Abstract
Colorectal cancer is an emerging public health problem in Indonesia and currently ranks among the three highest cancers. Lack of a colonoscopy screening and lifestyle changes might contribute to it. In the last few decades, there is an increasing interest towards the contribution of genetic-environment interaction in colorectal carcinogenesis. Some studies have indicated that CRC might develop through several different pathways; the three major routes are chromosomal instability (CIN), microsatellite instability (MSI), and inflammatory pathways. An earlier study on clinical epidemiology of CRC in Indonesia showed that the majority of patients were diagnosed between 45 and 50 years old, with a mean age around 47 years old. Further studies showed that most young Indonesian cases of CRC do not have hereditary characteristics; however, the CRC did not follow the conventional pathways of sporadic CRC (the CIN) pathway. Rather, it is a mixed of MSI and inflammatory pathways. Immunohistochemical studies showed that the proportion of patients with negative mismatch repair proteins was 43.5% for MSH2 and 83.5% for MLH1. Along the sporadic colorectal carcinogenesis pathway, there was a specific role of cyclooxygenase-2 (COX-2) enzyme during the polyp formation. COX-2 expression was reported in about 80% CRC cases worldwide. However, our study found only 49% of COX-2 expression among the CRC patients. Interestingly, an inflammatory marker, the nucleus factor κB (NF-κB), was expressed in about 73.5% cases, in line with a previous study. More recently, KRAS has been used as a potential tumor marker to select treatment and its expression was reported to be as high as 30%-40% worldwide. However, we found that KRAS gene expression was only 16.3%. Our findings support that CRC patients in Indonesian might follow a distinct pathway, a hypothesis that deserves further exploration.
Keywords: Colorectal cancer, Molecular profile, Indonesia
Background
Colorectal cancer (CRC) is an emerging public health problem in Indonesia and currently ranks among the three highest cancers. The age-standardized incidence rates of CRC per 100.000 populations in Indonesia were 19.1 for men and 15.6 for women (1). These rates are much lower than the incidence rates in Australia, New Zealand and Western Europe, but the number of cases is high because Indonesia ranks the fourth most populated country in the world with more than 235 million populations.
Previous epidemiological studies have shown that CRC patients in Indonesia are younger than patients in developed countries. More than 30% of cases were 40 years old or younger (2), while patients younger than 50 years in developed countries accounted for only 2-8% (3, 4). In addition, whereas young or early onset CRC in developed countries are often associated with familial cases (5, 6), most Indonesian CRC patients are determined to be sporadic. With the increasing number of cases and the occurrence of CRC during a person's productive years, it is obvious that CRC poses a major public health problem in Indonesia.
Early onset CRC in developed countries showed typical characteristics such as proximal localization (ascending colon), low pathological stage, rarely metastasize, and has better prognosis (7). However, our patients showed otherwise characteristics, i.e. distal localization (rectum), high clinical stage, and poor survival (unpublished data). All these observations have raised questions regarding the carcinogenesis pathways of CRCs in Indonesia.
Genomic instability in colorectal cancer
Classically, CRC is believed to develop from an adenoma, the so-called adenoma-carcinoma sequence (8, 9). Epithelial malignant transformation occurs after the cell underwent genomic instability, which promotes the accumulation of genetic and epigenetic alterations without obvious phenotypic changes (10). Genomic instability is the prerequisite of CRC development which allows multiple, gradual genetic mutations (11). There are two distinct pathways that marked the genomic instability, i.e. the microsatellite instability (MSI) and the chromosomal instability (CIN) pathways.
MSI is recognized as genome-wide alterations in repetitive DNA sequence (12). It is found in almost all malignancies from patients with hereditary non-polyposis colorectal cancer (HNPCC) and approximately 15% of the sporadic cancers (13). MSI is caused by defects in the nucleotide mismatch repair (MMR) mechanisms. Mismatch repair enzymes normally recognize errors in nucleotide matching of complementary chromosome strands and initiate segmental excision of the newly synthesized strand to ensure faithful strand replication.
CIN is the most common type of genomic instability and is associated with loss of heterozygosity in various tumor suppressor loci, especially 18q (14). Alterations range from different types of chromosomal rearrangements, to loss and gain of large fragments of the genome and may have abnormal number of chromosomes or DNA content (aneuploidy).
The role of epigenetic alterations
DNA methylation refers to the methyation of cytosine (C) nucleotide preceding guanine (G) nucleotide (CpG islands) throughout the genome, preferentially in gene promoter regions. This alteration may inactivate tumor suppressor genes, thereby promoting cancer. In colorectal cancer, DNA methylation may inactivate the MMR gene hMLH1 gene leading to microsatellite instability (15). Both hypomethylation of the genome and simultaneous hypermethylation of the promoter regions of strategic genes frequently occur (16).
Pathways to Colorectal Cancers
Colorectal cancer is a heterogeneous disease and may arise through at least three main pathways, namely sporadic, hereditary and inflammation-associated cancers. These pathways harbor distinct genetic alterations although they may share some common mutations.
Hereditary Non-Polyposis Colorectal Cancer (HNPCC)
HNPCC accounts for about 1-8% of all CRC cases based on clinical criteria, i.e. the Amsterdam criteria and the revised Bethesda guidelines (17–19). Pathogenesis of familial or hereditary CRC involved germ-line mutations of mismatch repair (MMR) gene family members and was named microsatellite instability (MSI) pathway (20, 21). Mutation of MMR genes such as hMLH1 and hMSH2 range from 0.3% to 3% of total CRC burden (22). However, hMLH1 and hMSH2 gene mutations account for more than 90% of HNPCC cases, with the remaining mutations occur in other MMR genes such as hMSH6, hPMS1, and hPMS2 (23).
Sporadic Colorectal Cancer
Most sporadic CRC follow the path of chromosomal instability (CIN) (24) that involved somatic mutation in adenomatous poliposis coli (APC) gene and other mutations which have been described as vogelstein adenoma-carcinoma sequel. This pathway is also known as the classical pathway, which involves mutation in APC, KRAS, and p53 genes, loss of 18q and deletion of 17p which harbor tumor suppressor gene p53 locus (8).
Mutations in the APC gene on chromosome 5q21 locus are considered the earliest events of colorectal carcinogenesis and constitutes between 60% and 80% of sporadic colorectal carcinoma and adenomas (25). Allelic loss of chromosome 18q21, which contains DCC, SMAD2, and SMAD4, is found in up to 60% of CRCs. Mutation of SMAD4 may be more influential in colorectal carcinogenesis and marked the divergence of chromosomal instability (CIN) from the microsatellite instability (MSI) pathway (26). A small proportion of sporadic CRCs, however, contains mutation in MMR genes and showed high MSI (MSI-H).
The reported incidences of KRAS and BRAF mutations in CRC were 37% and 13% of respectively (27, 28). The frequency of KRAS mutation was different among HNPCC, MSS, and MSI-H CRC cases. HNPCC and sporadic MSI-H tumors without hMLH1 hypermethylation shared similar KRAS mutation frequency. These findings suggested that KRAS mutation frequency depends on the genetic or epigenetic mechanism (29). Mutation in BRAF gene, another member of the RAS/RAF/MAPK pathway, is also crucial in colorectal tumorigenesis and co-occurrence with KRAS mutation in the same individual patient is a rare case (30, 31). The frequency of V600E mutation in BRAF has been described in about 35% of sporadic MSI-H CRCs and about 6% in CRC without MMR deficiency or microsatellite stable (MSS) CRCs (32–34). Moreover, BRAF mutation is associated with hMLH1 promoter hypermethylation (34, 35). While KRAS gene is mutated in about 20% of sporadic MSI-H and of 35% of MSS CRCs negative for the BRAF V600E mutation (32, 33, 36).
Colitis-Associated Cancer
Inflammation has been associated with cancer since the observation of leukocyte infiltrations in neoplastic tissues by Rudolf Virchow in 1863 (37). Inflammation could alter the dynamics of colonic epithelial cells growth, either their promoting survival or preventing apoptosis (38). Studies have shown a relationship between colonic inflammation and cancer incidence as indicated in context of ulcerative colitis and Crohn disease, the so-called inflammatory bowel disease (IBD). Colorectal cancer arising from IBD mostly occurs from a dysplastic precursor forming a flat adenoma, instead of polyp (39, 40).
Besides genes classically known to be involved in sporadic colorectal carcinogenesis, patients with IBD also showed overexpression of inflammatory genes, such as the cyclooxygenase 2 (COX-2) genes (41) and nuclear factor kappa B (NF-κB) (42). COX-2 enzyme plays important role in producing prostaglandin E2 (PGE2), which stimulates cascades of signaling pathways leading to tumor growth, such as activation of phosphotidilinositol-3-kinase (PI3) and Wnt/β-catenin pathways (43). COX-2 is expressed in 40% of colorectal adenomas and 85% of sporadic CRC (44). Early molecular studies showed that colorectal tumors expressed high level of mRNA (86%), while in normal colon mucosa, its expression is low or absent (45, 46). High expression of COX-2 in sporadic CRC indicated its important role in colorectal carcinogenesis, which probably occurs early during the formation of adenomatous polyp (47).
Activation of NF-κB has been described in Crohn's disease, ulcerative colitis, self-limited colitis, and experimental colitis (48, 49). Nuclear factor kappa B is the most important component of inflammatory signaling pathway that may promote tumorigenesis. It is a central transcription factor activated by inflammatory signals in response to infectious agents, cytokines, and necrotic cellular remnants (50). Activation of NF-κB leads to the overexpression of cell cycle genes, apoptosis inhibitor and proteases; all of which may trigger invasive phenotypes. Immunohistochemical staining of RelA protein, the p65 component of NF-κB, found a significant difference of NF-κB expression among normal colorectal mucosa (9.3%), colorectal adenoma (54.0%), and colorectal adenocarcinoma (71.9%) (51).
Molecular characteristics of CRC in Indonesia
MSI and CIN pathways
Most CRC patients in Indonesia are sporadic, even though they were 40 years old or less. Early study showed that the expression of MMR proteins did not differ between young (40 years or less) and old (60 years or more) patients. Moreover, MSH2 and MLH1 were not expressed in 43.5% and 83.5% of cases, respectively hinting toward a defect in DNA mismatch repair system which may promote microsatellite instability (52). However, when tested using BAT26, a surrogate markers of MSI our samples showed very low frequency of MSI that is consistent with sporadic cancer feature. The low expression of normal MMR protein may be caused by epigenetic silencing event of DNA methylation in the promoter regions of MMR genes, which are known to occur in sporadic CRC patients (53). If this is true, then young CRC patients in Indonesia conform to sporadic molecular feature and may not use the MSI pathway, in spite of the large proportion of patients showing low expression of MMR genes. To test this hypothesis, we have evaluated the same specimens for SMAD4 protein expression. None of these tumors stained positive for SMAD4, either young or older patients, confirming that they are not following the MSI pathway.(52)
Oncogenic mutation of KRAS and BRAF genes
Our preliminary study found that only 16.3% of of 43 sporadic CRC cases harbored KRAS mutation and no BRAF mutation (54). This low KRAS mutation may indicate a different pattern of genetic instability in these colorectal tumors. The absence of BRAF gene mutation suggested that the samples may not belongs to the MSI-H group, which also associated with high methylation of hMLH1 gene promoter. However, we have not analyzed the methylation pattern of our samples, while studies assessing the mutation loci of the KRAS gene are still ongoing.
Inflammatory pathways
Using immunohistochemistry technique, we found that NF-κB was expressed in 73.5% of CRC specimens, whereas COX-2 expression was found in only 49.0% cases (55). The COX-2 expression may be considered as low since most studies in sporadic CRC found at least 80% of cases expressed COX-2 in the cytoplasm (44–46, 56). Our further analysis found that there was a significant association between COX-2 and NF-κB expressions (bivariate analysis p < 0.001 and degree of agreement of 74%). The intensity score was also significantly correlated (r = 0.941; p < 0.001) (57). Other study also found a high correlation between COX-2 and RelA (p65) component of NF-κB expression (r = 0.83; p < 0.05) (58). The NF-κB is an important transcription factor to activate COX-2 gene expression. The 5’-UTR region of COX-2 gene promoter contains regulatory binding sites of several transcription factors, including two NF-κB binding site motifs (59).
Our study supports the hypothesis that inflammation may play roles in Indonesian CRC patients. However, the role of IBD in our subjects has not been proven yet. COX-2 was also expressed in moderate percentage (23.9%) in normal mucosa of our specimens (57), indicating the presence of chronic inflammation of unknown etiology (non-specific colitis). The role of normal gut flora as the etiology of CRC is difficult to be shown. However, experiment in mice suggested that the normal gut flora may play a role in colitis-associated carcinoma (60).
Is there another pathway?
Studies elucidating the molecular pathway of colorectal carcinogenesis in Indonesia have just begun. Besides the three main pathways mentioned above, there are several minor pathways. Serrated pathway is believed to arise in serrated adenoma and genetically associated with BRAF gene mutations exhibiting extensive DNA methylation but lacking APC gene mutations (61). The serrated adenoma is a precursor lesion of sporadic CRC with high microsatellite instability (MSI-H), which constitutes approximately 15% of sporadic colon cancers (62). However, the absence of BRAF mutation in our preliminary study does not support the notion that our CRC patients might take this pathway. Studies are still ongoing to tract their genetic alteration patterns. The summary of clinicopathological aspects and genetic mutation associated with various CRC pathways is given in Table 1 along with the proposed unanswered question on Indonesian CRCs. It seems that colorectal carcinogenesis in our population is still far beyond from common understanding and more genetic studies are needed to reveal the heterogeneous mechanisms of its process.
Table 1.
Sporadic | HNPCC | CAC | Serrated pathway | Sporadic indonesia? | |
---|---|---|---|---|---|
Age | >60 years | <40 years | Young | Young | |
Location | Distal | Proximal | Proximal | Distal | |
Pathway | Adenoma-carcinoma | Adenoma-carcinoma | Inflammation-dysplasia-cancer | Hyperplastic polyp- cancer | Adenoma-carcinoma |
CIN | No | ||||
Defective MMR protein | No | Yes | No | Yes | Yes |
MSI | No | Yes | No | Yes | No |
Inflammation | No | No | IBD | No | Non-specific? |
Adenoma | Polyp | Polyp | No | Serrated adenoma | Polyp |
KRAS mutation | Yes | Yes | Yes | Yes | No |
BRAF mutation | No | No | No | Yes | No |
COX-2 | No | High | |||
NF-κB | Yes | No | Yes | ? | Yes |
(Please cite as: Abdullah M, Sudoyo AW, Utomo AR, Fauzi A, Rani AA. Molecular profile of colorectal cancer in Indonesia: is there another pathway? Gastroenterol Hepatol Bed Bench 2012;5(2):71-78.)
References
- 1.Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010 doi: 10.1002/ijc.25516. in press. [DOI] [PubMed] [Google Scholar]
- 2.Departemen Kesehatan RI. Laporan Tahunan Kanker: Direktorat Jendral Pelayanan Medik dan Perhimpunan Patologi Anatomik Indonesia. Jakarta: Depkes RI; 1995. [Google Scholar]
- 3.Parramore JB, Wei JP, Yeh KA. Colorectal cancer in patients under forty: presentation and outcome. Am Surg. 1998;64:563–67. [PubMed] [Google Scholar]
- 4.Lee PY, Fletcher WS, Sullivan ES, Vetto JT. Colorectal cancer in young patients: characteristics and outcome. Am Surg. 1994;60:607–12. [PubMed] [Google Scholar]
- 5.Turkiewicz D, Fletcher WS, Sullivan ES, Vetto JT. Colorectal cancer in young patients: characteristics and outcome. Am Surg. 1994;60:607–12. [PubMed] [Google Scholar]
- 6.Chung YF, Eu KW, Machin D, Ho JM, Nyam DC, Leong AF, et al. Young age is not a poor prognostic marker in colorectal cancer. Br J Surg. 1998;85:1255–9. doi: 10.1046/j.1365-2168.1998.00805.x. [DOI] [PubMed] [Google Scholar]
- 7.Muta H, Noguchi M, Perrucho M, Ushio K, Sugihara D, Ochiai A, et al. Clinical implications of microsatellite instability in colorectal cancer. Cancer. 1996;77:265–70. doi: 10.1002/(SICI)1097-0142(19960115)77:2<265::AID-CNCR7>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
- 8.Vogelstein B. Genetic alterations during colorectal tumor development. N Engl J Med. 1988;319:525–32. doi: 10.1056/NEJM198809013190901. [DOI] [PubMed] [Google Scholar]
- 9.Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759–67. doi: 10.1016/0092-8674(90)90186-i. [DOI] [PubMed] [Google Scholar]
- 10.Schneider BL, Kulesz-Martin M. Destructive cycles: the role of genomic instability and adaptation in carcinogenesis. Carcinogenesis. 2004;25:2033–44. doi: 10.1093/carcin/bgh204. [DOI] [PubMed] [Google Scholar]
- 11.Stoler DL, Chen N, Basik M, Kahlenberg MS, Rodiguez-Bigas MA, Petrelli NJ, et al. The onset and extent of genomic instability in sporadic colorectal tumor progression. Proc Natl Acad Sci USA. 1999;96:15121–26. doi: 10.1073/pnas.96.26.15121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lothe RA. Microsatellite instability in human solid tumors. Mol Med Today. 1997;3:61–68. doi: 10.1016/S1357-4310(96)10055-1. [DOI] [PubMed] [Google Scholar]
- 13.Breivik J, Gaudernack G. Genomic instability, DNA methylation, and natural selection in colorectal carcinogenesis. Cancer Biol. 1999;9:245–54. doi: 10.1006/scbi.1999.0123. [DOI] [PubMed] [Google Scholar]
- 14.Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. Nature. 1997;386:623–7. doi: 10.1038/386623a0. [DOI] [PubMed] [Google Scholar]
- 15.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;57:808–11. [PubMed] [Google Scholar]
- 16.Bariol C, Suter C, Cheong K, Ku SL, Meagher A, Hawkins N, et al. The relationship between hypomethylation and CpG island methylation in colorectal neoplasia. Am J Pathol. 2003;162:1361–71. doi: 10.1016/S0002-9440(10)63932-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Aaltonen LA, Sankila R, Mecklin JP, Jarvinen H, Pukkala E, Peltomaki P, et al. A novel approach to estimate the proportion of hereditary nonpolyposis colorectal cancer of total colorectal cancer burden. Cancer Detect Prev. 1994;18:57–63. [PubMed] [Google Scholar]
- 18.Vasen H. Clinical diagnosis and management of hereditary colorectal cancer syndromes. J Clin Oncol. 2000;18:81s–92s. [PubMed] [Google Scholar]
- 19.Cravo ML. Validation and simplification of Bethesda guidelines for identifying apparently sporadic forms of colorectal carcinoma with microsatellite instability. Cancer. 1999;85:779–85. [PubMed] [Google Scholar]
- 20.Lynch HT, de-la-Chapelle A. Genetic susceptibility to non-polyposis colorectal cancer. J Med Genet. 1999;36:801–18. [PMC free article] [PubMed] [Google Scholar]
- 21.Salovaara R, Loukola A, Kristo P, Kaariainen H, Ahtola H, Eskelinen M, et al. Population-based molecular detection of hereditary nonpolyposis colorectal cancer. J Clin Oncol. 2000;18:2193–200. doi: 10.1200/JCO.2000.18.11.2193. [DOI] [PubMed] [Google Scholar]
- 22.Aaltonen LA, Salovaara R, Kristo P, Canzian F, Hemminki A, Peltomaki P, et al. Incidence of hereditary nonpolyposis colorectal cancer and the feasibility of molecular screening for the disease. N Engl J Med. 1998;338:1481–87. doi: 10.1056/NEJM199805213382101. [DOI] [PubMed] [Google Scholar]
- 23.Mitchell RJ, Farrington SM, Dunlop MG, Campbell H. Mismatch repair genes hMLH1 and hMSH2 and colorectal cancer: A huge review. Am J Epidemiol. 2002;156:885–902. doi: 10.1093/aje/kwf139. [DOI] [PubMed] [Google Scholar]
- 24.Breivik J. The evolutionary origin of genetic instability in cancer development. Semin Cancer Biol. 2005;15:51–60. doi: 10.1016/j.semcancer.2004.09.008. [DOI] [PubMed] [Google Scholar]
- 25.Narayan S, Roy D. Role of APC and DNA mismatch repair genes in the development of colorectal cancer. Mol Cancer. 2003;2(41):1–15. doi: 10.1186/1476-4598-2-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Woodford-Richens KL, Rowan AJ, Gorman P, Halford S, Bicknell DC, Wasan HS, et al. SMAD4 mutations in colorectal cancer probably occur before chromosomal instability, but after divergence of the microsatellite instability pathway. Proc Natl Acad Sci USA. 2001;98:9713–23. doi: 10.1073/pnas.171321498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 28.Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogestein B, Velculescu VE. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 2002;418:934. doi: 10.1038/418934a. [DOI] [PubMed] [Google Scholar]
- 29.Oliveira C, Westera JL, Arango D, Ollikainen M, Domingo E, Ferreira A, et al. Distinct patterns of KRAS mutations in colorectal carcinomas according to germline mismatch repair defects and hMLH1 methylation status. Hum Mol Genet. 2004;13:2303–11. doi: 10.1093/hmg/ddh238. [DOI] [PubMed] [Google Scholar]
- 30.Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell. 1996;87:159–70. doi: 10.1016/s0092-8674(00)81333-1. [DOI] [PubMed] [Google Scholar]
- 31.Yuen ST, Davies H, Chan TL, Ho JW, Bignel GR, Cox C, et al. Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res. 2002;62:6451–5. [PubMed] [Google Scholar]
- 32.Oliveira C, Pinto M, Duval A, Brennetot C, Domingo E, Espin E, et al. BRAF mutations characterize colon but not gastric cancer with mismatch repair deficiency. Oncogene. 2003;22:9192–96. doi: 10.1038/sj.onc.1207061. [DOI] [PubMed] [Google Scholar]
- 33.Koinuma K, Shitoh K, Miyakura Y, Furukawa T, Yamashita Y, Ota J, et al. Mutations of BRAF are associated with extensive MLH1 promoter methylation in sporadic colorectal carcinomas. Int J Cancer. 2004;108:237–42. doi: 10.1002/ijc.11523. [DOI] [PubMed] [Google Scholar]
- 34.Wang L, Cunningham JM, Winters JL, Guenther JC, French AJ, Boardman LA, et al. BRAF mutations in colon cancer are not likely attributable to defective DNA mismatch repair. Cancer Res. 2003;63:5209–12. [PubMed] [Google Scholar]
- 35.Deng G, Bell I, Crawley S, Gum J, Terdiman JP, Allen BA, et al. BRAF mutation is frequently present in sporadic colorectal cancer with methylated MLH1 but not in hereditary nonpolyposis colorectal cancer. Clin Cancer Res. 2004;10:191–95. doi: 10.1158/1078-0432.ccr-1118-3. [DOI] [PubMed] [Google Scholar]
- 36.Fujiwara T, Stolker JM, Watanabe T, Rashid A, Longo P, Eshleman JR, et al. Accumulated clonal genetic alterations in familial and sporadic colorectal carcinomas with widespread instability in microsatellite sequences. Am J Pathol. 1998;153:1063–78. doi: 10.1016/S0002-9440(10)65651-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–45. doi: 10.1016/S0140-6736(00)04046-0. [DOI] [PubMed] [Google Scholar]
- 38.Shanmugathasan M, Jothy S. Apoptosis, anoikis and their relevance to the pathobiology of colon cancer. Pathol Int. 2000;50:273–79. doi: 10.1046/j.1440-1827.2000.01047.x. [DOI] [PubMed] [Google Scholar]
- 39.Itzkowitz SH, Yio X. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004;287:G7–G17. doi: 10.1152/ajpgi.00079.2004. [DOI] [PubMed] [Google Scholar]
- 40.Itzkowitz SH. Molecular biology of dysplasia and cancer in inflammatory bowel disease. Gastroenterol Clin N Am. 2006;35:553–71. doi: 10.1016/j.gtc.2006.07.002. [DOI] [PubMed] [Google Scholar]
- 41.Agoff SN, Brentnall TA, Crispin DA, Taylor SL, Raaka S, Haggitt RC, et al. The role of cyclooxygenase 2 in ulcerative colitis-associated neoplasia. Am J Pathol. 2000;157:737–45. doi: 10.1016/S0002-9440(10)64587-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rogler G, Brand K, Vogl D, Page S, Hofmeister R, Andus T, et al. Nuclear factor κB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology. 1998;115:357–69. doi: 10.1016/s0016-5085(98)70202-1. [DOI] [PubMed] [Google Scholar]
- 43.Markowitz SD. Aspirin and colon cancer - targeting prevention? N Engl J Med. 2007;356:2195–8. doi: 10.1056/NEJMe078044. [DOI] [PubMed] [Google Scholar]
- 44.Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci USA. 1997;94:3336–40. doi: 10.1073/pnas.94.7.3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, Dubois RN. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology. 1994;107:1183–88. doi: 10.1016/0016-5085(94)90246-1. [DOI] [PubMed] [Google Scholar]
- 46.Sano H, Kawahito Y, Wilder RL, Hashiramoto A, Mukai S, Asai K, et al. Expression of cyclooxygenase-1 and -2 in human colorectal cancer. Cancer Rres. 1995;55:3785–89. [PubMed] [Google Scholar]
- 47.Cappell MS. From colonic polyps to colon cancer: pathophysiology, clinical presentation, and diagnosis. Clin Lab Med. 2005;25:135–77. doi: 10.1016/j.cll.2004.12.010. [DOI] [PubMed] [Google Scholar]
- 48.Schreiber S, Nikolaus S, Hampe H. Activation of nuclear factor-κB in inflammatory bowel disases. Gut. 1998;42:477–84. doi: 10.1136/gut.42.4.477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Neurath M, Petterson S, Meyer-Zum-Buschenfelde KH, Strober W. Local administration of antisense phosphotihioate oligonucleotides to the p65 subunit of NF-κB abrogates established experimental colitis in mice. Nat Med. 1996;2:998–1004. doi: 10.1038/nm0996-998. [DOI] [PubMed] [Google Scholar]
- 50.Karin M, Cao Y, Greten FR, Li ZW. NF-kB in cancer: from innocent bystander to major culprit. Nature Rev Cancer. 2002;2:301–10. doi: 10.1038/nrc780. [DOI] [PubMed] [Google Scholar]
- 51.Yu L-L, Yu H-G, Yu J-P, Luo H-S, Xu X-M, Li J-H. Nuclear factor-κB p65 (RelA) transcription factor is constitutively activated in human colorectal carcinoma tissue. World J Gastroenterol. 2004;10:3255–60. doi: 10.3748/wjg.v10.i22.3255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sudoyo AW. Karakteristik Molekular Kanker Kolorektal pada Usia Muda: Ekspresi MLH1 dan MSH2 sebagai Indikator Instabilitas Mikrosatelit, in Departemen Ilmu Penyakit Dalam; Jakarta: Universitas Indonesia; 2004. [Google Scholar]
- 53.Xiong Z, Wu AH, Bender CM, Tsao J-L, Blake C, Shibata D, et al. Mismatch repair deficiency and CpG island hypermethylation in sporadic colon adenocarcinomas. Cancer Epidemiol Biomarkers Prev. 2001;10:799–803. [PubMed] [Google Scholar]
- 54.Abdullah M, Utomo AR, Handjari DR, Sjam AF, Fauzi A, Sastranegara F, et al. Asian Oncology Summit; Bali, Indonesia: 2010. Apr 9-11, [Google Scholar]
- 55.Abdullah M, Sudoyo AW, Pranowo BS, Rini D, Sutrisna B, Rani AA. Expression of NF-κB and COX-2 in young versus older patients with sporadic colorectal cancer. Acta Med Indones – Indones. J Intern Med. 2009;41:70–74. [PubMed] [Google Scholar]
- 56.Hao X, Bishop AE, Wallace M, Wang H, Willcocks TC, Maclouf J, et al. Early expression of cyclo-oxygenase-2 during sporadic colorectal carcinogenesis. J Pathol. 1999;187:295–301. doi: 10.1002/(SICI)1096-9896(199902)187:3<295::AID-PATH254>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- 57.Abdullah M. Jalur inflamasi pada karsinogenesis kolorektal sporadic di Indonesia: Peran NF-κB dan COX-2 serta hubungannya dengan karakteristik klinikopatologis; Jakarta, Indonesia: Fakultas Pascasarjana Universitas; 2009. [Disertasi] [Google Scholar]
- 58.Charalambous MP, Maihöfner C, Bhambra U, Lightfoot T, Gooderham NJ. Upregulation of cyclooxygenase-2 is accompanied by increased expression of nuclear factor-κB and IκB kinase-α in human colorectal cancer epithelial cells. Br J Cancer. 2003;88:1598–604. doi: 10.1038/sj.bjc.6600927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Appleby SB, Ristimaki A, Neilson K, Narko K, Hla T. Structure of the human cyclooxygenase-2 gene. Biochem J. 1994;302:723–27. doi: 10.1042/bj3020723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Engle SJ, Hoying JB, Boivin GP, Ormsby I, Gartside PS, Doetschman T. Transforming growth factor beta-1 suppresses non-metastatic colon cancer at an early stage of tumorigenesis. Cancer Res. 1999;59:3379–86. [PubMed] [Google Scholar]
- 61.Spring KJ, Zhao ZZ, Karamatic R, Walsh MD, Whitehall VL, Pike T, et al. High prevalence of sessile serrated adenomas with BRAF mutations: a prospective study of patients undergoing colonoscopy. Gastroenterology. 2006;131:1400–407. doi: 10.1053/j.gastro.2006.08.038. [DOI] [PubMed] [Google Scholar]
- 62.Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the proximal colon. Science. 1993;260:816–19. doi: 10.1126/science.8484122. [DOI] [PubMed] [Google Scholar]