inflammatory bowel disease and familial adenomatous polyposis || ibd: cancer risk and surveillance

5
Introduction Patients with inflammatory bowel disease (IBD) are at an increased risk for colorectal cancer (CRC) [14], but the quantisation of the risk of CRC in this specific population varies widely in different studies. However, CRC complicating ulcerative colitis (UC) and Crohn’s disease (CD) only accounts for 12% of all cases of CRC in the general population. Most cases of CRC are either sporadic (6585%) or familial (1030%; Fig. 1) [34]. CRC is considered a serious complication of the disease and accounts for approximately 15% of all deaths in IBD patients [5]. The mortality in patients diagnosed with CRC who are also diagnosed with IBD is higher than for sporadic CRC [6]. However, much more is known about the risk in UC. The inci- dence of colorectal cancer in patients with ulcerative colitis is higher than in the general population. In a meta-analysis, the overall prevalence of CRC in any UC patient, based on 116 studies, was estimated to be 3.7%. The incidence rate corresponded to cumulative probabilities of 2% by 10 years, 8% by 20 years and 18% by 30 years [7]. The major risk factors for the development of CRC include young age at onset of IBD [1], extensive dis- ease [8] and long disease duration [5]. It is biologi- cally plausible that the excess cancer risk is second- ary to chronic inflammation and it is recognised that duration of colitis is an important risk factor for CRC. Rutter et al. [9] showed for the first time that increasing severity of colonic inflammation is associ- ated with an increase in the rate of colorectal neopla- sia in UC. Patients with disease extending to the hepatic flexure or more proximally have the greatest risk of CRC [1012]. Most studies have found that the risk of CRC increases after 15 to 20 years; approxi- mately one decade later than in pancolitis in patients with colitis confined to the left colon [8]. Patients with ulcerative proctitis and proctosigmoiditis are probably not at increased risk for CRC [13]. An increased risk of CRC has been observed in patients with UC complicated by primary sclerosing cholangi- tis. In these cases, cancer was more likely to be in the right colon, suggesting a possible role of bile acids in oncogenesis [14]. The evidence for other potential risk factors is scarce. Smoke [15], folate depletion [1617] and a positive family history of colon cancer [18] may affect the occurrence of CRC. A hypothesis has been put forward according to which UC, CD and CRC occur in predisposed patients because of a mixture of genetic and environmental factors. One study has shown that relatives of patients with both IBD and CRC have an 80% increased risk of CRC [19]. In our study, there is no statistically significant difference between IBD and control cases of family members as far as prevalence of malignant colorectal, digestive extra-colonic or extra-digestive tract tumours [20]. Another recent study has investigated the prevalence of all malignancies in first-degree relatives of CD patients. The result showed a higher prevalence of breast cancer in female relatives, mainly in mothers, of CD patients compared with controls. The presence of breast cancer was not associated with any specific phenotype of the CD [21]. As in UC, Crohn’s disease of the colon carries an increased risk of CRC. Ekbom et al. [1] found a rela- tive risk of 5.6% for CRC among patients with CD, whereas in another Swedish patient cohort study, Persson et al. [22] did not find that the population relative risk increased. Similarly, Fireman et al. [23] from Israel and Jess et al. [24] in the Danish cohort, did not find an increased risk of CRC among their IBD: Cancer Risk and Surveillance Gabriele Riegler, Ilaria Esposito Fig. 1. Distribution of CRC

Upload: gian-gaetano

Post on 05-Dec-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Introduction

Patients with inflammatory bowel disease (IBD) areat an increased risk for colorectal cancer (CRC)[1–4], but the quantisation of the risk of CRC in thisspecific population varies widely in different studies.However, CRC complicating ulcerative colitis (UC)and Crohn’s disease (CD) only accounts for 1–2% ofall cases of CRC in the general population. Most casesof CRC are either sporadic (65–85%) or familial(10–30%; Fig. 1) [3–4].

CRC is considered a serious complication of thedisease and accounts for approximately 15% of alldeaths in IBD patients [5]. The mortality in patientsdiagnosed with CRC who are also diagnosed withIBD is higher than for sporadic CRC [6]. However,much more is known about the risk in UC. The inci-dence of colorectal cancer in patients with ulcerativecolitis is higher than in the general population. In ameta-analysis, the overall prevalence of CRC in anyUC patient, based on 116 studies, was estimated to be3.7%. The incidence rate corresponded to cumulativeprobabilities of 2% by 10 years, 8% by 20 years and18% by 30 years [7].

The major risk factors for the development of CRCinclude young age at onset of IBD [1], extensive dis-ease [8] and long disease duration [5]. It is biologi-cally plausible that the excess cancer risk is second-ary to chronic inflammation and it is recognised thatduration of colitis is an important risk factor forCRC. Rutter et al. [9] showed for the first time that

increasing severity of colonic inflammation is associ-ated with an increase in the rate of colorectal neopla-sia in UC. Patients with disease extending to thehepatic flexure or more proximally have the greatestrisk of CRC [10–12]. Most studies have found that therisk of CRC increases after 15 to 20 years; approxi-mately one decade later than in pancolitis in patientswith colitis confined to the left colon [8]. Patientswith ulcerative proctitis and proctosigmoiditis areprobably not at increased risk for CRC [13]. Anincreased risk of CRC has been observed in patientswith UC complicated by primary sclerosing cholangi-tis. In these cases, cancer was more likely to be in theright colon, suggesting a possible role of bile acids inoncogenesis [14].

The evidence for other potential risk factors isscarce. Smoke [15], folate depletion [16–17] and apositive family history of colon cancer [18] mayaffect the occurrence of CRC. A hypothesis has beenput forward according to which UC, CD and CRCoccur in predisposed patients because of a mixture ofgenetic and environmental factors. One study hasshown that relatives of patients with both IBD andCRC have an 80% increased risk of CRC [19]. In ourstudy, there is no statistically significant differencebetween IBD and control cases of family members asfar as prevalence of malignant colorectal, digestiveextra-colonic or extra-digestive tract tumours [20].Another recent study has investigated the prevalenceof all malignancies in first-degree relatives of CDpatients. The result showed a higher prevalence ofbreast cancer in female relatives, mainly in mothers,of CD patients compared with controls. The presenceof breast cancer was not associated with any specificphenotype of the CD [21].

As in UC, Crohn’s disease of the colon carries anincreased risk of CRC. Ekbom et al. [1] found a rela-tive risk of 5.6% for CRC among patients with CD,whereas in another Swedish patient cohort study,Persson et al. [22] did not find that the populationrelative risk increased. Similarly, Fireman et al. [23]from Israel and Jess et al. [24] in the Danish cohort,did not find an increased risk of CRC among their

IBD: Cancer Risk and Surveillance

Gabriele Riegler, Ilaria Esposito

Fig. 1. Distribution of CRC

46 G. Riegler, I. Esposito

patients with CD. Although analyzing subgroupswithin the CD study meant that there were few can-cers identified, when patients with CD of >10-yearduration were analyzed, the relative risk for CRC was4.8%. For those patients with at least 10 years of dis-ease and no surgery within the first 10 years, the rel-ative risk was 8.3%. Thus, even among this reported-ly negative study, when variables such as diseaseduration and possibly disease extent (based on nosurgery) are analysed, the relative risk increases [5].In the study of Gillen et al. [25], however, it appearsthat the malignant potential in CD and UC is of thesame order of magnitude. Cancer developed ininflamed areas in both diseases, and was thus locatedin the right colon in 49% of patients with CD com-pared with 36% with UC, reflecting the difference ininflammatory sites in the two cases. Adenocarcino-mas develop only in affected segments of the smallintestine, and are difficult to diagnose at an earlystage because the radiological appearance is similarto that of stricturing CD.

Pathogenesis of CRC in IBD

The pathogenesis of CRC in IBD is poorly under-stood. However, several lines of evidence suggest thatthe pathobiology is different than sporadic CRC:1. the mean age of developing CRC in the setting of

IBD is lower than sporadic CRC (40–50 vs. 60years).

2. Dysplasia in UC is preceded by a long history ofchronic inflammation and can be found at distantsites from the cancer. In contrast, dysplasia in spo-radic CRC is usually associated with a discretepolyp without inflammation.

3. Mutations in the ras protooncogene are present in40–60% of sporadic CRC and are probably an earlyevent; in contrast, these mutations are less fre-quently observed in cancer associated with UC,and are probably a late event [26–27].

4. Loss of heterozygosity for the p53 gene and srcactivation occur earlier in cancers associated withIBD than in sporadic CRC. Src activity in UC cor-relates with the degree of dysplasia [28].

5. Abnormalities of the p53 locus are absent in non-dysplastic mucosa of patients with sporadic CRC.In contrast, non-dysplastic mucosa in UC fre-quently has aneuploid DNA content and mayshow clones of cells with loss of heterozygosity ofthe p53 gene [29].It is generally accepted that CRC in IBD is preced-

ed by dysplasia, which is defined as unequivocal neo-plastic epithelium and is currently the most impor-tant and best-defined marker of an increased risk ofmalignancy. Dysplasia is present in >70% of UC

patients with carcinoma. Although it may occur inany portion of the colon, it typically parallels thelocation of cancer arising from chronically inflamedmucosa [30–32]. From an endoscopic viewpoint, dys-plasia is characterised as flat (endoscopically invisi-ble but detected in mucosal specimens) or raised(endoscopically visible), in which case “dysplasiaassociated lesions or mass” (DALM) is applied, aterm that was coined by Blackstone et al. in 1981 [33].In their study, 12 of 112 patients with long-standingUC were found to have a DALM, and of these, seven(58%) had carcinoma. Given the strong associationwith cancer, the presence of DALM constituted astrong indication for colectomy, which has becomethe standard therapy for this type of lesion. DALMare a heterogeneous population of tumours that mayendoscopically appear as a plaque, mass (irregular,broad-based or strictured lesions), a discrete sessilenodule, or polyp. The cancer risk is not equal amongthese various subtypes. Unfortunately, most of theprevious studies of UC associated DALMs failed toevaluate these lesions in relation to their grossappearance. For instance, there is one specific sub-type of discrete DALM that endoscopically and histo-logically resembles a sporadic adenoma (isolateddysplastic nodule or polyp) and, as such, poses a dif-ficult diagnostic challenge both to clinicians and topathologists [34]. This is a critically important dis-tinction, because an adenoma-like DALM is atumour that arises as a result of UC and, thus, is anindication for colectomy, whereas an adenoma whichis also by definition a “polypoid dysplastic lesion”,the development of which is unrelated to the under-lying chronic colitis (but coincidentally exists withit), is usually treated by polypectomy.

Since both UC and sporadic adenomas (SA) arenot uncommon disorders, it is not surprising thatgastroenterologists regularly encounter patients whohave both conditions. Sporadic adenomas coexistwith UC if they are located next (proximal) to thecolitis because UC-related dysplasia does not developfrom non-inflamed epithelium. UC associated ade-noma-like DALMs were classified as lesions that werelocated within histologically proven areas of colitisand were associated with either synchronous ormetachronous flat dysplasia or adenocarcinoma [35].

A number of clinical, histologic and molecular fea-tures have been studied to help make a distinctionbetween DALM and SA:1. patients with non-adenoma-like DALM are more

likely to be younger and have a longer duration ofdisease, more extensive disease, and larger lesions[36].

2. Lesions that appear endoscopically as adenomas(pedunculated or sessile) rather than having othercharacteristics (such as flat, ulcerated, or plaque-

IBD: Cancer Risk and Surveillance 47

like appearance), even if found within an area ofhistologic colitis, may have a favourable progno-sis with endoscopic removal and close follow-up[35, 37, 38].In addition to the clinical and histologic features

described above, several molecular markers havebeen proposed for distinguishing DALM from SA.Two of these are beta catenin and p53 [39–40]. Betacatenin is a cell membrane protein that accumulatesmore frequently in the nuclei of cells within sporadiccolon cancer as compared to DALM. Mutations withp53 (a tumour suppressor gene) occur more fre-quently in DALM than sporadic adenomas. Severalstudies have provided evidence that p53 polymor-phism at codon 72 (Arg and Pro alleles) may be asso-ciated with a high risk of malignancies. A recentstudy investigated Arg72Pro polymorphism in UCand found that p53 Pro homozygosis was more fre-quent in patients who had a continuous disease and,therefore, may also favour the progression from dys-plasia to colon cancer [41].

The predictive value of dysplasia has been studiedin UC. Although dysplasia is a marker for future orconcurrent malignancy, it can also regress or remainstable for long periods. Most patients with dysplasiado not have cancer and dysplasia is absent in thecolonic regions distant from the malignancy in25–30% of patients [42-43].

The association between dysplasia and CRC in CDappears to be similar to that in UC. Dysplasia is pres-ent in 83% of patients diagnosed with CRC, and dys-plasia distant from the cancer is found in 23–70%. Incontrast, dysplasia is much less common in colecto-my specimens of patients with CD than without CRC,and has occurred in only 2% of the specimens in oneseries [44–45].

Interpretation of biopsy samples may be con-founded by interobserver variation in the recogni-tion and grading of dysplasia. A uniform terminolo-gy for dysplasia in IBD has been proposed [29]. Theclassification categories histology as:1. negative.2. Indefinite (with subgroups of probably negative,

unknown, and probably positive.3. Positive (with subgroups of low grade and high-

grade dysplasia).Dysplasia may be difficult to distinguish from

inflammation and regeneration on histologic sec-tions. As a result, the presence of dysplasia should beconfirmed by an experienced pathologist. The crite-ria for dysplasia stress the uniform clonal nature ofdysplastic changes, which affects equally all parts ofthe crypt and surface epithelium. In contrast, regen-erative changes are usually most prominent at thebase of the crypts and show evidence of maturationas they migrate toward the crypt surface.

Other architectural and cytological abnormalitiesseen in regards to dysplastic epithelium include [29]:increased epithelium proliferation and mitoses;increased epithelial height; branching of crypts;back-to-back glandular formation; variation in thesize and shape of nuclei; increased nuclear/cytoplas-matic ratio; altered nuclear polarity globet cells.

Surveillance

Dysplastic epithelium may be a marker for coexistingmalignancy, and provides the rationale for surveil-lance. The optimal surveillance strategy remains con-troversial [45]. Surveillance colonoscopy in IBD isadvocated for early diagnosis of neoplasia, but isimperfect because some patients develop cancerdespite surveillance. In effect, a few reports haveshown conflicting results and these studies suggestthat surveillance leads to the detection of early-stagecancer in only a minority of patients and a significantnumber of patients develop cancer at an advancedstage despite surveillance.

The American Gastroenterological Association(AGA) recommends that colonoscopic surveillanceshould begin after 8 years in patients with pancolitisand 15 years in patients with colitis involving the leftcolon. Colonoscopy should be repeated every 1–2 years.

The American College of Gastroenterology (ACG)recommends annual surveillance colonoscopy begin-ning after 8–10 years of disease. Multiple biopsiesshould be obtained at regular intervals. The findingof definite dysplasia is an indication for colectomy.Patients whose biopsies are indefinite for dysplasiashould undergo repeat surveillance colonoscopy at ashorter interval.

The American Society for GastrointestinalEndoscopy (ASGE) recommends that patients withUC who have pancolitis should begin surveillancecolonoscopy after 8 years of disease. Four biopsiesshould be obtained every 10 cm from the cecum tothe rectum. In addiction, any suspicious lesions ormasses should be biopsied. Colonoscopy should berepeated every 1–3 years. The finding of carcinomaor high-grade dysplasia is an indication for colecto-my. Colectomy is also indicated for any degree ofdysplasia associated with lesion or mass. However, inpatients in whom colectomy is not feasible or is unac-ceptable, frequent surveillance, every 3–6 months, isconsidered an acceptable alternative. For patientswith left-sided colitis, the ASGE recommends thatsurveillance should begin after 15 years of disease.Surveillance is not indicated in ulcerative proctitis. InCD, the risk of colorectal cancer is increased only inregard to Crohn’s colitis. Surveillance colonoscopyand biopsy for dysplasia should be offered to patients

48 G. Riegler, I. Esposito

with longstanding disease. Furthermore, a reductionin mortality due to surveillance has not yet beenestablished.

On the other hand, prophylactic colectomy is amethod to prevent the development of CRC in IBD.The development of the stapling of the ileal pouch-anal anastomosis with preservation of the anal tran-sitional zone is an important advancement in surgi-cal treatment, but remains controversial because ofconcerns about the potential risk of dysplasia andcancer. The risk factor for carcinoma is inflammationin the small intestinal and rectal mucosa. Pouchitis isthe most frequent late complication and clearly relat-ed to a worse outcome. The etiology of pouchitisremains unknown. Possible causes are fecal stasisresulting in bacterial overgrowth and infection [46],microbial imbalance [47], production of volatile fattyacids, ischemia [48], oxygen-free radical injury [49],nitric oxide [50] and deprivation of short chain fattyacids [51]. Penna et al. [52] reported a strong corre-lation between primary sclerosing cholangitis andpouchitis, suggesting a common link in their patho-genesis. Teixeira et al. [53] showed that pouchitis wasmore frequent in patients with extra-intestinal mani-festations. Acute pouchitis was more frequent thanchronic pouchitis described by others.

Conclusion

In conclusion, patients with UC who undergo an ilealanal pouch operation also require surveillance. Morerecent research has focused on the chemopreventionof CRC in IBD using aminosalicylate (5-ASA) prepa-rations. Several retrospective correlative studies havesuggested that the long-term use of 5-ASA in IBDpatients may significantly reduce the risk of develop-ment of CRC. Moody et al. [54] demonstrated thatpatients with UC who did not comply with 5-ASAtherapies were significantly more likely to developCRC than their counterparts. Eaden et al. [55] foundthat regular 5-ASA therapy reduces cancer risk by75% in IBD patients. Mesalazine at a dose of 1.2 g/dayor greater reduced CRC risk by 91% compared to notreatment and was also supportive when taken atlower doses. They concluded that the benefit of regu-lar consumption of 5-ASA was equal to frequent vis-its to a hospital physician.

References

1. Ekbom A, Helmick C, Zack M et al (1990) Increasedrisk of large bowel cancer in Crohn’s disease withcolonic involvement. Lancet 336:357–359

2. Langholz E, Munkholm P, Davidsen M et al (1992)

Colorectal cancer risk and mortality in patients withulcerative colitis. Gastroenterology 103:1444–1451

3. Choi PM, Zelig MP (1994) Similarity of colorectal can-cer in CD and UC. Gut 35:950–954

4. Gyde S, Prior P, Dew MJ et al (1982) Mortality in ulcer-ative colitis. Gastroenterology 83:36–43

5. Munkholm P, Langholz E, Davidsen M, Binder V(1993) Intestinal cancer risk and mortality in patientswith Crohn’s disease. Gastroenterology 105:1716–1723

6. Jess T, Winther KV, Munkholm P et al (2002) Mortali-ty and causes of death in Crohn’s disease: follow-up ofa population-based cohort in Copenhagen County,Denmark. Gastroenterology 122:1808–1814

7. Eaden JA, Abrams KR, Mayberry JF (2001) The risk ofcolorectal cancer in ulcerative colitis: a meta analysis.Gut 48:526–535

8. Ekbom A, Helmick C, Zack M, Ademi HO (1990)Ulcerative colitis and colorectal cancer. N Eng J Med323:1228–1233

9. Rutter M, Saunders B, Wilkinson K et al (2004) Sever-ity of inflammation is a risk factor for colorectal neo-plasia in ulcerative colitis. Gastroenterology126:451–459

10. Gyde SN, Prior P, Allan RN et al (1988) Colorectal can-cer in ulcerative colitis: a cohort study of primaryreferrals from three centers. Gut 29:206

11. Lennard-Jones JE (1985) Cancer risk in ulcerative coli-tis: surveillance or surgery. Br J Surg 72(Suppl):84

12. Collins RH, Feldman M, Fordtran JS (1987) Colon can-cer, dysplasia and surveillance in patients with ulcera-tive colitis: a critical review. N Engl J Med 316:1654

13. Levin B (1992) Inflammatory bowel disease and coloncancer. Cancer 70:1313

14. Marchesa P, Lashner BA, Lavery IC et al (1997) Therisk of cancer and dysplasia among ulcerative colitispatients with primary sclerosing cholangitis. Am JGastroenterol 92:1285

15. Pinczowski D, Ekbom A, Baron J, et al (1994) Risk fac-tors for colorectal cancer in patients with ulcertivecolitis: a case-control study. Gastroenterology 107:117–120

16. Lashner BA, Provencher KS, Seidner DL et al (1997)The effect of folic acid supplementation on the risk forcancer or dysplasia in ulcerative colitis. Gastroenterol-ogy 112:29–32

17. Lashner BA, Heindereich PA, Su GL et al (1989) Effectof folate supplementation on the incidence of dyspla-sia and cancer in chronic ulcerative colitis. Gastroen-terology 97:255–259

18. Nuako KW, Ahlquist DA, Mahoney DW et al (1998)Familial predisposition for colorectal cancer in chron-ic ulcerative colitis: a case-control study. Gastroen-terology 115:1079–1083

19. Askling J, Dickman PW, Karlen P et al (2001) Colorec-tal cancer rates among first-degree relatives of patientswith inflammatory bowel disease: a population-basedstudy. Lancet 357:262–266

20. Riegler G, Carratù R, Tartaglione MT et al (1988)Prevalence and relative risk of malignancy in relativesof inflammatory bowel disease patients an controlsubjects. J Clin Gastroenterol 27(3):211–214

21. Riegler G, Caserta L, Castiglione F et al (2006)Increased risk of breast cancer in first-degree relativesof Crohn's disease patients: an IG-IBD study. Dig Liv

IBD: Cancer Risk and Surveillance 49

Dis 38(1):18–2322. Persson PG, Karlen P, Bernell O et al (1994) Crohn’s

disease and cancer: a population-based cohort study.Gastroenterology 107:1675–1679

23. Fireman Z, Grossman A, Lilos P et al (1989) Intestinalcancer in patients with Cron’s disease: a populationstudy in cental Israel. Scand J Gastroenterol24:346–350

24. Jess T, Winther K, Munkholm P et al (2000) Is there anincreased risk of intestinal and extra intestinal cancerin patients with Crohn’s disease? A population basedcohort followed from 1962 to 1997. Gastroenterology118(Suppl 2):1472

25. Gillen CD, Walmsley RS, Prior R et al (1994) Ulcerativecolitis and Crohn’s disease: a comparison of the col-orectal cancer risk in extensive colitis. Gut 35:651–5

26. Vogelstein B, Fearon ER, Hamilton SR et al (1988)Genetic alteration during colorectal tumour develop-ment. N Engl J Med 319:525

27. Itzkowitz SH, Greenwald B, Meltzer SJ (1995) Coloncarcinogenesis in inflammatory bowel disease.Inflamm Bowel Dis 1:142

28. Burner GC, Rabinovitch PS, Haggit RC et al (1992)Neoplastic progression in ulcerative colitis: histology,DNA content, and loss of a p53 allele. Gastroenterolo-gy 103:1602

29. Riddell RH, Golman H, Ramsohoff DF et al (1983)Dysplasia in inflammatory bowel disease: standard-ized classification with provisional clinical applica-tion. Hum Pathol 14:931–968

30. Willenbucher RF (1996) Inflammatory bowel disease.Semin Gastrointest Dis 7:94–104

31. Lennard-Jones JE, Nelville DM, Morson BC et al (1990)Precancer and cancer in extensive ulcerative colitis:findings among 401 patients over 22 years. Gut31:800–806

32. Goldman H (1996) Significance and detection of dys-plasia in chronic colitis. Cancer 78:2261–2263

33. Blackstone MO, Riddell RH, Roger BHG et al (1982)Dysplasia-associated lesion or mass (DALM) detectedby colonscopy in long-standing ulcerative colitis: anindication for colectomy. Gastroenterology 80:366–374

34. Torres C, Antonioli D, Odze RD et al (1998) Polypoiddysplasia and adenomas in inflammatory bowel dis-ease. Am J Surg Pathol 22:275–284

35. Odze RD (1999) Adenomas and adenomas-likeDALMs in chronic ulcerative colitis: a clinical, patho-logical and molecular review. Am J Gastroenterol94:1746–1750

36. Suzuki K, Muto T, Shinozaki M et al (1998) Differen-tial diagnoses of dysplasia associated lesion or massand coincidental adenoma in ulcerative colitis. DisColon Rectum 41:322

37. Engelsgjerd M, Farraye FA, Odze RD (1999) Polypec-tomy may be adequate treatment for adenoma-likedysplastic lesions in chronic ulcerative colitis. Gas-troenterology 117:1288

38. Rubin PH, Friedman S, Harpaz N et al (1999)Colonoscopy polypectomy in chronic colitis: conser-vative management after endoscopic resection of dys-plastic polyps. Gastroenterology 117:1295

39. Morin PJ, Sparks AB, Korinek V et al (1997) Activation

of B-catenin-Tcf signaling in colon cancer by muta-tions in B-catenin or APC. Science 275:1787

40. Munemitsu S, Albert I, Souza B et al (1995) Regulationof intracellular beta-catenin levels by adenomatouspolyposis coli (APC) tumour suppressor protein. ProcNatl Acad Sci USA 92:3046

41. Vietri MT, Riegler G, Pappalardo D et al (2006) TheArg72Pro p53 polymorphism influences the clinicaloutcome of ulcerative colitis. Lab Invest (In press)

42. Ramsohoff DF, Riddell RH, Levin B (1985) Ulcerativecolitis and colonic cancer: problems in assessing thediagnostic usefulness of mucosal dysplasia. Dis ColonRectum 28:383

43. Taylor BA, Pemberton JH, Carpenter HA et al (1992)Dysplasia in chronic ulcerative colitis: implications forcolonoscopic surveillance. Dis Colon Rectum 35:950

44. Warren R, Barwick KW (1983) Crohn’s colitis withadenocarcinoma and dysplasia: report of a case andreview of 100 small and large bowel resection forCrohn’s disease to detect incidence of dysplasia. Am JSurg Pathol 7:151

45. Provenzale D, Wong JB, Onken JE, Lipscomb J (1998)Performing a cost effectiveness analysis: surveillanceof patients with ulcerative colitis. Am J Gastroenterol93:872

46. Fonkalsrud EW, Phillips JD (1990) Reconstruction ofmalfunctioning ileoanal pouch procedures as an alter-native to permanent ileostomy. Am J Surg 160:245–251

47. Ruseler-Van Embden JGH, Schouten WR (1994) Pou-chitis: result of microbial imbalance? Gut 35:658–664

48. Chaussade S, Denizot Y, Valleur P (1991) Presence ofPaf-acether in stool of patients with pouch ileoanalanastomosis and pouchitis. Gastroenterology100:1509–1514

49. Levin KE, Pemberton JH, Phillips SF (1992) Role ofoxygen free radicals in the etiology of pouchitis. DisColon Rectum 35:452–456

50. Kroesen AJ, John M, Schofelder G (1997) Expression ofinducible nitric oxide syntase (iNOS) in pouchitis afterileoanal pouch surgery for ulcerative colitis. Langen-becks Archiv fur Chirurgie 114:451–454

51. Clausen MR, Tvede M, Mortensen PB (1992) Short-chain fatty acids in pouch contents from patients withand without pouchitis after ileal pouch-anal anasto-mosis. Gastroenterology 103:1144–1153

52. Penna C, Dozois RR, Tremaine W (1996) Pouchitisafter ileal pouch anal anastomosis for chronic ulcera-tive colitis occur with increased frequency in patientswith associated primary sclerosing cholangitis. Gut38:234–239

53. Teixeira WGJ, Silva JH, Texeira MG (1999) Pouchitis:extracolonic manifestation of ulcerative colitis? RevHosp Clin Fac Med S Paulo 54:155–158

54. Moody GA, Javanthi V, Probert CS et al (1996) Long-term therapy with sulphasalazine protects against col-orectal cancer in ulcerative colitis: a retrospectivestudy of colorectal cancer risk and compliance withtreatment in Leicestershire. Eur J Gastroenterol Hepa-tol 8:1179–1183

55. Eaden J, Abrams K, Ekbom A et al (2000) Colorectalcancer prevention in ulcerative colitis: a case-controlstudy. Aliment Pharmacol Ther 14:145–153