Background: Colorectal cancer (CRC) is the second most common cancer in the Kingdom of Saudi Arabia with ever increasing incidence rates. DNA methylation is a common event in CRC where it is now considered an important phenomenon in CRC carcinogenesis and useful for the classification and prognosis of CRC.

Methods: To gain insight into the molecular mechanisms underpinning CRC in Saudi Arabian patients, we profiled the DNA methylation frequency of key genes (MLH1, MSH2, RASSF1A, SLIT2, HIC1, MGMT, SFRP1, MYOD1, APC, CDKN2A, as well as five CIMP markers) in 120 sporadic CRC cases. CRC tumors originating from the rectum, left, and right colons are represented in this cohort of formalin-fixed paraffin-embedded tissues.

Results: The most common methylation frequency was detected in the polycomb group target genes (PCGT) including SFRP1 (70%), MYOD1 (60.8%), HIC1 (61.7%), and SLIT2 (56.7%). In addition, MGMT methylation was detected at a high frequency (68.3%). RASSF1A, APC, and CDKN2A methylation frequencies were 42.5%, 25%, and 32.8%, respectively. K-means clustering analysis of the methylation events results in the clustering of the CRC samples into three groups depending on the level of methylation detected.

Conclusion: Group II (PCGT methylation and CIMP-negative) methylation signature carried a favorable prognosis for male patients, whereas older patients with group I rare methylation signature have a potentially poorer clinical outcome.

Impact: Methylation of the PCGT genes along with RASSF1A, APC, and MGMT can be potentially used as a new biomarker for the classification and prognosis of CRC tumors and independently of where the tumor has originated. Cancer Epidemiol Biomarkers Prev; 21(11); 2069–75. ©2012 AACR.

CRC is the third most common type of cancer in the worldwide and the most common type in males in the Kingdom of Saudi Arabia (1). The age-standardized incidence rates (per 100,000) of CRC in Kingdom of Saudi Arabia vary between 9.8 in females to 14.3 in males (1) Although low compared with Western countries, CRC incidence rate in Kingdom of Saudi Arabia has almost tripled in less than 8 years (2). This increase in CRC incidence rates coincides with a shift towards Western world lifestyle including diet and daily activities (3, 4).

CRC is a heterogeneous disease with different molecular characteristics associated with the sites from which the tumors originate. Such heterogeneity is compounded by the multitude of genetic and epigenetic variations acting as passengers or drivers of the tumor (5). Majority of CRC develop via chromosomal instability (CIN) pathway. CIN is often exacerbated by inactivation of the Wnt signaling pathway “master regulator” APC gene (6), activating mutations of KRAS or BRAF oncogenes (7), or deletions of the 18q (8), and 17p (9) chromosomal regions with deleterious effects on the tumor suppressor genes TP53 and DCC. Defective mismatch repair (MMR) pathway results in a subtler form of genomic instability, namely microsatellite instability (MSI). High levels of MSI (or MSI-H) in sporadic CRC are usually caused by hypermethylation of the MLH1 promoter (10). In terms of methylation, the CpG island methylator phenotype (CIMP) pathway is the second most common pathway in sporadic CRC (11).

CRC tumors with high levels of CIN have a poor prognosis, especially if they are in stage II or III (12). Conversely, MSI-H tumors have a better clinical outcome compared with microsatellite-stable (MSS) tumors (13). CIMP-positive (CIMP+) CRC tumors are usually associated with the proximal colon of older females. CIMP+ tumors also show positive association with BRAF mutations (14). CIMP+ CRC tumors have better prognosis if the tumors are also MSI-H. However, CIMP+ CRC tumors that are MSS have poor clinical outcome.

In this study, we investigate the methylation frequency of several genes including the CIMP markers (IGF2, CACNA1G, NEUROG1, RUNX3, and SOCS1), the MMR genes (MSH2 and MLH1), tumor suppressor genes (APC, RASSF1A, and CDKN2A) in addition to MGMT DNA repair gene. The hypermethylation-mediated silencing of the polycomb group target (PCGT) genes (15) in cancer has been recently shown to be a hallmark of carcinogenesis (16). Although the methylation of such genes have been shown in ageing normal colon mucosa, their methylation is much more widespread and pronounced in cancerous samples (16–18). We have recently shown that methylation of SLIT2, SFRP1, HIC1, and MYOD1 is frequent in sporadic breast cancer from Saudi Arabia with methylation of the latter a possible marker for poor prognosis (19). The contribution of the inactivation of MMR genes in CRC for this population is yet to be shown. Similarly, such information is lacking for the other genes analyzed in this cohort.

Patients

The material of the present study consist of a series of 120 CRC specimens, retrospectively collected from the archives of Anatomical Pathology Laboratory in King Abdulaziz University Hospital (Jeddah, Kingdom of Saudi Arabia), covering the period from January 2005 to December 2009. Serial sections were cut from paraffin blocks, stained with hematoxylin and eosin for routine histologic examination, classification, grading, and staging following the American Joint Committee on Cancer (AJCC) staging system (20). The pertinent clinicopathologic data (gender, age, grade, and lymph node status), and follow-up results were retrieved from the patients' records after obtaining the relevant ethical approvals. DNA was extracted from 10 μm thin formalin-fixed paraffin-embedded slices using the Qiagen QIAMP Formalin-fixed Paraffin-embedded Tissue DNA extraction kit, following the manufacturer's guidelines. KRAS and BRAF mutational status were determined according to the previously published reports (21). The microsatellite instability was determined according to Berg and colleagues (22).

Bisulfite DNA modification and MethyLight assay

Up to 0.5 microgram of DNA was used for bisulfite conversion using the Qiagen Epitect Bisulfite Conversion kit. DNA methylation analysis was conducted using MethyLight as described elsewhere (23). The methylation levels of RASSF1A, APC, MGMT, CDKN2A, SLIT2, SFRP1, MYOD1, HIC1, MSH2, MLH1 and the CIMP markers IGF2, SOCS1, RUNX3, CACNA1G, NEUROG1 were analyzed using the primer-probe combinations listed in Table 1 which were made according to previously published reports (24–27). A probe targeting bisulfite-modified Alu repeat sequences was used to normalize for input DNA. The specificity of the reaction was ascertained using sssl-treated and bisulfite-modified positive control DNA (Qiagen) and the negative control DNA (Qiagen). The percentage of fully methylated reference (PMR) was calculated by dividing the gene:Alu ratio of a sample by the gene:Alu ratio of the positive control DNA and multiplying by 100. Samples with PMR more than 10 were considered positive for methylation, whereas samples with PMR less than 10 were considered negative (i.e., unmethylated). The PMR more than 10 is considered positive as it indicates a very likely hypermethylation-mediated loss of expression for the genes analyzed.

Table 1.

Primer and probe sequences used in this study

GeneForward primer sequenceReverse primer sequenceProbe oligo sequence
SFRP1 CAACTCCCGACGAAACGAA CGCGAGGGAGGCGATT 6FAM-CACTCGTTACCACGTCCGTCACCG-BHQ1 
MYOD1 GAGCGCGCGTAGTTAGCG TCCGACACGCCCTTTCC 6FAM-CTCCAACACCCGACTACTATATCCGCGAAA-BHQ1 
HIC1 GTTAGGCGGTTAGGGCGTC CCGAACGCCTCCATCGTAT 6FAM-CAACATCGTCTACCCAACACACTCTCCTACG-BHQ1 
RASSF1A ATTGAGTTGCGGGAGTT GGT ACACGCTCCAACCGAATA CG 6FAM-CCCTTCCCAACGCGCCCA-BHQ1 
CDKN2A TGGAATTTTCGGTTGATT GGTT AACAACGTCCGCACCTC CT 6FAM-ACCCGACCCCGAACCGCG-BHQ1 
SLIT2 CAATTCTAAAAACGCACGACTCTAAA CGGGAGATCGCGAGGAT 6FAM-CGACCTCTCCCTCGCCCTCGACT-BHQ1 
IGF2 GAGCGGTTTCGGTGTCGTTA CCAACTCGATTTAAACCGACG 6FAM-CCCTCTACCGTCGCGAACCCGA-BHQ1 
NEUROG1 CGTGTAGCGTTCGGGTATTTGTA CGATAATTACGAACACACTCCGAAT 6FAM-CGATAACGACCTCCCGCGAACATAAA-BHQ1 
RUNX3 CGTTCGATGGTGGACGTGT GACGAACAACGTCTTATTACAACGC 6FAM-CGCACGAACTCGCCTACGTAATCCG-BHQ1 
SOCS1 GCGTCGAGTTCGTGGGTATTT CCGAAACCATCTTCACGCTAA 6FAM-ACAATTCCGCTAACGACTATCGCGCA-BHQ1 
CACNA1G TTTTTTCGTTTCGCGTTTAGGT CTCGAAACGACTTCGCCG 6FAM-AAATAACGCCGAATCCGACAACCGA-BHQ1 
MGMT CTAACGTATAACGAAAATCGTAACAACC AGTATGAAGGGTAGGAAGAATTCGG 6FAM-CCTTACCTCTAAATACCAACCCCAAACCCG-BHQ1 
APC TTATATGTCGGTTACGTGCGTTTATAT GAACCAAAACGCTCCCCAT 6FAM-CCCGTCGAAAACCCGCCGATTA-BHQ1 
MSH2 TTTTAGTGCGGAGGTACGGG AAACGATCCTCCGAAACCAAA 6FAM-CCGCACAAACACCAACGTTCCG-BHQ1 
MLH1 AGGAAGAGCGGATAGCGATTT TCTTCGTCCCTCCCTAAAACG 6FAM-CCCGCTACCTAAAAAAATATACGCTTACGCG-BHQ1 
ALU GGTTAGGTATAGTGGTTTATATTTGTAATTTTAGTA ATTAACTAAACTAATCTTAAACTCCTAACCTCA VIC-CCTACCTTAACCTCCC-MGBNFQ 
GeneForward primer sequenceReverse primer sequenceProbe oligo sequence
SFRP1 CAACTCCCGACGAAACGAA CGCGAGGGAGGCGATT 6FAM-CACTCGTTACCACGTCCGTCACCG-BHQ1 
MYOD1 GAGCGCGCGTAGTTAGCG TCCGACACGCCCTTTCC 6FAM-CTCCAACACCCGACTACTATATCCGCGAAA-BHQ1 
HIC1 GTTAGGCGGTTAGGGCGTC CCGAACGCCTCCATCGTAT 6FAM-CAACATCGTCTACCCAACACACTCTCCTACG-BHQ1 
RASSF1A ATTGAGTTGCGGGAGTT GGT ACACGCTCCAACCGAATA CG 6FAM-CCCTTCCCAACGCGCCCA-BHQ1 
CDKN2A TGGAATTTTCGGTTGATT GGTT AACAACGTCCGCACCTC CT 6FAM-ACCCGACCCCGAACCGCG-BHQ1 
SLIT2 CAATTCTAAAAACGCACGACTCTAAA CGGGAGATCGCGAGGAT 6FAM-CGACCTCTCCCTCGCCCTCGACT-BHQ1 
IGF2 GAGCGGTTTCGGTGTCGTTA CCAACTCGATTTAAACCGACG 6FAM-CCCTCTACCGTCGCGAACCCGA-BHQ1 
NEUROG1 CGTGTAGCGTTCGGGTATTTGTA CGATAATTACGAACACACTCCGAAT 6FAM-CGATAACGACCTCCCGCGAACATAAA-BHQ1 
RUNX3 CGTTCGATGGTGGACGTGT GACGAACAACGTCTTATTACAACGC 6FAM-CGCACGAACTCGCCTACGTAATCCG-BHQ1 
SOCS1 GCGTCGAGTTCGTGGGTATTT CCGAAACCATCTTCACGCTAA 6FAM-ACAATTCCGCTAACGACTATCGCGCA-BHQ1 
CACNA1G TTTTTTCGTTTCGCGTTTAGGT CTCGAAACGACTTCGCCG 6FAM-AAATAACGCCGAATCCGACAACCGA-BHQ1 
MGMT CTAACGTATAACGAAAATCGTAACAACC AGTATGAAGGGTAGGAAGAATTCGG 6FAM-CCTTACCTCTAAATACCAACCCCAAACCCG-BHQ1 
APC TTATATGTCGGTTACGTGCGTTTATAT GAACCAAAACGCTCCCCAT 6FAM-CCCGTCGAAAACCCGCCGATTA-BHQ1 
MSH2 TTTTAGTGCGGAGGTACGGG AAACGATCCTCCGAAACCAAA 6FAM-CCGCACAAACACCAACGTTCCG-BHQ1 
MLH1 AGGAAGAGCGGATAGCGATTT TCTTCGTCCCTCCCTAAAACG 6FAM-CCCGCTACCTAAAAAAATATACGCTTACGCG-BHQ1 
ALU GGTTAGGTATAGTGGTTTATATTTGTAATTTTAGTA ATTAACTAAACTAATCTTAAACTCCTAACCTCA VIC-CCTACCTTAACCTCCC-MGBNFQ 

Statistical analysis

All statistical tests were carried out using IBM SPSS Statistics version 19. Fisher exact test was used to identify statistical significance of correlation between methylation events and clinicopathologic factors. The primary endpoints of the study included overall disease-free survival (DFS) calculated from the date of diagnosis to the appearance of disease recurrence or the last recorded date of being alive or death caused by CRC. In calculating DFS, patients who died of other or unknown causes were excluded. All survival times were calculated by univariate Kaplan–Meier analysis, and equality of the survival functions between the strata was tested by log-rank (Mantel–Cox) test. Multivariate Cox regression analysis was conducted to disclose independent predictors of DFS. All tests were 2-sided, and P values < 0.05 were considered statistically significant. K-means clustering was conducted using the Gene CLUSTER 3.0 program and visualized using JavaTree software (28).

We analyzed 120 patients with colorectal cancer selected on the basis of the availability of tissue material and clinical data. Mean age was 58 years (range, 24–96 years) with 34 patients (28.3%) being under 50 years. This cohort consisted of 72 male patients (60%) and 48 female patients (40%). Thirty eight (31.7%) tumors were in the right colon, 36 (30%) tumors were in the left colon, and 46 (38.3%) tumors were in the rectum. There is a significant association between the male gender and tumors from the right colon (P = 0.016). In addition, tumors from the left colon were more predominant in females (P = 0.027). Tumors from the right side are more likely to involve the lymph nodes (P = 0.039). There are no other significant differences between right, left, and rectal colon cancer in terms of age, grade, or recurrence.

The mutation status of BRAF codon 600 and KRAS at codon 12,13 was determined by sequencing. BRAF mutations were rare in our cohort (n = 120, 2.5%). KRAS mutations were more frequent (n = 108, 24.1%). There was no significant association between KRAS mutation and tumor location, age, sex, or grade. However, metastatic tumors are more likely to harbor a KRAS mutation (P = 0.007). The microsatellite instability (MSI) status was determined for 66 cases. A total of 22.7% patients had microsatellite stable (MSS) tumors, whereas 34.8% exhibited MSI-low status and 42.4% were MSI-high. In this cohort, it is more likely for the rectal cancer cases to be microsatellite stable (P = 0.012). We have determined the CpG island methylator phenotype (CIMP) by analyzing the methylation frequency of 5 genes; IGF2, SOCS1, RUNX3, CACNA1G, and NEUROG1. A case is considered CIMP+ if methylation of 2 or more genes can be detected. Overall, CIMP+ tumors were only 14.2% of the total cohort and are significantly associated with male patients (P = 0.014).

The methylation frequency of the 15 genes analyzed are shown in Fig. 1. The highest overall methylation frequency observed was for SFRP1 (70%) and the lowest was for MLH1 and MSH2 (1.2% and 0%, respectively). The methylation levels for the PCGT genes are consistently higher in tumorous tissues compared with matching nonmalignant counterparts (Supplementary Fig. S1). There is no significant association between the methylation of any gene and tumor location (with the exception of the CIMP markers). When stratified according to tumor location, metastasis is associated with SLIT2, SFRP1, and RASSF1A methylation in tumors originating from the rectum (P = 0.011, P = 0.003, and P = 0.039, respectively). Also in the rectal tumors, MYOD1 methylation is positively associated with KRAS mutations (P = 0.003). Rectal cancers from male patients exhibit a significant association with APC methylation (P = 0.007). Moreover, APC methylation positively associated with MSI-H rectal tumors (P = 0.012). Tumors originating from the left colon exhibit a positive association between SFRP1 methylation (P = 0.022), HIC1 methylation (P = 0.022), RASSF1A methylation (P = 0.018), and male patients. MSI-H right-sided tumors are positively associated with RASSF1A methylation (P = 0.019).

Figure 1.

Methylation frequency in CRC. A, percentage of samples originating either from the right colon, left colon, or rectum that exhibit methylation events in the selected genes. B, methylation frequency of the genes analyzed in samples originating either from the right colon, left colon, or rectum.

Figure 1.

Methylation frequency in CRC. A, percentage of samples originating either from the right colon, left colon, or rectum that exhibit methylation events in the selected genes. B, methylation frequency of the genes analyzed in samples originating either from the right colon, left colon, or rectum.

Close modal

We have conducted K-means clustering analysis based on the methylation status of 13 genes and CIMP status (Fig. 2) to distinguish the subgroups of our cohort based on methylation events. As shown in Fig. 2, 3 distinct subgroups can be identified by K-means clustering. Group I (methylation-low, n = 36) is characterized by over representation of rectal cancer (50%) and being MSS or MSI-low (combined percentage is 70%). Group II (n = 67) is characterized by the prominent methylation of the PCGT genes (SFRP1, SLIT2, HIC1, MYOD1) in addition to the hypermethylation of MGMT, RASSF1A, and APC. Male patients were 59.7% of group II patients (Table 2). Group III (methylation-high, n = 17) is characterized by the predominance of male patients who in addition to methylation of the PCGT genes show positive CIMP status and frequent methylation of RASSF1A, APC, MGMT, and CDKN2A.

Figure 2.

K-means clustering analysis based on methylation events shows 3 distinct subgroups. Black shades indicate positive methylation results, whereas gray shades indicate lack of methylation. White shades reflect missing data.

Figure 2.

K-means clustering analysis based on methylation events shows 3 distinct subgroups. Black shades indicate positive methylation results, whereas gray shades indicate lack of methylation. White shades reflect missing data.

Close modal
Figure 3.

Univariate Kaplan–Meier blots for overall DFS. A, poor overall survival in older patients exhibiting group III methylation pattern (solid line). B, significantly better outcome for male patients with group II methylation pattern (solid line).

Figure 3.

Univariate Kaplan–Meier blots for overall DFS. A, poor overall survival in older patients exhibiting group III methylation pattern (solid line). B, significantly better outcome for male patients with group II methylation pattern (solid line).

Close modal
Table 2.

Clinicopathologic characteristics of group II patients in relations to all other patients

Group IIOther groupsTotal
Number of cases 67 53 120 
 Males 40 32 72 
 Females 27 21 48 
Age 
 Less than 50 years old 17 17 34 
Tumor location 
 Right colon 21 17 38 
 Left colon 23 13 36 
 Rectum 23 23 46 
Lymph node status 
 LN+ 31 27 58 
 LN− 19 16 35 
Grade 
 Grade 1 (well differentiated) 11 19 
 Grade 2 (moderately differentiated) 47 30 77 
 Grade 3 (poorly differentiated) 
MSI status 
 MSS 15 
 MSI-L 10 13 23 
 MSI-H 19 28 
Recurrence 35 23 58 
KRAS mutation 16 10 26 
BRAF mutation 
CIMP+ 17 17 
Group IIOther groupsTotal
Number of cases 67 53 120 
 Males 40 32 72 
 Females 27 21 48 
Age 
 Less than 50 years old 17 17 34 
Tumor location 
 Right colon 21 17 38 
 Left colon 23 13 36 
 Rectum 23 23 46 
Lymph node status 
 LN+ 31 27 58 
 LN− 19 16 35 
Grade 
 Grade 1 (well differentiated) 11 19 
 Grade 2 (moderately differentiated) 47 30 77 
 Grade 3 (poorly differentiated) 
MSI status 
 MSS 15 
 MSI-L 10 13 23 
 MSI-H 19 28 
Recurrence 35 23 58 
KRAS mutation 16 10 26 
BRAF mutation 
CIMP+ 17 17 

Group I exhibits week association with the female gender (P = 0.07) and tend to be well-differentiated tumors (P = 0.052). In addition, KRAS mutations show negative association with group I cases (P = 0.053). Group II is weekly but significantly associated with MSI-H status (P = 0.048). Group III is strongly associated with the male gender (P = 0.014) and poorly differentiated tumors (P = 0.048). None of the new groups show statistically significant association with age, tumor location, or KRAS mutation.

Next, we evaluated overall DFS in the 3 groups by univariate Kaplan–Meier analysis. When conducted on all patients, there was no significant effect of belonging into any of the groups on DFS. However, when stratified into young (<50 years old) versus old (>50 years old), worse DFS could be seen if the patient is older than 50 years old and displays group III methylation pattern (P = 0.058), Fig. 3A. When stratified according to the gender, group I patients display worse DFS in males only (P = 0.073). Interestingly, being a male patient displaying group II methylation pattern carries a favorable prognosis as significantly better DFS is observed (P = 0.027; Fig. 3B).

Multivariate Cox regression analysis was conducted with the variables like KRAS status, metastasis, age, sex, and tumor location in addition to grouping by K-means cluster analysis. As expected, metastasis is the strongest poor prognosis indicator with P < 0.0001 and HR of 8.837 [95% confidence interval (CI), 3.787–20.619]. However, group II methylation pattern is a good prognosis indicator with P = 0.013 and HR of 0.269 (95% CI, 0.095–0.761).

In this study, we have analyzed 120 cases of sporadic colorectal cancer (CRC) originating from the right colon, left colon, and the rectum for the presence of KRAS, BRAF mutations, MSI, and CIMP. We have additionally analyzed the methylation frequency of the polycomb group target genes (PCGT) that are normally silenced in stem cells. Furthermore, we have determined the methylation frequency of RASSF1A, APC, CDKN2A, MGMT, MLH1, and MSH2 in the same cohort.

The mutation rates of KRAS (24.1%) and BRAF (2.5%) were found to be lower than worldwide average, which may reflect possible ethnic differences affecting the mutation rates of these 2 oncogenes (29). CIMP frequency (14.2%) is similar to previous reports (11). However, we could not detect any statistically significant association between CIMP+ status and mutations of BRAF, KRAS, or presence of MSI. Moreover, CIMP+ tumors were found to be associated with the male gender in our cohort (P = 0.014). Right-sided tumors were also more prominent in males (P = 0.016). Therefore, there is a correlation between CIMP and right-sided tumors although this association is not statistically significant (P = 0.164).

The most common events in our cohort were the methylation of the PCGT genes as well as the methylation of MGMT gene. Interestingly, it has been suggested the MGMT is also a target of the polycomb complex (15). The methylation of the MMR genes was rare in our cohort. This is could be because CRC follows a different route to tumorigenesis and we cannot exclude the presence of inactivating mutations in this pathway

We have conducted K-means clustering analysis to stratify our samples based on their methylation signature. Our cohort can be separated into 3 distinct groups, group I (low methylation), group II (intermediate methylation), and group III (high methylation). Low-methylation frequency in all the genes studied is the hallmark of group I which is mostly, but not exclusively, represented by tumors originating from the rectum. Group I cases are likely to be well-differentiated tumors harboring wild-type KRAS (P = 0.052 and P = 0.053, respectively). Group II cases are MSI-H (P = 0.048); however, they are exclusively CIMP negative. Group II cases do not show any statistically significant association with any other clinicopathologic parameter despite being the group with best representation in our cohort. CRC tumors exhibiting the most frequent methylation amongst the genes analyzed clustered together in Group III. These poorly differentiated and CIMP+ tumors (P = 0.048) originated mostly from male patients (P = 0.014) but had no other significant associations. Belonging to any of the 3 groups can serve as a potential prognostic marker for overall survival depending on age or sex. Patients with CRC who are more than 50 years old and display group III methylation pattern have worse overall DFS compared with younger counterparts or patients displaying other methylation patterns. This is in line with previous reports of poor prognosis in CIMP+ cases that are not MSI-H (30). A potentially poorer clinical outcome is observed for male patients displaying group I methylation pattern. The reason for this observation is unknown as the presence and nature of genetic alterations in these tumors is not determined in this study. Unfortunately, the sample size of this group is too small to allow for a more definitive conclusion. Interestingly, however, is the observation that male patients with group II methylation pattern have a better clinical outcome compared with patients displaying other methylation pattern. This is perhaps not entirely surprising as MSI-H tumors generally have good prognosis (30). Multivariate Cox regression analysis confirmed this observation.

No potential conflicts of interest were disclosed.

Conception and design: A. Dallol, J. Al-Maghrabi, A. Buhmeida, M.S. Al-Ahwal, M.H. Al-Qahtani

Development of methodology: A. Dallol, J. Al-Maghrabi, A.G. Chaudhary

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Dallol, J. Al-Maghrabi, A. Buhmeida, M.A. Gari, A.G. Chaudhary, M.S. Al-Ahwal, A. Sibiany

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Dallol, J. Al-Maghrabi, H.-J. Schulten

Writing, review, and/or revision of the manuscript: A. Dallol, J. Al-Maghrabi, A. Buhmeida, A.G. Chaudhary, M.S. Al-Ahwal, M.H. Al-Qahtani

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Dallol, J. Al-Maghrabi, A.M. Abuzenadah, M.S. Al-Ahwal, M.H. Al-Qahtani

Study supervision: A. Dallol, J. Al-Maghrabi, A. Buhmeida, A.M. Abuzenadah, M.S. Al-Ahwal, M.H. Al-Qahtani

The authors thank the Ministry of Higher Education and King Abdulaziz City for Science and Technology (KACST) and the Scientific Chair for Colorectal Cancer for their financial support to this research. The authors also thank Ms. Shylu Mathews and Ms. Manar Ata for their significant technical assistance.

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.

1.
Ferlay
SHR
,
Bray
F
,
Forman
D
,
Mathers
C
,
Parkin
DM
. 
GLOBOCAN 2008 v2.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]
.
Lyon, France
:
International Agency for Research on Cancer
; 
2010
[cited 2011 October 1].
Available from
: http://globocan.iarc.fr.
2.
Al-Eid
HS
,
Arteh
SO
. 
Cancer Incidence Report Saudi Arabia 1999–2000
.
Kingdom of Saudi Arabia
:
National Cancer Registry
; 
2004
.
Report No.: 6. Sponsored by the Ministry of Health
.
3.
Boyle
T
,
Fritschi
L
,
Heyworth
J
,
Bull
F
. 
Long-term sedentary work and the risk of subsite-specific colorectal cancer
.
Am J Epidemiol
2011
;
173
:
1183
91
.
4.
Almurshed
KS
. 
Colorectal cancer: case-control study of sociodemographic, lifestyle and anthropometric parameters in Riyadh
.
East Mediterr Health J
2009
;
15
:
817
26
.
5.
Lao
VV
,
Grady
WM
. 
Epigenetics and colorectal cancer
.
Nat Rev Gastroenterol Hepatol
2011
;
8
:
686
700
.
6.
Powell
SM
,
Zilz
N
,
Beazer-Barclay
Y
,
Bryan
TM
,
Hamilton
SR
,
Thibodeau
SN
, et al
APC mutations occur early during colorectal tumorigenesis
.
Nature
1992
;
359
:
235
7
.
7.
Yokota
T
. 
Are KRAS/BRAF mutations potent prognostic and/or predictive biomarkers in colorectal cancers?
Anticancer Agents Med Chem
2012
;
12
:
163
71
.
8.
Fearon
ER
,
Cho
KR
,
Nigro
JM
,
Kern
SE
,
Simons
JW
,
Ruppert
JM
, et al
Identification of a chromosome 18q gene that is altered in colorectal cancers
.
Science
1990
;
247
:
49
56
.
9.
Purdie
CA
,
Piris
J
,
Bird
CC
,
Wyllie
AH
. 
17q allele loss is associated with lymph node metastasis in locally aggressive human colorectal cancer
.
J Pathol
1995
;
175
:
297
302
.
10.
Herman
JG
,
Umar
A
,
Polyak
K
,
Graff
JR
,
Ahuja
N
,
Issa
JP
, et al
Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma
.
Proc Natl Acad Sci U S A
1998
;
95
:
6870
5
.
11.
Worthley
DL
,
Leggett
BA
. 
Colorectal cancer: molecular features and clinical opportunities
.
Clin Biochem Rev
2010
;
31
:
31
8
.
12.
Watanabe
T
,
Kobunai
T
,
Yamamoto
Y
,
Matsuda
K
,
Ishihara
S
,
Nozawa
K
, et al
Chromosomal instability (CIN) phenotype, CIN high or CIN low, predicts survival for colorectal cancer
.
J Clin Oncol
2012
;
30
:
2256
64
.
13.
Sinicrope
FA
,
Sargent
DJ
. 
Molecular pathways: microsatellite instability in colorectal cancer: prognostic, predictive, and therapeutic implications
.
Clin Cancer Res
2012
;
18
:
1506
12
.
14.
Nosho
K
,
Irahara
N
,
Shima
K
,
Kure
S
,
Kirkner
GJ
,
Schernhammer
ES
, et al
Comprehensive biostatistical analysis of CpG island methylator phenotype in colorectal cancer using a large population-based sample
.
PLoS ONE
2008
;
3
:
e3698
.
15.
Lee
TI
,
Jenner
RG
,
Boyer
LA
,
Guenther
MG
,
Levine
SS
,
Kumar
RM
, et al
Control of developmental regulators by Polycomb in human embryonic stem cells
.
Cell
2006
;
125
:
301
13
.
16.
Teschendorff
AE
,
Menon
U
,
Gentry-Maharaj
A
,
Ramus
SJ
,
Wesenberger
DJ
,
Shen
H
, et al
Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer
.
Genome Res
2010
;
20
:
440
6
.
17.
Dunwell
TL
,
Dickinson
RE
,
Stankovic
T
,
Dallol
A
,
Weston
V
,
Austen
B
, et al
Frequent epigenetic inactivation of the SLIT2 gene in chronic and acute lymphocytic leukemia
.
Epigenetics
2009
;
4
:
265
9
.
18.
Dallol
A
,
Morton
D
,
Maher
ER
,
Latif
F
. 
SLIT2 axon guidance molecule is frequently inactivated in colorectal cancer and suppresses growth of colorectal carcinoma cells
.
Cancer Res
2003
;
63
:
1054
8
.
19.
Buhmeida
A
,
Merdad
A
,
Al-Maghrabi
J
,
Al-Thobaiti
F
,
Ata
M
,
Bugis
A
, et al
RASSF1A methylation is predictive of poor prognosis in female breast cancer in a background of overall low methylation frequency
.
Anticancer Res
2011
;
31
:
2975
81
.
20.
Greene
FL
,
Balch
CM
,
Haller
DG
,
Morrow
M
,
editors
. 
American Joint Committee on Cancer (AJCC) Cancer Staging Manual
. 6th ed.
Berlin
:
Springer
; 
2002
.
21.
Schulten
HJ
,
Al-Maghrabi
J
,
Al-Ghamdi
K
,
Salama
S
,
Al-Muhayawi
S
,
Chaudhary
A
, et al
Mutational screening of RET, HRAS, KRAS, NRAS, BRAF, AKT1, and CTNNB1 in medullary thyroid carcinoma
.
Anticancer Res
2011
;
31
:
4179
83
.
22.
Berg
KD
,
Glaser
CL
,
Thompson
RE
,
Hamilton
SR
,
Griffin
CA
,
Eshleman
JR
. 
Detection of microsatellite instability by fluorescence multiplex polymerase chain reaction
.
J Mol Diagn
2000
;
2
:
20
8
.
23.
Dallol
A
,
Al-Ali
W
,
Al-Shaibani
A
,
Al-Mulla
F
. 
Analysis of DNA methylation in FFPE tissues using the MethyLight technology
.
Methods Mol Biol
2011
;
724
:
191
204
.
24.
Hawes
SE
,
Stern
JE
,
Feng
Q
,
Wiens
LW
,
Rasey
JS
,
Lu
H
, et al
DNA hypermethylation of tumors from non-small cell lung cancer (NSCLC) patients is associated with gender and histologic type
.
Lung Cancer
2010
;
69
:
172
9
.
25.
Houshdaran
S
,
Cortessis
VK
,
Siegmund
K
,
Yang
A
,
Laird
PW
,
Sokol
RZ
. 
Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm
.
PLoS ONE
2007
;
2
:
e1289
.
26.
Weisenberger
DJ
,
Siegmund
KD
,
Campan
M
,
Young
J
,
Long
TI
,
Faasse
MA
, 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
.
27.
Widschwendter
M
,
Apostolidou
S
,
Raum
E
,
Rothenbacher
D
,
Fiegl
H
,
Menon
U
, et al
Epigenotyping in peripheral blood cell DNA and breast cancer risk: a proof of principle study
.
PLoS ONE
2008
;
3
:
e2656
.
28.
Eisen
MB
,
Spellman
PT
,
Brown
PO
,
Botstein
D
. 
Cluster analysis and display of genome-wide expression patterns
.
Proc Natl Acad Sci U S A
1998
;
95
:
14863
8
29.
Arrieta
O
,
Cardona
AF
,
Federico Bramuglia
G
,
Gallo
A
,
Campos-Parra
AD
,
Serrano
S
, et al
Genotyping non-small cell lung cancer (NSCLC) in Latin America
.
J Thorac Oncol
2011
;
6
:
1955
9
.
30.
Jass
JR
. 
Classification of colorectal cancer based on correlation of clinical, morphological and molecular features
.
Histopathology
2007
;
50
:
113
30
.