Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309 Published online: July 18, 2014

© 2014 S. Karger AG, Basel 2296–3774/14/0013–0146$39.50/0 www.karger.com/gat

Mini-Review

Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance Zhen-Hua Wang

Jing-Yuan Fang

Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Institute of Digestive Disease; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; State Key Laboratory of Oncogene and Related Genes, Shanghai, China

Key Words Colorectal cancer · Crohn’s disease · Dysplasia-associated lesion or mass · Surveillance · Ulcerative colitis Abstract Background: Inflammatory bowel disease (IBD; including ulcerative colitis and Crohn’s disease) is associated with an increased risk for colorectal cancer (CRC). Chronic mucosal inflammation is a key factor in the onset of carcinogenesis in IBD patients. Although most gene alterations that cause sporadic CRCs also occur in patients with IBD-associated CRC, some gene sequences and mutation frequencies differ between sporadic CRCs and IBD-associated CRCs. Summary: This review explores the incidence of CRC in IBD patients, with the goal of identifying the risk and protective factors for CRC in order to facilitate dysplasia management via individualized surveillance strategies. Key Message: The incidence of CRC is higher among IBD patients. Identifying the risk and protective factors for CRC will facilitate dysplasia management via individualized surveillance strategies. Practical Implications: Several risk factors, including active inflammation, the coexistence of primary sclerosing cholangitis, a family history of sporadic CRC and the extent and duration of colonic disease, can lead to the development of CRC in patients with IBD. These risk factors should be utilized in individualized surveillance strategies to lower CRC incidence among IBD patients. Use of 5-aminosalicylic acid may play an important role in CRC prevention. Until newer, more reliable markers of IBDassociated CRC risk are found, dysplasia will continue to be the best marker of CRC risk in IBD. Dysplasia management continues to play a key role in preventing the progression of carcino© 2014 S. Karger AG, Basel genesis. Jing-Yuan Fang Renji Hospital, School of Medicine, Shanghai Jiaotong University 145 Shandong Rd Middle, Shanghai 200001 (China) E-Mail jingyuanfang2007 @ 126.com

147

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

Introduction

Patients with inflammatory bowel disease (IBD), i.e. ulcerative colitis (UC) or Crohn’s disease (CD), have an increased risk of developing colorectal cancer (CRC). Although most gene alterations that cause sporadic CRCs also occur in patients with IBD-associated CRC, the involved gene sequences and mutation frequencies differ between sporadic CRCs and IBDassociated CRCs. Chronic mucosal inflammation is a key factor in the CRC carcinogenesis in IBD patients, and hence 5-aminosalicylic acid (5-ASA), which is anti-inflammatory, acts as a chemopreventive agent for CRC in patients with IBD. Colonoscopy surveillance to detect dysplasia has been shown to decrease the incidence of CRC. This paper aims to explore the risk and protective factors for CRC development in IBD patients in order to facilitate the formulation of individualized CRC surveillance strategies and the proper management of dysplasia in this patient population. Epidemiology of IBD-Associated CRC

CRC Risk in UC The most recent meta-analysis of CRC, which was published in 2014, found that the overall risk of CRC in 181,923 UC patients was 1.69/1,000/year; this risk was 0.91/1,000/ year in the first decade after the diagnosis of UC, 4.07/1,000/year in the second decade and 4.55/1,000/year in the third decade [1]. These rates are lower than those reported by a metaanalysis published in 2001 [2]: 3/1,000/year overall and 2, 8 and 18% in the first, second and third decades after diagnosis, respectively. This decrease in risk may be attributable to the widespread use of aminosalicylates, which are believed to have a chemoprotective effect in IBD, to the more liberal and early use of colectomy for medically refractory UC and, possibly, to surveillance colonoscopy. However, the current CRC incidence rates among IBD patients remain considerably higher than those in the general population. The CRC incidence among the general population has been reported to range from 0.4/1,000/year in Australia, New Zealand, the USA and Western Europe to 0.1/1,000/year in Africa [3]. CRC Risk in CD The global incidence rate of CRC among patients with CD is 2.5%, which increases to 5.6% among patients with CD of the colon; the relative risk for CRC is 3.2 among patients with ileocolitis, and there is no increase in CRC risk (relative risk = 1) in CD patients with small bowel involvement only [4]. This increased risk of CRC among CD patients has been confirmed by a meta-analysis of 12 hospital- and population-based studies [5]. This meta-analysis found a relative risk of 4.5 (1.3–14.9) among CD patients with colonic disease, whereas the risk in CD patients with ileal disease only did not differ from the risk in the general population, with a relative risk of 1.1 (0.8–1.5) [5]. Regardless of CD distribution, the cumulative risk of CRC among CD patients was 2.9% 10 years after the diagnosis of CD, 5.6% after 20 years and 8.3% after 30 years [5]. Risk Factors for IBD-Associated CRC

The factors that have been found to increase the risk of CRC incidence among IBD patients are listed in table 1.

148

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

Table 1. Risk and protective factors for IBD-associated CRC

Positive risk factors

Negative risk factors and protective agents

Active inflammation [6] PSC [7 – 9] Family history of CRC [10, 11] Increased extent of disease [12] Prolonged disease duration [13]

Folic acid use [18] UDCA use (in patients with PSC) [22] 5-ASA treatment [14 – 16] Colectomy [24] Compliance with CRC surveillance guidelines

Active Inflammation Active inflammation was recently confirmed in clinical studies to play a key role in CRC carcinogenesis among IBD patients. A history of severe inflammatory conditions, such as pseudopolyps, a shortened or tubular colon and stricture formation, significantly increase the risk of CRC among IBD patients [6]. Primary Sclerosing Cholangitis Many clinical studies have demonstrated that primary sclerosing cholangitis (PSC) is associated with an increased risk of CRC in IBD [7]. For example, a case-control study performed by Broomé et al. [8] found a cumulative CRC risk of 9% after 10 years of disease progression, 21% after 20 years and 50% after 25 years among patients presenting with IBD complicated by PSC. The corresponding CRC risk among patients with UC only was 2, 5 and 10%, respectively [8]. Moreover, this increased risk persisted even after liver transplantation [9]. Family History of CRC A family history of sporadic CRC is also a risk factor for cancer. IBD patients with a firstdegree relative with CRC have twice the risk of developing CRC than IBD patients without a family history of CRC [10]. Moreover, among IBD patients who have a first-degree relative in whom CRC was diagnosed before the age of 50 years, the risk of IBD progression to CRC increases ninefold [11]. However, the presence of a first-degree relative with IBD does not increase the risk of CRC among healthy family members [11]. Extent and Duration of IBD The extent of colonic inflammation is a risk factor for CRC. UC patients with extensive disease (pancolitis and left-sided colitis) have a greater risk of developing CRC than do UC patients with only proctitis or proctosigmoiditis [12]. IBD duration is one of the most important risk factors for CRC among IBD patients. CRC risk is significantly increased after 8 years of IBD and continues to increase in subsequent years [13]. Factors Protecting against IBD-Associated CRC

5-ASA 5-ASA is the most commonly used drug for CRC prophylaxis among IBD patients. In 2005, Velayos et al. [14] performed a meta-analysis involving a total of 1,932 subjects, including 334 patients with CRC and 140 patients with dysplasia. Their study confirmed that the use of 5-ASA showed a protective association with CRC (odds ratio: 0.51; 95% confidence interval: 0.37–0.69) or a combined endpoint of CRC/dysplasia (odds ratio: 0.51; 95% confidence

149

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

interval: 0.38–0.69). Recently, two large population-based studies [15, 16] showed the following: (i) regular 5-ASA users had a lower risk for CRC than non-regular users, (ii) no significant prophylactic effect was found among sulfasalazine users, and (iii) among 5-ASA users, the protective effect was only significant among subjects who had been taking 5-ASA for more than 13 years. Folic Acid Although epidemiologic studies have demonstrated that folate deficiency may be a risk factor for CRC [17], the protective effect of folate supplementation against CRC among IBD patients remains controversial. Thus far, only one small-scale case-control study has found a significant protective effect of folate supplementation [18]. No statistically significant effect of folate supplementation was observed in other case-control studies [19–21]. Therefore, folate supplementation, though essential to correct folate deficiency, cannot be considered to be an effective prophylactic therapy for decreasing CRC risk among IBD patients. Ursodeoxycholic Acid Ursodeoxycholic acid (UDCA) has been suggested to play a protective role in UC complicated by PSC. Among PSC patients, those taking high doses of UDCA (13–15 mg/kg/day) had a more than 4-year increase in survival [22]. Despite the significant decrease in mortality among UDCA-treated PSC patients, UDCA did not decrease the risk of CRC or dysplasia in these patients [23]. Therefore, whether UDCA protects IBD patients with or without concomitant PSC from progression to CRC remains unknown. Total Colectomy As mentioned above, two meta-analyses [1, 2] from different time periods suggest that the incidence of IBD-associated CRC is in decline. A cohort study conducted in Denmark between 1962 and 1987 reported only 13 cases of CRC among 1,160 UC patients [24]. The annual risk was 0.06%. The 30-year cumulative CRC risk was 2.1%. The rate of surgery for UC in Denmark is among the highest reported worldwide, and this is a plausible explanation for the very low risk of CRC in this population. Pathophysiology

Sporadic CRC and IBD-associated CRC develop through different molecular carcinogenesis pathways. Sporadic CRC is the result of two important types of genomic instability: chromosomal instability (85%) and microsatellite instability (15%) [25]. Both these mechanisms operate in IBD-associated CRC, but at different times and with different frequencies [25]. APC gene function loss is typically an early event in sporadic CRCs; however, in IBDassociated CRCs, APC dysfunction is seldom seen, and if present, usually occurs very late [26– 28]. In contrast, the loss or mutation of p53 chromosomes often occurs in the early stages of IBD-associated CRC, but is believed to be a late event in the carcinogenesis of sporadic CRC. In IBD-associated CRCs, microsatellite instability affects carcinogenesis. In contrast, in sporadic CRCs, methylation of the hMLH1 promoter leading to transcriptional silencing of hMLH1 is the most frequent event; hMLH1 promoter methylation is unusual in IBD-associated CRCs [29]. Recently, an increasing body of evidence has shown that chronic inflammation plays an important role in CRC carcinogenesis by producing a suitable microenvironment for CRC formation and progression. Proinflammatory cytokines secreted by infiltrating inflammatory and immune cells, prostaglandins induced by cyclooxygenase in epithelial cells and enhanced

150

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

reactive oxygen and nitrogen species alter many molecules, including DNA, RNA, proteins and lipids. Moreover, the formation of DNA adducts is induced, which causes point mutations in genes and in CpG islands involved in DNA methylation. Finally, local cytokines and prostaglandins inhibit apoptosis and promote cell proliferation [30]. Surveillance Colonoscopy

To determine whether surveillance colonoscopy is effective in IBD patients, we must verify whether this strategy reduces CRC-related mortality. As yet, no randomized controlled trial has investigated the effectiveness of surveillance colonoscopy. Numerous case series [31–35] and three case-control studies [36–38] have demonstrated a benefit of surveillance colonoscopy in IBD patients. However, the Cochrane Group conducted a pooled analysis of the published studies on UC patients and concluded that there was no clear evidence that surveillance colonoscopy decreases mortality in patients with extensive UC [39]. They did find, however, that CRC tended to be detected at an earlier stage and had a better prognosis in patients who underwent surveillance than in those who did not [39]. Thus, there is indirect evidence that colonoscopy surveillance is effective at reducing the mortality of IBD-associated CRC. Therefore, patients should be advised to undergo surveillance colonoscopy, which enables the detection of dysplasia or early-stage CRC. Our current knowledge of the efficacy of colonoscopy surveillance is based on random biopsies of colorectal mucosa. Careful inspection of the mucosa along with a sufficient number of biopsy specimens from all anatomic segments of the colon is required. To detect dysplasia and/or cancer, rigorous colonoscopy surveillance, including the acquisition of at least 33 random biopsy specimens from all portions of the colon in patients with pancolitis, must be performed [40]. Since dysplasia and cancer are more common in the left colon, it is recommended that more extensive sampling be performed in the left colon, particularly in the rectum. Biopsy specimens should be obtained separately from areas of flat mucosa surrounding the bases of adenoma-like and non-adenoma-like dysplasia-associated lesions or masses (DALMs). Chromoendoscopy or other image-enhancing techniques are recommended for physicians with experience with these techniques. With the use of enhanced endoscopic techniques, targeted biopsies may be performed as an alternative to obtaining random biopsy specimens. Poor patient compliance with surveillance programs reduces the effectiveness of surveillance. The 2004 American College of Gastroenterology guidelines [41] recommend that after 8–10 years of colitis, surveillance colonoscopy should be performed once every year or every 2 years with multiple biopsies taken at regular intervals in patients with left-sided colitis or pancolitis. Patients with proctitis and proctosigmoiditis are not considered to be at an increased risk for cancer, and thus are recommended to undergo average-risk surveillance [41]. Dysplasia Management

Despite having some limitations as a prognostic marker, dysplasia is still considered the best marker of CRC risk in IBD. CRC carcinogenesis in IBD follows a sequential progression from inflammation to low-grade dysplasia (LGD) and then high-grade dysplasia (HGD). Although this model is conceptually useful and serves as a reasonable paradigm, it is by no means absolute. CRC can occur in patients without a history of dysplasia [42]. In addition, not all patients with LGD will progress to CRC by way of HGD [43]. Nevertheless, until newer,

151

Gastrointest Tumors 2014;1:146–154 © 2014 S. Karger AG, Basel www.karger.com/gat

DOI: 10.1159/000365309

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

DALM

Adenoma-like

Non-adenoma-like

Colectomy Located outside area of colitis

Polypectomy Regular surveillance

Located within area of colitis

Complete excision possible No dysplasia within lesion No dysplasia elsewhere in colon

Complete excision not possible Dysplasia within lesion Dysplasia elsewhere in colon

Polypectomy Intensive surveillance

Colectomy

Fig. 1. Algorithm for the treatment of DALMs in IBD patients.

more reliable markers of IBD-associated CRC risk are confirmed, clinical practice relies mainly on the endoscopic and pathologic identification of dysplasia. Raised Dysplastic Lesions Raised, endoscopically visible, dysplastic lesions in IBD have been referred to by the acronym ‘DALM’. Recent studies suggest that IBD-related DALMs may be broadly separated into those that appear similar to sporadic adenomas in non-IBD patients, referred to as adenoma-like DALMs, and those that do not resemble adenoma-like DALMs, referred to as non-adenoma-like DALMs [44–46]. Adenoma-like DALMs are well-circumscribed, smooth, papillary, non-necrotic, sessile or pedunculated polyps that are similar to sporadic adenomas; they are usually readily accessible and can be removed via routine endoscopic methods [44]. Other synonyms used to describe these lesions include adenoma-like polyps, adenoma-like dysplastic polyps, polypoid dysplasia and adenoma-like masses. Non-adenoma-like DALMs contain velvety patches, plaques, irregular bumps and nodules, wart-like thickenings, stricturing lesions and broadbased masses [47, 48]. Non-adenoma-like DALMs represent a heterogeneous group of dysplastic lesions that have a strong positive association with CRC. There is a high possibility of CRC in patients with endoscopically unresectable non-adenoma-like DALMs, ranging from 38 to 83% [33, 49, 50]. Therefore, it is recommended that IBD patients with an endoscopically unresectable, non-adenoma-like DALM, regardless of the dysplasia grade, undergo colectomy [51]. In contrast, adenoma-like DALMs located outside or proximal to areas of known colitis may be considered to be sporadic in origin and thus treated conservatively with polypectomy and continued active surveillance [52]. Similarly, adenoma-like DALMs located within areas of known colitis may also be treated conservatively with polypectomy and continued surveil-

152

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

lance if the lesion has been excised completely, there is no dysplasia at the margins of the specimen and there is no evidence of flat dysplasia elsewhere in the colon, either adjacent to or distant from the polypoid lesion [45, 51]. These recommendations apply to patients with UC, regardless of age and duration/extent of colitis [45, 51, 52]. A treatment algorithm for UC patients with adenoma-like or a non-adenoma-like DALMs is proposed in figure 1 [52]. Flat Dysplasia Flat dysplasia refers to lesions that are not raised, minimally raised or occasionally invisible. Concomitant CRC may be present in 42–67% of patients with flat HGD. If immediate colectomy is not conducted when HGD is first detected, CRC will develop in 25–32% of patients on long-term follow-up [53]. Therefore, although the evidence is considered to be of fair quality due to its retrospective character, colectomy is recommended for all patients with flat HGD. The management of patients with LGD is more controversial. Like patients with flat HGD, patients with flat LGD may have concomitant CRC; however, the rate is lower, though in general still considerable. The prevalence rate of concomitant CRC is 19–27% among patients who have undergone colectomy within a few months of a colonoscopy that showed LGD as the most advanced histologic abnormality [53]. One recent meta-analysis revealed that the positive predictive value of LGD for progression to HGD and/or CRC is approximately 18% [54]. Thus, the decision to undergo colectomy versus continued surveillance in the setting of flat LGD should be individualized and discussed at length among the patient, gastroenterologist and colorectal surgeon. Conclusions

UC and CD are associated with an increased risk of CRC. The risk factors for CRC among IBD patients include severe inflammation, coexisting PSC, family history of sporadic CRC, large extent of colonic involvement and long duration of disease. Tumorigenesis within the colon depends on unique environmental, genetic and immunologic factors in IBD patients. The chemoprevention of CRC remains an important goal, and retrospective data suggest that 5-ASA plays an important role in this context. Colonoscopy surveillance programs, with multiple biopsies performed every 1–2 years, are key components in the early detection of CRC in IBD patients. Newer methods such as chromoendoscopy are currently being investigated as complementary techniques to enhance the early detection of dysplasia and cancer in this high-risk population.

References  1  2  3  4  5  6  7

Castaño-Milla C, Chaparro M, Gisbert JP: Systematic review with meta-analysis: the declining risk of colorectal cancer in ulcerative colitis. Aliment Pharmacol Ther 2014;39:645–659. Eaden JA, Abrams KR, Mayberry JF: The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526–535. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, et al: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010;127:2893–2917. Ekbom A, Helmick C, Zack M, Adami HO: Increased risk of large-bowel cancer in Crohn’s disease with colonic involvement. Lancet 1990;336:357–359. Canavan C, Abrams KR, Mayberry J: Meta-analysis: colorectal and small bowel cancer risk in patients with Crohn’s disease. Aliment Pharmacol Ther 2006;23:1097–1104. Rutter MD, Saunders BP, Wilkinson KH, Rumbles S, Schofield G, et al: Cancer surveillance in longstanding ulcerative colitis: endoscopic appearances help predict cancer risk. Gut 2004;53:1813–1816. Smith MP, Loe RH: Sclerosing cholangitis; review of recent case reports and associated diseases and four new cases. Am J Surg 1965;110:239–246.

153

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

 8  9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38

Broomé U, Löfberg R, Veress B, Eriksson LS: Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995;22:1404–1408. Loftus EV Jr, Aguilar HI, Sandborn WJ, Tremaine WJ, Krom RA, et al: Risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis following orthotopic liver transplantation. Hepatology 1998;27:685–690. Nuako KW, Ahlquist DA, Mahoney DW, Schaid DJ, Siems DM, et al: Familial predisposition for colorectal cancer in chronic ulcerative colitis: a case-control study. Gastroenterology 1998;115:1079–1083. Askling J, Dickman PW, Karlén P, Broström O, Lapidus A, et al: Family history as a risk factor for colorectal cancer in inflammatory bowel disease. Gastroenterology 2001;120:1356–1362. Ekbom A, Helmick C, Zack M, Adami HO: Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med 1990;323:1228–1233. Rutter MD, Saunders BP, Wilkinson KH, Rumbles S, Schofield G, et al: Thirty-year analysis of a colonoscopic surveillance program for neoplasia in ulcerative colitis. Gastroenterology 2006;130:1030–1038. Velayos FS, Terdiman JP, Walsh JM: Effect of 5-aminosalicylate use on colorectal cancer and dysplasia risk: a systematic review and metaanalysis of observational studies. Am J Gastroenterol 2005;100:1345–1353. Terdiman JP, Steinbuch M, Blumentals WA, Ullman TA, Rubin DT: 5-Aminosalicylic acid therapy and the risk of colorectal cancer among patients with inflammatory bowel disease. Inflamm Bowel Dis 2007;13:367–371. Velayos FS, Loftus EV Jr, Jess T, Harmsen WS, Bida J, et al: Predictive and protective factors associated with colorectal cancer in ulcerative colitis: a case-control study. Gastroenterology 2006;130:1941–1949. Giovannucci E, Stampfer MJ, Colditz GA, Hunter DJ, Fuchs C, et al: Multivitamin use, folate, and colon cancer in women in the Nurses’ Health Study. Ann Intern Med 1998;129:517–524. Lashner BA: Red blood cell folate is associated with the development of dysplasia and cancer in ulcerative colitis. J Cancer Res Clin Oncol 1993;119:549–554. Rutter M, Saunders B, Wilkinson K, Rumbles S, Schofield G, et al: Severity of inflammation is a risk factor for colorectal neoplasia in ulcerative colitis. Gastroenterology 2004;126:451–459. Lashner BA, Heidenreich PA, Su GL, Kane SV, Hanauer SB: Effect of folate supplementation on the incidence of dysplasia and cancer in chronic ulcerative colitis. A case-control study. Gastroenterology 1989;97:255–259. Lashner BA, Provencher KS, Seidner DL, Knesebeck A, Brzezinski A: The effect of folic acid supplementation on the risk for cancer or dysplasia in ulcerative colitis. Gastroenterology 1997;112:29–32. Harnois DM, Angulo P, Jorgensen RA, Larusso NF, Lindor KD: High-dose ursodeoxycholic acid as a therapy for patients with primary sclerosing cholangitis. Am J Gastroenterol 2001;96:1558–1562. Wolf JM, Rybicki LA, Lashner BA: The impact of ursodeoxycholic acid on cancer, dysplasia and mortality in ulcerative colitis patients with primary sclerosing cholangitis. Aliment Pharmacol Ther 2005;22:783–788. Winther KV, Jess T, Langholz E, Munkholm P, Binder V: Long-term risk of cancer in ulcerative colitis: a population-based cohort study from Copenhagen County. Clin Gastroenterol Hepatol 2004;2:1088–1095. Willenbucher RF, Aust DE, Chang CG, Zelman SJ, Ferrell LD, et al: Genomic instability is an early event during the progression pathway of ulcerative-colitis-related neoplasia. Am J Pathol 1999;154:1825–1830. Redston MS, Papadopoulos N, Caldas C, Kinzler KW, Kern SE: Common occurrence of APC and K-ras gene mutations in the spectrum of colitis-associated neoplasias. Gastroenterology 1995;108:383–392. Tarmin L, Yin J, Harpaz N, Kozam M, Noordzij J, et al: Adenomatous polyposis coli gene mutations in ulcerative colitis-associated dysplasias and cancers versus sporadic colon neoplasms. Cancer Res 1995;55:2035–2038. Aust DE, Terdiman JP, Willenbucher RF, Chang CG, Molinaro-Clark A, et al: The APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer 2002;94:1421–1427. Svrcek M, El-Bchiri J, Chalastanis A, Capel E, Dumont S, et al: Specific clinical and biological features characterize inflammatory bowel disease associated colorectal cancers showing microsatellite instability. J Clin Oncol 2007;25:4231–4238. 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;157:737–745. Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J: Colorectal cancer prevention in ulcerative colitis: a casecontrol study. Aliment Pharmacol Ther 2000;14:145–153. Löfberg R, Broström O, Karlén P, Tribukait B, Ost A: Colonoscopic surveillance in long-standing total ulcerative colitis – a 15-year follow-up study. Gastroenterology 1990;99:1021–1031. Rosenstock E, Farmer RG, Petras R, Sivak MV Jr, Rankin GB, Sullivan BH: Surveillance for colonic carcinoma in ulcerative colitis. Gastroenterology 1985;89:1342–1346. Nugent FW, Haggitt RC, Gilpin PA: Cancer surveillance in ulcerative colitis. Gastroenterology 1991;100:1241– 1248. Jonsson B, Ahsgren L, Andersson LO, Stenling R, Rutegård J: Colorectal cancer surveillance in patients with ulcerative colitis. Br J Surg 1994;81:689–691. Karlén P, Kornfeld D, Broström O, Löfberg R, Persson PG, et al: Is colonoscopic surveillance reducing colorectal cancer mortality in ulcerative colitis? A population based case control study. Gut 1998;42:711–714. Lashner BA, Turner BC, Bostwick DG, Frank PH, Hanauer SB: Dysplasia and cancer complicating strictures in ulcerative colitis. Dig Dis Sci 1990;35:349–352. Choi PM, Nugent FW, Schoetz DJ Jr, Silverman ML, Haggitt RC: Colonoscopic surveillance reduces mortality from colorectal cancer in ulcerative colitis. Gastroenterology 1993;105:418–424.

154

Gastrointest Tumors 2014;1:146–154 DOI: 10.1159/000365309

© 2014 S. Karger AG, Basel www.karger.com/gat

Wang and Fang: Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance

39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54

Mpofu C, Watson AJ, Rhodes JM: Strategies for detecting colon cancer and/or dysplasia in patients with inflammatory bowel disease. Cochrane Database Syst Rev 2004;2:CD000279. Rubin CE, Haggitt RC, Burmer GC, Brentnall TA, Stevens AC, et al: DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology 1992;103:1611–1620. Kornbluth A, Sachar DB; Practice Parameters Committee of the American College of Gastroenterology: Ulcerative colitis practice guidelines in adults (update): American College of Gastroenterology, Practice Parameters Committee. Am J Gastroenterol 2004;99:1371–1385. Woolrich AJ, DaSilva MD, Korelitz BI: Surveillance in the routine management of ulcerative colitis: the predictive value of low-grade dysplasia. Gastroenterology 1992;103:431–438. Ullman T: Hold the bone: good news for young women with IBD. Inflamm Bowel Dis 2003;9:396–397. Odze RD, Farraye FA, Hecht JL, Hornick JL: Long-term follow-up after polypectomy treatment for adenomalike dysplastic lesions in ulcerative colitis. Clin Gastroenterol Hepatol 2004;2:534–541. Rubin PH, Friedman S, Harpaz N, Goldstein E, Weiser J, et al: Colonoscopic polypectomy in chronic colitis: conservative management after endoscopic resection of dysplastic polyps. Gastroenterology 1999;117:1295– 1300. Engelsgjerd M, Farraye FA, Odze RD: Polypectomy may be adequate treatment for adenoma-like dysplastic lesions in chronic ulcerative colitis. Gastroenterology 1999;117:1288–1294; discussion 1488–1491. Rutter MD, Saunders BP, Wilkinson KH, Kamm MA, Williams CB, et al: Most dysplasia in ulcerative colitis is visible at colonoscopy. Gastrointest Endosc 2004;60:334–339. Lennard-Jones JE, Melville DM, Morson BC, Ritchie JK, Williams CB: Precancer and cancer in extensive ulcerative colitis: findings among 401 patients over 22 years. Gut 1990;31:800–806. Blackstone MO, Riddell RH, Rogers BH, Levin B: Dysplasia-associated lesion or mass (DALM) detected by colonoscopy in long-standing ulcerative colitis: an indication for colectomy. Gastroenterology 1981;80:366–374. Itzkowitz SH, Harpaz N: Diagnosis and management of dysplasia in patients with inflammatory bowel diseases. Gastroenterology 2004;126:1634–1648. Odze RD: Adenomas and adenoma-like DALMs in chronic ulcerative colitis: a clinical, pathological, and molecular review. Am J Gastroenterol 1999;94:1746–1750. Friedman S, Odze RD, Farraye FA: Management of neoplastic polyps in inflammatory bowel disease. Inflamm Bowel Dis 2003;9:260–266. Bernstein CN, Shanahan F, Weinstein WM: Are we telling patients the truth about surveillance colonoscopy in ulcerative colitis? Lancet 1994;343:71–74. Thomas T, Abrams KA, Robinson RJ, Mayberry JF: Meta-analysis: cancer risk of low-grade dysplasia in chronic ulcerative colitis. Aliment Pharmacol Ther 2007;25:657–668.

Colorectal Cancer in Inflammatory Bowel Disease: Epidemiology, Pathogenesis and Surveillance.

Inflammatory bowel disease (IBD; including ulcerative colitis and Crohn's disease) is associated with an increased risk for colorectal cancer (CRC). C...
NAN Sizes 0 Downloads 9 Views