Yonsei Med J.  2009 Jun;50(3):309-321. 10.3349/ymj.2009.50.3.309.

Promoter Methylation in the Genesis of Gastrointestinal Cancer

Affiliations
  • 1Division of Gastroenterology, Department of Internal Medicine, Sammons Cancer Center, Baylor Research Institute, Baylor University Medical Center, Dallas, Texas, USA. RickBo@BaylorHealth.edu
  • 2Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.

Abstract

Colorectal cancers (CRC)-and probably all cancers-are caused by alterations in genes. This includes activation of oncogenes and inactivation of tumor suppressor genes (TSGs). There are many ways to achieve these alterations. Oncogenes are frequently activated by point mutation, gene amplification, or changes in the promoter (typically caused by chromosomal rearrangements). TSGs are typically inactivated by mutation, deletion, or promoter methylation, which silences gene expression. About 15% of CRC is associated with loss of the DNA mismatch repair system, and the resulting CRCs have a unique phenotype that is called microsatellite instability, or MSI. This paper reviews the types of genetic alterations that can be found in CRCs and hepatocellular carcinoma (HCC), and focuses upon the epigenetic alterations that result in promoter methylation and the CpG island methylator phenotype (CIMP). The challenge facing CRC research and clinical care at this time is to deal with the heterogeneity and complexity of these genetic and epigenetic alterations, and to use this information to direct rational prevention and treatment strategies.

Keyword

Colorectal cancer; promoter methylation; CIMP; Lynch syndrome; HNPCC; microsatellite instability; chromosomal instability; hepatocellular carcinoma

MeSH Terms

Colorectal Neoplasms/genetics
DNA Methylation/*genetics
Gastrointestinal Neoplasms/*etiology/*genetics
Humans
Microsatellite Instability
Promoter Regions, Genetic/*genetics

Figure

  • Fig. 1 The relationships among various forms of genomic instability in CRC. (A) The pie chart illustrates the characterization of 209 sporadic CRCs for MSI and CIN.2 As shown, 48% of tumors had evidence of LOH, while 14% of tumors had MSI, including a minority of tumors (3%) that overlapped with LOH. We were most interested in the observation that nearly one third of the tumors (38%) did not have either MSI or LOH. (B) The bars demonstrate the mean methylation index based upon the number of markers methylated in the three subsets of CRCs.80 Methylation alterations were analyzed at 12 markers including 6 canonical CIMP markers (MINT-1, -2, -31, p16, p14 and MLH1) and 6 additional tumors suppressor genes (PTEN, TIMP3, RUNX3, HIC1, APC, and RARβ2). The three vertical columns represent the mean methylation ratios in MSI (blue), MSI-/LOH-(red) and LOH (white) CRCs. The error bars denote the S.D. The filled circles (color matched with vertical columns) represent the 95% confidence interval (CI) of the mean methylation ratios. The rectangular boxes in the upper panels represent the pair-wise correlation between the mean methylation ratios in each subset of CRC; p-values were calculated by the Wilcoxon test. As shown in the three panels, analysis of the methylation ratios using only four CIMP-related markers (versus using 12 or 6 CIMP-related markers) demonstrates a significant positive association for MSI and MSI-/LOH-tumors, but an inverse correlation for LOH CRCs. (C) The relationship between methylation ratios calculated using all 12 methylation markers was compared with LOH ratios in the total cohort of 126 CRCs. As shown, an inverse correlation was observed (p = -0.3690; p < 0.0001) between the methylation ratio and LOH. CRC, colorectal cancers; MSI, microsatellite instability; LOH, loss of heterozygosity.

  • Fig. 2 KRAS and BRAF signaling play a role in aberrant methylation, and may contribute to epigenetic alterations in Lynch Syndrome cancers. (A) These 3 boxes show the average number of methylated loci in various subgroups of CRCs categorized by their BRAF/KRAS mutation status as published previously.31 The average number of methylated loci in each subset was calculated using all fourteen markers ("All Loci"), seven canonical CIMP markers ("CIMP") and the seven additional markers ("Non-CIMP"). The p values are based on Kruskal-Wallis one way analysis of variance on ranks, and represent the statistical differences among all three subsets of CRCs (BRAF-mutated, KRAS-mutated or wild type for both). BRAF-mutated cancers showed the highest degree of methylation at CIMP-related loci, and as a consequence, the highest degree of methylation when data were analyzed from "All Loci". KRAS mutated tumors exhibited methylation not only at the CIMP-markers, but interestingly, when data were analyzed from the additional (Non-CIMP) markers, there was a minimal difference in the number of markers methylated between BRAF- and KRAS-mutated cancers. (B) These 3 bar charts illustrate the average number of methylated loci in sporadic MSI (MSI-H), Lynch syndrome and non-MSI (MSI-L and MSS) CRCs.31 As expected, sporadic MSI tumors were most frequently methylated at all markers (CIMP and Non-CIMP). Lynch syndrome CRCs also demonstrated frequent methylation at canonical-CIMP markers, which was surprisingly slightly higher compared to the non-MSI tumors. CRC, colorectal cancers; CIMP, CpG island methylator phenotype; MSI, microsatellite instability; MSS, microsatellite stable.

  • Fig. 3 Hepatocellular carcinomas (HCCs) are classified based upon methylation and mutational status. In the upper figure, all 81 HCCs are classified by hierarchical clustering analysis based upon the degree of methylation and β-catenin or p53 mutations as published previously.39 The heat map (green-black-red) shows the tumors with a lower methylation density as green, and higher methylation density as red. The Y-axis shows the loci analyzed. A solid triangle represents a tumor with a β-catenin mutation, and an open triangle indicates a HCC with a p53 mutation. As depicted, β-catenin mutations were significantly associated with the Group 2 HCCs on the right (p < 0.0001). The table in the lower half of the figure highlights the significant association between higher methylation in Group 2 HCCs and β-catenin mutations. FAL indicates the "fractional allelic loss", or proportion of analyzed loci with allelic imbalance, or LOH.

  • Fig. 4 Treatment with a demethylating agent restores sensitivity to 5-FU toxicity in a MMR-deficient CRC cell line. (A) The top schematic summarizes the treatment plan and results from the in-vitro experiments. Four CRC cell lines - HCT116 and HCT116 + 2 (both MMR-deficient due to mutation in MLH1), HCT116 + 3 (MMR-proficient following restoration of MLH1 gene with chromosome 3 transfer in HCT116 cells) and SW48 (MMR-deficient due to hypermethylation of MLH1) - were cultivated in growth medium or exposed to 5-AZA for 24 h.66 Culture medium was exchanged and cells were allowed to form colonies over a period of 10-12 days. During this time, the cells were continuously exposed to increasing concentrations of 5-FU. Cells were washed, fixed, stained and colonies counted. MLH1 methylation status, its expression (mRNA and protein), and sensitivity to 5-FU sensitivity were monitored in each experiment. (B) A representative graph (in the left panel) depicts the mean ± S.D. of three different experiments for the colony forming assay from CRC cell lines exposed to different doses of 5-FU (1 µM, 2.5 µM, and 5 µM). Cells were allowed to form colonies for 10-12 days. The means and S.D. of three independently performed experiments are shown. As indicated, only HCT116 + 3 cells (MMR proficient cells) are sensitive to 5-FU, while all MMR-deficient cells demonstrate more colonies, indicating resistance to the chemotherapeutic drug. However, when SW48 cells were pretreated with 5 µM 5-Aza for 24 h before 5-FU exposure, MMR activity is restored, and they behave similarly to the MMR-proficient HCT116 + 3 cells (right panel). MMR, mismatch repair; CRC, colorectal cancers.

  • Fig. 5 An integrated model for different forms of genetic and epigenetic instability in CRC. Tumors can develop via chromosomal instability (CIN), microsatellite instability (MSI), or CpG island methylator phenotype (CIMP) pathways. MSI may develop either from Lynch syndrome (the hereditary form), or via the CIMP pathway (a presumably acquired form). Ultimately, all pathways converge pathologically as cancer. Based upon our current understanding, CIN and CIMP represent the two primary processes of genetic and epigenetic instability respectively, in the colon. In essence, MSI in sporadic CRCs originates because of CIMP, and once MLH1 becomes methylated, the tumors acquire MSI. Thus far the molecular mechanisms for CIN and CIMP are not clear, however, there are data to suggest that JC virus (JCV) may be a putative unifying mechanism for genetic and epigenetic instability in CRC. CRC, colorectal cancers; MMR, mismatch repair.


Cited by  2 articles

Clinical Implications of Microsatellite Instability in T1 Colorectal Cancer
Jeonghyun Kang, Hak Woo Lee, Im-kyung Kim, Nam Kyu Kim, Seung-Kook Sohn, Kang Young Lee
Yonsei Med J. 2015;56(1):175-181.    doi: 10.3349/ymj.2015.56.1.175.

Lack of Aberrant Methylation in an Adjacent Area of Left-Sided Colorectal Cancer
Otgontuya Sambuudash, Hyun-Soo Kim, Mee Yon Cho
Yonsei Med J. 2017;58(4):749-755.    doi: 10.3349/ymj.2017.58.4.749.


Reference

1. Vogelstein B, Fearon ER, Kern SE, Hamilton SR, Preisinger AC, Nakamura Y, et al. Allelotype of colorectal carcinomas. Science. 1989. 244:207–211.
Article
2. Goel A, Arnold CN, Niedzwiecki D, Chang DK, Ricciardiello L, Carethers JM, et al. Characterization of sporadic colon cancer by patterns of genomic instability. Cancer Res. 2003. 63:1608–1614.
3. Li R, Sonik A, Stindl R, Rasnick D, Duesberg P. Aneuploidy vs. gene mutation hypothesis of cancer: recent study claims mutation but is found to support aneuploidy. Proc Natl Acad Sci U S A. 2000. 97:3236–3241.
Article
4. Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. Nature. 1997. 386:623–627.
Article
5. Nishida N, Nagasaka T, Kashiwagi K, Boland CR, Goel A. High copy amplification of the Aurora-A gene is associated with chromosomal instability phenotype in human colorectal cancers. Cancer Biol Ther. 2007. 6:525–533.
Article
6. Rodriguez J, Frigola J, Vendrell E, Risques RA, Fraga MF, Morales C, et al. Chromosomal instability correlates with genome-wide DNA demethylation in human primary colorectal cancers. Cancer Res. 2006. 66:8462–9468.
Article
7. Sieber OM, Heinimann K, Gorman P, Lamlum H, Crabtree M, Simpson CA, et al. Analysis of chromosomal instability in human colorectal adenomas with two mutational hits at APC. Proc Natl Acad Sci U S A. 2002. 99:16910–16915.
Article
8. Boland CR, Komarova NL, Goel A. Chromosomal instability and cancer: not just one CINgle mechanism. Gut. 2009. 58:163–164.
Article
9. Muleris M, Chalastanis A, Meyer N, Lae M, Dutrillaux B, Sastre-Garau X, et al. Chromosomal instability in near-diploid colorectal cancer: a link between numbers and structure. PLoS ONE. 2008. 3:e1632.
Article
10. Duval A, Reperant M, Compoint A, Seruca R, Ranzani GN, Iacopetta B, et al. Target gene mutation profile differs between gastrointestinal and endometrial tumors with mismatch repair deficiency. Cancer Res. 2002. 62:1609–1612.
11. Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995. 268:1336–1338.
Article
12. Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC, et al. Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science. 1997. 275:967–969.
Article
13. Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology. 2008. 135:1079–1099.
Article
14. Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A. 1999. 96:8681–8686.
Article
15. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 2003. 349:2042–2054.
Article
16. Yamashita K, Dai T, Dai Y, Yamamoto F, Perucho M. Genetics supersedes epigenetics in colon cancer phenotype. Cancer Cell. 2003. 4:121–131.
Article
17. Ahuja N, Mohan AL, Li Q, Stolker JM, Herman JG, Hamilton SR, et al. Association between CpG island methylation and microsatellite instability in colorectal cancer. Cancer Res. 1997. 57:3370–3374.
18. Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, et al. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res. 1997. 57:808–811.
19. 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. 38:787–793.
Article
20. Eads CA, Danenberg KD, Kawakami K, Saltz LB, Danenberg PV, Laird PW. CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res. 1999. 59:2302–2306.
21. Samowitz WS, Albertsen H, Herrick J, Levin TR, Sweeney C, Murtaugh MA, et al. Evaluation of a large, population-based sample supports a CpG island methylator phenotype in colon cancer. Gastroenterology. 2005. 129:837–845.
22. Goel A, Nagasaka T, Arnold CN, Inoue T, Hamilton C, Niedzwiecki D, et al. The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology. 2007. 132:127–138.
23. Ogino S, Odze RD, Kawasaki T, Brahmandam M, Kirkner GJ, Laird PW, et al. 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.
24. Tuupanen S, Karhu A, Järvinen H, Mecklin JP, Launonen V, Aaltonen LA. No evidence for dual role of loss of heterozygosity in hereditary non-polyposis colorectal cancer. Oncogene. 2007. 26:2513–2517.
25. Kaz A, Kim YH, Dzieciatkowski S, Lynch H, Watson P, Kay Washington M, et al. Evidence for the role of aberrant DNA methylation in the pathogenesis of Lynch syndrome adenomas. Int J Cancer. 2007. 120:1922–1929.
26. Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007. 318:1108–1113.
27. Nagasaka T, Goel A, Notohara K, Takahata T, Sasamoto H, Uchida T, et al. Methylation pattern of the O6-methylguanine-DNA methyltransferase gene in colon during progressive colorectal tumorigenesis. Int J Cancer. 2008. 122:2429–2436.
Article
28. Forrester K, Almoguera C, Han K, Grizzle WE, Perucho M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature. 1987. 327:298–303.
Article
29. Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van Boom JH, van der Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature. 1987. 327:293–297.
Article
30. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002. 417:949–954.
Article
31. Nagasaka T, Koi M, Kloor M, Gebert J, Vilkin A, Nishida N, et al. Mutations in both KRAS and BRAF may contribute to the methylator phenotype in colon cancer. Gastroenterology. 2008. 134:1950.e1–1960.e1.
Article
32. Aaltonen LA, Peltomäki P, Mecklin JP, Järvinen H, Jass JR, Green JS, et al. Replication errors in benign and malignant tumors from hereditary nonpolyposis colorectal cancer patients. Cancer Res. 1994. 54:1645–1648.
33. Tannergård P, Liu T, Weger A, Nordenskjöld M, Lindblom A. Tumorigenesis in colorectal tumors from patients with hereditary non-polyposis colorectal cancer. Hum Genet. 1997. 101:51–55.
Article
34. Esteller M, Fraga MF, Guo M, Garcia-Foncillas J, Hedenfalk I, Godwin AK, et al. DNA methylation patterns in hereditary human cancers mimic sporadic tumorigenesis. Hum Mol Genet. 2001. 10:3001–3007.
Article
35. Sanchez de Abajo A, de la Hoya M, van Puijenbroek M, Godino J, Díaz-Rubio E, Morreau H, et al. Dual role of LOH at MMR loci in hereditary non-polyposis colorectal cancer? Oncogene. 2006. 25:2124–2130.
Article
36. Kim WS, Son HJ, Park JO, Song SY, Park C. Promoter methylation and down-regulation of DAPK is associated with gastric atrophy. Int J Mol Med. 2003. 12:827–830.
37. Garcia-Manero G, Bueso-Ramos C, Daniel J, Williamson J, Kantarjian HM, Issa JP. DNA methylation patterns at relapse in adult acute lymphocytic leukemia. Clin Cancer Res. 2002. 8:1897–1903.
38. Shen L, Ahuja N, Shen Y, Habib NA, Toyota M, Rashid A, et al. DNA methylation and environmental exposures in human hepatocellular carcinoma. J Natl Cancer Inst. 2002. 94:755–761.
Article
39. Nishida N, Nishimura T, Nagasaka T, Ikai I, Goel A, Boland CR. Extensive methylation is associated with beta-catenin mutations in hepatocellular carcinoma: evidence for two distinct pathways of human hepatocarcinogenesis. Cancer Res. 2007. 67:4586–4594.
Article
40. Nishida N, Nagasaka T, Nishimura T, Ikai I, Boland CR, Goel A. Aberrant methylation of multiple tumor suppressor genes in aging liver, chronic hepatitis, and hepatocellular carcinoma. Hepatology. 2008. 47:908–918.
41. Greger V, Passarge E, Höpping W, Messmer E, Horsthemke B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Hum Genet. 1989. 83:155–158.
Article
42. Robertson KD, Wolffe AP. DNA methylation in health and disease. Nat Rev Genet. 2000. 1:11–19.
Article
43. Li E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet. 2002. 3:662–673.
Article
44. Slattery ML, Curtin K, Sweeney C, Levin TR, Potter J, Wolff RK, et al. Diet and lifestyle factor associations with CpG island methylator phenotype and BRAF mutations in colon cancer. Int J Cancer. 2007. 120:656–663.
Article
45. Samowitz WS, Albertsen H, Sweeney C, Herrick J, Caan BJ, Anderson KE, et al. Association of smoking, CpG island methylator phenotype, and V600E BRAF mutations in colon cancer. J Natl Cancer Inst. 2006. 98:1731–1738.
Article
46. Kaneko Y, Sakurai S, Hironaka M, Sato S, Oguni S, Sakuma Y, et al. Distinct methylated profiles in Helicobacter pylori dependent and independent gastric MALT lymphomas. Gut. 2003. 52:641–646.
47. Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, et al. High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin Cancer Res. 2006. 12:989–995.
Article
48. Chan AO, Peng JZ, Lam SK, Lai KC, Yuen MF, Cheung HK, et al. Eradication of Helicobacter pylori infection reverses E-cadherin promoter hypermethylation. Gut. 2006. 55:463–468.
Article
49. Perri F, Cotugno R, Piepoli A, Merla A, Quitadamo M, Gentile A, et al. Aberrant DNA methylation in non-neoplastic gastric mucosa of H. Pylori infected patients and effect of eradication. Am J Gastroenterol. 2007. 102:1361–1371.
Article
50. Goel A, Li MS, Nagasaka T, Shin SK, Fuerst F, Ricciardiello L, et al. Association of JC virus T-antigen expression with the methylator phenotype in sporadic colorectal cancers. Gastroenterology. 2006. 130:1950–1961.
Article
51. Herath NI, Leggett BA, MacDonald GA. Review of genetic and epigenetic alterations in hepatocarcinogenesis. J Gastroenterol Hepatol. 2006. 21:15–21.
Article
52. Kang GH, Lee S, Kim WH, Lee HW, Kim JC, Rhyu MG, et al. Epstein-barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. Am J Pathol. 2002. 160:787–794.
Article
53. Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007. 39:237–242.
Article
54. Ohm JE, Baylin SB. Stem cell chromatin patterns: an instructive mechanism for DNA hypermethylation? Cell Cycle. 2007. 6:1040–1043.
Article
55. Rashid A, Shen L, Morris JS, Issa JP, Hamilton SR. CpG island methylation in colorectal adenomas. Am J Pathol. 2001. 159:1129–1135.
Article
56. Chan AO, Broaddus RR, Houlihan PS, Issa JP, Hamilton SR, Rashid A. CpG island methylation in aberrant crypt foci of the colorectum. Am J Pathol. 2002. 160:1823–1830.
Article
57. Shin SK, Nagasaka T, Jung BH, Matsubara N, Kim WH, Carethers JM, et al. Epigenetic and genetic alterations in Netrin-1 receptors UNC5C and DCC in human colon cancer. Gastroenterology. 2007. 133:1849–1857.
Article
58. Ahlquist DA, Skoletsky JE, Boynton KA, Harrington JJ, Mahoney DW, Pierceall WE, et al. Colorectal cancer screening by detection of altered human DNA in stool: feasibility of a multitarget assay panel. Gastroenterology. 2000. 119:1219–1227.
Article
59. Müller HM, Oberwalder M, Fiegl H, Morandell M, Goebel G, Zitt M, et al. Methylation changes in faecal DNA: a marker for colorectal cancer screening? Lancet. 2004. 363:1283–1285.
Article
60. Nagasaka T, Goel A, Matsubara N, Tanaka N. Detection of fecal DNA methylation for colorectal neoplasia: does it lead to an optimal screening test? Acta Med Okayama. 2006. 60:249–256.
61. Chen WD, Han ZJ, Skoletsky J, Olson J, Sah J, Myeroff L, et al. Detection in fecal DNA of colon cancer-specific methylation of the nonexpressed vimentin gene. J Natl Cancer Inst. 2005. 97:1124–1132.
Article
62. Zou H, Taylor WR, Harrington JJ, Hussain FT, Cao X, Loprinzi CL, et al. High detection rates of colorectal neoplasia by stool DNA testing with a novel digital melt curve assay. Gastroenterology. 2009. 136:459–470.
Article
63. Esteller M. Epigenetics in cancer. N Engl J Med. 2008. 358:1148–1159.
Article
64. Mitsuno M, Kitajima Y, Ide T, Ohtaka K, Tanaka M, Satoh S, et al. Aberrant methylation of p16 predicts candidates for 5-fluorouracil-based adjuvant therapy in gastric cancer patients. J Gastroenterol. 2007. 42:866–873.
Article
65. Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008. 123:8–13.
66. Arnold CN, Goel A, Boland CR. Role of hMLH1 promoter hypermethylation in drug resistance to 5-fluorouracil in colorectal cancer cell lines. Int J Cancer. 2003. 106:66–73.
Article
67. Oki Y, Issa JP. Review: recent clinical trials in epigenetic therapy. Rev Recent Clin Trials. 2006. 1:169–182.
Article
68. Kaminskas E, Farrell A, Abraham S, Baird A, Hsieh LS, Lee SL, et al. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res. 2005. 11:3604–3608.
Article
69. Hellebrekers DM, Griffioen AW, van Engeland M. Dual targeting of epigenetic therapy in cancer. Biochim Biophys Acta. 2007. 1775:76–91.
70. Walker DL, Padgett BL, ZuRhein GM, Albert AE, Marsh RF. Human papovavirus (JC): induction of brain tumors in hamsters. Science. 1973. 181:674–676.
71. Gordon J, Khalili K. The human polyomavirus, JCV, and neurological diseases (review). Int J Mol Med. 1998. 1:647–655.
Article
72. Boland CR, Luciani MG, Gasche C, Goel A. Infection, inflammation, and gastrointestinal cancer. Gut. 2005. 54:1321–1331.
Article
73. Laghi L, Randolph AE, Chauhan DP, Marra G, Major EO, Neel JV, et al. JC virus DNA is present in the mucosa of the human colon and in colorectal cancers. Proc Natl Acad Sci U S A. 1999. 96:7484–7489.
Article
74. Ricciardiello L, Laghi L, Ramamirtham P, Chang CL, Chang DK, Randolph AE, et al. JC virus DNA sequences are frequently present in the human upper and lower gastrointestinal tract. Gastroenterology. 2000. 119:1228–1235.
Article
75. Jung WT, Li MS, Goel A, Boland CR. JC virus T-antigen expression in sporadic adenomatous polyps of the colon. Cancer. 2008. 112:1028–1036.
Article
76. Enam S, Del Valle L, Lara C, Gan DD, Ortiz-Hidalgo C, Palazzo JP, et al. Association of human polyomavirus JCV with colon cancer: evidence for interaction of viral T-antigen and beta-catenin. Cancer Res. 2002. 62:7093–7101.
77. Casini B, Borgese L, Del Nonno F, Galati G, Izzo L, Caputo M, et al. Presence and incidence of DNA sequences of human polyomaviruses BKV and JCV in colorectal tumor tissues. Anticancer Res. 2005. 25:1079–1085.
78. Theodoropoulos G, Panoussopoulos D, Papaconstantinou I, Gazouli M, Perdiki M, Bramis J, et al. Assessment of JC polyoma virus in colon neoplasms. Dis Colon Rectum. 2005. 48:86–91.
Article
79. Ricciardiello L, Baglioni M, Giovannini C, Pariali M, Cenacchi G, Ripalti A, et al. Induction of chromosomal instability in colonic cells by the human polyomavirus JC virus. Cancer Res. 2003. 63:7256–7262.
80. Goel A, Nagasaka T, Arnold CN, Inoue T, Hamilton C, Niedzwiecki D, et al. The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology. 2007. 132:127–138.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr