p53-dependent apoptotic response of colorectal adenoma cells to DNA damage [10]. IGFBP3 is known to interact with the transforming growth factor-beta (TGF-β) pathway [11–13].

Transcriptional inactivation by cytosine methylation at promoter CpG islands of tumor suppressor genes is an important mechanism in human carcinogenesis [14]. A number of tumor suppressor genes can be silenced by promoter methylation in colorectal cancers [14]. A subset of colorectal cancers exhibit widespread promoter CpG island methylation, which is referred to as the CpG island methylator phenotype (CIMP) [15]. CIMP-high colorectal tumors have a distinct clinical, pathologic, and molecular profile, such as associations with proximal tumor location, female, poor differentiation, BRAF mutation, wild-type tumor protein p53 (TP53), and inactive WNT/CTNNB1 (β-catenin) [16–24], independent of microsatellite instability (MSI) status [19–23]. In addition, CIMP-high in microsatellite instability-high (MSI-H) colorectal cancer is correlated with the transforming growth factor β receptor type 2 gene (TGFBR2) mononucleotide mutation [25]. Molecular classification of colorectal cancer based on CIMP and MSI is increasingly important [26,27] because MSI and CIMP represent global genomic and epigenomic phenomena, respectively, in tumor cells, and largely determine pathologic and molecular features of colorectal cancer [27].

In this study, using quantitative DNA methylation analysis (MethyLight) and a large number of population-based colorectal cancers, we have examined the relationship between IGFBP3 promoter methylation and various molecular features in colorectal cancer, including MSI, CIMP, p53, and mutations in TGFBR2 and BCL2-associated X protein (BAX). Discovering molecular correlates is important in cancer research, because it may: 1) provide clues to pathogenesis; 2) propose or support the existence of a new molecular subtype; 3) alert investigators to be aware of potential confounding in association studies; and 4) suggest surrogate markers in clinical or research settings [27].

Materials and Methods

Study Group

We used the databases of two large prospective cohort studies: the Nurses' Health Study (N = 121,700 women followed since 1976) [28] and the Health Professionals Follow-Up Study (N = 51,500 men followed since 1986) [29]. Informed consent was obtained from all participants before inclusion in the cohorts. A subset of the cohort participants developed colorectal cancers during prospective follow-up. Thus, these colorectal cancers represented population-based, relatively unbiased samples (compared to retrospective or single-hospital-based samples). Previous studies on the cohorts have described baseline characteristics of cohort participants and incident colorectal cancer cases, and confirmed that our colorectal cancer cases were well-represented as a population-based sample [28,29]. We collected paraffin-embedded tissue blocks from hospitals where cohort participants with colorectal cancers had undergone resections of primary tumors. We excluded cases if adequate paraffin-embedded tumor tissue was not available at the time of the study. As a result, a total of 902 colorectal cancer cases (405 from men's cohort and 497 from women's cohort) were included. Among our cohort studies, there was no significant difference in demographic features between cases with tissue available and those without available tissue [30]. Many of the cases have been previously characterized for status of CIMP, MSI, KRAS, and BRAF [23]. However, no tumor has been examined for IGFBP3 methylation in our previous studies. Tissue collection and analyses were approved by the Dana-Farber/Harvard Cancer Center and Brigham and Women's Hospital Institutional Review Boards.

Histopathologic Evaluations

Hematoxylin and eosin (H&E)-stained tissue sections were examined under a light microscope by one of the investigators (S.O.) blinded from clinical and other laboratory data as previously described [22]. The following eight features were evaluated: 1) the presence and extent of extracellular mucin were categorized as negative (no mucin), < 50%, or ≥50% of the tumor volume; 2) the presence and extent of signet ring cells were categorized as negative no signet ring cells), < 50%, or ≥ 50% of the tumor volume; 3) degree of tumor differentiation was categorized as well/moderate (≥ 50% gland formation) versus poor (< 50% gland formation); 4) to 6) the degree of Crohn's-like lymphoid reaction (defined as transmural lymphoid aggregates), the degree of a peritumoral lymphocytic reaction (defined as a discrete lymphoid infiltrate surrounding tumor cell nests), and the degree of tumor-infiltrating lymphocytes were graded as absent/mild versus moderate/severe; 7) the extent of extraglandular tumor necrosis was graded as < 20% vs ≥ 20%; and 8) the type of tumor border was categorized as circumscribed versus infiltrative.

Genomic DNA Extraction and Whole Genome Amplification

Genomic DNA was extracted from dissected tumor tissue sections using QIAmp DNA Mini Kit (Qiagen, Valencia, CA) as previously described [31]. Normal DNA was obtained from colonic tissue at resection margins. Whole genome amplification of genomic DNA was performed by polymerase chain reaction (PCR) using random 15-mer primers for subsequent MSI and loss of heterozygosity (LOH) analyses and KRAS and BRAF sequencing [31]. Previous studies by us and others showed that whole genome amplification did not significantly affect subsequent genetic analysis [31,32].

Analyses for MSI and 18q LOH

Methods to analyze for MSI and TGFBR2 mutation have been previously described [25,33]. In addition to the recommended MSI panel consisting of D2S123, D5S346, D17S250, BAT25, and BAT26 [34], we also used BAT40, D18S55, D18S56, D18S67, and D18S487 (i.e., 10-marker panel) [33]. A high degree of MSI (MSI-H) was defined as the presence of instability in ≥ 30% of the markers. A low degree of MSI MSI-L) was defined as the presence of instability in < 30% of the markers, and microsatellite stable (MSS) tumors were defined as tumors without an unstable marker. PCR primers for BAX mononucleotide repeat were: BAX-F, FAM-5′-atc cag gat cga gca ggg cg-3′; BAX-R, 5′-act cgc tca gct tct tgg tg-3′. PCR cycles consisted of initial denaturing at 94°C for 2 minutes, followed by 45 cycles of 94°C for 30 seconds, 55°C for 30 seconds, and 72°C for 30 seconds; and a final extension at 72°C for 2 minutes. PCR products were analyzed by ABI 3730 Applied Biosystems, Foster City, CA). The presence of a peak at an altered size in tumor DNA compared to normal DNA was interpreted as positivity for BAX mutation.

For 18q LOH analysis using microsatellite markers D18S55, D18S56, D18S67, and D18S487, we duplicated PCR reaction and electrophoresis in each sample to exclude allele dropouts of one of two alleles [33]. LOH at each locus was defined as 40% or greater reduction of one of two allele peaks in tumor DNA relative to normal DNA. Overall 18q LOH positivity was defined as the presence of one or more markers with LOH, and overall 18q LOH negativity as the presence of two or more informative markers and the absence of LOH in all informative markers.

Sequencing of KRAS and BRAF

Methods of PCR and sequencing targeted for KRAS codons 12 and 13, and BRAF codon 600 have been previously described [31,35]. Pyrosequecing was performed using the PSQ96 HS System (Biotage AB and Biosystems, Uppsala, Sweden) according to the manufacturer's instructions.

Real-Time PCR (MethyLight) for Quantitative DNA Methylation Analysis

Sodium bisulfite treatment on genomic DNA was performed as previously described [36]. Real-time PCR to measure DNA methylation (MethyLight) was performed as previously described [37]. Using ABI 7300 (Applied Biosystems), we examined IGFBP3 promoter and eight other CIMP-specific promoters [calcium channel, voltage-dependent, T type alpha-1G subunit (CACNA1G), cyclin-dependent kinase inhibitor 2A (CDKN2A) (p16), cellular retinoic acid binding protein 1 (CRABP1), insulin-like growth factor 2 (IGF2), MLH1, neurogenin 1 (NEUROG1), runt-related transcription factor 3 (RUNX3), and suppressor of cytokine signaling 1 (SOCS1)] [20,24]. We have shown that these eight markers are sensitive and specific markers for CIMP-high [23]. The collagen 2A1 gene (COL2A1) was used to normalize for the amount of input bisulfite-converted DNA [36]. The primers and probe for IGFBP3 were (bisulfite-converted nucleotides are in italics): IGFBP3-F, 5′-GT T TCG GGC GTG AGT ACG A-3′ (Gen-Bank No. M35878, nucleotides 1692–1710); IGFBP3-R, 5′-GAA TCG ACG CAA ACA CGA CTA C-3′ (GenBank No. M35878, nucleotides 1789–1810); and IGFBP3-probe, 6FAM-5′-TCG GTT GT T TAG GGC GAA GTA CGG G-3′-BHQ-1 (GenBank No. M35878, nucleotides 1760–1784) [38]. Other primers and probes were previously described [24]. The PCR condition was as follows: initial denaturation at 95°C for 10 minutes followed by 45 cycles at 95°C for 15 seconds and 60°C for 1 minute. A standard curve was made for each PCR plate by duplicated PCR amplifications for COL2A1 on bisulfite-converted human genomic DNA at four different concentrations (in a 5-fold dilution series). The percentage of methylated reference (PMR; i.e., degree of methylation) at a specific locus was calculated by dividing the GENE/COL2A1 ratio of template amounts in a sample by the GENE/COL2A1 ratio of template amounts in SssI-treated human genomic DNA (presumably fully methylated) and multiplying this value by 100 [37]. A PMR cutoff value of 4 (except for 6 in CRABP1 and IGF2) was based on previously validated data [36]. Precision and performance characteristics of bisulfite conversion and subsequent MethyLight assays have been previously evaluated and the assays have been validated [36]. CIMP-high was defined as the presence of ≥ 6 of 8 methylated promoters, CIMP-low as 1 to 5 of 8 methylated promoters, and CIMP-0 as the absence (0 of 8) of methylated promoters, according to the previously established criteria [23].

Tissue Microarrays (TMAs) and Immunohistochemistry for p53 and p21 (CDKN1A)

Tissue microarrays were constructed as previously described [39]. TMAs were constructed using the Automated Arrayer (Beecher Instruments, Sun Prairie, WI). Briefly, two 0.6-mm tissue cores each from a tumor and normal colonic mucosa were placed in each TMA block. Each TMA block will have a total of approximately 400 cores (100 cases). We examined two to four tumor tissue cores for each marker. A previous validation study have shown that examining two TMA cores can yield comparable results to examining whole tissue sections in more than 95% of cases [40]. We examined whole tissue sections for p21 in all cases, and for p53 in cases for which no tissue block was available for TMAs or results were equivocal in TMAs. Immunohistochemistry for p53 and p21 was performed as previously described [41,42]. p53 positivity was defined as 50% or more of tumor cells with unequivocal strong nuclear staining, as this high threshold has been shown to improve specificity [43]. p21 loss was defined as less than 5% of tumor cells with nuclear staining. Appropriate positive and negative controls were included in each run of immunohistochemistry. All immunohistochemically stained slides were interpreted by one of the investigators (S.O.) blinded from any other clinical and laboratory data.

Statistical Analysis

In the statistical analysis, chi-square test (or Fisher's exact test when the number in any category was less than 10) was performed for categorical data, and kappa coefficients were calculated to determine the degree of agreement between two observers, using SAS program (Version 9.1, SAS Institute, Cary, NC). All P values were two-sided and statistical significance was set at P ≤ .05.

Results

IGFBP3 Promoter Methylation Is Correlated with CIMP-High

Using MethyLight technology, we quantified DNA methylation in IGFBP3 and a panel of eight promoters (CACNA1G, CDKN2A, CRABP1, IGF2, MLH1, NEUROG1, RUNX3, and SOCS1) [23,24]. The latter eight promoters constitute a sensitive and specific marker panel for CIMP [23]. Among the 902 tumors, 258 (29%) were positive for IGFBP3 promoter methylation. There was no significant difference in the frequencies of IGFBP3 methylation between men (28%) and women (30%). Sensitivity and specificity of IGFBP3 methylation for the diagnosis of CIMP-high (≥ 6 of 8 methylated promoters, not including IGFBP3) were 60% and 77%, respectively (Table 1). Thus, IGFBP3 methylation was not an excellent marker for CIMP-high, but was still positively correlated with CIMP-high (P < .0001). Because 5/8 methylated tumors showed borderline features between CIMP-high and CIMP-low [23], we excluded those tumors from further analyses.

Table 1.

Sensitivity and Specificity of IGFBP3 Methylation for the Diagnosis of CIMP-High.

CIMP-High (≥ 6/8 Methylated Promoters) Non-CIMP-High (≤ 5/8 Methylated Promoters) Total
IGFBP3 Methylation Positive 79 (sensitivity 60%*) 179 (23%) 258
Negative 53 (40%) 591 (specificity 77%) 644
Total 132 770 902
*

Sensitivity is defined as the number of IGFBP3-positive CIMP-high cases divided by the number of all CIMP-high cases.

Specificity is defined as the number of IGFBP3-negative non-CIMP-high cases divided by the number of all non-CIMP-high cases. CIMP, CpG island methylator phenotype.

We also quantified IGFBP3 methylation in normal colon tissue in four IGFBP3-methylated tumor cases and 12 IGFBP3-unmethylated tumor cases. Only one normal sample among the four IGFBP3-methylated tumor cases showed IGFBP3 methylation, and all of the other normal samples showed unmethylated IGFBP3.

Inverse Relationship between IGFBP3 and MSI in CIMP-High Tumors

Because molecular classification based on MSI and CIMP status is increasingly important [26], we stratified tumors into four categories according to MSI and CIMP status (Figure 1). Within CIMP-high tumors, the frequency of IGFBP3 methylation was significantly higher in MSI-L/MSS CIMP-high tumors (85%) than MSI-H CIMP-high tumors (49%, P < .0001), indicating an inverse relationship between IGFBP3 methylation and MSI in CIMP-high tumors. CIMP-low/0 tumors showed low frequencies (17–22%) of IGFBP3 methylation regardless of MSI status.

Figure 1.

Figure 1

Frequency of IGFBP3 methylation in four MSI/CIMP subtypes of colorectal cancer. CIMP, CpG island methylator phenotype; MSI, microsatellite instability; MSS, microsatellite stable.

IGFBP3 Methylation, CIMP, and Pathologic Features

Because IGFBP3 methylation is positively correlated with CIMP-high, we stratified tumors according to IGFBP3 and CIMP status in subsequent analyses (as in Tables 2 and 3). Then, we could examine the effect of IGFBP3 methylation on various pathologic and molecular features independent of CIMP status.

Table 2.

Frequencies of Specific Clinical and Pathologic Features in Colorectal Cancer according to IGFBP3 Methylation and CIMP Status.

Clinical and Pathologic Features N All Cases P CIMP-High P CIMP-Low/0 P



IGFBP3 Methylation IGFBP3 Methylation IGFBP3 Methylation



(+) (-) (+) (-) (+) (-)
All Cases 902 259 643 80 53 161 582
Men 405 112 293 29 14 74 275
Women 497 147 350 51 39 87 307
Tumor Location
Total Examined 522 148 374 48 33 87 334
Proximal 248 97 (66%) 151 (40%) < .0001 44 (92%) 31 (94%) 42 (48%) 117 (35%) .02
Distal 274 51 (34%) 223 (60%) 4 (8.3%) 2 (6.1%) 45 (52%) 217 (65%)
Tumor Differentiation
Total Examined 885 255 630 80 53 158 568
Well/Moderate 802 221 (87%) 581 (92%) .01 57 (71%) 36 (68%) 148 (94%) 538 (95%)
Poor 83 34 (13%) 49 (7.8%) 23 (29%) 17 (32%) 10 (6.3%) 30 (5.3%)
Mucinous/Signet Ring Cell Features
Total Examined 782 228 554 76 49 136 497
Nonmucinous Carcinoma 471 100 (44%) 371 (67%) < .0001 26 (34%) 20 (41%) 69 (51%) 346 (70%) < .0001
Mucinous
1100% 311 128 (56%) 183 (33%) 50 (66%) 29 (59%) 67 (49%) 151 (30%)
1–49% 191 78 (34%) 113 (20%) 24 (32%) 12 (24%) 45 (33%) 100 (20%)
≥ 50% 120 50 (22%) 70 (13%) 26 (34%) 17 (35%) 22 (16%) 51 (10%)
Nonsignet Ring Cell Carcinoma 721 207 (91%) 514 (93%) 62 (82%) 39 (80%) 130 (96%) 468 (94%)
Signet Ring Cells
1–100% 61 21 (9.2%) 40 (7.2%) 14 (18%) 10 (20%) 6 (4.4%) 29 (5.8%)
1–49% 46 17 (7.5%) 29 (5.2%) 12 (16%) 9 (18%) 4 (2.9%) 19 (3.8%)
≥ 50% 15 4 (1.8%) 11 (2.0%) 2 (2.6%) 1 (2.0%) 2 (1.5%) 10 (2.0%)
Tumor-Infiltrating Lymphocytes
Total Examined 877 251 626 79 51 155 566
Absent/Mild 778 210 (84%) 568 (91%) .003 50 (63%) 39 (57%) 145 (94%) 533 (94%)
Moderate/Severe 99 41 (16%) 58 (9.3%) 29 (37%) 22 (43%) 10 (6.5%) 33 (5.8%)
Crohn's-Like Reaction
Total Examined 674 198 476 58 41 124 428
Absent/Mild 611 172 (87%) 439 (92%) .03 38 (66%) 28 (68%) 120 (97%) 404 (94%)
Moderate/Severe 63 26 (13%) 37 (7.7%) 20 (34%) 13 (32%) 4 (3.2%) 24 (5.6%)
Peritumoral Lymphocytic Reaction
Total Examined 879 275 604 76 51 182 541
Absent/Mild 782 241 (88%) 541 (90%) 47 (63%) 39 (57%) 172 (95%) 493 (91%)
Moderate/Severe 97 34 (12%) 63 (10%) 29 (37%) 22 (43%) 10 (5.5%) 48 (8.9%)
Tumor Border
Total Examined 726 219 507 69 43 136 455
Circumscribed 262 74 (34%) 188 (37%) 26 (38%) 16 (37%) 45 (33%) 167 (37%)
Infiltrative 464 145 (66%) 319 (63%) 43 (62%) 27 (63%) 91 (67%) 288 (63%)
Extraglandular Necrosis
Total Examined 896 259 637 80 52 161 576
< 20% 802 230 (89%) 572 (90%) 64 (80%) 37 (71%) 149 (93%) 526 (91%)
< 20% 94 29 (11%) 65 (10%) 16 (20%) 15 (29%) 12 (7.5%) 50 (8.7%)

Only significant P values are described.

CIMP, CpG island methylator phenotype.

Table 3.

Frequencies of Specific Molecular Features in Colorectal Cancer according to IGFBP3 Methylation and CIMP Status.

Molecular Features N All Cases P CIMP-High P CIMP-Low/0 P



IGFBP3 Methylation IGFBP3 Methylation IGFBP3 Methylation



(+) (-) (+) (-) (+) (-)
MSI
Total Examined 874 255 619 79 52 158 558
MSI-H 127 50 (20%) 77 (12%) 44 (56%) 46 (88%) < .0001 6 (3.8%) 30 (5.4%)
MSI-L/MSS 747 205 (80%) 542 (88%) 35 (44%) 6 (12%) 152 (96%) 528 (95%)
KRAS
Total Examined 860 250 610 77 51 155 550
Mutant 313 102 (41%) 211 (35%) 14 (18%) 4 (7.8%) 79 (51%) 204 (37%) .002
Wild-Type 547 148 (59%) 399 (65%) 53 (82%) 47 (92%) 76 (49%) 346 (63%)
BRAF
Total Examined 860 250 610 77 51 155 550
Mutant 112 59 (24%) 53 (8.7%) < .0001 45 (58%) 32 (63%) 8 (5.2%) 18 (3.3%)
Wild-Type 748 191 (76%) 557 (91%) 32 (42%) 19 (37%) 147 (95%) 532 (97%)
18q LOH
Total Examined (Only Non-MSI-H Tumors) 540 151 389 29 5 107 377
(+) 353 85 (56%) 268 (69%) .006 18 (62%) 2 (40%) 63 (59%) 261 (69%) .04
(-) 187 66 (44%) 121 (31%) 11 (38%) 3 (60%) 44 (41%) 116 (31%)
p53*
Total Examined 891 257 634 80 53 159 573
(+) 386 109 (42%) 277 (44%) 21 (26%) 9 (17%) 81 (51%) 263 (46%)
(-) 505 148 (58%) 357 (56%) 59 (74%) 44 (83%) 78 (49%) 310 (54%)
p21*
Total Examined 864 251 613 79 50 155 555
Loss 508 124 (49%) 384 (63%) .0003 22 (28%) 9 (18%) 92 (59%) 371 (67%)
(+) 356 127 (51%) 229 (37%) 57 (72%) 41 (82%) 63 (41%) 184 (33%)
TGFBR2 Mutation
Total Examined (Only MSI-H Tumors) 127 50 77 44 46 6 30
(+) 94 37 (74%) 57 (74%) 37 (84%) 39 (85%) 0 18 (60%) .02
(-) 33 13 (26%) 20 (26%) 7 (16%) 7 (15%) 6 (100%) 12 (40%)
BAX Mutation
Total Examined (Only MSI-H Tumors) 126 50 76 44 45 6 30
(+) 32 18 (36%) 14 (18%) .03 18 (41%) 8 (18%) .02 0 5 (17%)
(-) 94 32 (64%) 62 (82%) 26 (59%) 37 (82%) 6 (100%) 25 (83%)
*

p53 and p21 status was determined by immunohistochemistry.

Only significant P values are described.

CIMP, CpG island methylator phenotype; LOH, loss of heterozygosity; MSI, microsatellite instability.

Table 2 summarizes the relations between IGFBP3 methylation and pathologic features in colorectal cancer. Proximal tumor location, poor differentiation, mucinous features, tumor-infiltrating lymphocytes, and Crohn's-like reaction were significantly associated with IGFBP3 methylation in all cases, but no significant correlations persisted after tumors were stratified by CIMP status. These findings indicate that those features are associated primarily with CIMP, but not directly with IGFBP3 methylation.

IGFBP3 Methylation, CIMP, and Other Molecular Features

Table 3 summarizes the relations between IGFBP3 methylation and other molecular features in colorectal cancer. Whereas MSI and IGFBP3 were not significantly correlated in all cases and CIMP-low/0 cases, MSI and IGFBP3 methylation were inversely correlated in CIMP-high tumors (P < .0001).

Interestingly, IGFBP3 methylation and TGFBR2 mutation were inversely correlated (P = .02) in MSI-H CIMP-low/0 tumors, and IGFBP3 methylation and BAX mutation were positively correlated (P = .02) in MSI-H CIMP-high tumors (Table 3). These results may suggest possible interactions between the pathways related to these molecules (Figure 2).

Figure 2.

Figure 2

The p53 pathway and IGFBP3.

BRAF mutation, 18q LOH, and p21 loss were correlated with IGFBP3 methylation in all cases, but the relations did not persist after tumors were stratified by CIMP status.

Relationship between IGFBP3 Methylation and MSI according to p53 or p21 Status

Because IGFBP3 is one of the downstream effectors of the p53 pathway (Figure 2), we examined the interrelationship between MSI, IGFBP3, and p53 (Figure 3). The inverse relationship between MSI and IGFBP3 methylation (in CIMP-high tumors) persisted in p53-negative tumors (P < .0001), but not in p53-positive tumors. These results suggest that IGFBP3 methylation may be more important in TP53 wildtype tumors than in TP53-mutated tumors, which may have already downregulated IGFBP3.

Figure 3.

Figure 3

Frequency of IGFBP3 methylation in CIMP-high tumors according to p53 and MSI status. Note that the inverse relationship between MSI and IGFBP3 methylation is present in p53-negative tumors, but not in p53-positive tumors. CIMP, CpG island methylator phenotype; MSI, microsatellite instability; MSS, microsatellite stable; NS, not significant.

We also examined the interrelationship between MSI, IGFBP3, and p21, one of the downstream effectors of p53 (Figure 4). The inverse relationship between MSI and IGFBP3 methylation persisted regardless of p21 status, suggesting that p21 and IGFBP3 functions were not directly linked.

Figure 4.

Figure 4

Frequency of IGFBP3 methylation in CIMP-high tumors according to p21 and MSI status. Note that the inverse relationship between MSI and IGFBP3 methylation is present regardless of p21 status. CIMP, CpG island methylator phenotype; MSI, microsatellite instability; MSS, microsatellite stable.

Discussion

We conducted this study to examine IGFBP3 methylation in colorectal cancer, particularly in relation to MSI, CIMP, and p53. Molecular classification of colorectal cancer according to MSI and CIMP is increasingly important [26], because MSI and CIMP represent global genomic and epigenomic phenomena, respectively, in tumor cells, and largely determine clinical, pathologic, and molecular features of colorectal cancer. We have found that IGFBP3 methylation is inversely associated with MSI in CIMP-high tumors, but not in CIMP-low/0 tumors, and that this inverse relationship is limited to p53-negative tumors, but not p53-positive tumors. Our findings imply the complex interrelationship between genomic/epigenomic phenomena (such as MSI and CIMP) and single molecular events, IGFBP3 methylation and p53 alteration, in colorectal cancer.

Transcriptional inactivation of tumor suppressor genes by promoter CpG island methylation is an important mechanism in human carcinogenesis [14]. Epigenetic aberrations have been reported in various tumor-related genes [14,44–47]. For quantitative DNA methylation analysis, we used Methy-Light, which is robust and can reproducibly differentiate low-level methylation from high-level methylation [36,48,49]. Our resource of a large number of colorectal cancers, derived from two large prospective cohorts (relatively unbiased samples compared to retrospective or single-hospital-based samples), has enabled us to precisely estimate the frequency of colorectal cancers with a specific molecular feature (e.g., IGFBP3 methylation, MSI-H, and so forth). The large number of samples has also provided a sufficient power to accurately estimate the frequency of IGFBP3 methylation in rare tumor subtypes, such as MSI-L/MSS CIMP-high, CIMP-high p53-positive, and so forth.

IGFBP3 is one of the important downstream effectors of the p53 pathway [8,10], and is also known to interact with the TGF-β pathway [11–13] (Figure 2). Thus, we have examined the interrelationship between IGFBP3 methylation, p53 positivity, and MSI, and found that the inverse correlation between IGFBP3 methylation and MSI in CIMP-high tumors Table 3. Frequencies of Specific Molecular Features in Colorectal Cancer according to IGFBP3 Methylation and CIMP Status. Molecular Features N All Cases P CIMP-High P CIMP-Low/0 P IGFBP3 Methylation IGFBP3 Methylation IGFBP3 Methylation (+) (MSI Total Examined 874 255 619 79 52 158 558 (+) 353 85 (56%) 210 (54%) p21* Total Examined 864 251 613 79 50 155 555 Loss 508 124 (49%) 384 (63%) .0003 22 (28%) 9 (18%) 92 (59%) 371 (67%) (+) 356 127 (51%) 229 (37%) 57 (72%) 41 (82%) 63 (41%) 184 (33%) TGFBR2 Mutation Total Examined (Only MSI-H Tumors) 127 50 77 44 46 6 30 (+) 94 37 (74%) 57 (74%) 37 (84%) 39 (85%) 0 18 (60%) .02 (BAX Mu is limited to p53-negative tumors. Thus, IGFBP3 methylation may be important in TP53 wild-type tumors, whereas TP53-mutated tumors may have already downregulated IGFBP3 and IGFBP3 methylation may be less relevant. We have also found an inverse correlation between IGFBP3 methylation and TGFBR2 mutation in MSI-H CIMP-low/0 tumors, as well as a positive correlation between IGFBP3 methylation and BAX mutation in MSI-H CIMP-high tumors. These complex correlations suggest the intricate relationship between global genomic/epigenomic phenomena (such as MSI and CIMP), these single molecular events (such as IGFBP3 methylation, TP53 mutation, TGFBR2 mutation, and so forth), and the related pathways in colorectal cancer.

In summary, IGFBP3 promoter methylation in colorectal cancer is inversely associated with MSI in CIMP-high colorectal cancer, and this inverse correlation is limited to p53-negative tumors. Further studies are necessary to elucidate the exact pathogenic role of IGFBP3 promoter methylation in colorectal cancer.

Acknowledgements

We deeply thank the Nurses' Health Study and Health Professionals Follow-Up Study cohort participants who have generously agreed to provide us with biologic specimens and information through responses to questionnaires; hospitals and pathology departments throughout the U.S. for providing us with tumor tissue materials; Walter Willett, Graham Colditz, Sue Hankinson, and many other staff members who implemented and have maintained the cohort studies.

Abbreviations

BAX

BCL2-associated X protein

CACNA1G

calcium channel, voltage-dependent, T type alpha-1G subunit

CDKN1A

cyclin-dependent kinase inhibitor 1A (p21, CIP1)

CDKN2A

cyclin-dependent kinase inhibitor 2A (p16, INK4A)

CIMP

CpG island methylator phenotype

CRABP1

cellular retinoic acid binding protein 1

CTNNB1

β-catenin

IGF2

insulin-like growth factor 2

IGFBP3

insulin-like growth factor binding protein 3

MSI

microsatellite instability

MSI-H

microsatellite instability-high

MSI-L

microsatellite instability-low

MSS

microsatellite stable

NEUROG1

neurogenin 1

PCR

polymerase chain reaction

PMR

percentage of methylated reference (degree of methylation)

RUNX3

runt-related transcription factor 3

SOCS1

suppressor of cytokine signaling 1

TGF

transforming growth factor

TGFBR2

transforming growth factor b receptor type 2

TMA

tissue microarray

TP53

tumor protein p53

Footnotes

This work was supported by the National Institutes of Health/National Cancer Institute grants P01 CA87969, P01 CA55075K07, and K07CA122826, and in part by grants from the Bennett Family Fund and from the Entertainment Industry Foundation (EIF) through the EIF National Colorectal Cancer Research Alliance. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institute of Health.

References

  • 1.Giovannucci E. Nutrition, insulin, insulin-like growth factors and cancer. Horm Metab Res. 2003;35:694–704. doi: 10.1055/s-2004-814147. [DOI] [PubMed] [Google Scholar]
  • 2.Perks CM, McCaig C, Clarke JB, Clemmons DR, Holly JM. A non-IGF binding mutant of IGFBP-3 modulates cell function in breast epithelial cells. Biochem Biophys Res Commun. 2002;294:988–994. doi: 10.1016/S0006-291X(02)00569-7. [DOI] [PubMed] [Google Scholar]
  • 3.Hong J, Zhang G, Dong F, Rechler MM. Insulin-like growth factor (IGF)-binding protein-3 mutants that do not bind IGF-I or IGF-II stimulate apoptosis in human prostate cancer cells. J Biol Chem. 2002;277:10489–10497. doi: 10.1074/jbc.M109604200. [DOI] [PubMed] [Google Scholar]
  • 4.Hanafusa T, Yumoto Y, Nouso K, Nakatsukasa H, Onishi T, Fujikawa T, Taniyama M, Nakamura S, Uemura M, Takuma Y, et al. Reduced expression of insulin-like growth factor binding protein-3 and its promoter hypermethylation in human hepatocellular carcinoma. Cancer Lett. 2002;176:149–158. doi: 10.1016/s0304-3835(01)00736-4. [DOI] [PubMed] [Google Scholar]
  • 5.Tomii K, Tsukuda K, Toyooka S, Dote H, Hanafusa T, Asano H, Naitou M, Doihara H, Kisimoto T, Katayama H, et al. Aberrant promoter methylation of insulin-like growth factor binding protein-3 gene in human cancers. Int J Cancer. 2007;120:566–573. doi: 10.1002/ijc.22341. [DOI] [PubMed] [Google Scholar]
  • 6.Chang YS, Wang L, Liu D, Mao L, Hong WK, Khuri FR, Lee HY. Correlation between insulin-like growth factor-binding protein-3 promoter methylation and prognosis of patients with stage I non-small cell lung cancer. Clin Cancer Res. 2002;8:3669–3675. [PubMed] [Google Scholar]
  • 7.Wiley A, Katsaros D, Fracchioli S, Yu H. Methylation of the insulin-like growth factor binding protein-3 gene and prognosis of epithelial ovarian cancer. Int J Gynecol Cancer. 2006;16:210–218. doi: 10.1111/j.1525-1438.2006.00299.x. [DOI] [PubMed] [Google Scholar]
  • 8.Buckbinder L, Talbott R, Velasco-Miguel S, Takenaka I, Faha B, Seizinger BR, Kley Nm. Induction of the growth inhibitor IGF-binding protein 3 by p53. Nature. 1995;377:646–649. doi: 10.1038/377646a0. [DOI] [PubMed] [Google Scholar]
  • 9.Hanafusa T, Shinji T, Shiraha H, Nouso K, Iwasaki Y, Yumoto E, Ono T, Koide N. Functional promoter upstream p53 regulatory sequence of IGFBP3 that is silenced by tumor specific methylation. BMC Cancer. 2005;5:9. doi: 10.1186/1471-2407-5-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Williams AC, Collard TJ, Perks CM, Newcomb P, Moorghen M, Holly JM, Paraskeva C. Increased p53-dependent apoptosis by the insulin-like growth factor binding protein IGFBP-3 in human colonic adenoma-derived cells. Cancer Res. 2000;60:22–27. [PubMed] [Google Scholar]
  • 11.Kansra S, Ewton DZ, Wang J, Friedman E. IGFBP-3 mediates TGF beta 1 proliferative response in colon cancer cells. Int J Cancer. 2000;87:373–378. doi: 10.1002/1097-0215(20000801)87:3<373::aid-ijc10>3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  • 12.Peters I, Tossidou I, Achenbach J, Woroniecki R, Mengel M, Park JK, Paschy M, de Groot K, Haller H, Schiffer M. IGF-binding protein-3 modulates TGF-beta/BMP-signaling in glomerular podocytes. J Am Soc Nephrol. 2006;17:1644–1656. doi: 10.1681/ASN.2005111209. [DOI] [PubMed] [Google Scholar]
  • 13.Rosendahl AH, Forsberg G. IGF-I and IGFBP-3 augment transforming growth factor-beta actions in human renal carcinoma cells. Kidney Int. 2006;70:1584–1590. doi: 10.1038/sj.ki.5001805. [DOI] [PubMed] [Google Scholar]
  • 14.Laird PW. Cancer epigenetics. Hum Mol Genet. 2005;14:R65–R76. doi: 10.1093/hmg/ddi113. (Spec No 1) [DOI] [PubMed] [Google Scholar]
  • 15.Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999;96:8681–8686. doi: 10.1073/pnas.96.15.8681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hawkins N, Norrie M, Cheong K, Mokany E, Ku SL, Meagher A, O′Connor T, Ward R. CpG island methylation in sporadic colorectal cancers and its relationship to microsatellite instability. Gastroenterology. 2002;122:1376–1387. doi: 10.1053/gast.2002.32997. [DOI] [PubMed] [Google Scholar]
  • 17.van Rijnsoever M, Grieu F, Elsaleh H, Joseph D, Iacopetta B. Characterisation of colorectal cancers showing hypermethylation at multiple CpG islands. Gut. 2002;51:797–802. doi: 10.1136/gut.51.6.797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Whitehall VL, Wynter CV, Walsh MD, Simms LA, Purdie D, Pandeya N, Young J, Meltzer SJ, Leggett BA, Jass JR. Morphological and molecular heterogeneity within nonmicrosatellite instability-high colorectal cancer. Cancer Res. 2002;62:6011–6014. [PubMed] [Google Scholar]
  • 19.Samowitz W, Albertsen H, Herrick J, Levin TR, Sweeney C, Murtaugh MA, Wolff RK, Slattery ML. Evaluation of a large, populationbased sample supports a CpG island methylator phenotype in colon cancer. Gastroenterology. 2005;129:837–845. doi: 10.1053/j.gastro.2005.06.020. [DOI] [PubMed] [Google Scholar]
  • 20.Ogino S, Cantor M, Kawasaki T, Brahmandam M, Kirkner G, Weisenberger DJ, Campan M, Laird PW, Loda M, Fuchs CS. CpG island methylator phenotype (CIMP) of colorectal cancer is best characterised by quantitative DNA methylation analysis and prospective cohort studies. Gut. 2006;55:1000–1006. doi: 10.1136/gut.2005.082933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kawasaki T, Nosho K, Ohnishi M, Suemoto Y, Kirkner GJ, Meyerhardt JA, Fuchs CS, Ogino S. Correlation of beta-catenin localization with cyclooxygenase-2 expression and CpG island methylator phenotype (CIMP) in colorectal cancer. Neoplasia. 2007;9:569–577. doi: 10.1593/neo.07334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ogino S, Odze RD, Kawasaki T, Brahmandam M, Kirkner GJ, Laird PW, Loda M, Fuchs CS. Correlation of pathologic features with CpG island methylator phenotype (CIMP) by quantitative DNA methylation analysis in colorectal carcinoma. Am J Surg Pathol. 2006;30:1175–1183. doi: 10.1097/01.pas.0000213266.84725.d0. [DOI] [PubMed] [Google Scholar]
  • 23.Ogino S, Kawasaki T, Kirkner GJ, Kraft P, Loda M, Fuchs CS. Evaluation of markers for CpG island methylator phenotype (CIMP) in colorectal cancer by a large population-based sample. J Mol Diagn. 2007;9:305–314. doi: 10.2353/jmoldx.2007.060170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38:787–793. doi: 10.1038/ng1834. [DOI] [PubMed] [Google Scholar]
  • 25.Ogino S, Kawasaki T, Ogawa A, Kirkner GJ, Loda M, Fuchs CS. TGFBR2 mutation is correlated with CpG island methylator phenotype in microsatellite instability-high colorectal cancer. Hum Pathol. 2007;38:614–620. doi: 10.1016/j.humpath.2006.10.005. [DOI] [PubMed] [Google Scholar]
  • 26.Jass JR. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology. 2007;50:113–130. doi: 10.1111/j.1365-2559.2006.02549.x. [DOI] [PubMed] [Google Scholar]
  • 27.Ogino S, Goel A. Molecular classification and correlates in colorectal cancer. J Mol Diagn. 2008 doi: 10.2353/jmoldx.2008.070082. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Colditz GA, Hankinson SE. The Nurses' Health Study: lifestyle and health among women. Nat Rev Cancer. 2005;5:388–396. doi: 10.1038/nrc1608. [DOI] [PubMed] [Google Scholar]
  • 29.Giovannucci E, Liu Y, Rimm EB, Hollis BW, Fuchs CS, Stampfer MJ, Willett WC. Prospective study of predictors of vitamin D status and cancer incidence and mortality in men. J Natl Cancer Inst. 2006;98:451–459. doi: 10.1093/jnci/djj101. [DOI] [PubMed] [Google Scholar]
  • 30.Chan AT, Ogino S, Fuchs CS. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. New Engl J Med. 2007;356:2131–2142. doi: 10.1056/NEJMoa067208. [DOI] [PubMed] [Google Scholar]
  • 31.Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, Namgyal C, Mino-Kenudson M, Lauwers GY, Loda M, Fuchs CS. Sensitive sequencing method for KRAS mutation detection by pyrosequencing. J Mol Diagn. 2005;7:413–421. doi: 10.1016/S1525-1578(10)60571-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dietmaier W, Hartmann A, Wallinger S, Heinmoller E, Kerner T, Endl E, Jauch KW, Hofstadter F, Ruschoff J. Multiple mutation analyses in single tumor cells with improved whole genome amplification. Am J Pathol. 1999;154:83–95. doi: 10.1016/S0002-9440(10)65254-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ogino S, Brahmandam M, Cantor M, Namgyal C, Kawasaki T, Kirkner G, Meyerhardt JA, Loda M, Fuchs CS. Distinct molecular features of colorectal carcinoma with signet ring cell component and colorectal carcinoma with mucinous component. Mod Pathol. 2006;19:59–68. doi: 10.1038/modpathol.3800482. [DOI] [PubMed] [Google Scholar]
  • 34.Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58:5248–5257. [PubMed] [Google Scholar]
  • 35.Ogino S, Kawasaki T, Kirkner GJ, Loda M, Fuchs CS. CpG island methylator phenotype-low (CIMP-low) in colorectal cancer: possible associations with male sex and KRAS mutations. J Mol Diagn. 2006;8:582–588. doi: 10.2353/jmoldx.2006.060082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ogino S, Kawasaki T, Brahmandam M, Cantor M, Kirkner GJ, Spiegelman D, Makrigiorgos GM, Weisenberger DJ, Laird PW, Loda M, et al. Precision and performance characteristics of bisulfite conversion and real-time PCR (MethyLight) for quantitative DNA methylation analysis. J Mol Diagn. 2006;8:209–217. doi: 10.2353/jmoldx.2006.050135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Eads CA, Danenberg KD, Kawakami K, Saltz LB, Blake C, Shibata D, Danenberg PV, Laird PW. MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res. 2000;28:E32. doi: 10.1093/nar/28.8.e32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ogino S, Meyerhardt JA, Kawasaki T, Clark JW, Ryan DP, Kulke MH, Enzinger PC, Wolpin BM, Loda M, Fuchs CS. CpG island methylation, response to combination chemotherapy, and patient survival in advanced microsatellite stable colorectal carcinoma. Virchows Arch. 2007;450:529–537. doi: 10.1007/s00428-007-0398-3. [DOI] [PubMed] [Google Scholar]
  • 39.Ogino S, Brahmandam M, Kawasaki T, Kirkner GJ, Loda M, Fuchs CS. Combined analysis of COX-2 and p53 expressions reveals synergistic inverse correlations with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Neoplasia. 2006;8:458–464. doi: 10.1593/neo.06247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Camp RL, Charette LA, Rimm DL. Validation of tissue microarray technology in breast carcinoma. Lab Invest. 2000;80:1943–1949. doi: 10.1038/labinvest.3780204. [DOI] [PubMed] [Google Scholar]
  • 41.Ogino S, Kawasaki T, Kirkner GJ, Yamaji T, Loda M, Fuchs CS. Loss of nuclear p27 (CDKN1B/KIP1) in colorectal cancer is correlated with microsatellite instability and CIMP. Mod Pathol. 2007;20:15–22. doi: 10.1038/modpathol.3800709. [DOI] [PubMed] [Google Scholar]
  • 42.Ogino S, Meyerhardt JA, Cantor M, Brahmandam M, Clark JW, Namgyal C, Kawasaki T, Kinsella K, Michelini AL, Enzinger PC, et al. Molecular alterations in tumors and response to combination chemotherapy with gefitinib for advanced colorectal cancer. Clin Cancer Res. 2005;11:6650–6656. doi: 10.1158/1078-0432.CCR-05-0738. [DOI] [PubMed] [Google Scholar]
  • 43.Hall PA, McCluggage WG. Assessing p53 in clinical contexts: unlearned lessons and new perspectives. J Pathol. 2006;208:1–6. doi: 10.1002/path.1913. [DOI] [PubMed] [Google Scholar]
  • 44.Litkouhi B, Kwong J, Lo CM, Smedley JG, III, McClane BA, Aponte M, Gao Z, Sarno JL, Hinners J, Welch WR, et al. Claudin-4 overexpression in epithelial ovarian cancer is associated with hypomethylation and is a potential target for modulation of tight junction barrier function using a C-terminal fragment of Clostridium perfringens enterotoxin. Neoplasia. 2007;9:304–314. doi: 10.1593/neo.07118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kuester D, Dar AA, Moskaluk CC, Krueger S, Meyer F, Hartig R, Stolte M, Malfertheiner P, Lippert H, Roessner A, et al. Early involvement of death-associated protein kinase promoter hypermethylation in the carcinogenesis of Barrett's esophageal adenocarcinoma and its association with clinical progression. Neoplasia. 2007;9:236–245. doi: 10.1593/neo.06802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kwong J, Lee JY, Wong KK, Zhou X, Wong DT, Lo KW, Welch WR, Berkowitz RS, Mok SC. Candidate tumor-suppressor gene DLEC1 is frequently downregulated by promoter hypermethylation and histone hypoacetylation in human epithelial ovarian cancer. Neoplasia. 2006;8:268–278. doi: 10.1593/neo.05502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Liu Y, Lan Q, Siegfried JM, Luketich JD, Keohavong P. Aberrant promoter methylation of p16 and MGMT genes in lung tumors from smoking and never-smoking lung cancer patients. Neoplasia. 2006;8:46–51. doi: 10.1593/neo.05586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chen J, Rocken C, Lofton-Day C, Schulz HU, Muller O, Kutzner N, Malfertheiner P, Ebert MP. Molecular analysis of APC promoter methylation and protein expression in colorectal cancer metastasis. Carcinogenesis. 2005;26:37–43. doi: 10.1093/carcin/bgh280. [DOI] [PubMed] [Google Scholar]
  • 49.Ebert MP, Mooney SH, Tonnes-Priddy L, Lograsso J, Hoffmann J, Chen J, Rocken C, Schulz HU, Malfertheiner P, Lofton-Day C. Hypermethylation of the TPEF/HPP1 gene in primary and metastatic colorectal cancers. Neoplasia. 2005;7:771–778. doi: 10.1593/neo.05235. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES