Open Access

DNA methylation of the p14ARF, RASSF1A and APC1A genes as an independent prognostic factor in colorectal cancer patients

  • Authors:
    • Torbjörn K. Nilsson
    • Zarah M. Löf-Öhlin
    • Xiao-Feng Sun
  • View Affiliations

  • Published online on: October 30, 2012     https://doi.org/10.3892/ijo.2012.1682
  • Pages: 127-133
  • Copyright: © Nilsson et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

We quantitated the methylated fraction of CpG sites in the promoter regions of O6-MGMT, p14ARF, p16INK4a, RASSF1A and APC1A in tumor tissue from patients with colorectal cancer (CRC) in order to determine if promoter hypermethylation of any of these genes predicts survival. DNA was isolated from 111 primary CRC and 46 matched normal colorectal mucosa samples from the same patients, obtained at primary surgery and DNA methylation was examined by Pyrosequencing®. Follow-up time was up to 20 years. Patients showed partial promoter methylation in the following frequencies: O6-MGMT, 34%; p14ARF, 29%; p16INK4a, 28%; RASSF1A, 14%; and APC1A, 27%. Normal mucosa was always unmethylated. CRC patients with methylated p14ARF gene promoter had significantly worse prognosis (p=0.036), whereas those with methylated O6-MGMT had significantly better prognosis through the first 60 months post-treatment (RR 0.36; p=0.023). Methylation of one or more of the genes from the set p14ARF, RASSF1A and APC1A, was significantly (p=0.021) associated with worse prognosis even adjusting for tumor stage and differentiation (RR 2.2, p=0.037). Thus, DNA methylation of the p14ARF, RASSF1A and APC1A genes, diagnosed by Pyrosequencing, defines a poor prognosis subset of CRC patients independently of both tumor stage and differentiation. O6-MGMT methylation may play a protective role.

Introduction

DNA methylation is a common feature often seen in tumor suppressor and DNA repair genes (1). Methylation of CpG sites in the promoters of these genes frequently causes loss of expression, affecting cell cycle regulation, cell adhesion or DNA reparation. Several genes have been reported to be involved, through DNA methylation, in sporadic colorectal cancer (CRC) (2,3). Here, we focus on five of these, O6-MGMT, p14ARF, p16INK4a, RASSF1A and APC1A(49). Studies on the methylation status of the promoter regions of these genes in CRC development reported frequent promoter hypermethylation in tumor tissue DNA, whereas normal tissue DNA remained unmethylated (10,11).

Methylation specific PCR, a common technique to study DNA methylation, assays the methylation status of a few CpG sites (i.e., those interfering with the PCR primer binding) and only gives a qualitative indication (methylated or not). Like all ‘allele specific’ PCR methods, it depends largely on a combination of the number of PCR optimization experiments and individual judgment of presence or absence of bands. We do not know which particular CpG sites will silence these suppressor genes when methylated because the standard methods assay only a few of the sometimes 100 or so CpG sites (12). These methodological concerns stall our understanding of the clinical role of DNA methylation.

We have therefore developed Pyrosequencing® assays (13) to get a DNA sequence-specific as well as quantitative measure of DNA methylation of the promoter CpG sites as well as the adjacent CpG sites, of the DNA repair gene O6-MGMT and the 4 tumor suppressor genes p14ARF, p16INK4a, RASSF1A and APC1A previously studied by MS-PCR (10,11). We report our data on tumor biopsies from subjects diagnosed with colorectal cancer or adenomas and adjacent normal mucosa, and furthermore we assessed if these assays had any long-term prognostic value.

Materials and methods

Subjects

The study included 111 randomly selected patients with primary colorectal adenocarcinoma who underwent surgical resection at Linköping Hospital, Linköping and Vrinnevi Hospital, Norköping, Sweden. In 46 of the patients, normal mucosa specimens taken from the margin of the resected tumor were also available. The study also included 10 patients with colorectal adenomas, from 7 of whom matched normal mucosa specimens were also available. The patients’ gender, age, tumor location and stage were obtained from surgical and/or pathological records at Linköping and Vrinnevi Hospitals. Tumor differentiation was graded into well moderately or poorly differentiated. The study was approved by the Regional Ethics Review Board in Linköping and an informed consent document was signed by participants.

DNA isolation and bisulfite treatment of tissue DNA

Genomic DNA was isolated from 20 mg of colorectal tumor tissue (n=111) by means of the Wizard® SV Genomic DNA Purification System according to the manufacturer’s instructions (Promega, Madison, WI, USA). For some of these patients (n=46) distant normal colorectal mucosa tissue was also available and DNA was isolated in the same way. Genomic DNA was also isolated from 20 mg of colorectal adenoma tissue (n=10 subjects), and for several of these subjects (n=7) also from 20 mg of distant normal colorectal mucosa, by means of the QIAamp DNA Mini Kit according to the manufacturer’s instructions (Qiagen Inc., Valencia, CA, USA).

Approximately 1,000 ng of isolated and precipitated DNA was used for the bisulfite treatment. The bisulfite treatment was performed with EZ DNA Methylation kit according to the instructions by the manufacturer (Zymo Research, Orange, CA, USA) except that the incubation time was shortened to 10 h. In short, DNA was diluted with M-Dilution buffer and was incubated for 15 min at 37°C. After the incubation CT conversion reagent was added and the samples were incubated at 50°C for 10 h. The samples were then incubated on ice for 10 min and then M-Binding buffer was added. The samples were centrifuged and then washed using centrifugation and M-Wash buffer. The bisulfite treated DNA was eluted in 10 μl M-Elution buffer and then diluted 4 times with TE buffer (10 mmol/l Tris-HCl, 0.05 mmol/l EDTA, pH 7.5).

PCR and Pyrosequencing

PCR and Pyrosequencing of the promoter regions of the O6-MGMT, p14ARF, p16INK4a, RASSF1A and APC1A genes was performed as previously described (13). Pyrosequencing technology was used to sequence-specifically quantitate each of the CpG sites, which are analyzed in practice as C/T-polymorphisms, where a 100% C-reading denotes a fully methylated C (MeC=100%) in the original gDNA sample whereas a 100% T-reading denotes that this locus was unmethylated (MeC=0%) in the original gDNA. Intermediate MeC percentages denote partial methylation at the level of the sample. Partial methylation, when present, is presumed to be partly owing to admixture of unmethylated non-neoplastic cell types present in the tissues to a varying extent.

Statistical analysis

The significance of the difference of promoter region hypermethylation on the O6-MGMT, p14ARF, p16INK4a, RASSF1A and APC1A genes in between normal mucosa samples and primary tumors was tested by χ2 or McNemar’s method. The relationships between the promoter region hypermethylation and other factors were examined by the χ2 test. The relationship between the expression and survival was tested using Cox’s Proportional Hazard Model. Survival curves were calculated using the Kaplan-Meier method. Two-sided p-values of <0.05 were considered as statistically significant.

Results

Prevalence and extent of DNA hypermethylation

Samples from 111 colorectal cancer tumors, 10 colorectal adenomas and 53 matched normal tissues from the same patients (46 from CRC patients, 7 from adenoma patients) were analyzed. Assay failure rate was 0% for all 5 genes studied. All the 53 available paired normal tissues, both from tumor and adenoma tissues were found to be 100% unmethylated on the promoter regions of all the genes. The pyrograms of many of the tumor samples, on the other hand, showed methylation peaks, amounting to a consistent but individual-specific mean percentage of methylated fraction of gene promoter CpG sites. In Table I the number of patients with methylation-positive tumors or adenomas, as well as their mean methylated fractions (%MeC) of CpG sites, are shown. Promoter hypermethylation in the CRC samples was commonest for the O6-MGMT gene (34% of the patients) and least frequent for RASSF1A (14%). Adenomas showed a similar pattern.

Table I.

Mean methylated fraction and standard deviation (SD) for all colorectal cancer and adenoma specimens that showed methylation.

Table I.

Mean methylated fraction and standard deviation (SD) for all colorectal cancer and adenoma specimens that showed methylation.

GeneCRC
Adenomas
No. (%)%MeC (Mean ± SD)%MeC (Range)No. (%)%MeC (Mean ± SD)%MeC (Range)
O6-MGMT38 (34)28±1113–563 (30)24±916–33
p14ARF32 (29)36±1515–900 (0)0±0-
p16INK4a31 (28)29±1012–511 (10)22±0-
RASSF1A16 (14)31±1016–610 (0)0±0-
APC1A30 (27)38±1121–672 (20)44±641–48

[i] The range of the methylation is also shown here.

The methylation pattern throughout the promoter regions of all the genes was consistent within each individual patient: if methylation was detected for a sample all the CpG sites in the entire promoter region of that gene were methylated at roughly the same proportion (%MeC, as obtained from the Pyrograms). Representative patients showing non-methylated normal mucosa and tumor tissues methylated to varying degrees are displayed for O6-MGMT and p14ARF in Figs. 1 and 2. While the occurrence of methylated promoter CpG sites varied (Table I), within each patient the methylated fraction (%MeC) varied very little between the different CpG sites of a particular gene. The mean methylated fractions stated in Table I are the inter-individual (total-sample) mean values of %MeC of CpG sites though the entire promoter regions of all methylation-positive tumor samples.

Concurrent methylation of two or more genes in the colorectal cancer tumors are summarized in Table II. We found 42 patients (38%) to be methylated on one gene, 18 (16%) were methylated on two genes, 15 (14%) on three genes, 5 (4.5%) were found to be methylated on four genes and 1 (0.9%) was methylated on all the genes studied. Out of the 111 tumors, 30 (27%) were thus found to be unmethylated on all genes.

Table II.

Number of colorectal cancer specimens simultaneously methylated on more than one gene.

Table II.

Number of colorectal cancer specimens simultaneously methylated on more than one gene.

O6-MGMT p14ARF p16INK4aRASSF1AAPC1A
O6-MGMT-15101012
p14ARF-16117
p16INK4a-79
RASSF1A-5
APC1A-
DNA hypermethylation and long-term outcome

Survival plots for patients with or without promoter methylation for each of the five genes were analysed. In univariate analysis two genes reached statistical significance. Hypermethylation of p14ARF was related to worse survival (p=0.036) but its significance was attenuated in multivariate analysis when adjusting for tumor stage and differentiation (p=0.065). Hypermethylation of O6-MGMT was associated with better survival through the first 60 months of follow-up, the risk ratio was 0.36 (95% CI 0.15–0.87, p= 0.049) and still remained significant after adjusting for tumor stage and differentiation (p=0.023), see Table III.

Table III.

Multivariate analysis of combined promoter methylation of 06-MGMT, tumor stage and differentiation in relation to patient survival through 60 months post-surgery.

Table III.

Multivariate analysis of combined promoter methylation of 06-MGMT, tumor stage and differentiation in relation to patient survival through 60 months post-surgery.

VariablesNo.Cancer death Rate ratio95% CIP-value
Methylation0.023
  No711.00-
  Yes350.360.15–0.87
Stage<0.0001
  I171.00-
  II464.350.55–34.28
  III248.651.08–69.08
  IV1940.054.98–321.7
Differentiation0.151
  Well261.00-
  Moderately620.680.29–1.58
  Poorly180.850.30–2.40

Since promoter hypermethylation of p14ARF, RASSF1A and APC1A all were associated with a similar tendency towards worse prognosis, and many patients were methylated on more than one gene (Table II), we examined whether methylation of any of the three genes (i.e., one or more), would improve prediction of survival. Indeed, hypermethylation of one or more of these three genes defined a set of patient with a significantly (p=0.021) worse long-term survival (Fig. 3), where only ∼45% were still alive in the methylated group by 20 years of follow-up, compared to ∼75% in the unmethylated group. Adjusting for tumor stage and differentiation did not attenuate this association (risk ratio 2.20; 95% CI, 1.05–4.62, p=0.037; Table IV). No association could be found between promoter hypermethylation of these three genes and the other clinicopathological factors including gender, age, tumor location, or tumor stage and differentiation (p>0.05 for all variables).

Table IV.

Multivariate analysis of combined promoter methylation of p14ARF, RASSF1A and APC1A, tumor stage and differentiation in relation to patient survival.

Table IV.

Multivariate analysis of combined promoter methylation of p14ARF, RASSF1A and APC1A, tumor stage and differentiation in relation to patient survival.

VariablesNo.Cancer death Rate ratio95% CIP-value
Methylation0.037
  No481.00-
  Yes582.201.05–4.62
Stage<0.0001
  I181.00-
  II463.820.48–30.2
  III247.720.96–61.8
  IV1831.503.99–248.6
Differentiation0.330
  Well281.00-
  Moderately620.780.34–1.78
  Poorly161.140.42–3.10

Discussion

Aging and environmental factors may lead to neoplasia and cancer. This process usually involves changed expression pattern of genes involved in adhesion, proliferation, differentiation, cell growth, migration and apoptosis, and can be due to mutations, genetic rearrangements, chromosomal instability or promoter hypermethylation (14). In this study we have focused on five such genes, previously suggested to be involved in the development of CRC; O6-MGMT, p14ARF, p16INK4a, RASSF1A and APC1A(312). Methylation of the promoter regions of some of these genes in tissues or serum from patients with CRC has been reported (1525), but data on the long-term prognostic implications of the whole set is limited.

By using Pyrosequencing, a technique that offers a unique opportunity to quantitate, site-specifically, the methylated fraction in partially methylated CpG sites, we demonstrated that the promoter regions of one or more of the genes analyzed are methylated in tumor tissue from a majority (73%) of patients diagnosed with CRC, range 14–34% for the different genes. None of these genes was methylated in the 46 paired normal mucosa samples. Interestingly, adenoma tissue also appeared to be methylated in about a third of the patients and the paired normal mucosal tissue unmethylated, although caution is prudent here since we have analysed rather few adenomas.

Prognosis and methylation of p14ARF, p16INK4a, APC1A and RASSF1A

When correlating the methylation of the promoter regions of these genes with survival of the CRC patients, we found that methylation of p14ARF was significantly associated with shorter survival compared to patients that has this gene unmethylated. When adjusting for tumor stage and tumor differentiation this significance was attenuated somewhat. We also saw clear trends towards poorer prognosis when methylation was found in the promoter regions of RASSF1A and APC1A.

In some of the subjects several of the genes had their promoter regions hypermethylated concurrently, implicating that signalling pathways such as Wnt where APC1A are involved, p16INK4a-Rb and p14ARF-p53, and normal cell mechanisms such as alkylation and mitotic progression could be altered all at once. Since it has been suggested that CRC evolves from alterations of several of these pathways (26), we thought it relevant to investigate whether this concurrent methylation could affect the outcome for the patient. Grouping together all individuals showing methylation of one or more of the p14ARF, RASSF1A and APC1A genes, we obtained an association between promoter hypermethylation and shorter survival which remained statistically significant even after adjusting for tumor stage and differentiation (Fig. 3, Table IV). Thus, promoter hypermethylation of one or more of the genes p14ARF, RASSF1A and APC1A, when defined by Pyrosequencing assays, might serve as a marker of poor prognosis which we suggest as a novel, relevant stratification factor in future prospective and interventional studies on CRC.

In other recent studies, poor survival has been reported for patients methylated on the p14ARF gene (25,27) in agreement with our findings and for APC1A one report based on measurements of methylation in plasma DNA reported worse prognosis (28) but another study claimed a better survival in patients with methylated tumor tissue APC1A(17). Several reports have claimed predictive value of p16INK4a methylation (19,2123,27,29,30) which we could not replicate in our cohort. Data on the prognostic implications of RASSF1A methylation are sparse, one study reported a higher prevalence of methylation in liver metastases than in the primary tumor (31).

Prognosis and methylation of O6-MGMT

Promoter methylation in the O6-MGMT gene has in some studies on glioblastoma been associated with longer survival especially in therapeutic trials using alkylating agents (3236). It is now proposed that tests for O6-MGMT methylation status should be included in all future clinical trials in malignant glioma if treatment includes alkylating agents, since it is anticipated that those tests may guide choice of future therapy (37). To asses O6-MGMT promoter methylation in the mentioned glioma studies, several different methods have been employed, as was recently pointed out by van den Bent et al(12), who stressed the point that the CpG island in that part of the gene actually contains almost 100 individual CpG sites and that the used methods only give information on the methylation status of a few of these sites. It has not been clear how many CpG loci, or which ones of them, have to be methylated to achieve O6-MGMT gene silencing. Our method utilizes a sequence-specific and quantitative assay, Pyrosequencing, to classify the methylation status (13) and thus provides a tool to answer this question. Our assays cover 25 different CpG sites, 18 of which have never been assayed before, and we show here that in the individual patient, the %MeC of each of these sites is a characteristic feature of each individual tumor sample, and varies very little from CpG site to CpG site within the same tumor tissue sample (Fig. 1; the same observation holds for p14ARF, cf. Fig. 2). On the other hand, a considerable inter-individual difference was seen which could be owing to differing degrees of admixture of unmethylated non-neoplastic cells, to true differences in the biology of the tumor, or to a combination of these factors.

There are a few reports on CRC prognosis in relation to methylation of O6-MGMT, claiming basically no relation (17,29,38). We show here for the first time that like in glioma, promoter methylation of O6-MGMT in CRC patients tended to confer better long-term survival (Table III), in a context where alkylating agents are rarely an option. This suggests that O6-MGMT testing might be of a more general interest and warrants to be included not only in planning of glioma treatment but in studies on other malignant neoplasias as well.

General observations and limitations

A number of methodological differences between the various studies need to be pointed out. Most of the data come from methylation-specific PCR-based methods, not from sequencing-based techniques. Many studies are made on DNA isolated from FFPE samples, which may have decayed due to harsh conditions. We used DNA isolated from fresh tumor tissue, and analysis was performed by bisulphite pyrosequencing (13). The fraction of patients showing methylation of our set of genes ranged from 14% to 34% for the different genes, figures which are both higher (29) and lower (17) than those reported earlier. For instance, one study claiming no prognostic value of O6-MGMT in CRC classified 60% as methylation-positive by a MS-PCR method (17), as against our figure of 34% using Pyrosequencing which agrees better with the figure of Ogino and coworkers of 38% (38). In future studies, more attention should be payed to the source of DNA and to methods used to classify promoter methylation status, and we contend that Pyrosequencing has earned a place among the methods of choice.

The concept of CIMP-positivity (CpG Island Methylator Phenotype) needs to be relativised based on our findings. The number of subjects simultaneously methylated on 2 or more of the selected set of genes was rather small (Table II) and any predictions based on such a subset of the patient population will therefore have a very limited utility. In contrast, we found a statistically significant negative relation with survival, adjusted for tumor stage and differentiation based on a combined methylation variable defined as having any one or more of the genes p14ARF, RASSF1A, APC1A methylated (Table IV, Fig. 3). Employing the same Pyrosequencing assays (13) as in the present paper, we recently showed that APC1A promoter methylation was associated with poor prognosis also in cervical cancer (39).

Moreover, methylation of one of the genes in our set, O6-MGMT, showed a significant association with better survival through the first 60 months following primary surgery. For all these reasons, the concept of a CIMP as a unified set of methylated genes accompanying poor prognosis appears to have limited prospects as a valid prognostic tool in most CRC patients. Perhaps it may ultimately be replaced by a more precise molecular signature associated with poor prognosis (40,41), possibly even tailor-made on the individual basis which might better reflect the random stochastic nature of the processes that characterise sporadic CRC. Such signatures would likely include both chromosomal instability, microsatellite instability, and methylation of a larger set of cancer-related genes than has been currently studied (28,29,30,42,43) as well as less explored features such as histone modifications, nucleosomal occupancy and remodeling, chromatin looping, and non-coding RNAs (44). Hopefully, DNA methylation data may in the future also be useful in guiding adjuvant treatment in CRC as suggested in a recent article (45).

In conclusion, this is the first study to report a significant correlation between CRC patient survival and promoter methylation of the genes p14ARF, RASSF1A and APC1A, as defined by Pyrosequencing assay (13), as well as a protective role of O6-MGMT methylation. Such biomarkers of prognosis in CRC could be utilized as a relevant stratification factor in future prospective and interventional studies on CRC, and might serve as a tool when tailoring treatment for the individual patient. Finally, we maintain that choice of assay methodology may have a determining effect on proper classification of methylation status in the individual patient.

Acknowledgements

Financial support by Örebro County Council, Nyckelfonden, Örebro, and Lions Cancerfond, Uppsala is gratefully acknowledged.

References

1. 

Das PM and Singal R: DNA methylation and cancer. J Clin Oncol. 22:4632–4642. 2004. View Article : Google Scholar : PubMed/NCBI

2. 

Esteller M, Corn PG, Baylin SB and Herman JG: A gene hyper-methylation profile of human cancer. Cancer Res. 61:3225–3229. 2001.PubMed/NCBI

3. 

van Engeland M, Roemen GM, Brink M, et al: K-ras mutations and RASSF1A promoter methylation in colorectal cancer. Oncogene. 21:3792–3795. 2002.

4. 

Preuss I, Eberhagen I, Haas S, Eibl RH, Kaufmann M, von Minckwitz G and Kaina B: O6-methylguanine-DNA methyltransferase activity in breast and brain tumors. Int J Cancer. 61:321–326. 1995.

5. 

Newcomb EW, Alonso M, Sung T and Miller DC: Incidence of p14ARF gene deletion in high-grade adult and pediatric astrocytomas. Hum Pathol. 31:115–119. 2000.

6. 

Serrano M, Hannon GJ and Beach D: A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature. 366:704–707. 1993. View Article : Google Scholar : PubMed/NCBI

7. 

Song MS, Song SJ, Ayad NG, et al: The tumor suppressor RASSF1A regulates mitosis by inhibiting the APC-Cdc20 complex. Nat Cell Biol. 6:129–137. 2006. View Article : Google Scholar : PubMed/NCBI

8. 

Rubinfeld B, Souza B, Albert I, et al: Association of the APC gene product with beta-catenin. Science. 262:1731–1734. 1993. View Article : Google Scholar : PubMed/NCBI

9. 

Su LK, Vogelstein B and Kinzler KW: Association of the APC tumor suppressor protein with catenins. Science. 262:1734–1737. 1993. View Article : Google Scholar : PubMed/NCBI

10. 

van den Donk M, Pellis L, Crott JW, et al: Folic acid and vitamin B-12 supplementation does not favorably influence uracil incorporation and promoter methylation in rectal mucosa DNA of subjects with previous colorectal adenomas. J Nutr. 137:2114–2120. 2007.

11. 

van Engeland M, Weijenberg MP, Roemen GM, et al: Effects of dietary folate and alcohol intake on promoter methylation in sporadic colorectal cancer: the Netherlands cohort study on diet and cancer. Cancer Res. 63:3133–3137. 2003.PubMed/NCBI

12. 

van den Bent MJ, Dubbink HJ, Sanson M, et al: MGMT promoter methylation is prognostic but not predictive for outcome to adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors: a report from EORTC Brain Tumor Group Study 26951. J Clin Oncol. 27:5881–5886. 2009.PubMed/NCBI

13. 

Löf-Öhlin ZM and Nilsson TK: Pyrosequencing assays to study promoter CpG site methylation of the O6-MGMT, hMLH1, p14ARF, p16INK4a, RASSF1A and APC1A genes. Oncol Rep. 21:721–729. 2009.PubMed/NCBI

14. 

Aoyagi H, Iida S, Uetake H, et al: Effect of classification based on combination of mutation and methylation in colorectal cancer prognosis. Oncol Rep. 25:789–794. 2011.PubMed/NCBI

15. 

Esteller M, Tortola S, Toyota M, Capella G, Peinado MA, Baylin SB and Herman JG: Hypermethylation-associated inactivation of p14(ARF) is independent of p16(INK4a) methylation and p53 mutational status. Cancer Res. 60:129–133. 2000.PubMed/NCBI

16. 

Kamiyama H, Noda H, Takata O, Suzuki K, Kawamura Y and Konishi F: Promoter hypermethylation of tumor-related genes in peritoneal lavage and the prognosis of patients with colorectal cancer. J Surg Oncol. 100:69–74. 2009. View Article : Google Scholar : PubMed/NCBI

17. 

Chen SP, Chiu SC, Wu CC, et al: The association of methylation in the promoter of APC and MGMT and the prognosis of Taiwanese CRC patients. Genet Test Mol Biomarkers. 13:67–71. 2009. View Article : Google Scholar : PubMed/NCBI

18. 

Noda H, Mashima R, Kamiyama H, Okada S, Kawamura YJ and Konishi F: Promoter hypermethylation of tumor-related genes in sporadic colorectal cancer in young patients. J Exp Clin Cancer Res. 26:521–526. 2007.PubMed/NCBI

19. 

Krtolica K, Krajnovic M, Usaj-Knezevic S, Babic D, Jovanovic D and Dimitrijevic B: Comethylation of p16 and MGMT genes in colorectal carcinoma: correlation with clinicopathological features and prognostic value. World J Gastroenterol. 13:1187–1194. 2007. View Article : Google Scholar : PubMed/NCBI

20. 

Kohonen-Corish MR, Daniel JJ, Chan C, et al: Low microsatellite instability is associated with poor prognosis in stage C colon cancer. J Clin Oncol. 23:2318–2324. 2005. View Article : Google Scholar : PubMed/NCBI

21. 

Nakayama G, Hibi K, Kodera Y, Koike M, Fujiwara M and Nakao A: P16 methylation in serum as a potential marker for the malignancy of colorectal carcinoma. Anticancer Res. 27:3367–3370. 2007.PubMed/NCBI

22. 

Lee M, Han WS, Kim OK, Sung SH, Cho MS, Lee SN and Koo H: Prognostic value of p16INK4a and p14ARF gene hypermethylation in human colon cancer. Pathol Res Pract. 202:415–424. 2006.

23. 

Sanz-Casla MT, Maestro ML, Vidaurreta M, Maestro C, Arroyo M and Cerdan J: p16 Gene methylation in colorectal tumors: correlation with clinicopathological features and prognostic value. Dig Dis. 23:151–155. 2005. View Article : Google Scholar : PubMed/NCBI

24. 

Zou HZ, Yu BM, Wang ZW, et al: Detection of aberrant p16 methylation in the serum of colorectal cancer patients. Clin Cancer Res. 8:188–191. 2002.

25. 

Dominguez G, Silva J, Garcia JM, et al: Prevalence of aberrant methylation of p14ARF over p16INK4a in some human primary tumors. Mutat Res. 530:9–17. 2003. View Article : Google Scholar : PubMed/NCBI

26. 

Olschwang S, Hamelin R, Laurent-Puig P, et al: Alternative genetic pathways in colorectal carcinogenesis. Proc Natl Acad Sci USA. 94:12122–12127. 1997. View Article : Google Scholar : PubMed/NCBI

27. 

Shen L, Catalano PJ, Benson AB III, O’Dwyer P, Hamilton SR and Issa JP: Association between DNA methylation and shortened survival in patients with advanced colorectal cancer treated with 5-fluorouracil based chemotherapy. Clin Cancer Res. 13:6093–6098. 2007. View Article : Google Scholar : PubMed/NCBI

28. 

Hoffmann AC, Vallböhmer D, Prenzel K, et al: Methylated DAPK and APC promoter DNA detection in peripheral blood is a significantly associated with apparent residual tumor and outcome. J Cancer Res Clin Oncol. 135:1231–1237. 2009. View Article : Google Scholar : PubMed/NCBI

29. 

Kim JC, Choi JS, Roh SA, Cho DH, Kim TW and Kim YS: Promoter methylation of specific genes is associated with the phenotype and progression of colorectal adenocarcinomas. Ann Surg Oncol. 17:1767–1776. 2010. View Article : Google Scholar : PubMed/NCBI

30. 

Wettergren Y, Odin E, Nilsson S, Carlsson G and Gustavsson B: p16INK4a gene promoter hypermethylation in mucosa as a prognostic factor for patients with colorectal cancer. Mol Med. 14:412–421. 2008. View Article : Google Scholar

31. 

Tommasi S, Pinto R, Petriella D, et al: Oncosuppressor methylation: a possible key role in colon metastatic progression. J Cell Physiol. 226:1934–1939. 2011. View Article : Google Scholar : PubMed/NCBI

32. 

Weller M, Felsberg J, Hartmann C, et al: Molecular predictors of progression-free and overall survival in patients with newly diagnosed glioblastoma: a prospective translational study of the German Glioma Network. J Clin Oncol. 27:5743–5750. 2009. View Article : Google Scholar

33. 

Hegi ME, Diserens AC, Gorlia T, et al: MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 352:997–1003. 2005. View Article : Google Scholar : PubMed/NCBI

34. 

Esteller M, Garcia-Foncillas J, Andion E, et al: Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 343:1350–1354. 2000. View Article : Google Scholar

35. 

Jaeckle KA, Eyre HJ, Townsend JJ, et al: Correlation of tumor O6 methylguanine-DNA methyltransferase levels with survival of malignant astrocytoma patients treated with bis-chloroethylnitrosourea: a Southwest Oncology Group study. J Clin Oncol. 16:3310–3315. 1998.

36. 

Hegi ME, Diserens AC, Godard S, et al: Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin Cancer Res. 10:1871–1874. 2004. View Article : Google Scholar : PubMed/NCBI

37. 

Stupp R and Hegi ME: Methylguanine methyltransferase testing in glioblastoma: when and how? J Clin Oncol. 25:1459–1460. 2007. View Article : Google Scholar : PubMed/NCBI

38. 

Shima K, Morikawa T, Baba Y, et al: MGMT promoter methylation, loss of expression and prognosis in 855 colorectal cancers. Cancer Causes Control. 22:301–309. 2011. View Article : Google Scholar : PubMed/NCBI

39. 

Löf-Öhlin ZM, Sorbe B, Wingren S and Nilsson TK: Hypermethylation of promoter regions of the APC1A and p16INK4a genes in relation to prognosis and tumor characteristics in cervical cancer patients. Int J Oncol. 39:683–688. 2011.

40. 

Carmona FJ and Esteller M: Moving closer to a prognostic DNA methylation signature in colon cancer. Clin Cancer Res. 17:1215–1217. 2011. View Article : Google Scholar : PubMed/NCBI

41. 

Yi JM, Dhir M, Van Neste L, et al: Genomic and epigenomic integration identifies a prognostic signature in colon cancer. Clin Cancer Res. 17:1535–1545. 2011. View Article : Google Scholar : PubMed/NCBI

42. 

Cui T, Chen Y, Yang L, Knösel T, Zöller K, Huber O and Petersen I: DSC3 expression is regulated by p53, and methylation of DSC3 DNA is a prognostic marker in human colorectal cancer. Br J Cancer. 104:1013–1019. 2011. View Article : Google Scholar : PubMed/NCBI

43. 

Tong JD, Jiao NL, Wang YX, Zhang YW and Han F: Downregulation of fibulin-3 gene by promoter methylation in colorectal cancer predicts adverse prognosis. Neoplasma. 58:441–448. 2011. View Article : Google Scholar : PubMed/NCBI

44. 

van Engeland M, Derks S, Smits KM, Meijer GA and Herman JG: Colorectal cancer epigenetics: complex simplicity. J Clin Oncol. 29:1382–1391. 2011.PubMed/NCBI

45. 

Jover R, Nguyen TP, Pérez-Carbonell L, et al: 5-Fluorouracil adjuvant chemotherapy does not increase survival in patients with CpG island methylator phenotype colorectal cancer. Gastroenterology. 140:1174–1181. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 42 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nilsson TK, Löf-Öhlin ZM and Sun X: DNA methylation of the p14ARF, RASSF1A and APC1A genes as an independent prognostic factor in colorectal cancer patients. Int J Oncol 42: 127-133, 2013
APA
Nilsson, T.K., Löf-Öhlin, Z.M., & Sun, X. (2013). DNA methylation of the p14ARF, RASSF1A and APC1A genes as an independent prognostic factor in colorectal cancer patients. International Journal of Oncology, 42, 127-133. https://doi.org/10.3892/ijo.2012.1682
MLA
Nilsson, T. K., Löf-Öhlin, Z. M., Sun, X."DNA methylation of the p14ARF, RASSF1A and APC1A genes as an independent prognostic factor in colorectal cancer patients". International Journal of Oncology 42.1 (2013): 127-133.
Chicago
Nilsson, T. K., Löf-Öhlin, Z. M., Sun, X."DNA methylation of the p14ARF, RASSF1A and APC1A genes as an independent prognostic factor in colorectal cancer patients". International Journal of Oncology 42, no. 1 (2013): 127-133. https://doi.org/10.3892/ijo.2012.1682