You are on page 1of 132

Colorectal carcinogenesis in patients with primary sclerosing cholangitis and inflammatory bowel disease

M.M.H. Claessen

proefschrift Marian.indd 1

10-10-2010 20:55:46

MMH Claessen was kindly supported by an unrestricted grant from Janssen-Cilag, Tilburg, the Netherlands. Financial support for the publication of this theses was kindly provided by:

Copyright Marian Claessen ISBN: 9789461081049 Lay out: www.wenzid.nl Cover: www.grootzus.nl Printed by: Gildeprint Dukkerijen BV

proefschrift Marian.indd 2

10-10-2010 20:55:46

Colorectal carcinogenesis in patients with primary sclerosing cholangitis and inflammatory bowel disease

Colorectale carcinogenese in patinten met primaire scleroserende cholangitis en inflammatoire darmziekten


(met een samenvatting in het Nederlands)

Proefschrift

ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag van de rector magnificus, prof.dr. J.C. Stoof, ingevolge het besluit van het college voor promoties in het openbaar te verdedigen op dinsdag 23 november 2010 des middags te 4.15 uur

door

Marian Maria Hubertina Claessen

geboren op 3 november 1981 te Heumen

proefschrift Marian.indd 3

10-10-2010 20:55:46

Promotoren: Prof.dr. P.D. Siersema Prof.dr. G.J.A. Offerhaus Co-promotor: Dr. F.P. Vleggaar

proefschrift Marian.indd 4

10-10-2010 20:55:47

Contents
Chapter 1 Chapter 2 Chapter 3 Chapter 4 General Introduction IBD-related Carcinoma and Lymphoma High Lifetime Risk of Cancer in Primary Sclerosing Cholangitis More Right-sided IBD-associated Colorectal Cancer in Patients with Primary Sclerosing Cholangitis Microsatellite Instability in Patients with Primary Sclerosing Cholangitis and Colitis-associated Colorectal Cancer Differences in Molecular Markers between Colitis-associated Colorectal Cancer with or without Primary Sclerosing Cholangitis and Sporadic Colorectal Cancer Methylation Profiles of Sporadic and Colitis-associated Colorectal Cancer Wnt-pathway Activation in IBD-associated Colorectal Carcinogenesis: Potential Biomarkers for Colonic Surveillance Summary and discussion Nederlandse Samenvatting Acknowledgements About the author 7 15 33 47

Chapter 5

59

Chapter 6

73

Chapter 7

89

Chapter 8

103

Chapter 9

117 123 129 131

proefschrift Marian.indd 5

10-10-2010 20:55:47

proefschrift Marian.indd 6

10-10-2010 20:55:48

CHaPter 1
General introduction

proefschrift Marian.indd 7

10-10-2010 20:55:49

| CHAPteR 1 Background
Inflammatory bowel disease (IBD), comprising ulcerative colitis (UC) and Crohns disease (CD), is a chronic inflammatory disorder of the gastrointestinal tract characterized by a relapsing course. Patients with IBD are at an increased risk of developing colorectal cancer (CRC). eaden et al. has shown cumulative risks of 2%, 8% and 18% after 10, 20, and 30 years of disease duration, respectively 1. A more recent meta-analysis, which censored for colectomy rate and critically appraised isolated small bowel Crohns disease and study population, reported cumulative CRC risks of 1%, 2%, and 5% after 10, 20 years, or more than 20 years in all IBD patients in population-based studies. In referral-centre studies and in patients with an IBD diagnosis before the third decade, this risk was significantly increased (Lutgens et al., unpublished results). Although IBD-related CRC only constitutes 1-2% of all CRCs, it is a frequent cause of death in this young patient group and IBD remains one of the three high-risk conditions for developing CRC along with familial adenomatous polyposis and Lynch syndrome. Risk factors for IBD-related CRC are, among others, disease duration, concomitant primary sclerosing cholangitis, positive family history, extent of the disease and severity of inflammation 2,3. these and other risk factors for IBD-related CRC are further described in a review shown in Chapter 2. Primary Sclerosing Cholangitis An identified risk factor for CRC in patients with IBD is concurrent primary sclerosing cholangitis (PSC). PSC is a chronic cholestatic liver disease, characterized by inflammation and fibrosis of the intra- and extra hepatic bile ducts 4. the pathogenesis and aetiology are unknown and a treatment to stop or reverse the progression of the disease is not available. Apart from the development of decompensated liver cirrhosis with transplantation as the only effective treatment, life expectancy of PSC patients is seriously threatened by the increased risk of developing cholangiocarcinoma (CCA) and CRC. In the literature, contradictory results are reported on the risk of CCA and CRC in this subgroup of IBD patients 5-7. therefore, we determined the risk of malignancies in a large cohort of Dutch PSC patients (Chapter 3). In Chapter 4, we compared patients with IBD-related CRC and concurrent PSC to patients with IBD-related CRC alone without involvement of the biliary tract with respect to clinical characteristics, endoscopic and histological findings, and prognosis. the results could potentially help defining improved clinical management strategies for individuals with IBD and concurrent PSC and guide future research in pathofysiological mechanisms in PSC-IBD-related CRC.

proefschrift Marian.indd 8

10-10-2010 20:55:49

Introduction | 9

Pathogenesis Colitis-associated CRC develops through the so-called inflammation-dysplasiacarcinoma sequence instead of the adenoma-carcinoma sequence in sporadic CRC 8 . Inflammation plays an important role in the development of cancer resulting in a high turnover of cells and an increased production of DNA-damaging agents 9. the gradual accumulation of (epi) genetic alterations causes damage to important cellular processes leading to transformation of normal mucosa to low-grade dysplasia (LGD), which may progress to high-grade dysplasia (HGD) and eventually to invasiveness. In the sporadic as well as the colitis-associated carcinogenetic pathway the same gene alterations are involved, only the timing and frequency of the mutations seem to differ (Figure 1). Apart from the two major pathways of genetic instability, i.e., the chromosomal instability pathway (CIN) and the microsatellite instability pathway (MSI), epigenetic alterations such as hypermethylation have also been shown to play a major role in carcinogenesis. the term DNA methylation describes the addition of a methyl group to the cytokine base of DNA. extensive methylation of cytosine bases is associated with promoter silencing resulting in a modification in gene expression. In this way,

Figure 1. Comparison between sporadic colon cancer and colitis-associated colon cancer

proefschrift Marian.indd 9

10-10-2010 20:55:50

10

| CHAPteR 1
hypermethylation causes silencing of multiple (tumour suppressor) genes and contributes to cancer initiation and progression 10. Chapters 5 -8 reports on molecular aspects of carcinogenesis in IBD-related CRC. Chapter 5 focuses on the molecular background of colorectal tumours in IBD with concurrent PSC. Microsatellite status was assessed in these tumours and compared to other subgroups of CRC, such as sporadic CRC, Lynch syndrome-related CRC and IBD-related CRC. the molecular background of PSC-IBD-related CRC was further elucidated in Chapter 6 by immunohistochemical expression and mutation-analysis of genes involved in carcinogenesis. In chapter 7 the methylation status of several tumour suppressor genes was assessed in inflammation-related CRC and non-inflammationrelated CRC. Involvement of an important carcinogenetic pathway, the Wnt-pathway, in inflammation-related colorectal carcinogenesis was evaluated in chapter 8. Surveillance the multi-step character of carcinogenesis in IBD makes this chronic disorder suitable for surveillance with the ultimate goal to detect neoplasia at a pre-cancerous stage or at least at a more favourable stage for performing a curative treatment. the guidelines, defined by the American Gastrointestinal Association (AGA) and the British Society for Gastroenterology (BSG) recommend screening colonoscopy after 8 years of disease duration in extensive IBD, comprising the whole colorectum followed by surveillance within 1-2 years after screening colonoscopy 3. Surveillance recommendations in patients with IBD and concomitant PSC are to initiate surveillance immediately after the diagnosis PSC by performing colonoscopy at a yearly basis. However, no clinical evidence is available to support these recommendations. Lundquist et al have shown that colitis often runs a more quiescent course in PSC-IBD patients than in UC patients without PSC 11. Furthermore, histological findings of colitis may precede clinical symptoms by as much as 7 years, which makes it more difficult to determine the exact duration of IBD 12. Several drawbacks of endoscopic surveillance have impeded its acceptance as a common practice performed by gastroenterologists on a worldwide basis. these include interobserver variability among pathologists when grading dysplasia and the disappearance of dysplasia on repeat biopsy after previously confirmed dysplasia 13,14. If this is combined with the requirement to take at least 33 random biopsies to rule out dysplasia with 90% certainty, this has led to considerable debate on the efficacy and the benefits of colonoscopic surveillance 15. In addition, the time- and cost-consuming effects as well as the burden of patients to undergo invasive endoscopy at regular intervals are important limitations of surveillance. Finally, it is still unclear whether colectomy should be advised to patients with LGD

proefschrift Marian.indd 10

10-10-2010 20:55:50

Introduction | 11

found in biopsies taken during surveillance endoscopy. the main reason for this is that the risk of progression of LGD to HGD or even adenocarcinoma is still subject of debate. Several reports studying the adherence of gastroenterologists to the international guidelines reveal a low adherence to these protocols, probably due to the above mentioned reasons 16,17. Despite all the scepticism, dysplasia has been unmistakably identified as a strong predictor of CRC in IBD patients and evidence of improved survival for patients who underwent surveillance is emerging. HGD is generally considered to be an indication for colectomy due to the high risk of synchronous or metachronous CRC 18. the progression rate of LGD however has for a long time provoked more controversy than HGD as an indication for proctocolectomy. A randomized controlled trial would be the ideal study design to assess whether surveillance really reduces IBD-CRC-related mortality. to date, such trial has not been conducted mainly because a very large population is needed and follow-up of this cohort should be done for a long period, i.e., at least 5-10 years. Case-control studies provide a more accessible insight. Few studies have assessed the question whether surveillance programs reduce CRC-related mortality 19-21. All publications so far point towards a beneficial effect of surveillance, with more favourable tumour stages being detected in patients included in a surveillance program. the most predominant limitations of the surveillance guidelines include sampling error at the time of biopsy, interobserver disagreement in histologically grading of dysplasia, i.e., LGD and disagreement on the optimal management of LGD, being either surveillance or colectomy. this indicates the need for continued research into the molecular pathogenesis of IBD-associated CRC, with the hope that finally targets for prevention will be identified. A more patient-tailored approach in surveillance is absolutely essential to keep such a programme cost-effective but also to keep the burden to patients as low as possible. Several studies have tried to unravel the molecular background of colitis-associated CRC in order to detect biomarkers that may indicate progression from normal to pre-neoplastic mucosa. these biomarkers in combination with advanced endoscopic techniques, aiming at better visualizing early stage CRC in IBD (dysplasia and early adenocarcinoma), may add to a more efficient and optimal surveillance program. Individual surveillance programs may increase patient care in several ways and eventually improve survival. the general and overall aim of this thesis is therefore to contribute to the elucidation of colitis-associated carcinogenesis, eventually leading to a more optimized surveillance program for patients with longstanding IBD and at risk of developing CRC.

proefschrift Marian.indd 11

10-10-2010 20:55:50

12

| CHAPteR 1 Reference List


1. eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526-535. 2. Farraye FA, Odze RD, eaden J, Itzkowitz SH, McCabe RP, Dassopoulos t, Lewis JD, Ullman tA, James t, III, McLeod R, Burgart LJ, Allen J, Brill JV. AGA medical position statement on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology 2010;138:738-745. 3. Farraye FA, Odze RD, eaden J, Itzkowitz SH. AGA technical review on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology 2010;138:746-74, 774. 4. Chapman RW, Arborgh BA, Rhodes JM, Summerfield JA, Dick R, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut 1980;21:870-877. 5. Broome U, Lofberg R, Veress B, eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995;22:1404-1408. 6. Gurbuz AK, Giardiello FM, Bayless tM. Colorectal neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Dis Colon Rectum 1995;38:37-41. 7. Loftus eV, Jr., Sandborn WJ, tremaine WJ, Mahoney DW, Zinsmeister AR, Offord KP, Melton LJ, III. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis. Gastroenterology 1996;110:432-440. 8. Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004;287:G7-17. 9. terzic J, Grivennikov S, Karin e, Karin M. Inflammation and colon cancer. Gastroenterology 2010;138:2101-2114. 10. Jass JR, Whitehall VL, Young J, Leggett BA. emerging concepts in colorectal neoplasia. Gastroenterology 2002;123:862-876. 11. Lundqvist K, Broome U. Differences in colonic disease activity in patients with ulcerative colitis with and without primary sclerosing cholangitis: a case control study. Dis Colon Rectum 1997;40:451-456. 12. Broome U, Lofberg R, Lundqvist K, Veress B. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum 1995;38:1301-1305. 13. Befrits R, Ljung t, Jaramillo e, Rubio C. Low-grade dysplasia in extensive, long-standing inflammatory bowel disease: a follow-up study. Dis Colon Rectum 2002;45:615-620. 14. eaden J, Abrams K, McKay H, Denley H, Mayberry J. Inter-observer variation between general and specialist gastrointestinal pathologists when grading dysplasia in ulcerative colitis. J Pathol 2001;194:152-157. 15. Rubin Ce, Haggitt RC, Burmer GC, Brentnall tA, Stevens AC, Levine DS, Dean PJ, Kimmey M, Perera DR, Rabinovitch PS. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology 1992;103:1611-1620. 16. eaden JA, Ward BA, Mayberry JF. How gastroenterologists screen for colonic cancer in ulcerative colitis: an analysis of performance. Gastrointest endosc 2000;51:123-128. 17. van Rijn AF, Fockens P, Siersema PD, Oldenburg B. Adherence to surveillance guidelines for dysplasia and colorectal carcinoma in ulcerative and Crohns colitis patients in the Netherlands. World J Gastroenterol 2009;15:226-230. 18. Bernstein CN, Shanahan F, Weinstein WM. Are we telling patients the truth about surveillance colonoscopy in ulcerative colitis? Lancet 1994;343:71-74. 19. 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. 20. Lashner BA, Kane SV, Hanauer SB. Colon cancer surveillance in chronic ulcerative colitis: historical cohort study. Am J Gastroenterol 1990;85:1083-1087.

proefschrift Marian.indd 12

10-10-2010 20:55:50

Introduction | 13

21. Lutgens MW, Oldenburg B, Siersema PD, van Bodegraven AA, Dijkstra G, Hommes DW, de Jong DJ, Stokkers PC, van der Woude CJ, Vleggaar FP. Colonoscopic surveillance improves survival after colorectal cancer diagnosis in inflammatory bowel disease. Br J Cancer 2009;101:1671-1675.

proefschrift Marian.indd 13

10-10-2010 20:55:50

14

proefschrift Marian.indd 14

10-10-2010 20:55:51

CHaPter 2
IBD-related carcinoma and lymphoma

MMH Claessen, PD Siersema, FP Vleggaar Department of Gastroenterology and Hepatology, University Medical Center Utrecht in press: Best Practice & Research: Clinical Gastroenterology

proefschrift Marian.indd 15

10-10-2010 20:55:53

16

| CHAPteR 2 Abstract
Patients with inflammatory bowel disease (IBD) have an increased risk of developing colorectal cancer (CRC). Risk factors are extent and severity of colonic inflammation, concurrent primary sclerosing cholangitis, and a positive family history of sporadic CRC. the chromosomal instability, microsatellite instability and hypermethylation pathways form the molecular background of IBD-related carcinogenesis, which is not different from sporadic CRC. the dysplasia-carcinoma sequence of IBD-related colorectal carcinogenesis makes patients suitable for endoscopic surveillance. In the future, new molecular biomarkers and endoscopic techniques may improve early detection of precursor lesions of IBD-related CRC. the potential of aminosalicylates and ursodeoxycholic acid as chemopreventive agents needs to be studied in randomized clinical trials. Patients with IBD who are being treated with thiopurines have a slightly increased risk of developing lymphoproliferative disorders, whereas patients with small bowel Crohns disease have a high relative risk and a small absolute risk of developing small bowel adenocarcinoma.

proefschrift Marian.indd 16

10-10-2010 20:55:53

IBD-related malignancies | 17

Introduction
Patients with inflammatory bowel disease (IBD), i.e. Crohns disease (CD) or ulcerative colitis (UC), have chronic relapsing inflammation of the colon, and in case of CD, also of the small bowel. In 1925, Crohn and Rosenberg were the first to report an association between IBD and colorectal carcinoma (CRC). Since then, it has become clear that patients with IBD located in the colon are at an increased risk of developing CRC. Although IBD-related CRC only constitutes 1-2% of all CRCs, it is a frequent cause of death in this relatively young patient group and remains one of the three high-risk conditions for developing CRC along with familial adenomatous polyposis (FAP) and Lynch syndrome. the dysplasia-carcinoma sequence of IBDrelated colorectal carcinogenesis makes patients suitable for endoscopic surveillance. Updated surveillance guidelines from the American Gastroenterological Association (AGA) and British Society for Gastroenterology (BSG) have recently been published (table 1) 1,2. Lymphoma and small bowel adenocarcinoma are other IBD-related malignancies that may occur in this group of patients. risk of developing CrC the magnitude of CRC risk in IBD remains a topic of debate. A landmark study on the risk of CRC in IBD patients is the meta-analysis of eaden et al. performed in 2001, showing risks of 2%, 8% and 18% after 10, 20, and 30 years of disease, respectively 3. these results were based on studies with highly variable methodologies or inclusion criteria and therefore need to be interpreted with caution. Since 2001, several population-based studies have been published indicating lower than previously assumed risks. Bernstein et al. calculated incidence rate ratios (IRR) for CRC in IBD patients matched with a non-IBD population and found an IRR of 2.64 (95%CI 1.69-4.12) in CD patients and 2.75 (95%CI 1.91-3.97) in UC patients 4. A recent study from eastern europe showed cumulative risks of 0.6%, 5.4%, and 7.5% after a disease duration of 10, 20, and 30 years, respectively 5. these studies also showed a decrease in CRC incidence over time. Speculations on the reasons for this decline include the use of maintenance therapy with possible chemopreventive effect, higher clinical awareness of precursor lesions, more wide-spread practice of endoscopic surveillance, and a more aggressive surgical approach towards dysplasia. risk factors for CrC Several risk factors for IBD-related CRC have been identified. First, disease duration was found to be an important risk factor. It is however difficult to determine the exact commencement of IBD in individual patients. eaden et al. reported a cumulative risk of only 2% after 10 years of UC 3. It is important to realise that some studies

proefschrift Marian.indd 17

10-10-2010 20:55:53

18

| CHAPteR 2
table 1. Summary of AGA Guidelines 2010. I. II. III. IV. V. VI. VII. Screening colonoscopy 8 years after onset of symptoms Left-sided/extensive colitis: start surveillance within 2 years after initial screening Repeat surveillance every 1-3 years Biopsies should be taken of each anatomic section of the colon Patients with PSC: start annual surveillance after this diagnosis Ideally, surveillance colonoscopy should be performed when remission is achieved A positive family history in first-degree relatives, ongoing active inflammation, anatomic abnormalities, or multiple inflammatory pseudopolyps may benefit from more frequent surveillance colonoscopies

included in this meta-analysis excluded all patients with CRC occurring within the first decade of IBD. Moreover, Lutgens et al. reported that 17-28% of IBD-related CRCs occur in the first decade after diagnosing IBD 6. therefore, one may question whether duration of IBD is important for evaluating the risk of CRC in individual patients. A second risk factor is the extent of colitis. An increasing gradient in CRC risk has been reported in IBD patients with extent of colitis ranging from proctitis to leftsided colitis to pancolitis, showing relative risks of 1.7, 2.8, and 14.8, respectively. Similar results have been reported in another study with relative risks of 19.2 and 2.8 for patients with pancolitis and left-sided disease, respectively 7,8. It is difficult to compare studies due to different definitions of colonic involvement and various methods that have been used to ascertain disease extent, varying from barium enema studies to endoscopic or histopathological techniques. Microscopic changes are superior in defining disease extent compared to endoscopic or radiographic changes, because neoplasia may arise in areas without endoscopic evidence of inflammation 9. Backwash ileitis, defined as involvement of the ileum in UC patients, has also been suggested as a risk factor for CRC. However, a more recent study with strict inclusion criteria (no CD patients) reported no increased risk in patients with backwash ileitis 10 . Severity of inflammation may also add to an increased risk. earlier studies may not have recognized severity of inflammation as risk factor because patients with severe inflammation used to undergo colectomy at an early stage. Furthermore, severity of inflammation may be an unreliable variable to assess in retrospective studies. More recent data did show that severity of inflammation is an independent risk factor. Rutter et al. reported a positive association between severity of inflammation and the development of CRC (OR 4.6 95%CI 2.1-10.5), later confirmed by Gupta et al. (OR 3.0 95%CI 1.4-6.3) 11,12. Recent AGA surveillance guidelines have been adjusted accordingly and surveillance at more frequent intervals is now advised in patients with severe inflammation.

proefschrift Marian.indd 18

10-10-2010 20:55:53

IBD-related malignancies | 19

A fourth risk factor is concurrent primary sclerosing cholangitis (PSC) 13,14. A recent Dutch PSC cohort study found 10-year and 20-year CRC risks of 14% and 31%, respectively 15. Remarkably, two-third of PSC-related CRCs were located in the proximal part of the colon 16. A meta-analysis by Soetikno et al. reported a four-fold higher risk of CRC in patients with concurrent PSC compared to patients with IBD alone 17. there is no clear explanation for this additional risk; colitis-related as well as PSC-related factors may be responsible. Guidelines advice to start annual surveillance for this specific subgroup directly after diagnosing PSC. A positive family history for CrC imparts a two-fold increased risk of CRC in IBD patients. A study by Askling et al. showed that almost 10% of all CRC cases occurred in IBD patients with a positive family history for CRC 18. Moreover, a positive family history for CRC was reported to be twice as common in UC-CRC patients compared to UC controls matched for extent and duration of colitis 19. the risk of CRC in relatives of IBD patients without CRC was not increased; however, first-degree relatives of patients with IBD-related CRC entailed an 80% increased risk of developing CRC 20. Another risk factor may be young age at onset of IBD. ekdom et al. reported a cumulative risk of 40% after 35 years of disease in patients diagnosed before the age of 15 years and 25% in patients diagnosed with IBD between 15-39 years of age 7. In contrast, Greenstein et al. showed an age-specific CRC incidence of 3.6 per 1,000 patient years in patients who were 10-19 years old at onset of IBD and 12.7 in patients who were between 30-39 years old at onset 21. It is important to realise that disease duration and a low risk of CRC at young age in the general population may be major confounders. Recent guidelines conclude that patients with IBD onset at early age have the same CRC risk as in adults with disease onset at older age, resulting in the advice to perform regular surveillance endoscopies also in this patient group. Dysplasia Dysplasia is the only currently available (histological) marker associated with progression to CRC. Dysplasia is defined as neoplastic alterations of the epithelium without invasiveness into the lamina propria. the diagnosis of dysplasia is divided into 3 subgroups: (a) negative for dysplasia, (b) indefinite for dysplasia (IND), and (c) positive for dysplasia, further subdivided into low-grade (LGD) and high-grade (HGD) dysplasia 22. the intra- and inter-observer agreement of diagnosing dysplasia among pathologists is low, especially in distinguishing LGD from IND or reactive mucosa 23. therefore, guidelines recommend that biopsies showing dysplasia should be reviewed by an experienced gastrointestinal pathologist. Several studies have reported a positive effect of endoscopic surveillance on survival of IBD patients 24-27. Lutgens et al. demonstrated in 149 patients with IBD-related

proefschrift Marian.indd 19

10-10-2010 20:55:53

20

| CHAPteR 2
CRC a significant reduction in CRC-related mortality in the surveillance group compared to the non-surveillance group (5-year survival rate of 100% and 74%, respectively). Moreover, a significantly higher proportion of early-stage tumours was found in the surveillance group. the outcomes of the above-mentioned studies need to be interpreted with caution because of non-randomized designs. Dysplastic lesions may differ in appearance varying from flat to raised and from unifocal to multifocal 22. As lesions are not always visible during endoscopy, Rubin et al. calculated that it is necessary to take at least 33 random biopsies, in order to detect dysplasia with 90% confidence 28. the detection of dysplasia can be improved with new endoscopic techniques, such as spraying the colonic mucosa with methylene blue or indigo carmine (chromoendoscopy). this technique increases sensitivity and specificity of endoscopic surveillance and questions the need for random biopsies. Kiesslich et al. randomized patients to conventional colonoscopy or chromoendoscopy using methylene blue. the rate of endoscopically detected dysplasia was significantly higher with methylene blue spraying compared to conventional endoscopy alone (32 vs 10 biopsies with neoplasia) 29. Similar results with chromoendoscopy have been reported by Marion et al. with 17 neoplastic lesions being detected with targeted chromoendoscopy compared to 3 lesions with random biopsies alone and 9 lesions with targeted biopsies using conventional colonoscopy 30. Another study reported an additional detection of 114 abnormalities in 55 patients, of which 7 were dysplastic, using indigo carmine spraying after conventional colonoscopy had already been performed 31. Consequently, the recently revised British Society for Gastroenterology (BSG) guidelines strongly advises using chromoendoscopy for surveillance in IBD. A recent study used a mathematical model to compare the sensitivity of random biopsy surveillance with that of enhanced endoscopy (for example chromoendoscopy), which can detect dysplastic fields of just 10 mm in diameter. Calculations showed that standard surveillance was insensitive to detect small areas of dysplasia. the number of random biopsies needed to achieve comparable sensitivity as with enhanced endoscopy was very high (n=4690). the clinical implications of detecting such small dysplastic fields, however, are unknown, particularly because the progression risk is unknown. Improvement of detection does not necessarily lead to an improved outcome. Moreover, it could lead to over-diagnosis, an increased number of endoscopic interventions or colectomies, resulting in additional morbidity and even mortality 32. It is important to consider the clinical consequences of diagnosing dysplasia. Bernstein et al. reported that 32% of patients with HGD progressed to CRC during follow-up. Furthermore, as HGD has been associated with a high rate of synchronous CRCs (42%), proctocolectomy is recommended in these patients. For those with LGD, the probability of eventually progressing was 16-29% 33. Connell et al. reported

proefschrift Marian.indd 20

10-10-2010 20:55:53

IBD-related malignancies | 21

a 5-year progression rate from LGD to HGD or CRC of 54%, which is similar to that reported by another group 34,35. A more recent study investigated flat LGD, reviewed by 3 experienced gastrointestinal pathologists and showed a 5-year progression risk of 37% 36. this wide range of progression rates probably reflects the large interobserver disagreement among pathologists. the 5-year progression risk for IND has been reported to vary between 5-28% 33,36. Due to the variability in progression rates, it is not clear what the therapeutic implications for a diagnosis of LGD are. In particular for unifocal dysplasia detected with random biopsies it is unclear what to advise the patient. In our experience, enhanced surveillance seems to be safe. It is suggested to consider all clinical, endoscopic, and histological risk factors and discuss these extensively with the patient before making the decision to perform either colectomy or enhanced surveillance. Figure 1 shows a flow chart of the management of dysplasia based on current guidelines. the time- and cost-consuming effects, the low inter-observer histological agreements as well as the uncertain progression rate of LGD stress the importance of identifying other factors that reliably identify patients at risk. Better understanding of the molecular carcinogenesis in an environment of chronic inflammation may provide molecular markers that could identify patients at risk for neoplastic progression. this could result in a more patient-tailored approach with individual risk stratification and adjusted surveillance intervals.

Figure 1. Flow chart of the management of dysplasia based on current guidelines

proefschrift Marian.indd 21

10-10-2010 20:55:54

22

| CHAPteR 2
IBD-related carcinogenesis It is known that IBD-related CRC develops through the so-called inflammationdysplasia-carcinoma sequence instead of the sporadic adenoma-carcinoma sequence 37. Inflammation plays an important role in the initiation and development of CRC 38. the gradual accumulation of (epi)genetic alterations causes damage to important cellular processes leading to transformation into LGD/HGD, which may progress to invasive CRC. Some features of the IBD-related molecular colorectal carcinogenesis are equal and some differ from its sporadic counterpart. the majority (85%) of sporadic CRCs arises due to problems in chromosomal stability and segregation, resulting in gain or loss of chromosomes, known as the chromosomal instability pathway (CIN). two key tumour suppressor genes involved in this pathway are adenomatous polyposis coli (APC) and p53. the timing and frequency of mutations in APC and p53 seem to differ between sporadic and IBD-related carcinogenesis. the second main carcinogenetic pathway is the microsatellite instability pathway (MSI), which involves loss of function of one of the mismatch repair (MMR) genes either by mutation or hypermethylation of the MLH1 promoter region. About 15% of the sporadic tumours develop through this pathway, in particular right-sided tumours. the same distribution of CIN and MSI is also found in IBD-related CRCs 37. Apart from the two major pathways of genetic instability, epigenetic alterations such as hypermethylation, leading to the addition of a methyl group to the cytokine base of DNA, have also been shown to play a role in carcinogenesis. Hypermethylation may cause silencing of multiple genes and contributes to cancer initiation and progression 38. Chromosomal instability pathway Aneuploidy Aneuploidy (abnormal DNA content) has been studied for several decades and has been shown to be an early event, even preceding dysplasia 28,39. It is one of the few markers that has been studied longitudinally. Lofberg et al. have shown that aneuploidy may also occur simultaneously with the development of dysplasia or even thereafter 40. Furthermore, CRCs may also develop without the development of aneuploidy 41. So, although aneuploidy may have additional value in cancer surveillance indicating a high risk of progression to neoplasia, it cannot be used as solitary marker of neoplastic progression. P53 the p53 protein arrests the entry of cells with DNA damage into the S phase, thereby providing an opportunity for DNA repair to proceed through normal repair mechanisms, whereas it triggers apoptosis in cells in which DNA damage is irreversible. Loss of these protective mechanisms predisposes to malignancy, especially under conditions

proefschrift Marian.indd 22

10-10-2010 20:55:54

IBD-related malignancies | 23

of persistent epithelial injury and high cell turnover, such as those prevailing in colitis 42. Most of the p53 mutations are missense mutations, producing an altered protein that accumulates in the cell nucleus which can be demonstrated by immunohistochemistry 43. Immunohistochemical expression, however, is an indirect manifestation of p53 inactivation. Some mutations, the so-called null-mutations, do not result in detectable expression of p53 protein. therefore, in case of a complete negative immunohistochemical expression, p53 mutation analysis should be performed. Several studies have shown that alterations in p53 seem to represent an early event in IBD-related neoplasia 44,45. Patients with longstanding UC without neoplasia and showing p53 expression develop up to 5 times more likely neoplasia than those without 46. Similarly, Holzmann et al. reported a higher frequency of p53 mutations in non-neoplastic epithelium in patients with neoplasia than those without neoplasia 47. However, a recent immunohistochemistry study did not report an early role for p53 expression, which was found only in already neoplastic mucosa 48. expression of p53 may be useful as a marker to distinguish between LGD and regenerative inflamed mucosa, although p53 expression also has been reported to be associated with inflammation. However, a study by Wong et al showed that a strong intensity of p53 staining was only found in dysplastic lesions 49. Wnt-pathway the -catenin mediated Wnt-pathway controls proliferation and differentiation in the colonic stem cell compartment and is involved in carcinogenesis 50. Loss of APC function results in activation of transcription of target genes such as C-myc and Cyclin D1. In general, Wnt-pathway activation dominates early sporadic carcinogenesis, whereas Wnt activation occurs less frequently, and, if occurring, usually at a late stage of IBD-related carcinogenesis 51. We recently reported, however, early and frequent Wnt activation in IBD-related neoplasia, based on nuclear -catenin and Cyclin D1 immunohistochemical staining. expression of these markers was found in 50% of non-dysplastic mucosa from IBD patients with colonic neoplasia elsewhere 48. Van Dekken et al. also found increased nucleo-cytoplasmic -catenin staining in 88% and 79% of dysplastic and cancerous lesions, respectively, whereas other studies have shown a low frequency of APC and -catenin mutations in IBDrelated CRC compared to sporadic CRC 52,53. Hence, available data on the role of Wnt activation in IBD-related carcinogenesis seem to be conflicting. Activation of the Wnt-pathway in early IBD-related carcinogenesis seems to be caused by other gene alterations than APC mutations.

proefschrift Marian.indd 23

10-10-2010 20:55:54

24

| CHAPteR 2
Kras Kras plays a pivotal role in the transmission of growth-promoting signals from the cell-surface to the nucleus and is assumed to be an early event in sporadic carcinogenesis. A low incidence of Kras mutations in IBD-related tumours (0-15%) compared with sporadic tumours (75%) has been reported and may indicate a minor role for this oncogene 54. Another explanation for the low incidence of Kras mutations in colitis-associated CRCs may be the occurrence of alterations in other Ras associated genes 55. DNA fingerprinting DNA fingerprinting is a technique that uses random primers to amplify the genome and to detect widespread genetic instability (allelic imbalances). Chen et al. have reported an increased genomic instability in adjacent histologically normal mucosa of IBD patients with neoplasia. More importantly, it was not found in colonic mucosa of patients without neoplasia or in non-UC controls 56. Further studies are needed to determine its potential role in biomarker-based surveillance. Microsatellite instability pathway the role of the DNA MisMatch Repair (MMR) system is to maintain genomic integrity by correcting mismatches that are generated by errors in base pairing occurring during normal DNA replication. Inactivation of MMR genes causes genomic instability, leading to a specific phenotype known as microsatellite instability (MSI). About 1520% of sporadic CRCs show MSI, with a high prevalence in the proximal colon 57. Instead of a germ line mutation as in Lynch syndrome, MSI arises in sporadic CRC through hypermethylation of the promoter region of hMLH1, one of the MMR genes, causing transcriptional silencing 58. the prevalence of MSI-High status in IBD-related CRCs varies between 9-15% 59-61, resulting in a similar distribution as in sporadic CRCs. Schulmann et al. did not find an association between MSI and clinical characteristics in IBD-related CRCs, such as a proximal tumour location, female gender, and a low tumour differentiation grade, as has been reported in sporadic tumours 62. A MSI-low phenotype has been reported in non-neoplastic actively inflamed mucosa of IBD patients 63. Some have hypothesized that the amount of DNA damage caused by oxidative stress accompanied by chronic inflammation may overwhelm the DNA MMR systems ability to repair small alterations, with as consequence a MSI-low status. In vitro studies have supported this hypothesis 64. the clinical significance of this finding is yet to be determined. CpG island methylation pathway Aberrant methylation of promoter region CpG islands is associated with transcriptional inactivation of tumour suppressor genes. In normal colorectal

proefschrift Marian.indd 24

10-10-2010 20:55:54

IBD-related malignancies | 25

epithelium, increased methylation has been found with aging, which is known as age-related methylation 65. Although methylation has been reported to be associated with inflammation, we found less frequent methylation in IBD-related CRC compared to sporadic CRC 66. Similar results have been reported by Konishi et al. who compared methylation levels in dysplastic and tumour samples of IBD patients with sporadic CRC tumour samples 67. they found a higher methylation level in sporadic CRC compared to IBD-related CRC. More importantly, a higher degree of methylation was detected in IBD-related dysplasia compared to IBD-related CRC samples, suggesting that genetic changes and not epigenetic alterations largely determine progression to malignancy in inflamed epithelium. two types of methylation have been reported in the colon, i.e. methylation starting in normal mucosa as a function of age (type A) and methylation that is restricted to tumours (type C). Issa et al. investigated the methylation status of 5 genes in nondysplastic and dysplastic colonic mucosa of 23 IBD patients. Remarkably, only increased methylation levels in type A genes were observed, suggesting that carcinogenesis may be a result of increased cell turnover due to chronic inflammation 68. Furthermore, methylation was also found in non-dysplastic mucosa from patients with colorectal neoplasia elsewhere, implicating that age-related methylation marks a field defect that reflects predisposition to colorectal neoplasia. In addition, the data suggest that chronic inflammation is associated with high levels of type A methylation and that colitis-related carcinogenesis can be viewed as a result of premature aging of colorectal epithelial cells. A recent study reported increased methylation of some genes, i.e. MINT1 and RUNX3 in non-adjacent normal mucosa of patients with IBD-related CRC compared with UC controls without CRC. these results suggest a possible role for methylation as a screening tool in cancer surveillance, thereby identifying those at increased risk of CRC in routinely taken biopsies 69. Chemoprevention the use of pharmacotherapy as a chemopreventive measure to reduce the risk of developing CRC has been studied extensively. the ultimate goal of chemoprevention is to reduce CRC risk, allowing for less frequent surveillance examinations and a reduction in incidence of invasive tumours. Aminosalicylates (5-ASA) the most widely studied agent for chemoprevention is 5-ASA. A meta-analysis of nine observational studies demonstrated a reduced risk of developing CRC or dysplasia with 5-ASA with an OR of 0.51 (95%CI 0.37-0.69) 70. this is in contrast to a retrospective case-control study in 25 patients with IBD and CRC, that found no

proefschrift Marian.indd 25

10-10-2010 20:55:54

26

| CHAPteR 2
association between 5-ASA use and reduction in CRC risk 71. As conflicting data are available, the US Preventive Services task Force has concluded that there is moderate certainty that the magnitude of net benefits is moderate. Ursodeoxycholic acid (UDCA) UDCA has been shown to significantly reduce CRC risk in patients with UC and concurrent PSC. In animal studies, UDCA was found to inhibit colorectal carcinogenesis by reducing the colonic concentration of secondary bile acids, inhibiting ras gene mutations and by showing anti-oxidant activity 72. two separate studies have also shown a chemopreventive effect of UDCA in IBD patients with concomitant PSC, resulting in a lower incidence of dysplasia (OR 0.18 and 0.26) 73,74. therefore, the use of UDCA in patients with IBD and concurrent PSC is encouraged; however, the role of UDCA as a chemopreventive agent in UC without PSC is unknown. Immunomodulators and Biologicals Use of azathioprine (AZA) or 6-mercaptopurine (6-MP) has not consistently been shown to be associated with lower rates of CRC. Any possible effect of tNF therapy as chemopreventive agent has not been studied yet. Folic acid, Calcium, Multivitamins & Corticosteroids Folic acid deficiency has been associated with an increased risk of developing CRC 75. Patients with chronic IBD are prone to folate deficiency due to inadequate nutritional intake and reduced intestinal absorption. Several studies have suggested a trend towards protection against CRC in folate users, although neither study demonstrated statistical significance 76. Oral or topical corticosteroids, while demonstrating anti-neoplastic effects in 2 studies, are associated with significant side effects and should not be used as chemopreventive agents 77. Calcium, multivitamins, and statins have not consistently been associated with lower rates of CRC. Lymphoproliferative disorders in patients with IBD An increased risk of lymphoproliferative disorders (LD) in patients with IBD has been a reported in several observational studies 78,79. However, population-based studies did not confirm these findings. Askling et al. did not find an increased risk of LD in 50,000 Swedish IBD patients 80. Similar results were reported by Lewis et al. 81. One large population-based study from Canada however, reported an increased risk of LD in male patients with CD 4. Palli et al. from Italy reported an excess risk of Hodgkin lymphoma in patients with IBD 82. these results have not been confirmed by others.

proefschrift Marian.indd 26

10-10-2010 20:55:54

IBD-related malignancies | 27

the development of LD may in fact be related to the use of immunosuppressives for the treatment of IBD or to the chronic course of IBD itself. Recent studies have reported a 5-fold increased risk of developing LD under AZA or 6-MP treatment 83. the possible role of anti-tNF therapy in having a relationship with lymphomas is difficult to determine, mainly because patients are often co-treated with thiopurines or other immunosuppressants, such as steroids. Small bowel adenocarcinoma in CD the risk of developing small bowel adenocarcinoma is increased in patients with CD with a relative risk of 28 compared to the general population. In patients with CD limited to the small bowel, the risk further increases to 159 84. Palascak-Juif et al. described cumulative risks of 0.2% at 10 years and 2.2% at 25 years in 1935 CD patients with small bowel involvement at diagnosis 85. Risk factors for small bowel adenocarcinoma in CD include small bowel strictures, chronic fistulous disease, CD located in the distal jejunum and ileum, a small bowel bypass loop, and immunosuppressive therapy with AZA, 6-MP or anti-tNF agents 86. the carcinogenesis of small bowel adenocarcinoma is still unknown due to the poor accessibility of the small bowel and the low overall incidence. Several hypotheses have been postulated to explain the reduced absolute carcinoma risk including a rapid transit time leading to a short exposure time of carcinogenic substances, the absence of secondary bile acids in the small bowel, and large quantities of lymphoid tissue present in the small bowel. Survival is poor for patients with small bowel adenocarcinoma with reported 5-year survival rates between 20-30%. Mortality after 1-2 years varies between 30% and 60% 87. Unfortunately, our diagnostic tools to investigate the small bowel are currently not adequate to detect small bowel adenocarcinoma at an early stage.

Summary
Patients with IBD have an increased risk of developing CRC, but the magnitude of the risk depends on the type of patient population studied. Identified risk factors include extent and severity of disease, concurrent PSC and a positive family history for sporadic CRC. endoscopic surveillance improves overall survival and decreases CRC-related mortality, most likely due to detection of early neoplastic lesions with a more favourable prognosis. Dysplasia is currently the only marker associated with progression to CRC. Patients with HGD are advised to undergo proctocolectomy. Clinical decision making in patients with LGD is impeded by the low interobserver agreement among pathologists and the wide variation of observed progression risks

proefschrift Marian.indd 27

10-10-2010 20:55:54

28

| CHAPteR 2
in LGD. Chromoendoscopy with methylene blue or indigo carmine increases sensitivity and specificity of dysplasia detection. the progression risk of small dysplastic lesions detected by enhanced endoscopy is however, not known yet. In the future, biomarkers may aid in risk stratification and clinical management of patients with IBD-related CRC. Drugs, such as 5-ASA and UDCA may have chemopreventive potential in IBD, but their efficacy still needs to be evaluated in randomized studies. Patients with IBD are also at risk for developing LD when they are on thiopurine treatment. Small bowel adenocarcinoma may develop in patients with CD located in the small bowel, although the absolute risk for this malignancy is low.

proefschrift Marian.indd 28

10-10-2010 20:55:54

IBD-related malignancies | 29

Reference List
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 Farraye FA, Odze RD, eaden J, Itzkowitz SH. AGA technical review on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology 2010;138:746-74. Cairns SR, Scholefield JH, Steele RJ, et al. Guidelines for colorectal cancer screening and surveillance in moderate and high risk groups (update from 2002). Gut 2010;59:666-89. eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526-35. Bernstein CN, Blanchard JF, Kliewer e, Wajda A. Cancer risk in patients with inflammatory bowel disease: a population-based study. Cancer 2001;91:854-62. Lakatos L, Lakatos PL. Is the incidence and prevalence of inflammatory bowel diseases increasing in eastern europe? Postgrad Med J 2006;82:332-7. Lutgens MW, Vleggaar FP, Schipper Me, et al. High Frequency of early Colorectal Cancer in Inflammatory Bowel Disease. Gut 2008;57:1246-51. ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer. A population-based study. N engl J Med 1990;323:1228-33. Gyde SN, Prior P, Allan RN, et al. Colorectal cancer in ulcerative colitis: a cohort study of primary referrals from three centres. Gut 1988;29:206-17. Mathy C, Schneider K, Chen YY, et al. Gross versus microscopic pancolitis and the occurrence of neoplasia in ulcerative colitis. Inflamm Bowel Dis 2003;9:351-5. Haskell H, Andrews CW, Jr., Reddy SI, et al. Pathologic features and clinical significance of backwash ileitis in ulcerative colitis. Am J Surg Pathol 2005;29:1472-81. Gupta RB, Harpaz N, Itzkowitz S, et al. Histologic inflammation is a risk factor for progression to colorectal neoplasia in ulcerative colitis: a cohort study. Gastroenterology 2007;133:1099-105. Rutter M, Saunders B, Wilkinson K, et al. Severity of inflammation is a risk factor for colorectal neoplasia in ulcerative colitis. Gastroenterology 2004;126:451-9. Broome U, Lofberg R, Veress B, eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995;22:1404-8. Kornfeld D, ekbom A, Ihre t. Is there an excess risk for colorectal cancer in patients with ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut 1997;41:522-5. Claessen MM, Vleggaar FP, tytgat KM, et al. High lifetime risk of cancer in primary sclerosing cholangitis. J Hepatol 2009;50:158-64. Claessen MM, Lutgens MW, van Buuren HR, et al. More right-sided IBD-associated colorectal cancer in patients with primary sclerosing cholangitis. Inflamm Bowel Dis 2009;15:1331-6. Soetikno RM, Lin OS, Heidenreich PA, et al. Increased risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis: a meta-analysis. Gastrointest endosc 2002;56:48-54. Askling J, Dickman PW, Karlen P, et al. Family history as a risk factor for colorectal cancer in inflammatory bowel disease. Gastroenterology 2001;120:1356-62. Nuako KW, Ahlquist DA, Mahoney DW, et al. Familial predisposition for colorectal cancer in chronic ulcerative colitis: a case-control study. Gastroenterology 1998;115:1079-83. Askling J, Dickman PW, Karlen P, et al. Colorectal cancer rates among first-degree relatives of patients with inflammatory bowel disease: a population-based cohort study. Lancet 2001;357:262-6. Greenstein AJ, Sachar DB, Smith H, et al. Cancer in universal and left-sided ulcerative colitis: factors determining risk. Gastroenterology 1979;77:290-4. Riddell RH, Goldman H, Ransohoff DF, et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983;14:931-68. eaden J, Abrams K, McKay H, et al. Inter-observer variation between general and specialist gastrointestinal pathologists when grading dysplasia in ulcerative colitis. J Pathol 2001;194:152-7.

18 19 20 21 22 23

proefschrift Marian.indd 29

10-10-2010 20:55:54

30

| CHAPteR 2
24 Choi PM, Nugent FW, Schoetz DJ, Jr., et al. Colonoscopic surveillance reduces mortality from colorectal cancer in ulcerative colitis. Gastroenterology 1993;105:418-24. 25 Karlen P, Lofberg R, Brostrom O, et al. Increased risk of cancer in ulcerative colitis: a population-based cohort study. Am J Gastroenterol 1999;94:1047-52. 26 Lashner BA, Kane SV, Hanauer SB. Colon cancer surveillance in chronic ulcerative colitis: historical cohort study. Am J Gastroenterol 1990;85:1083-7. 27 Lutgens MW, Oldenburg B, Siersema PD, et al. Colonoscopic surveillance improves survival after colorectal cancer diagnosis in inflammatory bowel disease. Br J Cancer 2009;101:1671-5. 28 Rubin Ce, Haggitt RC, Burmer GC, et al. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology 1992;103:1611-20. 29 Kiesslich R, Fritsch J, Holtmann M, et al. Methylene blue-aided chromoendoscopy for the detection of intraepithelial neoplasia and colon cancer in ulcerative colitis. Gastroenterology 2003;124:880-8. 30 Marion JF, Waye JD, Present DH, et al. Chromoendoscopy-targeted biopsies are superior to standard colonoscopic surveillance for detecting dysplasia in inflammatory bowel disease patients: a prospective endoscopic trial. Am J Gastroenterol 2008;103:2342-9. 31 Rutter MD, Saunders BP, Schofield G, et al. Pancolonic indigo carmine dye spraying for the detection of dysplasia in ulcerative colitis. Gut 2004;53:256-60. 32 Awais D, Siegel CA, Higgins PD. Modelling dysplasia detection in ulcerative colitis: clinical implications of surveillance intensity. Gut 2009;58:1498-503. 33 Bernstein CN, Shanahan F, Weinstein WM. Are we telling patients the truth about surveillance colonoscopy in ulcerative colitis? Lancet 1994;343:71-4. 34 Connell WR, Lennard-Jones Je, Williams CB, et al. Factors affecting the outcome of endoscopic surveillance for cancer in ulcerative colitis. Gastroenterology 1994;107:934-44. 35 Ullman t, Croog V, Harpaz N, et al. Progression of flat low-grade dysplasia to advanced neoplasia in patients with ulcerative colitis. Gastroenterology 2003;125:1311-9. 36 van Schaik FD, ten Kate F, Offerhaus GJA, et al. Substantial Increase of Progression Rate of Flat Low-Grade Dysplasia in Inflammatory Bowel Disease After Review by an expert Pathologist Panel. Gastroenterology 138[5], s-45. 2010. 37 Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004 ;287:G7-17. 38 terzic J, Grivennikov S, Karin e, Karin M. Inflammation and colon cancer. Gastroenterology 2010;138:2101-14. 39 Lindberg JO, Stenling RB, Rutegard JN. DNA aneuploidy as a marker of premalignancy in surveillance of patients with ulcerative colitis. Br J Surg 1999;86:947-50. 40 Lofberg R, Brostrom O, Karlen P, et al. DNA aneuploidy in ulcerative colitis: reproducibility, topographic distribution, and relation to dysplasia. Gastroenterology 1992;102:1149-54. 41 Lofberg R, Lindquist K, Veress B, tribukait B. Highly malignant carcinoma in chronic ulcerative colitis without preceding dysplasia or DNA aneuploidy. Report of a case. Dis Colon Rectum 1992;35:82-6. 42 Harpaz N, Peck AL, Yin J, et al. p53 protein expression in ulcerative colitis-associated colorectal dysplasia and carcinoma. Hum Pathol 1994;25:1069-74. 43 Bruwer M, Schmid KW, Senninger N, Schurmann G. Immunohistochemical expression of P53 and oncogenes in ulcerative colitis-associated colorectal carcinoma. World J Surg 2002;26:390-6. 44 Brentnall tA, Crispin DA, Rabinovitch PS, et al. Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology 1994;107:369-78. 45 Burmer GC, Rabinovitch PS, Haggitt RC, et al. Neoplastic progression in ulcerative colitis: histology, DNA content, and loss of a p53 allele. Gastroenterology 1992;103:1602-10. 46 Lashner BA, Shapiro BD, Husain A, Goldblum JR. evaluation of the usefulness of testing for p53 mutations in colorectal cancer surveillance for ulcerative colitis. Am J Gastroenterol 1999;94:456-62. 47 Holzmann K, Weis-Klemm M, Klump B, et al. Comparison of flow cytometry and histology with mutational screening for p53 and Ki-ras mutations in surveillance of patients with long-standing ulcerative colitis. Scand J Gastroenterol 2001 ;36:1320-6.

proefschrift Marian.indd 30

10-10-2010 20:55:54

IBD-related malignancies | 31
48 Claessen MM, Schipper Me, Oldenburg B, et al. WNt-pathway activation in IBD-associated colorectal carcinogenesis: potential biomarkers for colonic surveillance. Cell Oncol 2010;32:303-10. 49 Wong NA, Mayer NJ, MacKell S, et al. Immunohistochemical assessment of Ki67 and p53 expression assists the diagnosis and grading of ulcerative colitis-related dysplasia. Histopathology 2000;37:108-14. 50 Pinto D, Clevers H. Wnt control of stem cells and differentiation in the intestinal epithelium. exp Cell Res 2005;306:357-63. 51 tarmin L, Yin J, Harpaz N, et al. Adenomatous polyposis coli gene mutations in ulcerative colitis-associated dysplasias and cancers versus sporadic colon neoplasms. Cancer Res 1995;55:2035-8. 52 Aust De, terdiman JP, Willenbucher RF, et al. the APC/beta-catenin pathway in ulcerative colitisrelated colorectal carcinomas: a mutational analysis. Cancer 2002 ;94:1421-7. 53 van Dekken, Wink JC, Vissers KJ, et al. Wnt pathway-related gene expression during malignant progression in ulcerative colitis. Acta Histochem 2007;109:266-72. 54 Burmer GC, Levine DS, Kulander BG, et al. c-Ki-ras mutations in chronic ulcerative colitis and sporadic colon carcinoma. Gastroenterology 1990;99:416-20. 55 Haigis KM, Kendall KR, Wang Y, et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet 2008;40:600-8. 56 Chen R, Rabinovitch PS, Crispin DA, et al. DNA fingerprinting abnormalities can distinguish ulcerative colitis patients with dysplasia and cancer from those who are dysplasia/cancer-free. Am J Pathol 2003;162:665-72. 57 Deng G, Kakar S, tanaka H, et al. Proximal and distal colorectal cancers show distinct gene-specific methylation profiles and clinical and molecular characteristics. eur J Cancer 2008;44:1290-301. 58 Kuismanen SA, Holmberg Mt, Salovaara R, et al. Genetic and epigenetic modification of MLH1 accounts for a major share of microsatellite-unstable colorectal cancers. Am J Pathol 2000;156:1773-9. 59 Fleisher AS, esteller M, Harpaz N, et al. Microsatellite instability in inflammatory bowel diseaseassociated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1. Cancer Res 2000 ;60:4864-8. 60 Svrcek M, el-Bchiri J, Chalastanis A, et al. Specific clinical and biological features characterize inflammatory bowel disease associated colorectal cancers showing microsatellite instability. J Clin Oncol 2007;25:4231-8. 61 Willenbucher RF, Aust De, Chang CG, et al. Genomic instability is an early event during the progression pathway of ulcerative-colitis-related neoplasia. Am J Pathol 1999 ;154:1825-30. 62 Schulmann K, Mori Y, Croog V, et al. Molecular phenotype of inflammatory bowel disease-associated neoplasms with microsatellite instability. Gastroenterology 2005 ;129:74-85. 63 Itzkowitz SH. Molecular biology of dysplasia and cancer in inflammatory bowel disease. Gastroenterol Clin North Am 2006;35:553-71. 64 Chang CL, Marra G, Chauhan DP, et al. Oxidative stress inactivates the human DNA mismatch repair system. Am J Physiol Cell Physiol 2002;283:C148-C154. 65 Fujii S, Fujimori t, Chiba t, et al. efficacy of surveillance and molecular markers for detection of ulcerative colitis-associated colorectal neoplasia. J Gastroenterol 2003;38:1117-25. 66 Shen L, Ahuja N, Shen Y, et al. DNA methylation and environmental exposures in human hepatocellular carcinoma. J Natl Cancer Inst 2002;94:755-61. 67 Konishi K, Shen L, Wang S, et al. Rare CpG island methylator phenotype in ulcerative colitis-associated neoplasias. Gastroenterology 2007;132:1254-60. 68 Issa JP, Ahuja N, toyota M, et al. Accelerated age-related CpG island methylation in ulcerative colitis. Cancer Res 2001;61:3573-7. 69 Garrity-Park MM, Loftus eV, Jr., Sandborn WJ, et al. Methylation status of genes in non-neoplastic mucosa from patients with ulcerative colitis-associated colorectal cancer. Am J Gastroenterol 2010;105:1610-9.

proefschrift Marian.indd 31

10-10-2010 20:55:54

32

| CHAPteR 2
70 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;100:1345-53. 71 Bernstein CN, Blanchard JF, Metge C, Yogendran M. Does the use of 5-aminosalicylates in inflammatory bowel disease prevent the development of colorectal cancer? Am J Gastroenterol 2003;98:2784-8. 72 Alberts DS, Martinez Me, Hess LM, et al. Phase III trial of ursodeoxycholic acid to prevent colorectal adenoma recurrence. J Natl Cancer Inst 2005;97:846-53. 73 Pardi DS, Loftus eV, Jr., Kremers WK, et al. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology 2003;124:889-93. 74 tung BY, emond MJ, Haggitt RC, et al. Ursodiol use is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Ann Intern Med 2001;134:89-95. 75 Giovannucci e, Stampfer MJ, Colditz GA, et al. Folate, methionine, and alcohol intake and risk of colorectal adenoma. J Natl Cancer Inst 1993;85:875-84. 76 Lashner BA, Provencher KS, Seidner DL, et al. the effect of folic acid supplementation on the risk for cancer or dysplasia in ulcerative colitis. Gastroenterology 1997 ;112:29-32. 77 Velayos FS, Loftus eV, Jr., Jess t, et al. Predictive and protective factors associated with colorectal cancer in ulcerative colitis: A case-control study. Gastroenterology 2006;130:1941-9. 78 ekbom A, Helmick C, Zack M, et al. extracolonic malignancies in inflammatory bowel disease. Cancer 1991;67:2015-9. 79 Greenstein AJ, Gennuso R, Sachar DB, et al. extraintestinal cancers in inflammatory bowel disease. Cancer 1985;56:2914-21. 80 Askling J, Brandt L, Lapidus A, et al. Risk of haematopoietic cancer in patients with inflammatory bowel disease. Gut 2005;54:617-22. 81 Lewis JD, Bilker WB, Brensinger C, et al. Inflammatory bowel disease is not associated with an increased risk of lymphoma. Gastroenterology 2001;121:1080-7. 82 Palli D, trallori G, Bagnoli S, et al. Hodgkins disease risk is increased in patients with ulcerative colitis. Gastroenterology 2000;119:647-53. 83 Beaugerie L, Brousse N, Bouvier AM, et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Lancet 2009;374:1617-25. 84 von Roon AC, Reese G, teare J, et al. the risk of cancer in patients with Crohns disease. Dis Colon Rectum 2007;50:839-55. 85 Palascak-Juif V, Bouvier AM, Cosnes J, et al. Small bowel adenocarcinoma in patients with Crohns disease compared with small bowel adenocarcinoma de novo. Inflamm Bowel Dis 2005;11:828-32. 86 Feldstein RC, Sood S, Katz S. Small bowel adenocarcinoma in Crohns disease. Inflamm Bowel Dis 2008;14:1154-7. 87 Solem CA, Harmsen WS, Zinsmeister AR, Loftus eV, Jr. Small intestinal adenocarcinoma in Crohns disease: a case-control study. Inflamm Bowel Dis 2004 ;10:32-5.

proefschrift Marian.indd 32

10-10-2010 20:55:56

CHaPter 3
High lifetime risk of cancer in primary sclerosing cholangitis

MMH Claessen1, FP Vleggaar1, KM tytgat1, PD Siersema1, HR van Buuren2


1

Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Department of Gastroenterology and Hepatology, erasmus MC - University Medical Center Rotterdam Journal of Hepatology, 2009 50: 158-64

proefschrift Marian.indd 33

10-10-2010 20:55:57

34

| CHAPteR 3 Abstract
Background Primary sclerosing cholangitis (PSC) patients are at risk for developing cholangiocarcinoma (CCA) and colorectal carcinoma (CRC). aims & Methods Our aim was to assess the risk of malignancies and their influence on survival. Data from PSC patients diagnosed between 1980 and 2006 in two university hospitals were retrieved. the Kaplan-Meier method and a time-dependent Cox regression model were used to calculate risks of malignancies and their influence on survival. results 211 Patients were included, 143 (68%) were male and 126 (60%) had inflammatory bowel disease (IBD). Median transplantation-free survival was 14 years. the risk of CCA after 10 and 20 years was 9% and 9%, respectively. In patients with concomitant IBD the 10-year and 20-year risks for CRC were 14% and 31%, which was significantly higher than for patients without IBD (2% and 2% (p= 0.008)). CCA, cholangitis, and age at entry were independent risk factors for the combined endpoint death or liver transplantation. Risk factors for the endpoint death were CCA, CRC, age, and symptomatic presentation. Conclusions Patients with PSC and IBD have a high long-term risk of developing CRC and this risk is about threefold higher than the risk for CCA. Both malignancies are associated with decreased survival.

proefschrift Marian.indd 34

10-10-2010 20:55:57

Malignancies in PSC | 35

Introduction
Primary sclerosing cholangitis (PSC) is a chronic disease, characterized by inflammation and fibrosis of the intra- and extrahepatic bile ducts 1,2. the etiology is unknown and a therapy to stop or reverse progression of the disease is not available. Apart from the development of decompensated liver cirrhosis with transplantation as the only effective treatment, the life expectancy of PSC patients is seriously threatened by the unpredictable occurrence of cholangiocarcinoma (CCA) and colorectal cancer (CRC). the reported overall risk for CCA varies from 8% to 36% 3-6 and the 10-year cumulative incidence from 11% to 31% 3,7-9. these highly variable results are probably largely explained by differences in patient selection and duration of follow-up. Few studies have assessed the risk of CRC in PSC. Up to 90% of patients with PSC have concurrent inflammatory bowel disease (IBD), a major risk factor for CRC 10,11. It is still debated whether PSC carries an additional and independent risk for CRC. the frequent use of colorectal neoplasia (dysplasia and CRC) instead of CRC as clinical endpoint of studies contributes to the difficulty to appreciate the actual CRC risk. Broome et al. 12 found cumulative risks for colorectal neoplasia of 9%, 31%, and 50% after an IBD disease duration of 10, 20, and 25 years, respectively. In contrast, others reported no significant difference in CRC risk between patients with PSC-IBD or IBD alone 13-15. the aim of this study was to assess the risk of malignancies, in particular CRC and CCA, as well as their impact on survival in a large and long-term follow-up study of Dutch PSC patients.

Methods
Patients All patients diagnosed with PSC in the period January 1980 - May 2006 were identified from two databases (endobase III, Olympus and the central hospital patient registration system) at the University Medical Center of Utrecht (UMCU) and the erasmus MC University center, the latter being one of the three liver transplant centres in the Netherlands. PSC patients, seen at the UMCU, with an indication for liver transplantation were referred to Rotterdam. the diagnosis of PSC was based on typical findings on cholangiography 16, or the presence of characteristic histological liver abnormalities (peri-cholangiolar onionskin fibrosis, 17 in combination with an elevated serum alkaline phosphatase and the presence of IBD. the onset of PSC was designated as the date of the first abnormalities on cholangiography or liver histology. Patients with bile duct abnormalities attributable to other causes such as bile duct surgery, ischemia, portal vein thrombosis, or autoimmune pancreatico-cholangitis were excluded 18-20.

proefschrift Marian.indd 35

10-10-2010 20:55:57

36

| CHAPteR 3
Collection of data Data were retrieved from the medical records and computerized hospital databanks. Missing information was completed by contacting the patients physician. Data collected at the time of PSC diagnosis included: time of onset of symptoms, presence, extension, duration and type of IBD, asymptomatic or symptomatic (defined as: pruritus, jaundice, fatigue, cholangitis, ascites, variceal bleeding, hepatic encephalopathy or CCA) presentation, medical history, and laboratory data. the diagnosis of IBD was based on endoscopic and histological findings. During follow-up, the following data and events were collected: date and cause of death, liver transplantation, bacterial cholangitis, dominant bile duct strictures, diagnosis and therapy of malignancies, newly developed IBD with extent and duration of the disease, and participation in a CRC surveillance program. Histological criteria by Riddell et al. for diagnosing colitis-associated-dysplasia and carcinoma were applied 21. Dysplasia was not considered part of the diagnosis CRC. For this study surveillance was defined as colonic surveillance according to the AGA guidelines 22. therefore, patients who had (incidental) follow-up colonoscopies, but not according to a fixed time schedule and without multiple biopsies, were not considered as having participated in a surveillance program. A dominant bile duct stricture was defined as a stricture without malignant cause that required intervention which would lead to a 50% decrease of bilirubin within two months. end of follow up was either the end of the study (May 2006), death, or time of last visit in case patients were lost-to-follow-up. Statistical analysis Follow-up commenced at the time that a diagnosis of PSC was made. Primary endpoints of the study were CRC, CCA, and liver transplantation. Survival and risk analyses were performed using the Kaplan-Meier method 23. Groups were compared using the log-rank test. With respect to CCA, two separate analyses were made; one with and one without bile duct tumours diagnosed within six months after PSC was detected. For the cumulative risk of CRC, censoring was performed for colectomy. the prognostic significance of entry data was determined by univariate analysis (log rank and Cox regression). A time-dependent Cox regression model was used for multivariate analysis of both entry and follow-up variables independently related to the endpoints death or liver transplantation. In a separate analysis, the influence of malignancies on death alone was determined, also including patients after liver transplantation. A multivariate analysis with entry variables was performed to assess potential risk factors for CCA and CRC. All statistical analyses were done with SPSS 12.0. P-values <0.05 were considered statistically significant.

proefschrift Marian.indd 36

10-10-2010 20:55:57

Malignancies in PSC | 37

Results
Patient characteristics at diagnosis of PSC two hundred eleven patients (143 males) were included (table 1). the median age at diagnosis was 33 years (range 11-72) for males and 40 years (range 11-75) for females. Seventy-six (36%) patients were symptomatic at the time of diagnosis. the most prevalent symptom was jaundice (n=43). In most cases (84%) diagnosis of PSC was based on cholangiographic findings. Histology was available in 148 (70%) patients and showed cirrhosis in 23 cases. Concurrent IBD was present in 126 (60%) patients of which 23 (18%) had Crohns disease, all with colonic involvement. to rule out the diagnosis of IBD in asymptomatic patients (n=85), colonoscopy was performed in 62% of these patients. the median interval between the initial diagnosis IBD and subsequent PSC was 6.9 years (range 0-37.7).

table 1. entry characteristics of 211 patients with primary sclerosing cholangitis Variable Sex, male (%) Age at diagnosis PSC (yrs) (median (range))
*

n=211 143 (68) 35 (11-75) 76 (36) 23 (11) 126 (60) 85 (40) 93 (44) 23 (11) 10 (5) 6.9 (0-38) 11 (5) 17 (4-400) 276 (11-2325) 237 (14-1557) 68 (6-918) 96 (10-266) 40.8 (21.3-55) 263 (44-739)

Presentation, symptomatic (%) Histology, cirrhosis 1 (%) IBD (%)


*

- No

- Ulcerative colitis - Crohns disease - Indeterminate colitis Interval IBD-PSC (yrs) (median (range)) Previous colectomy Bilirubin (mol/L) (normal 3-21)
3

Alkaline phosphatase (U/L) (normal <120) Gt (U/L) (normal <55) ASt (U/L) (normal <35) ALt (U/L) (normal <45) Albumin (g/L) (normal 34-50) Platelet count (10 ) (normal 150-450)
9

PSC: primary sclerosing cholangitis; IBD: inflammatory bowel disease Histology available in 148 patients 2. endoscopically verified in 62.4% 3. Laboratory values given in median (range)
* * 1.

proefschrift Marian.indd 37

10-10-2010 20:55:57

38

| CHAPteR 3
Follow up the median follow-up was 9.0 years (range 0.3-25 years); 15% (n=32) of the patients were lost to follow-up. Forty-five (21%) patients died during follow up and 49 (23%) patients underwent liver transplantation. the estimated median transplantation-free survival of the entire cohort was 14 years. Patients with a symptomatic presentation had a significantly lower transplantation-free survival rate than those with an asymptomatic presentation (p=0.001) (Figure 1). IBD was newly diagnosed in 33/85 (39%) patients without concurrent IBD at entry. Consequently, the total prevalence of IBD in this cohort was 75%. the 10-year and 20-year risks for developing bacterial cholangitis were 42% and 57%, respectively. the risk of a dominant structure was 35% and 39% after disease duration of 10 and 20 years, respectively. Malignancies Malignancies were the most frequent (44%) cause of death (n=45) in our cohort. CCA and CRC were the cause of death in 11 and 5 patients, respectively. the second leading cause was liver failure (table 2). During follow up, 39 (19%) malignancies were diagnosed (table 3). Sixteen (41%) patients developed CRC and 15 (39%) CCA. Other malignancies included gallbladder cancer (n=2), pancreatic cancer (n=1), lymphoma (n=3), melanoma (n=1), and gastric cancer (n=1). All 16 CRC patients had concurrent IBD, although in one case IBD was diagnosed

Figure 1ab. Kaplan-Meier plot of the transplantation-free survival (+ 95% Confidence interval) for 211 patients with primary sclerosing cholangitis with (Figure 1a) or without (Figure 1b) a symptomatic presentation. A statistically significant difference was found between both groups (p=0.001, Log rank test)

proefschrift Marian.indd 38

10-10-2010 20:55:59

Malignancies in PSC | 39

six months after PSC had been diagnosed. Median interval between the diagnoses PSC and CRC was 8.3 years (range 0-18.2) and the IBD-CRC interval was 12.6 years (range 0-34.5). In patients with concurrent IBD the 10-year and 20-year risks for CRC were 14% and 31%, respectively, which was significantly higher than in patients without IBD (2.3% (p= 0.008)) (Figure 2). the estimated risk of CRC in the entire cohort of patients after 10 and 20 years was 9% and 22%, respectively. the majority of CRCs (63%) was located in the caecum and the ascending and transverse colon.

table 2. Causes of death in 45 patients with primary sclerosing cholangitis n=45 Liver failure Cholangiocarcinoma Colorectal carcinoma Other malignancies During/ within 30 days after liver transplantation Sepsis after colectomy Cardiac arrest Pulmonary embolism Unknown 13 11 5 4 4 4 2 1 1 (%) 29 24 11 9 9 9 4 2 2

Figure 2. Kaplan-Meier plot of the cumulative risk (+ 95% Confidence interval) of cholangiocarcinoma for 211 patients with primary sclerosing cholangitis

proefschrift Marian.indd 39

10-10-2010 20:56:00

40

| CHAPteR 3
Half of the patients who developed CRC did not participate in an endoscopic CRC surveillance program. Nineteen (9%) patients developed dysplasia, of which 12 underwent colectomy (63%). two of the 19 patients with low grade dysplasia underwent regular surveillance colonoscopy, whereas in 5 of the 19 patients dysplasia was found after colectomy had taken place for exacerbation of the colitis. the 10year and 20-year risk for dysplasia was 15% and 30% for patients with IBD, and 2.3% and 21% for patients without IBD at the time of diagnosis PSC, respectively (p=0.02).

table 3. Malignancies in 39 patients with primary sclerosing cholangitis n=39 Colorectal carcinoma Cholangiocarcinoma Non Hodgkin lymphoma Gall bladder cancer Pancreatic cancer Gastric cancer Melanoma 16 15 3 2 1 1 1 (%) 41 39 8 5 3 3 3

Figure 3. Kaplan-Meier plot of the cumulative risk (+ 95% Confidence interval) of colorectal cancer for 211 patients with primary sclerosing cholangitis with (- - -) or without () concurrent IBD. A statistically significant difference was found between both groups (p= 0.008, Log rank test)

proefschrift Marian.indd 40

10-10-2010 20:56:01

Malignancies in PSC | 41

Fifteen patients developed CCA. Median interval between the diagnosis PSC and CCA was 2.5 years (range 0-9.8 yrs). the estimated risk of CCA after 10 and 20 years was 9%. After excluding tumours diagnosed within six months of entry (n=4), the cumulative risk decreased to 7% (Figure 3). No significant differences in risk were found for patients with and without concurrent IBD (p= 0.386). the occurrence of CRC was significantly associated with the endpoint death (table 4). Other independent risk factors for this endpoint were occurrence of CCA, age at entry, and symptoms at presentation. Multivariate analysis including time-

table 4. Uni- and multivariate analysis including time-dependent variables for endpoint death, death or liver transplantation, CRC, or CCA, respectively, in 211 patients with primary sclerosing cholangitis endpoint Death Univariate Analysis Variable Colorectal carcinoma Cholangiocarcinoma Age Symptomatic presentation Bacterial cholangitis Comorbidity Variable Cholangiocarcinoma Bacterial cholangitis Age Symptomatic presentation Cholecystectomy extension IBD HR 4.9 198.2 1.1 2.2 3.0 4.6 HR 99.9 5.8 1.0 2.0 2.3 0.5 95% CI 2.2-10.8 77.3-508.5 1.0-1.1 1.2-4.0 1.6-5.6 1.6-13.3 95% CI 45.0-221.9 3.7-9.1 1.0-1.1 1.3-3.1 1.4-3.8 0.3-0.8 p-value 0.000 0.000 0.000 0.008 0.000 0.004 p-value 0.000 0.000 0.000 0.001 0.002 0.010 Multivariate Analysis HR 8.5 171.8 1.1 2.4 not in model not in model HR 43.2 3.9 1.0 not in model not in model not in model 95% CI 18.0-103.8 2.4-6.4 1.0-1.0 p-value 0.000 0.000 0.018 95% CI 3.2-22.7 57.1-517.4 1.0-1.1 1.2-5 p-value 0.000 0.000 0.000 0.016

endpoint Death or Liver transplantation

endpoint CrC Variable Concurrence of IBD extension of IBD Variable Age HR 8.6 3.4 HR 1.0 95% CI 1.1-64.8 1.3-9.0 95% CI 1.0-1.1 p-value 0.038 0.017 p-value HR not in model not in model HR 0.021 95% CI not in model p-value 95% CI p-value

endpoint CCa

HR: Hazard Ratio; 95% CI: 95% Confidence Interval

proefschrift Marian.indd 41

10-10-2010 20:56:02

42

| CHAPteR 3
dependent variables for the combined endpoint death or liver transplantation showed that age at entry, CCA, and bacterial cholangitis were independent risk factors. Cholecystectomy, serum bilirubin, hemoglobin level and symptoms of liver failure (ascites, encephalopathy, spontaneous bacterial peritonitis, hepatorenal syndrome) were also statistically significant in univariate analysis, but were excluded from the multivariate model because of the already known association between liver failure and liver transplantation. A similar analysis for the single endpoint CRC showed that concurrence and extent of IBD were of significant value (table 4). For the endpoint CCA, only age at presentation was a significant risk factor.

Discussion
this large and long-term cohort study showed that PSC-IBD patients have a high lifetime risk for CRC and that this risk is substantially higher than that for CCA. Furthermore, occurrence of CRC in these patients leads to reduced survival chances. Few and conflicting data on CRC risk in PSC have been reported. Loftus et al. 14 found an actuarial incidence at 10 years of only 4%, whereas Kornfeld et al. 24 reported a 10-year risk of 25%. the latter finding, however, was based on only five CRC cases. Comparison of our results with those of Broome et al. 12 is hampered by the fact that colorectal neoplasia, including dysplasia and CRC, was taken as study endpoint. If we would assume that colorectal dysplasia is the immediate precursor lesion of all IBD-associated CRCs, our study as well as the one by Broome et al. found a 20-year risk of developing CRC in PSC of approximately 31%. this percentage may be an underestimation as some of our patients underwent proctocolectomy when dysplasia was found during surveillance colonoscopy. Our high 20-year risk of CRC (31%) was significantly higher than the risk of 8% reported in a meta-analysis on CRC risk in IBD without PSC published in 2001 10. therefore, our observation provides further evidence that PSC promotes colorectal carcinogenesis in IBD 25-28. No new risk factors for CRC additional to the ones that are already known were found in the current study. Only concurrence and extent of IBD were found to be risk factors for developing CRC. three patients who did not have concurrent IBD at diagnosis PSC developed IBD and colorectal dysplasia, resulting in a 20-year dysplasia risk of 21% in patients without IBD at diagnosis PSC. the median interval between the diagnosis PSC and IBD in these three cases was 5.3 years (range 0.5-14.3 years). the 10-year and 20-year actuarial incidences of CCA (9% and 9%) were significantly lower than those for CRC. Furthermore, this risk of 9% after 10 years was lower than has been published previously 7. After excluding CCAs that were diagnosed within a six-month period after detection of PSC, the 10-year and 20-year risks were

proefschrift Marian.indd 42

10-10-2010 20:56:02

Malignancies in PSC | 43

7% and 7%, respectively. this is probably justified by our observations which make it likely that, in retrospect, these early CCAs probably were present at the time of the initial diagnostic evaluations, although we acknowledge that a period of six months is arbitrary. It was remarkable that despite the nearly equal absolute numbers of CRCs (n=16) and CCAs (n=15), the long-term risk was quite different. this is explained by the fact that nearly all CCAs presented within the first three years after PSC had been diagnosed. the majority of patients were not yet censored, thus still in follow-up at that time, which reduced the impact of a single CCA on the actuarial incidence. In contrast, the incidence of CRC was not limited to the first three years of follow-up. Our relatively low risk of CCA may be an underestimation, because autopsy was not performed in all cases dying from other causes. Besides age at presentation of PSC, no other independent risk factors for CCA were found. earlier studies reported an association between IBD and CCA 29,30. this may be due to the fact that the diagnosis PSC could not be detected accurately at a time when reliable non-invasive cholangiography was not available. Other, more recent studies did not find an association between IBD and CCA 31,32. this is the first study that demonstrates that occurrence of CRC in PSC patients reduces patient survival. Both the high risk of CRC and its impact on survival underline the suggested beneficial effect of colonic surveillance in PSC-IBD patients. Only half of the patients underwent surveillance colonoscopy according to a strict protocol. During the 1980s, the current accepted guidelines were not yet implemented which explains this relatively low number 22. this study did not have the correct design to evaluate the effect of surveillance on survival. In the multivariate analysis with the combined end-point liver transplantation and death (transplantation-free survival), CRC was not a statistically significant timedependent variable (table 4). An explanation for this may be that malignancy was a contraindication for liver transplantation within a period of five years after diagnosis. CCA remained significant in this analysis, due to the well-known nearly 100% fatal outcome of this malignancy 7,33,34. A high 10-year and 20-year risk, 42% and 57%, respectively, was observed for bacterial cholangitis. Most patients were diagnosed with PSC at a time that invasive endoscopic retrograde cholangiography, and not MRCP, was the main diagnostic method. However, considering the time intervals between diagnosis and the occurrence of bacterial cholangitis this seems not a satisfactory explanation for the incidence of this complication found in this study. Bacterial cholangitis was associated with the development of dominant bile duct stenoses in the majority of the patients (62%). this may be another explanation for the high risk of developing bacterial cholangitis. there are several limitations of this study including the lack of a control group. It is difficult to find a reliable control group that matches the study group because

proefschrift Marian.indd 43

10-10-2010 20:56:02

44

| CHAPteR 3
it is known that the course of IBD differs in patients with PSC from those without PSC 35,36. IBD in PSC patients is characterized by a high prevalence of pancolitis with rectal sparing, a mild and sometimes asymptomatic course and a lower colectomy rate. Secondly, the calculation of the cumulative risk of colorectal neoplasia, CRC and CCA is based on 30, 16 and 15 patients respectively. Nonetheless, as pointed out previously, this is still a far higher number than in other studies 12,24,36. Finally, all patients were treated in a tertiary referral hospital, which may have resulted in bias due to inclusion of patients with cancer risks differing from those in the general PSC population. In conclusion, PSC-IBD patients have a high risk of developing CRC over time and this has a negative impact on survival of these patients. therefore, it seems likely that this patient category in particular may benefit from colorectal surveillance endoscopy.

Acknowledgements
We thank R. Stellato, statistician, for her assistance with the analyses.

proefschrift Marian.indd 44

10-10-2010 20:56:02

Malignancies in PSC | 45

Reference List
1 Chapman RW, Arborgh BA, Rhodes JM, Summerfield JA, Dick R, Scheuer PJ, et al. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut 1980;21: 870-877. thorpe Me, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis, the biliary tree, and ulcerative colitis. Gut 1967;8: 435-448. Bergquist A, ekbom A, Olsson R, Kornfeldt D, Loof L, Danielsson A, et al. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol 2002;36: 321-327. Broome U, Olsson R, Loof L, Bodemar G, Hultcrantz R, Danielsson A, et al. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut 1996;38: 610-615. Miros M, Kerlin P, Walker N, Harper J, Lynch S, Strong R. Predicting cholangiocarcinoma in patients with primary sclerosing cholangitis before transplantation. Gut 1991;32:1369-1373. Nashan B, Schlitt HJ, tusch G, Oldhafer KJ, Ringe B, Wagner S, et al. Biliary malignancies in primary sclerosing cholangitis: timing for liver transplantation. Hepatology 1996;23: 1105-1111. Ahrendt SA, Pitt HA, Nakeeb A, Klein AS, Lillemoe KD, Kalloo AN, et al. Diagnosis and management of cholangiocarcinoma in primary sclerosing cholangitis. J Gastrointest Surg 1999;3: 357-367. Farges O, Malassagne B, Sebagh M, Bismuth H. Primary sclerosing cholangitis: liver transplantation or biliary surgery. Surgery 1995;117: 146-155. Kornfeld D, ekbom A, Ihre t. Survival and risk of cholangiocarcinoma in patients with primary sclerosing cholangitis. A population-based study. Scand J Gastroenterol 1997; 32: 1042-1045. eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48: 526-535. Fausa O, Schrumpf e, elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis 1991;11: 31-39. Broome U, Lofberg R, Veress B, eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995;22: 1404-1408. Gurbuz AK, Giardiello FM, Bayless tM. Colorectal neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Dis Colon Rectum 1995;38: 37-41. Loftus eV, Jr., Sandborn WJ, tremaine WJ, Mahoney DW, Zinsmeister AR, Offord KP, et al. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis. Gastroenterology 1996;110: 432-440. Nuako KW, Ahlquist DA, Sandborn WJ, Mahoney DW, Siems DM, Zinsmeister AR. Primary sclerosing cholangitis and colorectal carcinoma in patients with chronic ulcerative colitis: a case-control study. Cancer 1998;82: 822-826. MacCarty RL, LaRusso NF, Wiesner RH, Ludwig J. Primary sclerosing cholangitis: findings on cholangiography and pancreatography. Radiology 1983;149: 39-44. Ludwig J, Barham SS, LaRusso NF, elveback LR, Wiesner RH, McCall Jt. Morphologic features of chronic hepatitis associated with primary sclerosing cholangitis and chronic ulcerative colitis. Hepatology 1981;1: 632-640. Dilawari JB, Chawla YK. Pseudosclerosing cholangitis in extrahepatic portal venous obstruction. Gut 1992;33: 272-276. erkelens GW, Vleggaar FP, Lesterhuis W, van Buuren HR, van der Werf SD. Sclerosing pancreatocholangitis responsive to steroid therapy. Lancet 1999;354: 43-44. Gelbmann CM, Rummele P, Wimmer M, Hofstadter F, Gohlmann B, endlicher e, et al. Ischemic-like cholangiopathy with secondary sclerosing cholangitis in critically ill patients. Am J Gastroenterol 2007;102: 1221-1229. Riddell RH, Goldman H, Ransohoff DF, Appelman HD, Fenoglio CM, Haggitt RC, et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983;14: 931-968.

2 3 4 5 6 7 8 9 10 11 12 13 14 15

16 17

18 19 20

21

proefschrift Marian.indd 45

10-10-2010 20:56:02

46

| CHAPteR 3
22 Winawer S, Fletcher R, Rex D, Bond J, Burt R, Ferrucci J, et al. Colorectal cancer screening and surveillance: clinical guidelines and rationale-Update based on new evidence. Gastroenterology 2003;124: 544-560. 23 Parmar M, Machin D. Survival Analysis, a practical approach. Cambridge: John Wiley & Sons,1995. 24 Kornfeld D, ekbom A, Ihre t. Is there an excess risk for colorectal cancer in patients with ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut 1997;41: 522-525. 25 Brentnall tA, Haggitt RC, Rabinovitch PS, Kimmey MB, Bronner MP, Levine DS, et al. Risk and natural history of colonic neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis. Gastroenterology 1996;110: 331-338. 26 Broome U, Lindberg G, Lofberg R. Primary sclerosing cholangitis in ulcerative colitis--a risk factor for the development of dysplasia and DNA aneuploidy? Gastroenterology 1992;102: 1877-1880. 27 Marchesa P, Lashner BA, Lavery IC, Milsom J, Hull tL, Strong SA, et al. the risk of cancer and dysplasia among ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol 1997;92: 1285-1288. 28 DHaens GR, Lashner BA, Hanauer SB. Pericholangitis and sclerosing cholangitis are risk factors for dysplasia and cancer in ulcerative colitis. Am J Gastroenterol 1993;88: 1174-1178. 29 Mir-Madjlessi SH, Farmer RG, Sivak MV, Jr. Bile duct carcinoma in patients with ulcerative colitis. Relationship to sclerosing cholangitis: report of six cases and review of the literature. Dig Dis Sci 1987;32: 145-154. 30 Wee A, Ludwig J, Coffey RJ, Jr., LaRusso NF, Wiesner RH. Hepatobiliary carcinoma associated with primary sclerosing cholangitis and chronic ulcerative colitis. Hum Pathol 1985;16: 719-726. 31 Bergquist A, Glaumann H, Persson B, Broome U. Risk factors and clinical presentation of hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a case-control study. Hepatology 1998;27: 311-316. 32 Chalasani N, Baluyut A, Ismail A, Zaman A, Sood G, Ghalib R, et al. Cholangiocarcinoma in patients with primary sclerosing cholangitis: a multicenter case-control study. Hepatology 2000;31: 7-11. 33 Rosen CB, Nagorney DM, Wiesner RH, Coffey RJ, Jr., LaRusso NF. Cholangiocarcinoma complicating primary sclerosing cholangitis. Ann Surg 1991;213: :21-25. 34 Kaya M, de Groen PC, Angulo P, Nagorney DM, Gunderson LL, Gores GJ, et al. treatment of cholangiocarcinoma complicating primary sclerosing cholangitis: the Mayo Clinic experience. Am J Gastroenterol 2001;96: 1164-1169. 35 Broome U, Lofberg R, Lundqvist K, Veress B. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum 1995;38: 1301-1305. 36 Loftus eV, Jr., Harewood GC, Loftus CG, tremaine WJ, Harmsen WS, Zinsmeister AR, et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005;54: 91-96.

proefschrift Marian.indd 46

10-10-2010 20:56:03

CHaPter 4
More right-sided IBD-associated colorectal cancer in patients with primary sclerosing cholangitis

MMH Claessen1, MWMD Lutgens1, HR van Buuren2, B Oldenburg1, PCF Stokkers3, CJ van der Woude2, DW Hommes4, DJ de Jong5, G Dijkstra6, AA van Bodegraven7, PD Siersema1, FP Vleggaar1

1 2

Department of Gastroenterology and Hepatology, University Medical Center Utrecht,

Department of Gastroenterology and Hepatology, erasmus MC University Medical Center Rotterdam, 3Department of Gastroenterology and Hepatology, Academic Medical Center

Amsterdam, 4Department of Gastroenterology and Hepatology, Leiden University Medical Center, 5Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Center, 6Department of Gastroenterology and Hepatology, University Medical Center Groningen, 7Department of Gastroenterology and Hepatology, VU University Medical Center Amsterdam Inflammatory Bowel Diseases, 2009; 15: 1331-1336

proefschrift Marian.indd 47

10-10-2010 20:56:05

48

| CHAPteR 4 Abstract
Background Patients with inflammatory bowel disease (IBD) and concurrent primary sclerosing cholangitis (PSC) have a higher risk of developing colorectal cancer (CRC) than IBD patients without PSC. aims & Methods the aim of this study was to investigate potential clinical differences between patients with CRC in IBD and those with CRC in IBD and PSC, as this may lead to improved knowledge of underlying patho-physiological mechanisms of CRC development. A retrospective study was performed, from 1980-2006 involving 7 Dutch university medical centres. Clinical data were retrieved from cases identified using the national pathology database (PALGA). results 27 IBD-CRC patients with PSC (70% male) and 127 IBD-CRC patients without PSC (59% male) were included. CRC-related mortality was not different between groups (30% vs. 19%, p=0.32), however, survival for cases with PSC after diagnosing CRC was lower (5-year survival: 40% vs. 75% p=<0.001). Right-sided tumours were more prevalent in the PSC group (67% vs. 36%, p=0.006); adjusted for age, sex and extent of IBD, this difference remained significant (Odds Ratio: 4.8 (95% CI 2.0-11.8). In addition, tumours in individuals with PSC were significantly more advanced. Conclusions the right colon is the predilection site for development of colonic malignancies in patients with PSC and IBD. When such patients are diagnosed with cancer they tend to have more advanced tumours than patients with IBD without concurrent PSC, and the overall prognosis is worse. Furthermore, the higher frequency of rightsided tumours in patients with PSC suggests a different pathogenesis between patients with PSC and IBD and those with IBD alone.

proefschrift Marian.indd 48

10-10-2010 20:56:05

Right-sided IBD-associated CRC

49

Introduction
Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disorder, characterized by inflammation and fibrosis of the intra- and extrahepatic bile ducts 1,2. Up to 90% of the patients with PSC have inflammatory bowel disease (IBD) as well, usually ulcerative colitis, while IBD is accompanied by PSC in only 5% of IBD patients 3. Patients with IBD have an increased risk of developing colorectal cancer (CRC). Recent meta-analyses show 10-year and 20-year risks of 1-2% and 4-8%, respectively 4,5 Several studies have shown that in patients with IBD concurrent PSC increases the risk of developing colitis-associated CRC 6,7. Our group found 10-year and 20-year risks of 14% and 31%, respectively, of developing CRC in such individuals 8. An explanation for this high CRC risk is not available. Several hypotheses have been postulated, such as an abnormal bile acid composition in patients with PSC resulting in an increased concentration of secondary bile acids which may have a carcinogenic effect on the colonic mucosa 9,10. Another explanation may be the altered course of IBD in PSC patients. A more extensive colitis with at the same time a mild or even asymptomatic course, resulting in diagnostic delay and a lower colectomy rate, may contribute to an increased risk of developing CRC 11. the aim of this study was to compare patients with CRC and IBD with and without PSC with respect to clinical characteristics, endoscopic and histological findings, and prognosis. Potentially, the results may help to define improved clinical management strategies for individuals with PSC and IBD and guide future research in patho-physiological mechanisms in PSC-IBD-associated CRC.

Methods
Patients the Dutch nationwide network and registry of histo-and cytopathology reports (PALGA) was used to select patients with IBD-associated CRC 12. A search in this database from January 1990 until June 2006, for cases diagnosed, in Dutch university medical centres, with synchronous or metachronous diagnoses of IBD and CRC was performed 13. Patients with PSC-IBD and CRC were also selected from this search and from a long-term cohort study containing all PSC patients followed in the period 1980-2006 in 2 university medical centres (University Medical Center Utrecht and erasmus Medical Center Rotterdam) 8. the diagnosis PSC was based on typical findings on cholangiography 14 or the presence of characteristic histological liver abnormalities (pericholangiolar onionskin fibrosis), in combination with elevated serum alkaline phosphatase levels and

proefschrift Marian.indd 49

10-10-2010 20:56:05

50

| CHAPteR 4
the presence of IBD 15. the onset of PSC was designated as the date of the first abnormalities on cholangiography or liver histology. Patients with bile duct abnormalities attributable to other causes such as bile duct surgery, ischemia, portal vein thrombosis, or autoimmune pancreatico-cholangitis were excluded. the diagnoses IBD and CRC were based on a combination of endoscopic and histological findings 16. Patients with mere dysplasia were not eligible. Colonic surveillance was based on the AGA or BSG guidelines and defined as surveillance after 8-10 years of disease duration in cases of Crohns disease or extensive ulcerative colitis, after 15-20 years of disease duration in cases of left-sided ulcerative colitis, and directly after diagnosing PSC 17,18. Data collection Data were retrieved from medical records and computerized hospital databases and completed by contacting the patients physician. Collected data included date of diagnosis of IBD, PSC, and CRC, time of onset of symptoms, medical history, location and stage of the CRC, cause of death, use of 5-aminosalicylic acid (5-ASA) and/or ursodeoxycholic acid (UCDA), and information on phenotype of IBD (extent, duration, and type). extensive colitis was defined as a colitis proximal of the splenic flexure in patients with UC or >50% involved colonic mucosa in patients with Crohns disease. end of follow-up was the end of the study (June 2006), death, or time of last visit in case patients were lost-to-follow-up. Apart from location and stage of the CRC, information on synchronous and metachronous tumours was collected as well. Right-sided or proximal tumours were defined as tumours located proximally of the splenic flexure. tumour stages were defined according to the American Joint Committee on Cancer (AJCC) 19. Statistical analysis Follow-up started at the time when CRC was diagnosed. Differences between patients with PSC and IBD and those with IBD alone were analysed using Students t-test and the Chi square test, when appropriate. A separate analysis was performed, excluding the patients with Crohns disease to rule out that possible differences were based on the underlying type of inflammatory bowel disease. Multivariate analysis was done using binary logistic regression to determine whether there were independent risk factors for tumour location. these variables included age at diagnosis CRC, gender, presence of PSC, and extent of colitis. Survival analyses were performed using the Kaplan-Meier method; differences between groups were analysed with the log-rank test. SPSS 12.0 Statistical Software (Chicago, USA) was used for all statistical analyses. the Kaplan-Meier plots were created in MedCalc for Windows, version 9.3. (MedCalc Software, Mariakerke, Belgium). P-values <0.05 were considered statistically significant.

proefschrift Marian.indd 50

10-10-2010 20:56:05

Right-sided IBD-associated CRC

51

Results
Clinical characteristics twenty-seven IBD-CRC patients with PSC (70% male) and 127 IBD-CRC patients without PSC (60% male) were included. Median age at diagnosis of IBD was 33 years (range 3-46) and 28 years (range 6-82) in the groups with and without concurrent PSC, respectively (p=0.5) (table 1). Median age at diagnosis of PSC was 39 years (range 11-59). the majority of patients had extensive colitis, i.e., 89% and 71% of patients with and without PSC, respectively (p=0.1). Ulcerative colitis (UC) was diagnosed in 89% of the patients with PSC, and in 56% of the patients without PSC (p=0.003). All patients with Crohns disease had colonic involvement, an extensive colitis (>50% inflammation) was found in 100% and 63% of the patients with and without PSC, respectively. Sixty-nine percent (n=31) of the non-PSC patients with Crohns disease also had ileitis compared to 33% (n=1) of the Crohns patients in the PSC group. the majority (86%) of all patients used 5-ASA. UDCA was used by 15 PSC patients (56%) and by 1 patient without PSC. the latter patient used it for cholelithiasis. After excluding all patients with Crohns disease 24 UC-CRC patients with PSC (67% male) and 72 UC-CRC patients without PSC (67% male) remained. No changes in age at diagnosis IBD or PSC were found. Almost all patients in the PSC-UC-CRC group had an extensive colitis (92%) compared to 79% in the UC-CRC group (p=0.2). Follow-up the median follow-up period after diagnosing CRC was 2.3 years (range 0-17.3) and 2.7 years (0-19.5) for the PSC and non-PSC group, respectively (p=0.09). Sixty

table 1. Patient characteristics of 27 PSC-IBD-CRC patients and 127 IBD-CRC patients PSC-IBD-CRC (n=27) Gender, (% male) Median age at diagnosis IBD, yrs (range) Median age at diagnosis PSC, yrs (range) Median age at diagnosis CRC, yrs (range) extent of IBD (%), (extensive) type of IBD (%), (UC) Median IBD-CRC interval, yrs (range) 19 (70) 33 (3-46) 39 (11-59) 47 (27-67) 24 (89) 24 (89) 13 (0-35) IBD-CRC (n=127) 75 (59) 28 (6-82) na 49 (20-84) 80 (63 71 (56) 17 (0-43) 0.4 0.5 na 0.06 0.1 0.003 0.2 p-value

PSC: primary sclerosing cholangitis, IBD: inflammatory bowel disease, CRC: colorectal cancer, UC: ulcerative colitis, na: not applicable

proefschrift Marian.indd 51

10-10-2010 20:56:05

52

| CHAPteR 4
percent (n=16) of the patients in the PSC group died versus 26% (n=33) in the nonPSC group (p=0.002). CRC-related mortality was not different between the two groups, i.e., 30% and 19% in the PSC-IBD-CRC and IBD-CRC group (p= 0.324). Other causes of death are shown in table 2. twenty-one percent of all patients were lost-to-follow-up. Differences in mortality rates remained statistically significant after excluding the Crohns disease patients (63% and 23% in the PSC-UC-CRC patients and UC-CRC patients, respectively). In the PSC-UC-CRC group 47% of the deaths were CRC-related versus 81% in the UC-CRC group (p= 0.044) three patients with PSC had undergone liver transplantation before developing CRC. these patients were not enrolled in a surveillance program, but underwent colonoscopy at an irregular basis without taking the required number of biopsies. the median interval between liver transplantation and detection of CRC was 13 months (range 2-20). Liver transplantation following a prior diagnosis of CRC was not performed. the estimated overall 5-year survival rate after the diagnosis CRC was lower for patients with PSC (40%) than for those without PSC (75%, p=<0.001) (Figure 1ab). CRC-related mortality was not statistically significant different, with a 5-year survival of 62% and 82% for patients with and without PSC, respectively (p= 0.1) (Figure 2ab). Colorectal cancer Patients were younger at CRC diagnosis in the PSC group than in the non-PSC group (median age: 47 years (range 27-67) vs. 49 years (range 20-84), p=0.064). the median interval between the diagnosis IBD and the development of CRC was not different, i.e., 13 years (range 0-35) in the PSC group and 18 years (range 0-43) in the non-PSC

table 2. Causes of death in 16 PSC-IBD-CRC patients and 32 IBD-CRC patients PSC-IBD-CRC n= 16 (%) Colorectal cancer Cholangiocarcinoma Other malignancy Colectomy Liver failure Other causes Unknown 8 2 0 2 1 1** 2 (50) (13) (0) (13) (6) (6) (13) 0 3* 3 0 2*** 0 IBD-CRC n= 32 (%) 24 (75) (0) (9) (9) (0) (6) (0)

* gastric, urothelial, and renal cell cancer, ** ileus, *** sepsis and gastrointestinal bleeding of unknown cause, IBD: inflammatory bowel disease, PSC: primary sclerosing cholangitis, CRC: colorectal cancer

proefschrift Marian.indd 52

10-10-2010 20:56:05

Right-sided IBD-associated CRC

53

group (p=0.2). Patients with PSC had significantly more tumours with an AJCC tumourstage of 3A or higher compared to patients with IBD alone (61.5% and 38.5%, p=0.003) (table 3). eleven patients (9%) in the non-PSC group had a synchronous CRC and three (2.4%) patients had even 3 tumours diagnosed at the same time. Synchronous tumours were not detected in the PSC group. No statistically significant differences were found in the AJCC tumour stages when comparing PSC-UC-CRC

Figure 1 ab. Kaplan-Meier plot of survival (+95% confidence interval) after diagnosing CRC of 27 patients with primary sclerosing cholangitis and concurrent inflammatory bowel disease (Figure 1a) and 127 patients with inflammatory bowel disease alone (Figure 1b) (p= 0.002, Log rank test).

Figure 2 ab. Kaplan-Meier plot of the CRC-related mortality (+95% confidence interval) for 27 patients with primary sclerosing cholangitis and concurrent inflammatory bowel disease (Figure 2a) and 127 patients with inflammatory bowel disease alone (Figure 2b) (p= 0.324, Log rank test).

proefschrift Marian.indd 53

10-10-2010 20:56:13

54

| CHAPteR 4
table 3. AJCC tumour stages AJCC 0 1 2A 2B 3A** 3B** 3C** 4** tumour Stage t in situ t1, t2. N0, M0 t3, N0, M0 t4, N0, M0 t1, t2, N1, M0 t3, t4, N1, M0 Any t, N2, M0 M1 PSC-IBD-CRC n= 26 (%)* 2 4 4 0 1 5 2 8 (8) (15) (15) (0) (4) (19) (8) (31) IBD-CRC n= 122 (%)* 7 26 40 2 5 14 10 18 (6) (21) (33) (2) (4) (12) (8) (15)

* 6 tumours (1 in PSC-IBD-CRC group, 5 in IBD-CRC group) were not classifiable due to unknown tumour stages, ** PSC-IBD-CRC patients had statistically significant more advanced tumours (tumour stage: 3A, 3B, 3C, or 4) compared to the IBD-CRC group (p= 0.03).

to UC-CRC patients (tumour stage of 3A or more: 61% of the PSC-UC-CRC patients and in 41% of the UC-CRC patients, respectively, p= 0.091) the tumours were located proximally of the splenic flexure in 18 (67%) patients with PSC and in 52 (36%) patients without PSC (p=0.006). After excluding patients with Crohns disease this difference remained statistically significant (p=0.020). Multivariate analysis, including age at diagnosis of CRC, gender and extent of colitis showed that PSC was an independent risk factor for (right-sided) tumour location with an Odds Ratio of 4.8 (95% CI 2.0-11.8). Only a small number of patients underwent colonic surveillance according to the AGA/ BSG guidelines, i.e. 33% and 17% patients with and without PSC, respectively. No difference in AJCC tumour stage was found between patients with or without colonic surveillance (p= 0.128).

Discussion
this study shows a significantly higher prevalence of right-sided CRCs in patients with PSC and IBD compared to patients with IBD alone. these findings are in line with results from previous series. However, the studies of Marchesa et al. 20, Lindberg et al. 21, and Shetty et al. 10 were all based on relatively small numbers of CRC patients with IBD with or without concomitant PSC. In addition, we found that CRC in patients with PSC and IBD was associated with a decreased survival compared to patients with IBD alone. the difference in CRC-related 5-year survival (62% vs 82%) between the two groups, however, was not statistically significant.

proefschrift Marian.indd 54

10-10-2010 20:56:13

Right-sided IBD-associated CRC

55

the high frequency of right-sided colorectal tumours in PSC patients fits well with the hypothesis that an altered bile acid composition is implicated in the increased frequency of CRCs in these patients 10. In this respect, an increased concentration of secondary bile acids and higher serum and bile levels of deoxycholic acid and lithocholic acid both might play a role 22-24. Interestingly, specific binding sites for deoxycholic acid were identified in 31% of the sporadic CRCs compared to only 3% of normal mucosa samples (p<0.05) 25. the same has been observed in colitisassociated carcinogenesis by Hill et al. who reported an increased concentration of total fecal bile acids in UC patients with dysplasia or carcinoma as well. 9 the promising results of UDCA as chemopreventive agent in the development of CRC in UC patients with concurrent PSC also suggest a role for bile acids in colorectal carcinogenesis 26,27. On the other hand, the sparse data available on bile composition in patients with PSC do not corroborate this hypothesis: bile acid excretion in cholestatic patients, including PSC patients, is decreased and the serum concentration of secondary bile acids was not increased in patients with PSC when compared to healthy controls 28,31,32. In addition, the risk of developing CRC remains increased after liver transplantation, which is likely to normalize bile acid composition 33. Another explanation for the predominance of right-sided CRCs in PSC may be offered by an alternative molecular pathogenesis 34,35. A parallel might be drawn with patients with Lynch syndrome in whom a high frequency of right-sided tumours occurring at a relatively young age is observed as well 36. the pathophysiological mechanism of this hereditary disease is based on a defect in the DNA mismatch repair genes. In colitis-associated carcinogenesis, data regarding defects in DNA mismatch repair genes and the associated molecular phenotype of microsatellite instability (MSI) are conflicting. the frequency of MSI varies between 1% and 45%, depending on the types of tissue analysed and, in particular, the number and choice of microsatellite markers 37-41. the frequency of MSI in patients with IBD-associated CRC and concurrent PSC is presently unknown. A remarkable observation of our study was that the IBD-CRC interval between patients with and without PSC did not differ. Although PSC is known to be an additional risk factor for CRC, our results do not support an accelerated progression to CRC in these patients 6-8. However, as many PSC patients seem to have asymptomatic or mild colitis, the exact date of onset of IBD is difficult to establish 11,42. this might have led to an underestimation of the duration of the colitis. A minority of patients (20%) were enrolled in a strict surveillance colonoscopy program. During the 1980s and 1990s, the currently accepted guidelines were not yet implemented, which might explain this low percentage 17,18. In addition, the fact that three patients developed CRC relatively shortly after liver transplantation, also illustrates that the current standard of surveillance before and after liver transplantation was not yet introduced at that time.

proefschrift Marian.indd 55

10-10-2010 20:56:13

56

| CHAPteR 4
Patients with PSC had a decreased survival in comparison with the non-PSC group, although CRC-related mortality did not differ. Most patients in both groups died from CRC. Although statistically not significant, the difference in 5-year survival rate of 62% and 82% for CRC patients with and without PSC, respectively, is noteworthy. the observed higher frequency of more advanced AJCC tumour stages in the PSC group may explain this lower survival rate. the lack of significance may be ascribed to the relatively small patient population (type II error). Several limitations of this study are worth considering. First of all, data were collected retrospectively. Second, the controls were not case-matched. Due to the asymptomatic course of IBD in patients with PSC it is difficult to match patients with regard to IBD duration and extent. In the multivariate analysis, we corrected for the extent of the colitis as potential confounder. Finally, in the IBD-CRC group more patients with Crohns disease were included compared to the PSC-CRC group, which may have affected the results. However, all patients with Crohns disease had colonic involvement and after excluding the patients with Crohns disease from analysis the differences remained statistically significant. In summary, our study shows that CRCs develop predominantly in the proximal colon of PSC patients with IBD. Additional studies are needed to explore the underlying molecular mechanisms to explain these findings.

Acknowledgements
the authors would like to thank dr. M. Casparie (Prismant, Utrecht, the Netherlands) for help with the PALGA data search.

proefschrift Marian.indd 56

10-10-2010 20:56:13

Right-sided IBD-associated CRC

57

Reference List
1 2 3 4 5 6 7 8 9 10 11 12 Chapman RW, Arborgh BA, Rhodes JM, et al. Primary sclerosing cholangitis: a review of its clinical features, cholangiography, and hepatic histology. Gut. 1980; 21:870-7. thorpe Me, Scheuer PJ, Sherlock S. Primary sclerosing cholangitis, the biliary tree, and ulcerative colitis. Gut. 1967;8:435-48. Fausa O, Schrumpf e, elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis. 1991;11:31-9. eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut. 2001;48:526-35. M.W.Lutgens, G.J.van der Heijden, F.P.Vleggaar, et al. A comprehensive meta-analysis of the risk of colorectal cancer in ulcerative colitis and Crohns disease. Gastroenterology. 2008;134, A-33-A-34. Broome U, Lofberg R, Veress B, et al. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology. 1995;22:1404-8. Kornfeld D, ekbom A, Ihre t. Is there an excess risk for colorectal cancer in patients with ulcerative colitis and concomitant primary sclerosing cholangitis? A population based study. Gut. 1997;41:522-5. Claessen MMH, Vleggaar FP, tytgat KMAJ, et al. High life-time risk of cancer in primary sclerosing cholangitis. J Hepatol. 2008;50:158-64. Hill MJ, Melville DM, Lennard-Jones Je, et al. Faecal bile acids, dysplasia, and carcinoma in ulcerative colitis. Lancet. 1987;25:185-6. Shetty K, Rybicki L, Brzezinski A, et al. the risk for cancer or dysplasia in ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol. 1999;94:1643-9. Loftus eV, Jr., Harewood GC, Loftus CG, et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut. 2005;54:91-6. Casparie M, tiebosch At, Burger G, et al. Pathology databanking and biobanking in the Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive. Cell Oncol. 2007;29:19-24. Lutgens MW, Vleggaar FP, Schipper Me, et al.High Frequency of early Colorectal Cancer in Inflammatory Bowel Disease. Gut. 2008;57:1246-51 MacCarty RL, LaRusso NF, Wiesner RH, et al. Primary sclerosing cholangitis: findings on cholangiography and pancreatography. Radiology. 1983;149:39-44. Ludwig J, Barham SS, LaRusso NF, et al. Morphologic features of chronic hepatitis associated with primary sclerosing cholangitis and chronic ulcerative colitis. Hepatology. 1981;1:632-40. Riddell RH, Goldman H, Ransohoff DF, et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol. 1983;14:931-68. eaden JA, Mayberry JF. Guidelines for screening and surveillance of asymptomatic colorectal cancer in patients with inflammatory bowel disease. Gut. 2002;51 Suppl 5:V10-V12. Winawer S, Fletcher R, Rex D, et al. Colorectal cancer screening and surveillance: clinical guidelines and rationale-Update based on new evidence. Gastroenterology. 2003;124:544-60. AJCC cancer staging manual. 6th ed. New York (NY): 2002. Marchesa P, Lashner BA, Lavery IC, et al. the risk of cancer and dysplasia among ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol. 1997;92:1285-8. Lindberg BU, Broome U, Persson B. Proximal colorectal dysplasia or cancer in ulcerative colitis. the impact of primary sclerosing cholangitis and sulfasalazine: results from a 20-year surveillance study. Dis Colon Rectum. 2001;44:77-85. Nagengast FM, Grubben MJ, van Munster, I. Role of bile acids in colorectal carcinogenesis. eur J Cancer. 1995;31A:1067-70. Bayerdorffer e, Mannes GA, Ochsenkuhn t, et al. Unconjugated secondary bile acids in the serum of patients with colorectal adenomas. Gut. 1995;36:268-73.

13 14 15 16 17 18 19 20 21

22 23

proefschrift Marian.indd 57

10-10-2010 20:56:13

58

| CHAPteR 4
24 Kishida t, taguchi F, Feng L, et al. Analysis of bile acids in colon residual liquid or fecal material in patients with colorectal neoplasia and control subjects. J Gastroenterol. 1997;32:306-11. 25 Summerton J, Flynn M, Cooke t, et al. Bile acid receptors in colorectal cancer. Br J Surg. 1983;70:549-51. 26 Pardi DS, Loftus eV, Jr., Kremers WK, et al. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology. 2003;124:889-93. 27 tung BY, emond MJ, Haggitt RC, et al. Ursodiol use is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Ann Intern Med. 2001;16:89-95. 28 Jazrawi RP, de Caestecker JS, Goggin PM, et al. Kinetics of hepatic bile acid handling in cholestatic liver disease: effect of ursodeoxycholic acid. Gastroenterology. 1994;106:134-42. 29 Paumgartner G, Beuers U. Ursodeoxycholic acid in cholestatic liver disease: mechanisms of action and therapeutic use revisited. Hepatology. 2002;36:525-31. 30 Rost D, Rudolph G, Kloeters-Plachky P, et al. effect of high-dose ursodeoxycholic acid on its biliary enrichment in primary sclerosing cholangitis. Hepatology. 2004;40:693-8. 31 Stiehl A, Rudolph G, Sauer P, et al. Biliary secretion of bile acids and lipids in primary sclerosing cholangitis. Influence of cholestasis and effect of ursodeoxycholic acid treatment. J Hepatol. 1995;23:283-9. 32 Beuers U, Spengler U, Zwiebel FM, et al. effect of ursodeoxycholic acid on the kinetics of the major hydrophobic bile acids in health and in chronic cholestatic liver disease. Hepatology. 1992;5:603-8. 33 Loftus eV, Jr., Aguilar HI, Sandborn WJ, et al. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis following orthotopic liver transplantation. Hepatology. 1998;27:685-90. 34 Gervaz P, Bouzourene H, Cerottini JP, et al. Dukes B colorectal cancer: distinct genetic categories and clinical outcome based on proximal or distal tumor location. Dis Colon Rectum. 2001;44:364-72. 35 Sugai t, Habano W, Jiao YF, et al. Analysis of molecular alterations in left- and right-sided colorectal carcinomas reveals distinct pathways of carcinogenesis: proposal for new molecular profile of colorectal carcinomas. J Mol Diagn. 2006;8:193-201. 36 Vasen HF, Watson P, Mecklin JP, et al. New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology. 1999;116:1453-6. 37 Brentnall tA, Crispin DA, Bronner MP, et al. Microsatellite instability in nonneoplastic mucosa from patients with chronic ulcerative colitis. Cancer Res. 1996;15:1237-40. 38 Itzkowitz SH. Microsatellite instability in colitis associated colorectal cancer. Gut. 2000;46:304-5. 39 Lyda MH, Noffsinger A, Belli J, et al. Microsatellite instability and K-ras mutations in patients with ulcerative colitis. Hum Pathol. 2000;31:665-71. 40 Suzuki H, Harpaz N, tarmin L, et al.Microsatellite instability in ulcerative colitis-associated colorectal dysplasias and cancers. Cancer Res. 1994;15:4841-4. 41 Willenbucher RF, Aust De, Chang CG, et al. Genomic instability is an early event during the progression pathway of ulcerative-colitis-related neoplasia. Am J Pathol. 1999;154:1825-30. 42 Broome U, Lofberg R, Lundqvist K, et al. Subclinical time span of inflammatory bowel disease in patients with primary sclerosing cholangitis. Dis Colon Rectum. 1995;38:1301-5.

proefschrift Marian.indd 58

10-10-2010 20:56:14

CHaPter 5
Microsatellite instability in patients with primary sclerosing cholangitis and colitis-associated colorectal cancer

MMH Claessen1, FP Vleggaar1, MeI Schipper2, FHM Morsink2, JWJ Hinrichs2, WtM Blokland2, PD Siersema1, GJA Offerhaus2
1

Department of Gastroenterology and Hepatology, Univeristy Medical Center Utrecht,


2

Department of Pathology, Univeristy Medical Center Utrecht Submitted to British journal of Cancer

proefschrift Marian.indd 59

10-10-2010 20:56:16

60

| CHAPteR 5 Abstract
Background Proximal colorectal cancer (CRC) may develop through different molecular mechanisms compared to distal tumours and regularly displays microsatellite instability (MSI). Proximal CRC is more frequently found in patients with primary sclerosing cholangitis (PSC) and inflammatory bowel disease (IBD) than in patients with IBD alone. aim & Methods the aim of this study was to compare MSI status of PSC-IBD-related CRC with other subtypes of CRCs. therefore, a tissue micro-array was constructed with colonic samples from 7 groups: (1) healthy subjects (n=17), (2) IBD (n=15), (3) IBD-related CRC (n=17), (4) PSC-IBDrelated CRC (n=10), (5) Lynch syndrome-related CRC (n=8), (6) sporadic left-sided CRC (n=16), and (7) sporadic right-sided CRC (n=19). Immunohistochemistry was performed using antibodies for MLH1, MSH2, MSH6, and PMS2. Staining was scored positive or negative. tumour and normal DNA were isolated to examine MSI status using 6 microsatellite markers. tumours were classified MSI-High if 2 or more markers showed instability. results Median age of all patients was 55 years (range 12-92), with 61% being male. Of all IBD patients (groups 2-4), ulcerative colitis was present in 53% of cases. In groups 3-5, 29%, 60%, and 50% of CRCs, respectively, were proximally located, (p=0.3). In 55% of all tumours, CRC had metastasized. MSI-High was found in 0%, 0%, 12%, 0%, 88%, 0% and 32% of patients in groups 1-7, respectively. MLH1 and PMS2 were negative in 67% and 73% of the MSI-High cases, respectively, whereas 20% and 27% had no MSH2 and MSH6 expression. Conclusions MSI does not seem to be a major factor in colorectal carcinogenesis in patients with PSC and IBD, despite a relatively high frequency of proximal tumours in these patients.

proefschrift Marian.indd 60

10-10-2010 20:56:16

MSI-status in PSC-IBD-related CRC | 61

Introduction
Patients with inflammatory bowel disease (IBD) have an increased risk of developing colorectal cancer (CRC). Recent meta-analyses have shown 10-year and 20-year risks of 1-2% and 4-8%, respectively 1,2. Five percent of the patients with IBD also develop primary sclerosing cholangitis (PSC) 3. PSC is a chronic cholestatic liver disease, characterized by inflammation and fibrosis of the intra- and extra hepatic bile ducts. Patients with IBD and concurrent PSC have an even higher risk of developing CRC, with 10-year and 20-year risks of 14% and 31%, respectively 4. there is no clear explanation for the increased risk of CRC in IBD with concurrent PSC. It has been suggested that IBD-related factors contribute to a higher risk of developing CRC. For example, pancolitis with an asymptomatic or mild course is frequently observed in patients with IBD and concurrent PSC, possibly resulting in a diagnostic delay of the diagnosis IBD and a lower colectomy rate 5. Another more PSC-related hypothesis suggests that an altered bile composition might have a carcinogenic effect on the colonic mucosa 6. A recent study by our group has suggested that patients with PSC and concurrent IBD more frequently have right-sided CRC compared to patients with IBD alone 7. A predominance for the proximal colon as tumour location is also found in Lynch syndrome-related CRC. Lynch syndrome is characterized by an increased risk of developing gastrointestinal and extra-intestinal tumours. It is caused by a germline mutation in one of several DNA MisMatch Repair (MMR) genes 8. the role of the DNA MMR system is to maintain genomic integrity by correcting base substitution mismatches and small insertion-deletion mismatches that are generated by errors in base pairing during DNA replication. Inactivation of the MMR genes causes genomic instability as a result of failure to repair the DNA mismatches that occur commonly during normal DNA synthesis. this genomic instability will lead to a specific phenotype known as microsatellite instability (MSI). About 20% of sporadic CRCs also show MSI, with a high prevalence in the proximal colon 9,10. Instead of a germline mutation, MSI arises through hypermethylation of the promoter region of a MMR gene causing transcriptional silencing 11. the role of MSI was determined in PSC-IBD-related tumours in order to elucidate the molecular background of these tumours. MSI-status of PSC-IBD-related tumours was compared to other subtypes of CRCs.

proefschrift Marian.indd 61

10-10-2010 20:56:16

62

| CHAPteR 5 Methods
Patients and tissue samples Paraffin embedded tissue specimens from surgical resection material were obtained from the Department of Pathology, University Medical Center (UMC) Utrecht, the Netherlands. the study was carried out with approval of and in accordance with the ethical guidelines of the research review committee of our institution. None of the patients had received chemotherapy or radiotherapy prior to surgery. tissue samples of seven different patient groups were collected. Group 1 (C) consisted of healthy controls who underwent sigmoid resection for symptomatic diverticular disease (n=17). Histological examination only showed diverticular disease. In group 2 (IBD) IBD-patients with chronic actively inflamed colonic mucosa were included (n=15). Group 3 (IBD-CRC) consisted of patients with IBDrelated CRC (n=17) and group 4 of patients with PSC-IBD-related CRC (PSC-CRC; n=10). In group 5, patients with Lynch syndrome-related CRC were included (LYNCH-CRC; n=8), with all patients having a confirmed mutation in one of the MMR genes. Group 6 (CRC-LeFt; n=16) consisted of left-sided sporadic CRC, defined as a tumour located distal to the splenic flexure, whereas in group 7 (CRCRIGHt; n=19) patients with sporadic CRC located proximal to the splenic flexure were included. except for the LYNCH-CRC group, no other patient had a family history of colorectal, gastric, or endometrial cancer suggestive of Lynch syndrome. All inflammation-related tumours developed in a background of chronic inflammation. the histological criteria by Riddell et al. for colitis-associated carcinoma were applied 12. Since colorectal carcinogenesis has not been shown to be different between Crohns disease (CD) and ulcerative colitis (UC), specimens from patients with either disease were included in this study 13. tumour stage was defined according the American Joint Committee on Cancer (AJCC) 14. tissue Micro array (tMa) A tMA was constructed according to a standard protocol described by Kononen et al 15. Specific tumour parts were selected on corresponding haematoxylin and eosin (H&e) stained slides after which at least three core biopsies (diameter 0.6 mm) were taken from each sample and punched into a recipient block using a tissue microarrayer (MtA-I, Beecher Instruments Inc ,Sun Prairie, USA). the slides were used for immunohistochemistry. DNa extraction for MSI DNA was isolated from paraffin embedded tissue. the H&e slides were used to select specific tumour regions. the tissue was deparaffinated with xylene for

proefschrift Marian.indd 62

10-10-2010 20:56:16

MSI-status in PSC-IBD-related CRC | 63

10 minutes followed by ethanol 100% and 70%, respectively. enriched tumour tissue (>75% tumour cells) was used to obtain DNA using 20 l of proteinase K (10 mg/ml) in 100 l buffer (0.5% tween 20, 50 mmol/ L tris (pH=9), 1mmol/L NaCl, 2 mmol/L eDtA). In order to isolate DNA from normal mucosa, non-neoplastic mucosa, that was not adjacent to the tumour site, was selected. In case of inflammationassociated tumours the normal mucosa contained no inflammation. No DNA tests were performed on group 1 (C). After overnight incubation at 56 C, tubes were incubated at 95 C for 10 minutes to inactivate the proteinase K and cooled on ice afterwards. the supernatant was transferred to a second tube after centrifugation. Samples were stored at -20C. Microsatellite analysis MSI was evaluated in genomic DNA extracted from microdissected tumour tissue and non-neoplastic mucosa. A panel of six fluorescence-labelled microsatellite markers was evaluated: 3 mononucleotide repeats BAt25, BAt26, and BAt 40; 3 dinucleotide repeats D17S250, D5S346, and D2S123 16. the primer sets used to amplify BAt25, BAt26, BAt40, D17S250, and D5S346 have been described previously 17. For D2S123, a newly designed primer set, suitable for damaged DNA template (D2S123 3 GACTTTCCACCTATGGGACT) was used. Analysis was performed using a 3130 xl Genetic Analyzer (Applied Biosystems, Foster City, CA) with a GeneScan 350ROX size standard (Applied Biosystems) and the manufacturers GeneScan 3.7 software. tumours were categorized according to criteria from the National Cancer Institute conference on MSI 18. tumours were classified as MSI-High if 2 or more markers (at least 1 BAt-marker) showed instability. MSI-Low was defined as 1 unstable marker and MS-Stable tumours had no shifts. All cases that showed MSI and lacked immunohistochemical expression of the MLH1 protein were further analyzed by performing hypermethylation analysis of the promoter region of the MLH1 gene by Methylation-Specific Multiplex Ligation-dependent Probe Amplification (MS-MLPA) (MRC Holland, Amsterdam, the Netherlands) 19. Immunohistochemistry Immunohistochemistry was performed using monoclonal antibodies to the mismatch repair proteins MLH1, MSH2, MSH6, and PMS2. table 1 shows details of the used antibodies in this study. Briefly, 4m sections were deparaffinated, blocked for endogenous peroxidase activity by immersion with 0.3% H2O2 in methanol for 20 min. Antigen retrieval was performed in tris/eDtA buffer (10 mM/1 mM; pH 9.0) for 10 min at 120 C. After cooling for 10 minutes and washing in PBS, non-specific binding sites were blocked in PBS with 5% normal goat serum for 10 minutes, followed by antibody incubation as indicated previously. Antibody binding was visualized using the Powervision+ poly-HRP detection system

proefschrift Marian.indd 63

10-10-2010 20:56:16

64

| CHAPteR 5
(Immunologic, Duiven, the Netherlands), with PowerDAB (Immunologic) as chromogen. Sections were counterstained with haematoxylin. Positive controls were included for each antibody. Staining interpretation the staining was considered negative if the tumour cells completely lacked the brown nuclear staining seen in the surrounding non-neoplastic cells. All slides were evaluated by three observers conjointly (MMHC, FHMM and GJAO). A minimum of 2 evaluable cores per case needed to be present; otherwise the case was excluded from further interpretation. Statistical analysis Statistical analysis was performed using SPSS for Windows (version 15.0). Fishers exact test or the chi-square test was used to determine possible statistical significant differences between the groups, as appropriate. Mean age was compared using an analysis of variance test, followed by a Games-Howell post-hoc test. A p-value of <0.05 was interpreted as statistically significant.

Results
Clinicopathological characteristics the clinicopathological characteristics of all patients are summarized in table 2. Patients with sporadic CRC were significantly older compared to other groups (p=0.000). the type of IBD was also different among the different IBD groups, with only patients in the PSC-CRC group all having UC (p=0.009). Apart from groups 1 and 2 (C and IBD) having a lower mortality rate, mortality was not different between the other patient groups (groups 3-7). tumour stage was statistically significantly different, with more advanced tumours (AJCC>2b) in the sporadic CRC groups 6 and 7 (p=0.000). MSI analysis MSI-High status was found in 0%, 12%, 0%, 88%, 0%, and 32% of the cases in the IBD, IBD-CRC, PSC-CRC, LYNCH-CRC, CRC-LeFt, and CRC-RIGHt groups, respectively. When the PSC-CRC group and IBD-CRC group were compared, no statistically significant differences were found (p=0.512), however comparing the PSC-CRC group with the CRC-RIGHt group showed a borderline significant difference (p= 0.068) (table 3). After combining all tumour groups into two categories: a chronic inflammationrelated cancer group (IBD-CRC and PSC-CRC) and a non-inflammation-related cancer

proefschrift Marian.indd 64

10-10-2010 20:56:16

MSI-status in PSC-IBD-related CRC | 65

group, (CRC-LeFt, CRC-RIGHt, and LYNCH-CRC), MSI-High was found in 7% and 30%, respectively (p=0.023). However, after excluding the LYNCH-CRC group from the non-inflammation cancer group, only 17% of the non-inflammation-related cases showed MSI-High, with this difference being not statistically significant (p=0.447).

table 1. Specification of antibodies used and details of tissue processing Antibody hMLH1 MSH2 MSH6 PMS2 Clone & Code G168-15/ 13271A Fe11/NA27 44/610919 A16-4/556415 Antigen Retrieval ARS1 ARS ARS ARS Dilution 1:50 1:200 1:400 1:500 Incubation time ON ON ON ON Detection Powervis/DAB+2 Powervis/DAB+ Powervis/DAB+ Powervis/DAB+

Source hMLH1& PMS2: Pharmingen, BD Biosciences Pharmingen, San Diego, CA, USA; MSH2: Oncogene Research Products, San Diego, CA, USA; MSH6: BD transduction laboratories, Lexington, KY, USA. 1 ARS: Antigen Retrieval System with tris/eDtA buffer pH 9.0, 10 min /120 C/ 10 min cool down 2 Powervis/DAB+: Powervision and DAB+, Immunologic, Duiven, the Netherlands ON: Overnight incubation. Positive control for all groups: normal colon mucosa, Staining pattern: nuclear

table 2. Baseline characteristics C Variables Gender (,%) Age, yrs IBD (UC,%) Location (%) Death (%) AJCC stage (%) (n=17) 8 (47) 59 (11) na na 1 (6) na IBD (n=15) 11 (73) 32 (14) 7 (47) na 0 na IBD-CRC (n=17) 11 (65) 46 (9) 6 (38) 5 (29) 6 (35) 4 (24) PSC-CRC (n=10) 4 (40) 45 (14) 8 (100) 6 (60) 6 (60) 4 (50) LYNCHCRC (n=8) 3 (38) 49 (24) na 4 (50) 1 (17) 1 (17) CRCLeFt (n=16) 9 (56) 65 (12) na na 4 (27) 12 (75) CRCRIGHt (n=19) 15 (79) 72 (9) na 19 (100) 7 (37) 16 (84)

C= healthy controls, IBD= inflammatory bowel disease, IBD-CRC= colitis-associated colorectal carcinoma, PSC-CRC= colitis-associated colorectal carcinoma and concurrent primary sclerosing cholangitis, LYNCHCRC= Lynch syndrome-associated colorectal carcinoma, CRC-LeFt= left-sided sporadic colorectal carcinoma, CRC-RIGHt= right-sided sporadic colorectal carcinoma Gender: p=0.16 (Fishers exact test) Age: mean age and (sd) are shown; p=0.00 (ANOVA, Games-Howell) IBD: p=0.009 (Fishers exact test) Location CRC: percentage of proximally located tumours is shown Death: p=0.003 (Fishers exact test) AJCC stage: tumour stage according to AJCC stages, all tumours with nodal or distant metastasis are shown; p=0.000 (Fishers exact test)

proefschrift Marian.indd 65

10-10-2010 20:56:16

66

| CHAPteR 5
Immunohistochemistry Of the MSI-High CRCs 50%, 43%, and 100% of the IBD-CRC, LYNCH-CRC, and CRC-RIGHt group, respectively, showed no MLH1 protein expression. A negative MSH2 and MSH6 expression was only found in the Lynch group, in 43% and 57% of the MSI-High tumours, respectively. PMS2 expression was negative in 100%, 43%, and 100% of the IBD-CRC, LYNCH-CRC, and CRC-RIGHt groups, respectively.

table 3. MSI-High cases per group in detail MSI-High Cases in tumour Groups (%) IBD-CRC (12%) #1 #2 CRC RIGHt (32%) #1 #2 #3 #4 #5 #6 CRC LYNCH (88%) #1 #2 #3 #4 #5 #6 #7 PSC-CRC (0%) CRC LeFt (0%) MSH2 & MSH6 MSH2 & MSH6 MSH2 & MSH6 MSH6 MLH1 & PMS2 MLH1 & PMS2 MLH1 & PMS2 na na na na na na na na na na na MLH1 & PMS2 MLH1 & PMS2 MLH1 & PMS2 MLH1 & PMS2 MLH1 & PMS2 MLH1 & PMS2 MLH1: 32% MLH1: 100% MLH1: 33% MLH1: 53% MLH1: 70% MLH1: 63% MLH1 & PMS2 PMS2 AbsentB PMS2: 16% Absent MMR-Protein expression Hypermethylation Promoter RegionA

C= healthy controls, IBD= inflammatory bowel disease, IBD-CRC= colitis-associated colorectal carcinoma, PSC-CRC= colitis-associated colorectal carcinoma and concurrent primary sclerosing cholangitis, LYNCHCRC= Lynch syndrome-associated colorectal carcinoma, CRC-LeFt= left-sided sporadic colorectal carcinoma, CRC-RIGHt= right-sided sporadic colorectal carcinoma A Hypermethylation is shown in percentages. Cut-off value for definition hypermethylation is 20%. B this case was also analyzed for the presence of a Braf mutation, which was not present.

proefschrift Marian.indd 66

10-10-2010 20:56:16

MSI-status in PSC-IBD-related CRC | 67

MLH1 promoter hypermethylation All cases (except those of the LYNCH-CRC group) with a MSI-High phenotype and an absent MLH1 protein expression were further analyzed for the presence of hypermethylation at the promoter region of MLH1. Of the 7 MSI-High cases, 6 showed MLH1 hypermethylation; these were all right-sided sporadic CRCs. One case in the IBD-CRC group did not show hypermethylation of MLH1. PMS2 hypermethylation analysis was performed in one specific IBD-CRC case with MSI-H status lacking PMS2 protein expression. tumour tissue showed 16% hypermethylation in the promoterregion of PMS2, indicating hypermethylation of the promoterregion of PMS2.

Discussion
In the current study, no evidence for MSI was found in PSC-IBD-related CRCs. Although PSC-IBD-related tumours show resemblance to Lynch-associated CRCs with regard to the more commonly seen proximal tumour location in the colon and onset at young age, we showed that the molecular pathogenetic origin is likely to be different. In more detail, the molecular pathway involving alteration of the MMR genes, either by gene mutation or by transcriptional silencing through hypermethylation of the promoter region, does not seem to be a major factor involved in PSC-IBDassociated carcinogenesis. Until now, no other studies have specifically addressed the presence of MSI in PSCrelated CRC. the prevalence of MSI in colitis-associated tumours without PSC varies however strongly, i.e., between 9-45% in the literature 20-22. this wide range could be attributed to different methodologies used by previous studies to determine MSI status. therefore, a reference marker panel, as determined by the International Committee on MSI, is now in use to allow comparison between studies 23. the prevalence of MSI-High status in colitis-associated tumours, according to these guidelines, has been reported to be lower, varying between 9-15% 24-26. the results of our study are in agreement with these data. the two MSI-High cases in the IBD-CRC group of this study were found in a patient with CD and another with UC. One of these tumours was located in the proximal colon, whereas the other was found in the distal colon. Of note, both tumours had the typical morphology of a well differentiated tubulo-glandular carcinoma as has been reported in association with IBD 27. theoretically, these findings can be assigned to a de novo mutation of one of the MMR genes, caused by an increased inflammation-induced mutation frequency, or by concurrent Lynch syndrome and IBD 28. Scvreck et al. also reported five MSI-High IBD-CRC cases with absent MMR protein expression and concluded that it was unlikely that all these 5 patients

proefschrift Marian.indd 67

10-10-2010 20:56:16

68

| CHAPteR 5
suffered from two underlying CRC-predisposing disorders at the same time 29. the microscopic appearance of the two tumours in our study is also an argument against this explanation. edwards et al. recently provided an alternative theory. they found that chronic inflammation-induced hypoxia in the murine colon may lead to a specific tumour phenotype with similar histopathological characteristics as in Lynch syndrome-related tumours. Immunohistochemical expression of MLH1 and PMS2 was absent in these tumours caused by epigenetic silencing of MLH1, however, not through promoter hypermethylation, but by decreased histone acetylation initiated due to hypoxia 30. One MSI-High IBD-CRC case in our study displayed no MLH1 and PMS2 protein expression, which was not based on promoter hypermethylation of MLH1, nor had the tumour a BRAF mutation. epigenetic silencing through decreased histone acetylation caused by inflammation-induced hypoxia may be a plausible explanation for our findings. the other MSI-High IBD-CRC case was also based on epigenetic silencing through PMS2 hypermethylation. the prevalence of MSI in colitis-associated tumours corresponds to that of sporadic CRCs (1-15%) 31. We also found a non-significant difference in MSI-High status between inflammation-related and non-inflammation-related CRC of 7% and 19%, respectively (p=0.279), provided that the LYNCH-CRC group, causing a statistically significant difference when comparing all groups, was excluded. Not all cases in the LYNCH-CRC group showed a MSI-High phenotype. Despite the fact that three mononucleotide markers were used, one case in the LYNCH-CRC group, with a confirmed germline MSH6 mutation, showed neither MSI nor absence of immuno-expression. It is known that one of the major functions of MSH6 is to correct base-base mismatches and that this not necessarily results in MSI. the positive MSH6 protein expression in this case may be due to a missense mutation with a relatively small conformational change but nevertheless loss of function 32. Despite the higher prevalence of right-sided tumours in patients with PSC and concurrent IBD compared to IBD alone, microsatellite instability did not seem to play a prominent role in the colorectal carcinogenesis of these patients. Although only 10 CRC-patients with PSC and IBD were included in this study, which may have resulted in a type II error, it is more likely that another explanation accounts for the predominantly right-sided colorectal carcinogenesis, for example an altered bile composition in these patients 33,34. In sporadic CRCs, several studies have shown that bile acids do play a role in the development of CRC 35. In patients with cholestatic diseases, the role of bile acids on colorectal carcinogenesis remains speculative. Bile acids have been associated with the potential of inducing colonic preneoplastic lesions, so-called aberrant crypt foci (ACF) 36. the hydrophobic bile acids (CDCA and DCA) are potentially the most cytotoxic and may cause liver damage in cholestatic patients 37. the exact alterations in bile composition in cholestatic patients however, remain unclear. No studies have reported an increase

proefschrift Marian.indd 68

10-10-2010 20:56:16

MSI-status in PSC-IBD-related CRC | 69

of hydrophobic bile acids in PSC patients 38,39. It is well known that an increased amount of primary bile acids and fewer secondary bile acids in the bile pool are present in cholestatic patients, mainly due to a decreased secretion of the latter 40. An argument in favour of the bile alteration hypothesis is the suggested chemopreventive effect of UDCA on the colon in PSC patients and the predominantly right-sided tumour location 41. However, the persistent high risk of developing CRC even after liver transplantation is contradictory to this hypothesis 42. More research is needed to study the effect of bile acids on the colonic epithelium and to elucidate the underlying molecular mechanism in this specific subgroup of colitis-associated CRC patients.

Acknowledgements
the authors like to thank R Stellato, statistician for her help with the analyses and K. Suijkerbuijk for her assistance with the MSI analyses. All analyses were performed in the Molecular Pathology Lab of the University Medical Centre Utrecht.

proefschrift Marian.indd 69

10-10-2010 20:56:17

70

| CHAPteR 5 Reference List


1 2 eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001 Apr;48(4):526-35. M.W.Lutgens, G.J.van der Heijden, F.P.Vleggaar, B.Oldenburg. A comprehensive meta-analysis of the risk of colorectal cancer in ulcerative colitis and Crohns disease. Gastroenterology1344,A33-A-34.1-4-2008. Fausa O, Schrumpf e, elgjo K. Relationship of inflammatory bowel disease and primary sclerosing cholangitis. Semin Liver Dis 1991 Feb;11(1):31-9. Claessen MM, Vleggaar FP, tytgat KM, Siersema PD, van Buuren HR. High lifetime risk of cancer in primary sclerosing cholangitis. J Hepatol 2009 Jan;50(1):158-64. Loftus eV, Jr., Harewood GC, Loftus CG, tremaine WJ, Harmsen WS, Zinsmeister AR, et al. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005 Jan;54(1):91-6. Shetty K, Rybicki L, Brzezinski A, Carey WD, Lashner BA. the risk for cancer or dysplasia in ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol 1999 Jun;94(6):1643-9. Claessen MM, Lutgens MW, van Buuren HR, Oldenburg B, Stokkers PC, van der Woude CJ, et al. More right-sided IBD-associated colorectal cancer in patients with primary sclerosing cholangitis. Inflamm Bowel Dis 2009 Sep;15(9):1331-6. Lynch Ht, Smyrk tC, Watson P, Lanspa SJ, Lynch JF, Lynch PM, et al. Genetics, natural history, tumour spectrum, and pathology of hereditary nonpolyposis colorectal cancer: an updated review. Gastroenterology 1993 May;104(5):1535-49. Breivik J, Lothe RA, Meling GI, Rognum tO, Borresen-Dale AL, Gaudernack G. Different genetic pathways to proximal and distal colorectal cancer influenced by sex-related factors. Int J Cancer 1997 Dec 19;74(6):664-9. Deng G, Kakar S, tanaka H, Matsuzaki K, Miura S, Sleisenger MH, et al. Proximal and distal colorectal cancers show distinct gene-specific methylation profiles and clinical and molecular characteristics. eur J Cancer 2008 Jun;44(9):1290-301. Kuismanen SA, Holmberg Mt, Salovaara R, de la CA, Peltomaki P. Genetic and epigenetic modification of MLH1 accounts for a major share of microsatellite-unstable colorectal cancers. Am J Pathol 2000 May;156(5):1773-9. Riddell RH, Goldman H, Ransohoff DF, Appelman HD, Fenoglio CM, Haggitt RC, et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983 Nov;14(11):931-68. Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004 Jul;287(1):G7-17. AJCC cancer staging manual. 6th ed. New York (NY): 2002. Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, et al. tissue microarrays for high-throughput molecular profiling of tumour specimens. Nat Med 1998 Jul;4(7):844-7. Boland CR, thibodeau SN, Hamilton SR, Sidransky D, eshleman JR, Burt RW, et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 1998 Nov 15;58(22):5248-57. Dietmaier W, Wallinger S, Bocker t, Kullmann F, Fishel R, Ruschoff J. Diagnostic microsatellite instability: definition and correlation with mismatch repair protein expression. Cancer Res 1997 Nov 1;57(21):4749-56. Umar A, Boland CR, terdiman JP, Syngal S, de la CA, Ruschoff J, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 2004 Feb 18;96(4):261-8.

3 4 5

10

11

12

13 14 15 16

17

18

proefschrift Marian.indd 70

10-10-2010 20:56:17

MSI-status in PSC-IBD-related CRC | 71


19 Schouten JP, Mcelgunn CJ, Waaijer R, Zwijnenburg D, Diepvens F, Pals G. Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res 2002 Jun 15;30(12):e57. 20 Fujiwara I, Yashiro M, Kubo N, Maeda K, Hirakawa K. Ulcerative Colitis-Associated Colorectal Cancer is Frequently Associated with the Microsatellite Instability Pathway. Dis Colon Rectum 2008 Jun 11. 21 Schulmann K, Mori Y, Croog V, Yin J, Olaru A, Sterian A, et al. Molecular phenotype of inflammatory bowel disease-associated neoplasms with microsatellite instability. Gastroenterology 2005 Jul;129(1):74-85. 22 Suzuki H, Harpaz N, tarmin L, Yin J, Jiang HY, Bell JD, et al. Microsatellite instability in ulcerative colitis-associated colorectal dysplasias and cancers. Cancer Res 1994 Sep 15;54(18):4841-4. 23 Boland CR, thibodeau SN, Hamilton SR, Sidransky D, eshleman JR, Burt RW, et al. A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 1998 Nov 15;58(22):5248-57. 24 Fleisher AS, esteller M, Harpaz N, Leytin A, Rashid A, Xu Y, et al. Microsatellite instability in inflammatory bowel disease-associated neoplastic lesions is associated with hypermethylation and diminished expression of the DNA mismatch repair gene, hMLH1. Cancer Res 2000 Sep 1;60(17):4864-8. 25 Svrcek M, el-Bchiri J, Chalastanis A, Capel e, Dumont S, Buhard O, et al. Specific clinical and biological features characterize inflammatory bowel disease associated colorectal cancers showing microsatellite instability. J Clin Oncol 2007 Sep 20;25(27):4231-8. 26 Willenbucher RF, Aust De, Chang CG, Zelman SJ, Ferrell LD, Moore DH, et al. Genomic instability is an early event during the progression pathway of ulcerative-colitis-related neoplasia. Am J Pathol 1999 Jun;154(6):1825-30. 27 Levi GS, Harpaz N. Intestinal low-grade tubuloglandular adenocarcinoma in inflammatory bowel disease. Am J Surg Pathol 2006 Aug;30(8):1022-9. 28 Schulmann K, Mori Y, Croog V, Yin J, Olaru A, Sterian A, et al. Molecular phenotype of inflammatory bowel disease-associated neoplasms with microsatellite instability. Gastroenterology 2005 Jul;129(1):74-85. 29 Svrcek M, el-Bchiri J, Chalastanis A, Capel e, Dumont S, Buhard O, et al. Specific clinical and biological features characterize inflammatory bowel disease associated colorectal cancers showing microsatellite instability. J Clin Oncol 2007 Sep 20;25(27):4231-8. 30 edwards RA, Witherspoon M, Wang K, Afrasiabi K, Pham t, Birnbaumer L, et al. epigenetic repression of DNA mismatch repair by inflammation and hypoxia in inflammatory bowel disease-associated colorectal cancer. Cancer Res 2009 Aug 15;69(16):6423-9. 31 Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004 Jul;287(1):G7-17. 32 Berends MJ, Wu Y, Sijmons RH, Mensink RG, van der St, Hordijk-Hos JM, et al. Molecular and clinical characteristics of MSH6 variants: an analysis of 25 index carriers of a germline variant. Am J Hum Genet 2002 Jan;70(1):26-37. 33 Bayerdorffer e, Mannes GA, Ochsenkuhn t, Dirschedl P, Wiebecke B, Paumgartner G. Unconjugated secondary bile acids in the serum of patients with colorectal adenomas. Gut 1995 Feb;36(2):268-73. 34 Nagengast FM, Grubben MJ, van M, I. Role of bile acids in colorectal carcinogenesis. eur J Cancer 1995 Jul;31A(7-8):1067-70. 35 tong JL, Ran ZH, Shen J, Fan GQ, Xiao SD. Association between fecal bile acids and colorectal cancer: a meta-analysis of observational studies. Yonsei Med J 2008 Oct 31;49(5):792-803. 36 Peterlik M. Role of bile acid secretion in human colorectal cancer. Wien Med Wochenschr 2008;158(1920):539-41. 37 Hofmann AF, Hagey LR. Bile acids: chemistry, pathochemistry, biology, pathobiology, and therapeutics. Cell Mol Life Sci 2008 Aug;65(16):2461-83.

proefschrift Marian.indd 71

10-10-2010 20:56:17

72

| CHAPteR 5
38 Beuers U, Spengler U, Zwiebel FM, Pauletzki J, Fischer S, Paumgartner G. effect of ursodeoxycholic acid on the kinetics of the major hydrophobic bile acids in health and in chronic cholestatic liver disease. Hepatology 1992 Apr;15(4):603-8. 39 Stiehl A, Rudolph G, Sauer P, theilmann L. Biliary secretion of bile acids and lipids in primary sclerosing cholangitis. Influence of cholestasis and effect of ursodeoxycholic acid treatment. J Hepatol 1995 Sep;23(3):283-9. 40 van de Meeberg PC, Wolfhagen FH, Van Berge-Henegouwen GP, Salemans JM, tangerman A, van Buuren HR, et al. Single or multiple dose ursodeoxycholic acid for cholestatic liver disease: biliary enrichment and biochemical response. J Hepatol 1996 Dec;25(6):887-94. 41 tung BY, emond MJ, Haggitt RC, Bronner MP, Kimmey MB, Kowdley KV, et al. Ursodiol use is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Ann Intern Med 2001 Jan 16;134(2):89-95. 42 Loftus eV, Jr., Aguilar HI, Sandborn WJ, tremaine WJ, Krom RA, Zinsmeister AR, et al. Risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis following orthotopic liver transplantation. Hepatology 1998 Mar;27(3):685-90.

proefschrift Marian.indd 72

10-10-2010 20:56:18

CHaPter 6
Differences in molecular markers between colitisassociated colorectal cancer with or without primary sclerosing cholangitis and sporadic colorectal cancer

MMH Claessen1, FP Vleggaar1, MeI Schipper2, FHM Morsink2, JWJ Hinrichs2, PD Siersema1, GJA Offerhaus2
1

Department of Gastroenterology and Hepatology, 2Department of Pathology , University Medical Center Utrecht submitted to: Journal of Clinical Pathology

proefschrift Marian.indd 73

10-10-2010 20:56:19

74

| CHAPteR 6 Abstract
Background Patients with inflammatory bowel disease (IBD) and concurrent primary sclerosing cholangitis (PSC) have a significantly higher risk of developing colorectal cancer (CRC) than IBD-patients without PSC. Although the molecular background behind colorectal carcinogenesis is gradually elucidated, still little is known about this specific subgroup. aims & Methods to compare specific gene and protein profiles involved in the carcinogenetic pathway of PSC-IBD-CRC with those of other subtypes of CRC, i.e., IBD-CRC, and sporadic CRC. A tissue micro-array was constructed with colonic samples from 6 groups: (1) healthy subjects (n=17), (2) IBD-patients (n=15), (3) IBD-related CRC (n=17), (4) PSC-IBD-related CRC (n=10), (5) sporadic left-sided CRC (n=16), and (6) sporadic right-sided CRC (n=20). Immunohistochemistry was performed using monoclonal antibodies against -catenin, Cyclin D1, CD44V6, p53, SMAD4, and COX2. In addition, Kras and Braf mutation analysis was performed. results Median age of all patients was 55 years (range 12-92 years), with 61% being male. Ulcerative colitis was present in 53% of IBD cases (groups 2-4). Aberrant expression of p53 was detected in 60% of CRCs with no difference between inflammation-related (18/27 (67%) and sporadic (20/36 (55%)) CRCs (p=0.45). Cyclin D1 overexpression was found in 15/27 (56%) and 31/36 (86%) of inflammation-related and sporadic CRCs, respectively, which was significantly different after correction for tumour stage and age (p=0.038). Loss of SMAD4 expression was twice as high in inflammatory-related CRCs compared to sporadic CRCs, 26% and 11%, respectively, which was however not significant (p=0.18). No differences were found in expression of nuclear -catenin, CD44V6, and COX-2. A significant difference was found in Kras or Braf mutations between the inflammation-related and sporadic tumour groups, which was no longer significant after correction for age and tumour location (p=0.06). Conclusion Of all molecular features investigated, only Cyclin D1 overexpression occurred less frequently and a trend towards loss of SMAD4 expression was seen in colitis-related CRC compared to the sporadic CRCs. No differences between IBD-CRC patients with or without PSC were observed. Our results suggest that carcinogenetic pathways in subgroups only differ in small details whenever CRC is already clinically apparent.

proefschrift Marian.indd 74

10-10-2010 20:56:19

Gene expression in CRC | 75

Introduction
Patients with Inflammatory Bowel Disease (IBD) are at increased risk for colorectal cancer (CRC) when compared to the normal population 1. It is known that some features of the molecular background of colorectal carcinogenesis are equal and some differ between sporadic CRCs and its colitis-associated counterparts. the majority of sporadic tumours arise from a precursor lesion initiated by a loss of function of adenomatous polyposis coli (APC), whereas p53, the second key tumour suppressor gene loses its function during progression from adenoma to carcinoma. these genetic alterations are part of the chromosomal instability pathway (CIN), through which the majority of CRCs (85%) arise. the second main pathway is the microsatellite instability pathway (MSI) which involves loss of function of one of the mismatch repair (MMR) genes either by mutation or hypermethylation of the MLH1 promoter region. About 15% of the sporadic tumours develop through this pathway, in particular proximally located tumours. the same distribution of CIN and MSI is also found in colitis-associated CRCs 2. In contrast, the timing and frequency of genetic alterations involved in the CIN pathway vary between sporadic and colitis-induced colorectal carcinogenesis. the sequence of genetic alterations is almost reversed in colitis-related CRC development compared to sporadic CRC development, with p53 acting as a gatekeeper and APC mutations at the end of the pathway 2. However, the exact timing of mutations during carcinogenesis is still point of discussion. Our group recently reported indications for an activated Wnt-pathway in normal colonic mucosa of UC patients who had neoplasia elsewhere in their colon, implying an early role for the Wnt-pathway in colitis-associated carcinogenesis 3. A small subgroup of patients with IBD also has concurrent primary sclerosing cholangitis (PSC), a chronic inflammatory disorder of the bile duct system. these patients have an even higher risk of developing CRC than patients with IBD alone, with 10- and 20-year risks of 14% and 31%, respectively 4-7. Several hypotheses have been suggested to explain this increased risk, including IBD-related as well as PSC-related factors. Although the molecular background of colorectal carcinogenesis is gradually elucidated, much less is known of PSC-IBD-related CRCs. Only a few studies have examined the molecular background of these tumours 8,9. Previous studies have shown that patients with PSC and IBD exhibit more frequent aneuploidy in non-CRCrelated colorectal biopsies than patients with UC alone 10,11. Our study group recently studied the MSI status in patients with PSC, but we did not find evidence for instable markers 12. Because of the exceptionally high CRC risk in these patients, we hypothesized that the molecular background of colorectal carcinogenesis in PSCIBD-related CRC might well differ from that of IBD-related and sporadic CRC.

proefschrift Marian.indd 75

10-10-2010 20:56:19

76

| CHAPteR 6
the aim of this study was to compare the expression of proteins involved in various carcinogenetic pathways (p53, -catenin, CD44V6, Cyclin D1, COX-2, SMAD4) and to perform mutation analysis of 2 important oncogenes (Kras and Braf) in sporadic CRC and IBD-associated CRC, with a specific focus on PSC-IBD patients.

Patients and methods


Patients and tissue samples Paraffin embedded tissue specimens from surgical resection material were obtained from the Department of Pathology of the University Medical Center Utrecht, the Netherlands. the study was carried out in concordance with the ethical guidelines and approval of the research review committee of our institution 13. tissue samples of six groups of patients were collected. the first group (C) consisted of healthy controls who underwent sigmoid resection because of diverticular disease (n=17). Histological examination showed no inflammation or other pathology except diverticular disease. In the second group (IBD), IBD-patients with chronic active inflammation of colonic mucosa were included (n=15). the third group (IBD-CRC) consisted of IBD-related CRCs (n=17), the fourth group PSC-IBD-related CRCs (PSC-CRC; n=10), the fifth group (CRC-LeFt; n=16) left-sided sporadic CRC, defined as a tumour distal from the splenic flexure, and the sixth group (CRC-RIGHt; n=20) sporadic CRCs located in the proximal colon, defined as a tumour proximal from the splenic flexure. the histological criteria by Riddell et al. for colitis-associated dysplasia-adenocarcinoma were applied 14. Since there is no clear evidence for a difference in carcinogenesis between Crohns disease (CD) and ulcerative colitis (UC), specimens from patients with either disease were included in this study. tumour stages were defined according to the American Joint Committee on Cancer (AJCC) 15. tissue Micro array A tissue micro array (tMA) was constructed according to a standardized protocol described by Kononen et al. 16. Briefly, at least three core biopsies (diameter 0.6 mm) were taken from each sample and punched into a recipient block using a tissue micro-arrayer (MtA-I, Beecher Instruments Inc.,Sun Prairie, USA), which was guided by the MtA Booster (Alphelys, Plaisir, France). the distribution and position of the cores was determined in advance with tMA-Designer Software (Version 1.6.8, Alphelys).

proefschrift Marian.indd 76

10-10-2010 20:56:20

Gene expression in CRC | 77

Immunohistochemistry Immunohistochemistry (IHC) was performed on the tMA using monoclonal antibodies to p53, -catenin, CD44V6, Cyclin D1, COX2, and SMAD4. table 1 shows the details of all antibodies, dilutions, incubation times and antigen retrieval methods applied in this study. Briefly, 4m sections were deparaffinized, blocked for endogenous peroxidase activity by immersion with 0.3% H2O2 in methanol for 20 min. Antigen retrieval was performed in tris/eDtA buffer (10 mM/1 mM; pH 9.0) for 10 min at 120 C. After cooling for 10 minutes and washing in PBS, non-specific binding sites were blocked in PBS with 5% normal goat serum for 10 minutes, followed by antibody incubation as indicated. Antibody binding was visualized using Powervision+ poly-HRP detection system (Immunologic, Duiven, the Netherlands), with 3.3-DiAmino Benzidine tetrachloride (DAB) (Sigma, Missouri, USA) and PowerDAB (Immunologic, Duiven, the Netherlands) as chromogen for -catenin, CD44V6, p53, and Cyclin D1, respectively. Sections were counterstained with haematoxylin. Positive controls were included for each antibody. DNa isolation for Kras and Braf mutation analysis DNA was isolated from paraffin embedded tissue from all tumour groups. the tissue was deparaffinized with xylene for 10 minutes followed by ethanol. to obtain DNA from enriched tumour tissue (>75% tumour cells), 20 l of proteinase K was used (10 mg/ml) in 100 l buffer (0.5% tween 20, 50 mmol/ L tris (pH=9), 1mmol/L NaCl, 2 mmol/L eDtA). After overnight incubation in 56 C, tubes were incubated at 95 C for 10 minutes to inactivate the proteinase K and cooled on ice afterwards. the supernatant was transferred to a second tube after centrifugation. Samples were stored at -20C. Kras mutation analysis Genomic DNA was amplified by PCR using Platinum taq polymerase (Invitrogen Corporation, Carlsbad, CA, USA) and specific primers (for codons 12 and 13 in exon 1 and codon 61 in exon 2). PCR products were sequenced in both sense and antisense directions using the BigDye terminator v1.1 sequencing kit on ABI 3130 (Applied Biosystems, Foster City, CA, USA) according to the manufacturers instructions. Primers used for amplification were exon 1 forward: CTG GTG GAG TAT TTG ATA GT, reverse: ATG GTC CTG CAC CAG TAA TA and exon 2 forward: GGT GCA CTG TAA TAA TCC AGA C, reverse: CCA CCT ATA ATG GTG AAT ATC T. Sequence primers were exon 1 forward: GTG TGA CAT GGT CTA AT, reverse: CCT GCA CCA GTA ATA TGC and exon 2 forward: TGT GTT TCT CCC TTC TC and reverse: CTT CAA ATG ATT TAG TAT TA. Samples that that could not be processed with the above described method due to a low DNA quality or quantity were analyzed for codons 12 and 13 with a high resolution melting (HRM) technology-based assay according to Kramer et al. 17.

proefschrift Marian.indd 77

10-10-2010 20:56:20

78

table 1. Specification of antibodies used and details of tissue processing Source Neomarkers BD Biosciences Bender MedSystems Neomarkers Cayman Chemical Santa Cruz DPC4 ARS pH 9 1:300 1 hr 160112 ARS pH 9 1:200 overnight Powervision/AeC Powervision/DAB++ SP4 rabbit ARS pH 9 1:100 overnight Powervision/DAB++ VFF18 ARS pH 9 1:300 1 hr Powervision/DAB 14 ARS pH 9 1:5000 overnight Powervision/DAB DO-7+BP53-12 ARS pH 9 1:2000 1 hr Powervision/DAB Clone/Code Antigen retrieval* Dilution Incubation time Detection Positive control Colon tumour Colon tumour Colon tumour Mamma tumour Stomach tumour Pancreatic tumour

proefschrift Marian.indd 78

Antibody

Staining pattern

| CHAPteR 6

P53

Nuclear

-catenin

Nuclear/Membr

CD44 V6

Membr/Cytoplasm

Cyclin D1

Nuclear

COX2

Cytoplasm

SMAD4

Nuclear

* ARS: Antigen Retrieval System with tris/eDtA buffer pH9.0 or Sodium Citrate pH6, 10min 120 C / 10 minutes cool down

10-10-2010 20:56:20

Gene expression in CRC | 79

Braf mutation analysis Genomic DNA was amplified by PCR using Amplitaq Gold (Applied Biosystems, Foster City, CA, USA) and specific primers (for exon 15, hotspot V600e mutation). PCR products were sequenced in both sense and antisense directions using the BigDye terminator v1.1 sequencing kit on ABI 3130 (Applied Biosystems, Foster City, CA, USA) according to the manufacturers instructions. Primers used for amplification were forward: 5 CAt-AAt-GCt-tGC-tCt-GAt-AG 3 and reverse: 5 AtC-CAG-ACA-ACt-Gtt-CAA-AC 3. Primers used for sequencing were forward: 5-GCt-CtG-AtA-GGA-AAA-tGA-GAt-3 and reverse: 5 tGA-tGG-GAC-CCA-CtCCAt 3. Staining interpretation All slides were evaluated by three observers conjointly (MC, FM and JO) and the final staining result of each slide was based on a combined opinion. the specimens were considered positive for p53 if the epithelial cells exhibited intense brown nuclear staining in the entire tissue sample or in segments but always more than 20% positive cells 18,19. Interpretation of Cyclin D1 was based on intensity and divided into 3 categories: negative or weakly positive (-), positive (+), or strongly positive (++). Both the positive and strongly positive samples were regarded as Cyclin D1 overexpression 20. -catenin was evaluated in the nucleus, with the percentage of stained cells being semi-quantitatively analyzed and divided into three categories: low (<10% of cells), moderate (10-50%), or high (>50% of cells). If more than 10% of cells showed positive nuclear staining, this was interpreted as indicative of Wntpathway activation 21-23. Intensity of the staining was also taken into account. Some slides showed only vague staining in the base of the crypts; this was regarded as a physiological phenomenon. CD44V6 was scored positive when membranous or cytoplasmatic staining was present. Loss of SMAD4 expression in tumours was recorded. For the interpretation of COX2 expression, only cases with an intense positive cytoplasm were scored as positive. As a rule, a minimum of 2 cores per case had to be present; otherwise the case was excluded from further interpretation. Figure 1 illustrates the expression profiles of all antibodies. Statistical analysis Statistical analysis was performed using SPSS for Windows (version 15.0). Fishers exact or chi square tests were used to determine differences that were statistically significant between the groups. We used binary logistic regression including a Hosmer-Lemshow Goodness of Fit test to assess how well the chosen model fitted the data and to correct for possible confounders. A p-value of <0.05 was interpreted as statistically significant.

proefschrift Marian.indd 79

10-10-2010 20:56:20

80

| CHAPteR 6

Figure 1. Images illustrating expression profiles of p53 (a, b), -catenin (c, d), Cyclin D1 (e, f), SMAD4 (g, h), COX2 (i, j), CD44V6 (k, l). Magnification 80x and 400x. Sample (a) shows nuclear p53 expression whereas sample (b) shows negative staining for p53. Sample (c) demonstrates nuclear expression of -catenin and sample (d) membranous expression. Sample (e) shows increased expression of Cyclin D1 and sample (f) a negative staining pattern for Cyclin D1. Loss of SMAD4 expression is shown in sample (g) whereas sample (h) shows normal SMAD4 expression. Sample (i) demonstrates membranous COX2 staining and sample (j) negative COX2 staining. Sample (k) and sample (l) show positive membranous expression and negative expression for CD44V6, respectively.

proefschrift Marian.indd 80

10-10-2010 20:56:24

Gene expression in CRC | 81

Results
Clinicopathological characteristics Clinicopathological characteristics of all patients are shown in table 2. All patients in the PSC-CRC group had UC as concurrent disease, whereas in the other IBDrelated groups also patients with CD were included (p=0.009). Patients with an inflammation-related CRC (IBD-CRC & PSC-CRC) were significantly younger compared to patients with a sporadic CRC (CRC-RIGHt & CRC-LeFt) (p=0.000). the mortality rates did not differ among the tumour groups. tumour stages according AJCC were statistically different with more advanced tumours in the sporadic CRC groups (p=0.001). Immunohistochemical analysis Results of immunohistochemistry are shown in table 3 (separate patient groups) and table 4 (inflammation-related vs sporadic CRCs). No significant differences in p53 overexpression were found between the different CRC subgroups. Comparison of p53 overexpression between the inflammationrelated and sporadic CRCs also showed no significant differences (p=0.45). Positive nuclear -catenin staining was not different between the inflammationrelated and sporadic CRCs, 63% and 61%, respectively. In the left-sided sporadic CRC group (88%), nuclear -catenin expression was significantly higher than in the PSC-CRC (50%) and CRC-RIGHt (40%) groups. the frequency of Cyclin D1 overexpression was significantly lower in inflammationrelated CRCs (56%) compared to sporadic CRCs (86%) (p=0.003). this difference remained significant after correction for age and tumour stage (p=0.038). No significant differences in Cyclin D1 overexpression were found between PSC-CRCs and IBD-CRCs.
table 2. Baseline patient characteristics Variables (%) Gender () Age (yrs) (mean; SD) IBD (UC) Location CRC (prox)* Death tumour stage (>2b)** Healthy n=17 8 (47%) 59 (11) na na 1 (6%) na IBD n=15 11 (73%) 32 (14) 7 (47%) na 0 na IBD-CRC n=17 11 (65%) 46 (9) 6 (35%) 5 (29%) 6 (35%) 4 (24%) PSC-CRC n=10 4 (40%) 45 (14) 8 (80%) 6 (60%) 6 (60%) 4 (40%) CRC-LeFt n=16 9 (56%) 65 (12) na na 4 (25%) 12 (75%) CRC-RIGHt n=20 16 (80%) 72 (9) na na 8 (40%) 17 (85%)

na: not applicable, UC: ulcerative colitis, *Location CRC: percentage of CRC proximally located tumours is shown, **tumour stage according to the AJCC tumour staging system, tumours with nodal or distant metastasis are shown

proefschrift Marian.indd 81

10-10-2010 20:56:24

82

| CHAPteR 6
Loss of expression of SMAD4 was found in 26% and 11% in the inflammationrelated and sporadic groups, respectively (p=0.15). CD44V6 and COX-2 expression were not different between the subgroups of CRC.

table 3. Immunohistochemical expression and mutation analysis in each patient group expression (%) P53 -catenin Cyclin D1 CD44V6 COX2 SMAD4 Krasb Braf
c a

Healthy n=17 0* 0* 0* 0* 0* na na na na

IBD n=15 0* 3 (20%) 7 (47%) 11 (73%) 0* na na na na

IBD-CRC n=17 10 (59%) 12 (71%) 8 (47%) 10 (59%) 9 (53%) 5 (29%) 3 (18%) 0 3 (18%)

PSC-CRC n=10 8 (80%) 5 (50%) 7 (70%) 5 (50%) 5 (50%) 2 (20%) 0 0 0

CRC-LeFt n=16 8 (50%) 14 (88%)* 14 (88%) 10 (63%) 11 (69%) 2 (13%) 3 (19%) 1 (6%) 4 (25%)

CRC-RIGHt n=20 12 (60%) 8 (40%) 17 (85%) 13 (65%) 9 (45%) 2 (10%) 8 (40%)* 7 (35%)** 15 (75%) *

Kras or Brafd

*Significant p-value compared with the PSC-CRC group (p<0.05), **Borderline significant compared with PSC-CRC group (p=0.064), aloss of SMAD4 expression, bKras mutation, cBraf mutation, dKras or Braf mutation

table 4. Immunohistochemical expression and mutation analysis in inflammation-related (PSC-CRC & IBD-CRC) and sporadic CRC (CRC-LeFt & CRC-RIGHt) expression (%) P53 -catenin Cyclin D1 CD44V6 COX2 SMAD4a Kras Braf
b c

Inflammation-related CRC n=27 18 (67%) 17 (63%) 15 (56%) 15 (56%) 14 (52%) 7 (26%) 3 (11%) 0 3 (11%)
b c

Sporadic n=36 20 (55%) 22 (61%) 31 (86%) 23 (64%) 20 (56%) 4 (11%) 11 (31%) 8 (22%) 19 (53%)
d

p-value 0.45 0.99 0.003 0.39 0.68 0.18 0.07 0.008 0.001

Kras or Brafd
a

: loss of SMAD4 expression, : Kras mutation, : Braf mutation, : Kras or Braf mutation

proefschrift Marian.indd 82

10-10-2010 20:56:24

Gene expression in CRC | 83

Kras and Braf mutation analysis In the PSC-CRC group only wild-type Kras and wild-type Braf were found (table 3). Moreover, in the IBD-CRC group also no mutations in Braf were found, whereas 18% of IBD-CRCs showed a mutation in Kras. A statistically significant difference was found for Braf mutations comparing the inflammation-related CRCs (IBD-CRC & PSC-CRC) and sporadic CRCs, 0% vs. 22%, respectively (p=0.008) (table 4). this difference was no longer statistically significant after correction for age and tumour location. Comparison of inflammation-related CRCs with sporadic CRCs showed Kras mutations in 11% and 31%, respectively (p=0.079). No statistically significant differences in Kras or Braf mutation frequency were found between the PSC-CRC and the IBD-CRC groups. Cumulative analysis (Kras and Braf taken together) showed a statistically significant difference between inflammation-related CRCs and sporadic CRCs with 11% and 53% of the cases, respectively, with a mutation in one of the genes (p=0.001). After correction for age and tumour location, this difference was borderline significant (p=0.06).

Discussion
this study was conducted to elucidate the role of key carcinogenetic pathways in inflammation-related and sporadic CRCs. the most remarkable findings were a less frequent Cyclin D1 overexpression in IBD-related CRC and a trend towards loss of SMAD4 expression in IBD-related CRC compared to sporadic CRC. In addition, no differences were found between IBD-CRC with or without PSC. the expression of Cyclin D1 was significantly lower in IBD-related CRC compared to sporadic CRC, with overexpression in 56% and 89%, respectively. Cyclin D1, an essential factor in the cell cycle control and promoter of progression through the G1-S-phase, plays a pivotal role in the development of several human cancers including CRC 24. Furthermore, Cyclin D1 is a target gene of the Wnt-signaling pathway 25. this may well provide an explanation for the reported difference in expression. Lower incidences of nuclear -catenin and APC mutations have also been reported in colitis-associated CRCs compared to sporadic CRCs 20,26. the expression profile of Cyclin D1 in IBD-related CRC could be attributed to these corresponding lower incidences. Furthermore, overexpression of Cyclin D1 was also found in half of the IBD patients without neoplasm, which corresponds with earlier reports and may be associated with an increased cell turnover, DNA damage, and enhanced apoptosis in active colitis 27,28. Another interesting finding was the loss of expression of SMAD4, a tumour suppressor gene which plays an essential role in the downstream conduction of the transforming growth factor (tGF) signaling pathway. this signaling cascade leads

proefschrift Marian.indd 83

10-10-2010 20:56:24

84

| CHAPteR 6
to activation of genes responsible for regulation of growth, differentiation, and embryonic development 29. the reduction in SMAD4 expression was twice as high in inflammation-related CRCs compared to sporadic CRCs. Although this difference was not statistically significant, loss of SMAD4 expression in a high percentage of inflammation-related CRCs suggests a role in the colitis-associated pathogenesis of CRC as compared to CRCs that arise in a sporadic setting. terdiman et al. performed a high resolution analysis of chromosome 18 which indicated that 18q21, the location of the SMAD4 gene, was a target of loss in colitis-associated cancer and suggested a major role for SMAD4 in colitis-associated carcinogenesis 30. However, Hill et al found a higher prevalence of loss of SMAD4 expression in sporadic CRCs compared to colitis-related CRCs of 70% and 40%, respectively 31. Loss of SMAD4 protein expression varies between 9% and 66% in sporadic CRCs. this variation may be the result of differentiation grade and tumour stage. A higher loss of SMAD4 protein expression has been observed in poorly differentiated CRCs and signet-cell carcinomas as well as in CRCs at more advanced stages of malignant progression32. Inflammation-related CRCs are often poorly differentiated and may reveal signet-cell histology; which may explain our findings of a relatively high prevalence of SMAD4 protein reduction. We found no evidence for Braf and Kras mutations in the PSC-related tumour group. Although Kras and Braf mutations seemed to occur more frequently in sporadic CRCs than in inflammation-related CRCs, this difference was only borderline significant after adjustment for age and tumour location. Apparently, the Mitogenactivated Protein Kinase (MAPK) pathway, with Kras as well as Braf being part of it, is not to a larger extent involved in inflammation (IBD with or without PSC)-related CRCs compared to sporadic CRCs. It might however be that activation of the MAPKpathway occurs through other genes, which may explain the low rates of Kras and Braf mutations IBD- and PSC-IBD-CRCs 33. In contrast, Aust et al. reported that 27% of colitis-associated tumours showed a Kras or Braf mutation and concluded that the MAPK pathway does play an important role 34. It is known that Braf mutations occur frequently in tumours that exhibit microsatellite instability through promoter hypermethylation of MLH1 35. In an earlier study, we reported that all PSC-related CRCs that we studied were microsatellite stable, which may explain the absence of Braf alterations in our study 12. the low incidence of Kras mutations in inflammation-related CRCs is in concordance with previous reports 36,37. Furthermore, the association between a proximal tumour location and Kras or Braf mutation is also supported by others 38-42. Kras plays a pivotal role in the transmission of growth-promoting signals from the cell-surface to the nucleus and is assumed to be an early event in the classical-adenoma-carcinoma pathway, contributing to growth and dysplastic changes in adenomas 43. A lower frequency of Kras mutations in inflammation-related CRCs may indicate that this

proefschrift Marian.indd 84

10-10-2010 20:56:24

Gene expression in CRC | 85

classical pathway occurs less frequently in this subgroup of CRCs. We recognize that an impeding factor in studying rare subgroups of CRC is to collect enough samples. therefore, it was not possible to draw firm conclusions about the PSC-related group. Although the complete lack of Braf and Kras mutations remain a remarkable finding, further larger studies are needed to verify these findings. In conclusion, of all molecular features investigated, only Cyclin D1 overexpression occurred less frequently and a trend towards loss of SMAD4 expression was seen in colitis-related CRC, with or without PSC, compared to sporadic CRC. In addition, we showed that Kras and Braf mutations were not a frequent event in IBD-related carcinogenesis, including IBD-PSC-related CRCs. Our results suggest that based on this study, carcinogenetic pathways differ only in small details when subgroups of clinically-evident CRC are compared. It could well be that subgroups of CRCs differ at an earlier stage, i.e., at a stage when the initial neoplastic changes occur leading to dysplasia and finally CRC.

Acknowledgements
the authors like to thank R Stellato, statistician for her help with the analyses. All analyses were performed in the Molecular Pathology Lab of the University Medical Center Utrecht.

proefschrift Marian.indd 85

10-10-2010 20:56:24

86

| CHAPteR 6 Reference List


1. eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526-535. 2. Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004;287:G7-17. 3. Claessen MM, Schipper Me, Oldenburg B, Siersema PD, Offerhaus GJ, Vleggaar FP. WNt-pathway activation in IBD-associated colorectal carcinogenesis: potential biomarkers for colonic surveillance. Cell Oncol 2010;32:303-310. 4. Brentnall tA, Haggitt RC, Rabinovitch PS, Kimmey MB, Bronner MP, Levine DS, Kowdley KV, Stevens AC, Crispin DA, emond M, Rubin Ce. Risk and natural history of colonic neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis. Gastroenterology 1996;110:331-338. 5. Broome U, Lofberg R, Veress B, eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology 1995;22:1404-1408. 6. Claessen MMH, Vleggaar FP, tytgat KMAJ, Siersema PD, van Buuren HR. High life-time risk of cancer in primary sclerosing cholangitis. J Hepatol 2008;50:15-64. 7. Soetikno RM, Lin OS, Heidenreich PA, Young HS, Blackstone MO. Increased risk of colorectal neoplasia in patients with primary sclerosing cholangitis and ulcerative colitis: a meta-analysis. Gastrointest endosc 2002;56:48-54. 8. Loftus eV, Jr., Harewood GC, Loftus CG, tremaine WJ, Harmsen WS, Zinsmeister AR, Jewell DA, Sandborn WJ. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut 2005;54:91-96. 9. Sokol H, Cosnes J, Chazouilleres O, Beaugerie L, tiret e, Poupon R, Seksik P. Disease activity and cancer risk in inflammatory bowel disease associated with primary sclerosing cholangitis. World J Gastroenterol 2008;14:3497-3503. 10. Broome U, Lindberg G, Lofberg R. Primary sclerosing cholangitis in ulcerative colitis--a risk factor for the development of dysplasia and DNA aneuploidy? Gastroenterology 1992;102:1877-1880. 11. Holzmann K, Klump B, Borchard F, Gregor M, Porschen R. Flow cytometric and histologic evaluation in a large cohort of patients with ulcerative colitis: correlation with clinical characteristics and impact on surveillance. Dis Colon Rectum 2001;44:1446-1455. 12. Claessen MMH, Vleggaar FP, Schipper MeI, Morsink F, Hinrichs JW, Siersema PD, Offerhaus GJA. Colorectal Cancer Microsatellite Status in Patients With Primary Sclerosing Cholangitis and Concurrent Inflammatory Bowel Disease. 138 ed. 2010:S-443. 13. van Diest PJ. No consent should be needed for using leftover body material for scientific purposes. BMJ 2002;325:648-651. 14. Riddell RH, Goldman H, Ransohoff DF, Appelman HD, Fenoglio CM, Haggitt RC, Ahren C, Correa P, Hamilton SR, Morson BC, . Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983;14:931-968. 15. AJCC cancer staging manual. New York (NY): 2002. 16. Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 1998;4:844-847. 17. Kramer D, thunnissen FB, Gallegos-Ruiz MI, Smit eF, Postmus Pe, Meijer CJ, Snijders PJ, Heideman DA. A fast, sensitive and accurate high resolution melting (HRM) technology-based assay to screen for common K-ras mutations. Cell Oncol 2009;31:161-167. 18. Kaserer K, Schmaus J, Bethge U, Migschitz B, Fasching S, Walch A, Herbst F, teleky B, Wrba F. Staining patterns of p53 immunohistochemistry and their biological significance in colorectal cancer. J Pathol 2000;190:450-456. 19. torhorst J, Bucher C, Kononen J, Haas P, Zuber M, Kochli OR, Mross F, Dieterich H, Moch H, Mihatsch M, Kallioniemi OP, Sauter G. tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol 2001;159:2249-2256.

proefschrift Marian.indd 86

10-10-2010 20:56:24

Gene expression in CRC | 87


20. Wong NA, Morris RG, McCondochie A, Bader S, Jodrell DI, Harrison DJ. Cyclin D1 overexpression in colorectal carcinoma in vivo is dependent on beta-catenin protein dysregulation, but not k-ras mutation. J Pathol 2002;197:128-135. 21. Joo Ye, Rew JS, Park CS, Kim SJ. expression of e-cadherin, alpha- and beta-catenins in patients with pancreatic adenocarcinoma. Pancreatology 2002;2:129-137. 22. Kim MJ, Jang SJ, Yu e. Loss of e-cadherin and cytoplasmic-nuclear expression of beta-catenin are the most useful immunoprofiles in the diagnosis of solid-pseudopapillary neoplasm of the pancreas. Hum Pathol 2008;39:251-258. 23. Li CC, Xu B, Hirokawa M, Qian Z, Yoshimoto K, Horiguchi H, tashiro t, Sano t. Alterations of e-cadherin, alpha-catenin and beta-catenin expression in neuroendocrine tumors of the gastrointestinal tract. Virchows Arch 2002;440:145-154. 24. Fu M, Wang C, Li Z, Sakamaki t, Pestell RG. Minireview: Cyclin D1: normal and abnormal functions. endocrinology 2004;145:5439-5447. 25. Nosho K, Kawasaki t, Chan At, Ohnishi M, Suemoto Y, Kirkner GJ, Fuchs CS, Ogino S. Cyclin D1 is frequently overexpressed in microsatellite unstable colorectal cancer, independent of CpG island methylator phenotype. Histopathology 2008;53:588-598. 26. Aust De, terdiman JP, Willenbucher RF, Chang CG, Molinaro-Clark A, Baretton GB, Loehrs U, Waldman FM. the APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer 2002;94:1421-1427. 27. Ioachim ee, Katsanos KH, Michael MC, tsianos eV, Agnantis NJ. Immunohistochemical expression of cyclin D1, cyclin e, p21/waf1 and p27/kip1 in inflammatory bowel disease: correlation with other cell-cycle-related proteins (Rb, p53, ki-67 and PCNA) and clinicopathological features. Int J Colorectal Dis 2004;19:325-333. 28. Wong NA, Mayer NJ, Anderson Ce, McKenzie HC, Morris RG, Diebold J, Mayr D, Brock IW, Royds JA, Gilmour HM, Harrison DJ. Cyclin D1 and p21 in ulcerative colitis-related inflammation and epithelial neoplasia: a study of aberrant expression and underlying mechanisms. Hum Pathol 2003;34:580-588. 29. Xu X, Brodie SG, Yang X, Im YH, Parks Wt, Chen L, Zhou YX, Weinstein M, Kim SJ, Deng CX. Haploid loss of the tumor suppressor Smad4/Dpc4 initiates gastric polyposis and cancer in mice. Oncogene 2000;19:1868-1874. 30. terdiman JP, Aust De, Chang CG, Willenbucher RF, Baretton GB, Waldman FM. High resolution analysis of chromosome 18 alterations in ulcerative colitis-related colorectal cancer. Cancer Genet Cytogenet 2002;136:129-137. 31. Hill MJ, Melville DM, Lennard-Jones Je, Neale K, Ritchie JK. Faecal bile acids, dysplasia, and carcinoma in ulcerative colitis. Lancet 1987;2:185-186. 32. Miyaki M, Kuroki t. Role of Smad4 (DPC4) inactivation in human cancer. Biochem Biophys Res Commun 2003;306:799-804. 33. Haigis KM, Kendall KR, Wang Y, Cheung A, Haigis MC, Glickman JN, Niwa-Kawakita M, Sweet-Cordero A, Sebolt-Leopold J, Shannon KM, Settleman J, Giovannini M, Jacks t. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet 2008;40:600-608. 34. Aust De, Haase M, Dobryden L, Markwarth A, Lohrs U, Wittekind C, Baretton GB, tannapfel A. Mutations of the BRAF gene in ulcerative colitis-related colorectal carcinoma. Int J Cancer 2005;115:673-677. 35. Deng G, Bell I, Crawley S, Gum J, terdiman JP, Allen BA, truta B, Sleisenger MH, Kim YS. BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer. Clin Cancer Res 2004;10:191-195. 36. Burmer GC, Levine DS, Kulander BG, Haggitt RC, Rubin Ce, Rabinovitch PS. c-Ki-ras mutations in chronic ulcerative colitis and sporadic colon carcinoma. Gastroenterology 1990;99:416-420. 37. Delattre O, Olschwang S, Law DJ, Melot t, Remvikos Y, Salmon RJ, Sastre X, Validire P, Feinberg AP, thomas G. Multiple genetic alterations in distal and proximal colorectal cancer. Lancet 1989;2:353-356.

proefschrift Marian.indd 87

10-10-2010 20:56:24

88

| CHAPteR 6
38. Bleeker WA, Hayes VM, Karrenbeld A, Hofstra RM, Hermans J, Buys CC, Plukker Jt. Impact of KRAS and tP53 mutations on survival in patients with left- and right-sided Dukes C colon cancer. Am J Gastroenterol 2000;95:2953-2957. 39. Deng G, Kakar S, tanaka H, Matsuzaki K, Miura S, Sleisenger MH, Kim YS. Proximal and distal colorectal cancers show distinct gene-specific methylation profiles and clinical and molecular characteristics. eur J Cancer 2008;44:1290-1301. 40. elnatan J, Goh HS, Smith DR. C-KI-RAS activation and the biological behaviour of proximal and distal colonic adenocarcinomas. eur J Cancer 1996;32A:491-497. 41. Nagasaka t, Koi M, Kloor M, Gebert J, Vilkin A, Nishida N, Shin SK, Sasamoto H, tanaka N, Matsubara N, Boland CR, Goel A. Mutations in both KRAS and BRAF may contribute to the methylator phenotype in colon cancer. Gastroenterology 2008;134:1950-60, 1960. 42. toyota M, Ohe-toyota M, Ahuja N, Issa JP. Distinct genetic profiles in colorectal tumors with or without the CpG island methylator phenotype. Proc Natl Acad Sci U S A 2000;97:710-715. 43. Umetani N, Sasaki S, Masaki t, Watanabe t, Matsuda K, Muto t. Involvement of APC and K-ras mutation in non-polypoid colorectal tumorigenesis. Br J Cancer 2000;82:9-15.

proefschrift Marian.indd 88

10-10-2010 20:56:26

CHaPter 7
Methylation profiles of sporadic and colitis-associated colorectal cancer

MMH Claessen1, FP Vleggaar1, MeI Schipper2, JWJ Hinrichs, R Radersma2, PD Siersema1, GJA Offerhaus2
1

Department of Gastroenterology and Hepatology, University Medical Center Utrecht,


2

Department of Pathology, University Medical Center Utrecht manuscript in preparation

proefschrift Marian.indd 89

10-10-2010 20:56:27

90

| CHAPteR 7 Abstract
Introduction Patients with inflammatory bowel disease (IBD) are at an increased risk of developing colorectal cancer (CRC), and therefore undergo regular surveillance. Detailed knowledge on the carcinogenetic pathway will likely improve surveillance strategies and enables the development of biomarkers for risk stratification. aims & Methods the aim of this study was to determine differences in epigenetic alterations between IBD-related and sporadic CRCs. tissue samples from sporadic (n=17) and IBD-related (n=15) CRCs were collected. Following DNA isolation, the methylation percentage was determined using a quantitative method: Methylation-Specific Multiplex Ligationdependent Probe Amplification (MS-MLPA). Median methylation percentages were determined with a cut-off value of more than 15% being considered as methylation. results No differences were found in baseline characteristics between groups except that the IBD-CRC group was significantly younger compared to the sporadic group, 46 vs. 66 yrs (p<0.001). the number of samples with methylation as well as the median methylation percentages for ESR1, CDKN2B, and CDH13 were significantly higher in the sporadic than in the IBD-CRC group (p<0.001). A borderline significance was found for CD44 with 20% of the IBD-CRC samples showing >15% methylation compared to none of the sporadic group (p=0.092). the median cumulative methylation index was 197% and 326% in the IBD and sporadic tumour groups, respectively (p=0.051); however, this difference disappeared after age-adjustment (p=0.34). Conclusions epigenetic alterations occur less frequently in colitis-associated CRC compared to sporadic CRC. these results may well indicate a more important role for genetic than for epigenetic alterations in colitis-associated tumorigenesis

proefschrift Marian.indd 90

10-10-2010 20:56:27

Methylation profiles in CRC | 91

Introduction
Patients with inflammatory bowel disease (IBD) are at an increased risk of developing colorectal cancer (CRC). the inflammation-dysplasia-carcinoma sequence through which colitis-associated CRCs develops makes this cancer type suitable for surveillance programs with the ultimate goal early detection of neoplasia, thereby reducing CRC-related mortality. So far, the only histological marker indicating neoplastic transformation is dysplasia. Patients with longstanding colitis therefore undergo regular surveillance colonoscopy with random and targeted biopsies. Despite these surveillance strategies, an appreciable number of CRCs are missed or detected at an advanced stage 1. Moreover, endoscopic surveillance is a burden to patients and results in high healthcare costs 2. Furthermore, histological classification of dysplasia, especially low-grade dysplasia (LGD), has a high interobserver variability 3. the molecular process of colitis-associated carcinogenesis is an area of intensive research as it may provide biomarkers which potentially could help to identify patients at risk for CRC. Not only genetic alterations but also epigenetic phenomena play a prominent role in carcinogenesis. the best studied epigenetic modification is DNA methylation of cytosine residues by DNA methyltransferases. Over 50% of the protein encoding genes have at least one CpG island within or near their promoters, which are unmethylated in normal cells. In cancer, however, hypermethylation of these promoter regions is currently known as the most well-categorized epigenetic change, found in virtually every type of human neoplasm 4-6. Promoter hypermethylation leads to inactivation of genes involved in the cell cycle, cell adherence, DNA repair, and apoptosis. Its reversible nature makes methylation an attractive therapeutic target 7. In addition, methylation has been shown to have prognostic value and predicts treatment response. Above all, as methylation plays an early role in tumour development, a significant clinical benefit could be expected of methylation as a biomarker for early detection 8,9. earlier studies have reported a relation between inflammation and methylation in various tumours 10,11. the aim of this study was to determine the methylation profiles of colitis-associated CRC and sporadic CRC by quantitative analysis of a large gene panel.

Methods
Patients and tissue samples Paraffin embedded tissue specimens from surgical resection material were obtained from the Department of Pathology, University Medical Center Utrecht, the Netherlands. the study was carried out in accordance with the ethical guidelines

proefschrift Marian.indd 91

10-10-2010 20:56:27

92

| CHAPteR 7
and with approval of the research review committee of our institution 12. None of the patients had received chemotherapy or radiotherapy prior to surgery. tissue samples from sporadic CRC and colitis-associated CRC were collected. All inflammation-related tumours had developed in a background of chronic inflammation. A subgroup of these tumours was from patients who also have concurrent primary sclerosing cholangitis (PSC). the histological criteria by Riddell et al. for colitisassociated carcinoma were applied 13. Since colorectal carcinogenesis has not been shown to be different between Crohns disease (CD) and ulcerative colitis (UC), specimens from patients with either disease were included in this study 14. tumour stage was defined according the American Joint Committee on Cancer (AJCC) 15. Haematoxylin and eosin stained slides were reviewed by 2 pathologists to confirm diagnosis and to select representative cancer cell rich areas. DNa extraction the tissue was deparaffinized with xylene for 10 minutes followed by ethanol 100% and 70%, respectively. to obtain DNA from enriched tumour tissue (>75% tumour cells) 20 l of proteinase K (10 mg/ml) in 100 l buffer (0.5% tween 20, 50 mmol/ L tris (pH=9), 1mmol/L NaCl and 2 mmol/L eDtA) was used. After overnight incubation at 56 C, samples were incubated at 95 C for 10 minutes to inactivate the proteinase K and cooled on ice afterwards. the supernatant was transferred into a second tube after centrifugation. Samples were stored at -20 C. Methylation-Specific Multiplex Ligation-dependent Probe amplification (MSMLPa) MS-MLPA is a quantitative method for methylation profiling and has been described and validated previously 16. the SALSA MS-MLPA Me001-C1 lot 0808 kit (MRC Holland, Amsterdam, the Netherlands) contains 26 MS-MLPA probes with a digestion site for the methylation-specific Hha1 restriction enzyme which can be used to detect the methylation status of promoter regions of 24 different tumour suppressor genes (table 1). these tumour suppressor genes are frequently silenced by methylation in tumours, but are unmethylated in blood-derived DNA from healthy individuals. In addition, 15 reference probes are included in the kit which are not influenced by the Hha1 digestion. All MS-MLPA reactions were carried out and results were analysed according to the manufacturers instructions (www.mrcholland.com). In brief, approximately 100 ng isolated DNA in 5 l te buffer (10 mM tris 1 mM eDtA pH 7.5) was heat-denatured (10 minutes at 98 C) and incubated with the MLPA buffer and probe mix to hybridize to their respective targets at 60 C for 16 hours. After adding ligase buffer, samples were divided into two series of tubes and heated to 49 C. Ligase was added to the first series, whereas ligase together with

proefschrift Marian.indd 92

10-10-2010 20:56:27

Methylation profiles in CRC | 93

table 1. Genes in the MS-MLPa kit tumour suppressor-1 (MrC Holland) Gene TIMP3 APC CDKN2A MLH1 ATM RARB CDKN2B HIC1 CHFR BRCA1 CASP8 CDKN1B Name tissue inhibitor of metalloproteinase 3 Adenomatous polyposis coli Cyclin-dependent kinase inhibitor 2a (p14-ARF) MutL protein homolog 1 Ataxia telangiectasia mutated Retinoic acid receptor, beta Cyclin-dependent kinase inhibitor 2b (p15) Hypermethylated in cancer 1 Checkpoint with forkhead & ring finger domains Breast cancer 1, early onset Caspase 8 Cyclin-dependent kinase inhibitor 1B (p27, Kip1) Gene PTEN BRCA2 CD44 RASSF1 DAPK1 VHL ESR1 TP73 FHIT IGSF4 CDH13 GSTP1 Name Phosphatase and tensin homolog Breast cancer 2, early onset CD44 (Indian blood group) Ras associated domain family member 1 Death-associated protein kinase 1 Von Hippel-Lindau estrogen receptor 1 tumour protein p73 Fragile histidine triad gene Cell adhesion molecule 1 (CADM1) Cadherin 13, H-cadherin Glutathione S-transferase pi 1

the Hha1 restriction enzyme (5U) (Promega Corporation, Madison, WI, USA) was added to the second series. Simultaneous ligation and digestion (second series only) reactions were performed by incubating at 49 C for 30 minutes followed by heat inactivation at 98 C for 5 minutes. PCR buffer, PCR primers, enzyme dilution buffer and polymerase were added to 5 l of each ligation or digestion sample and amplified by 35 PCR cycles (95 C: 30 sec, 60 C: 30 sec, 72 C:1 minute, finished by 20 minutes at 72 C). Positive and negative controls were included. Samples were run on a 3130 xl Genetic Analyzer (Applied Biosystems, Foster City, CA, USA). the ratio of the two reactions per sample was converted to a methylation percentage, using the Coffalyser software (http://old.mlpa.com/coffalyser/). Statistical analysis All tumour samples with more than 15% of methylation were interpreted as hypermethylation (further denoted as methylation). this cut-off value is based on titration experiments in cell lines 17. Fishers exact test was applied to compare the baseline variables and percentage of samples with methylation between groups. Age was compared between groups using a non-parametric test (MannWhitney U test). A cumulative methylation index (CMI) was calculated by the sum of methylation percentages of all 24 genes shown in table 1 18. Differences in methylation

proefschrift Marian.indd 93

10-10-2010 20:56:27

94

| CHAPteR 7
percentages of individual genes and the CMI were analysed by the independent t-test (Student of Welch) or a non-parametric test (Mann Whitney U test), as appropriate. Correlation coefficients (Pearsons or Spearmans, as appropriate) were computed for assessing the association between methylation and other continuous variables. Linear regression was used for age-adjustment. A two-sided p-value <0.05 was interpreted as statistically significant. All statistical analyses were carried out with use of SPSS 15.0 for Windows.

Results
Clinicopathological characteristics the clinicopathological characteristics of all patients are summarized in table 2. Patients with sporadic CRC were significantly older than the inflammation-related group (p=0.000). No differences between groups were found in gender, proximal tumour location, or death. the sporadic group contained more tumours with an advanced tumour stage (AJCC >2b), however, this difference was not statistically significant. the majority of inflammation-related patients had ulcerative colitis (UC) in an extensive form. Primary sclerosing cholangitis (PSC) was present in 40% of the patients in the inflammation-related group. Methylation in colitis-associated compared to sporadic tumours the numbers of samples with more than 15% methylation are shown in table 3. All sporadic tumours showed more than 15% methylation of the estrogen receptor 1 (ESR1), which was significantly different from the inflammation-related samples (p=0.038). Identical results were found for Cyclin-dependent Kinase Inhibitor 2b (CDKN2B) (p=0.038). CD44 showed borderline significance with 20% of the inflammation-related samples showing methylation versus 0% in the sporadic group (p=0.092). None of the other genes showed differences in methylation between the two groups. table 4 shows the extent of methylation of the various genes in the two groups. the median CMI of the sporadic tumour group was 326% (range 151-688) which was (borderline) significantly higher compared to the inflammation-related group, 197% (range 72-536) (p=0.051). Again the individual genes, ESR1 and CDKN2B showed a significant difference in methylation with a median of 58.5% and 45.5% in the sporadic tumour group, and 28.3% and 28.5% in the inflammation-related group, respectively (p=0.000 and p=0.003). CDH13 methylation was also significantly higher in the sporadic group with a median of 47.4% versus 31.8% in IBD-CRCs (p=0.037). No other genes showed significant differences.

proefschrift Marian.indd 94

10-10-2010 20:56:27

Methylation profiles in CRC | 95

Methylation and clinicopathological characteristics the extent of methylation (CMI) was positively correlated to age (Pearsons correlation coefficient: .339; p=0.057). After age-adjustment using linear regression the statistically significant difference in CMI between colitis-associated and sporadic CRCs disappeared (p=0.34). the mean difference in CMI between groups was 94.6% and after age-adjustment 59.7 %. the ESR1 gene remained however statistically significant after age-adjustment with a mean difference of 30% before and 22% after age-adjustment (p=0.034). CDKN2B also remained significantly different with a mean difference of 16% after age correction (p=0.032). CDH13 was not statistically different after age-correction with a mean difference of 13% (p=0.19). table 5 shows median methylation percentages of all samples and clinicopathological characteristics. Methylation was not influenced by gender. Some genes were potentially more methylated with borderline p-values in the PSC group. Several genes and the CMI showed more methylation in proximally located tumours (p-value= 0.006). tumour stage did not clearly influence methylation, although CD44 showed slightly more methylation in lower stages (p= 0.011).

table 2. Baseline characteristics of inflammation-related and sporadic colorectal tumours Inflammation-CRC n=15 Age, yrs. (SD) Gender (male, %) AJCC stage >21 Proximal tumour location (%) IBD (UC2) Pancolitis PSC (%) Death (%)
a

Sporadic CRC n=17 65 10 15 8 na na na 4 (25%) (11) (59%) (88%) (47%)

total n=32 56 19 20 14 na na na 10 (32%) (15) (59%) (67%) (44%)

p-value <0.01 a 1.00 b 0.07 b 0.74 b na na na 0.46 b

46 9 5 6 9 67% 6 6

(12) (60%) (39%) (40%) (60%) (40%) (40%)

Independent t-test, b Fishers exact, 1 AJCC >2: advanced tumour stages. 2 Ulcerative Colitis (%). na: not applicable

proefschrift Marian.indd 95

10-10-2010 20:56:27

96

| CHAPteR 7
table 3. Number of samples with more than 15% methylation of specific genes per group. Only genes showing more than 15% methylation in at least one tumour are shown Genes InflammationCRC (%) n=15 TP73 CASP8 RARB MLH1 FHIT APC ESR1 CDKN2A CDKN2B DAPK1 PTEN CD44 GSTP1 ATM IGSF4 CHFR BRCA2 CDH13 HIC1 BRCA1 TIMP73
a

Sporadic CRC (%) n=17 2 0 4 4 0 8 17 1 17 4 0 0 3 0 5 8 0 15 2 0 4 (24%) (88%) (12%) (29%) (47%) (18%) (47%) (100%) (6%) (100%) (24%) (24%) (12%) (12%)

total (%) n=32 3 2 8 5 1 13 28 2 28 7 2 3 5 2 8 12 1 27 3 1 8 (9%) (3%) (25%) (8%) (3%) (41%) (88%) (6%) (88%) (22%) (6%) (9%) (16%) (6%) (25%) (38%) (3%) (84%) (9%) (3%) (25%)

p-value a

1 1 4 1 1 5 11 1 11 3 2 3 2 2 3 4 1 12 1 1 4

(7%) (7%) (27%) (3%) (7%) (33%) (73%) (7%) (73%) (20%) (13%) (20%) (13%) (13%) (20%) (27%) (7%) (80%) (7%) (7%) (27%)

1.00 0.47 1.00 0.36 0.47 0.49 0.038 1.00 0.038 1.00 0.21 0.092 1.00 0.21 0.69 0.29 0.47 0.65 1.00 0.47 1.00

Fishers exact test

proefschrift Marian.indd 96

10-10-2010 20:56:27

Methylation profiles in CRC | 97

table 4. Median promoter methylation percentage (range) by MS-MLPA for 24 tumour suppressor genes in inflammation-related and sporadic tumours and sporadic tumours Inflammation-CRC, n=15 Genes TP73 CASP8 VHL RARB MLH1 RASSF1 FHIT APC ESR1 CDKN2A CDKN2B DAPK1 PTEN CD44 GSTP1 ATM IGSF4 CDKN1B CHFR BRCA2 CDH13 HIC1 BRCA1 TIMP73 CMI Mean (SD) 8.2 (15.5) 5.4 (5.3) 1.8 (3.9) 12.6 (7.6) 4.5 (5.3) 3.3 (4.2) 4.1 (13.7) 12.7 (15.5) 28.3 (20.5) 7.9 (6.7) 28.5 (15.3) 6.3 (10.2) 9.8 (5.6) 8.5 (7.6) 9.1 (14.0) 6.8 (7.6) 7.5 (12.3) 2.5 (4.1) 7 (10.5) 6.7 (5.8) 31.8 (16.5) 5.1 (4.8) 4.3 (5.9) 8.4 (13.4) 238.8 (124.5) Median (range) 0 (0-61) 5 (0-20) 0 (0-12) 9 (4-28) 2.5 (0-19) 0 (0-11) 0 (0-53) 10 (0-53) 30 (0-72) 7 (0-28) 25 (8-59) 0 (0-28) 8 (4-25) 7 (0-24) 8 (0-53) 4 (0-25) 0 (0-38) 0 (0-12) 0 (0.31) 6 (0-23) 32 (0-62) 5 (0-15) 3 (0-23) 3 (0-45) 197 (72-536) Sporadic CRC, n=17 Mean (SD) 8.4 (13.6) 2.8 (2.0) 0.5 (1.13) 17.8 (22.5) 9.3 (17.5) 4.2 (3.6) 0.2 (0.73) 19.3 (19.7) 58.5 (22.0) 6.9 (4.8) 45.5 (14.3) 7.5 (9.6) 7.0 (2.7) 6.2 (3.9) 8.8 (7.2) 4.3 (3.0) 14.7 (18.2) 1.3 (2.1) 21.5 (26.6) 5.4 (2.6) 47.4 (23.0) 8.8 (12.5) 2.9 (2.3) 10.8 (14.1) 333.4 (137.5) Median (range) 7 (0-57) 3 (0-6) 0 (0-4) 8 (0-78) 4.5 (0-72) 5 (0-12) 0 (0-3) 11 (0-74) 53 (21-96) 6 (0-21) 40 (24-76) 4 (0-33) 7 (0-13) 6 (0-11) 9 (0-26) 4 (0-10) 8 (0-59) 0 (0-6) 9 (0-77) 5 (0-10) 50 (11-89) 6 (0-45) 3 (0-6) 9 (0-55) 326 (151-688) p-valuea 0.81 0.088 0.68 0.47 0.37 0.29 0.35 0.31 0.000 0.61 0.003 0.57 0.08 0.28 0.94 0.25 0.12 0.77 0.16 0.42 0.037 0.29 0.40 0.63 0.051

a differences in methylation between groups were analysed by unpaired t-test (student or Welch t-test) or nonparametric test (mann-whitney U test) as appropriate.

proefschrift Marian.indd 97

10-10-2010 20:56:28

98

| CHAPteR 7
table 5. Median methylation percentage (range) and clinicopathological characteristics of patients with inflammation-related and sporadic CRC
Genes Gender Male (n=19) Female (n=13) PSC No (n=26) Yes (n=6) p-value Location Distal (n=18) 0 (0-12) 3 (0-20) 4 (0-9) NS 0 (0-12) 0 (0-8) NS 8 (0-28) 10 (4-78) 0.065 0 (0-3) 0 (0-53) NS 9 (0-53) 11 (0-74) NS 36 (0-87) 53 (27-96) 0.089 7 (0-28) 7 (0-13) NS 36 (8-76) 35 (9-63) NS 3 (0-57) 10 (0-61) NS 3 (0-20) 9 (0-9) 0.047 0 (0-7) 0 (0-12) 0.079 8 (0-78) 11 (7-24) NS 0 (0-3) 0 (0-53) 0.030 9 (0-74) 13 (7-37) NS 44 (0-96) 38 (0-72) NS 7 (0-28) 8 (6-10) NS 35 (8-76) 37 (30-59) NS 7 (0-61) 0 (0-13) 4 (0-9) 3 (0-20) 0 (0-12) 0 (0-8) 9 (4-78) 8 (0-59) 0 (0-3) 0 (0-53) 9 (0-74) 11 (0-40) 37 (0-96) 41 (0-87) 7 (0-21) 7 (0-28) 34 (8-63) 45 (12-76) TP73 CASP8 VHL RARB FHIT APC ESR1 CDKN2A CDKN2B

Proximal (n=14) 8 (0-61) p-value tumour stage <2b (n=10) >2b (n=20) p-value 8 (0-61) 0 (0-57) NS NS

4 (0-20) 3 (0-8) NS

0 (0-7) 0 (0-12) NS

9 (4-23) 8 (0-78) NS

na 0 (0-53) NS

7 (0-40) 11 (0-74) NS

33 (0-72) 51 (0-96) NS

8 (0-28) 7 (0-13) NS

30 (9-76) 40 (8-63) NS

Genes Gender Male (n=19) Female (n=13) PSC No (n=26) Yes (n=6) p-value Location Distal (n=18)

DAPK1

PTEN

CD44

GSTP1

ATM

IGSF4

CKN1B

CHFR

BRCA2

3 (0-25) 0 (0-33)

8 (4-10) 8 (0-25)

7 (0-24) 5 (0-22)

8 (0-17) 9 (0-53)

5 (0-25) 3 (0-22)

4 (0-59) 3 (0-40)

0 (0-12) 0 (0-10)

3 (0-77) 15 (0-51)

7 (0-10) 4 (0-23)

0 (0-33) 5 (0-28) NS

8 (0-25) 8 (5-19) NS

6 (0-22) 8 (0-24) NS

9 (0-26) 12 (0-53) 0.063

4 (0-25) 6 (0-12) NS

3 (0-59) 9 (0-38) NS

0 (0-6) 4 (0-12) 0.072

2 (0-77) 11 (0-31) NS

6 (0-23) 7 (0-13) NS

2 (0-33)

8 (0-25) 8 (4-19) NS

7 (0-22) 6 (4-24) NS

0 (0-26) 11 (0-53) 0.049

4 (0-25) 5 (0-12) NS

2 (0-21) 16 (0-59) 0.057

0 (0-12) 0 (0-10) NS

0 (0-24) 21 (0-77) 0.011

6 (0-23) 7 (0-13) NS

Proximal (n=14) 0 (0-28) p-value tumour stage <2b (n=10) >2b (n=20) p-value 2 (0-33) 0 (0-20) NS NS

8 (4-25) 8 (0-14) NS

8 (4-24) 5 (0-11) 0.011

8 (0-12) 9 (0-53) NS

4 (0-22) 5 (0-25) NS

4 (0-38) 3 (0-59) NS

0 (0-7) 0 (0-12) NS

2 (0-24) 5 (0-77) NS

6 (0-23) 6 (0-10) NS

proefschrift Marian.indd 98

10-10-2010 20:56:28

Methylation profiles in CRC | 99

Genes Gender Male (n=19) Female (n=13) PSC No (n=26) Yes (n=6) p-value Location Distal (n=18) Proximal (n=14) p-value tumour stage <2b (n=10) >2b (n=20) p-value

CDH13

HIC1

BRCA1

TIMP3

SUM

40 (0-89) 37 (14-71)

6 (0-45) 5 (0-15)

4 (0-6) 3 (0-23)

5 (0-55) 0 (0-45)

243 (72-688) 262 (151-536)

39 (0-89) 38 (17-52) NS

5 (0-45) 6 (0-15) NS

3 (0-23) 4 (0-11) NS

4 (0-55) 9 (0-45) NS

237 (72-688) 279 (183-536) NS

32 (0-69) 50 (24-89) 0.004

4 (0-12) 6 (0-45) NS

2 (0-23) 4 (0-11) 0.073

3 (0-15) 10 (0-55) NS

215 (72-369) 358 (107-688) 0.006

35 (10-62) 42 (0-89) NS

6 (0-12) 5 (0-45) NS

3 (0-23) 3 (0-6) NS

7 (0-31) 3 (0-55) NS

252 (107-466) 263 (72-688) NS

Discussion
In this study, we found a statistically borderline significant difference in methylation detected by MS-MLPA using a large gene panel when comparing sporadic and inflammation-related tumours, which disappeared after age-adjustment. Although methylation has been reported to be associated with inflammation, we found less methylation in inflammation-related tumours 19,20. Similar results were reported by Konishi et al. who compared methylation levels in dysplastic and tumour samples of IBD patients with those of sporadic CRC 21. they also found a higher methylation level in sporadic CRCs compared to IBD-related CRCs. More importantly, a higher degree of methylation was detected in dysplastic IBD-mucosa compared to the IBD-related CRCs samples. Apparently, genetic changes and not epigenetic alterations largely determine progression to malignancy in inflamed epithelium. All samples from the sporadic tumour group showed methylation of the ESR1 gene and also, their median methylation percentage was significantly higher than the colitis-associated group. the ESR1 gene is one of the genes, that shows increased methylation with aging, which is also known as age-related methylation 22. this gene encodes an estrogen receptor, a ligand-activated transcription factor composed of several domains which are important for hormone binding, DNA binding and activation of transcription 23. Age-related methylation predisposes genes to DNA methylationassociated inactivation 8. Several studies have reported an increase, varying from 25-40%, in methylation of the ESR1 gene in colitis-associated cancers and adjacent

proefschrift Marian.indd 99

10-10-2010 20:56:28

100

| CHAPteR 7
normal tissue 24,25. In our study, we found less methylation of the ESR1 gene in IBDrelated tumours in contrast to expectations that in UC epigenetic damage would be more pronounced as a consequence of chronic inflammation and the associated increased levels of exogenous carcinogens and reactive oxygen species. We found that even after age-adjustment the difference remained significant. Another significant difference in methylation was found in Cyclin-dependent Kinase Inhibitor 2B (CDKN2B, also known as p15INK4b). CDKN2B blocks progression in the G1 phase of the cell cycle by binding to Cyclin-dependent Kinases 4 and 6 (CDK4/6). When inactivated, CDK4/6 binds to Cyclin D1 and activates phosphorylation and progression into the S-phase 26. Methylation of CDKN2B is well documented in haematological malignancies, but is less consistently reported in CRC. Several studies have not found evidence for aberrant methylation of CDKN2B in CRC 27-30, whereas other studies reported high numbers of CRCs with extensive methylation of CDKN2B 31,32. It is known that age influences methylation and indeed, when adjusted for age, the differences in CMI were no longer significant. the mean difference in methylation, however, remained still 60%, even after correction, indicating that sample size may have been a limitation in the current study. A larger study would also enable to look more carefully to the influence of tumour location since our results suggest that methylation decreases from the proximal towards the distal colorectum, a gradient that is well known in the normal colorectum as well. Another interesting finding is the possible association of an increased methylation percentage in patients with PSC compared to non-PSC samples. Hypothetically speaking, concurrent PSC may well influence methylation profiles through an altered bile acid composition. It is important to emphasize that in this study a quantitative technique (MS-MLPA) was used. It was therefore possible to evaluate the level and incidence of methylation of a single gene, as well as that of a gene panel, whereas methylation-specific PCR (MSP), which has been used widely in previous studies, is limited by the fact that it provides only binary qualitative results. Due to this technical limitation, MSP cannot reliably distinguish between low and high levels of methylation. Suijkerbuijk et al. reported a poor concordance between quantitative techniques and the nonquantitative technique of MSP, which questions the validity of MSP for accurate methylation detection in clinical samples and makes comparison between studies difficult (Suijkerbuijk et al. (unpublished results)33. the different methodology may have contributed to the discrepant results obtained. In summary, the extent of methylation in a commonly used gene panel is lower in colitis-associated than in sporadic tumours. these results may well imply that the role of methylation in colitis-associated carcinogenesis appears less significant than in sporadic CRCs.

proefschrift Marian.indd 100

10-10-2010 20:56:28

Methylation profiles in CRC | 101

Reference List
1. Lutgens MW, Vleggaar FP, Schipper Me, Stokkers PC, van der WJ, Hommes DW, de Jong DJ, Dijkstra G, van Bodegraven AA, Oldenburg B, Samsom M. High Frequency of early Colorectal Cancer in Inflammatory Bowel Disease. Gut 2008. 2. Mitchell PJ, Salmo e, Haboubi NY. Inflammatory bowel disease: the problems of dysplasia and surveillance. tech Coloproctol 2007;11:299-309. 3. eaden J, Abrams K, McKay H, Denley H, Mayberry J. Inter-observer variation between general and specialist gastrointestinal pathologists when grading dysplasia in ulcerative colitis. J Pathol 2001;194:152-157. 4. Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP. Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res 1998;72:141-196. 5. Baylin SB, Herman JG. DNA hypermethylation in tumorigenesis: epigenetics joins genetics. trends Genet 2000;16:168-174. 6. Jones PA, Laird PW. Cancer epigenetics comes of age. Nat Genet 1999;21:163-167. 7. Mulero-Navarro S, esteller M. epigenetic biomarkers for human cancer: the time is now. Crit Rev Oncol Hematol 2008;68:1-11. 8. Issa JP, Ahuja N, toyota M, Bronner MP, Brentnall tA. Accelerated age-related CpG island methylation in ulcerative colitis. Cancer Res 2001;61:3573-3577. 9. Suijkerbuijk KP, van Diest PJ, van der We. Improving early breast cancer detection: focus on methylation. Ann Oncol 2010. 10. eads CA, Lord RV, Wickramasinghe K, Long tI, Kurumboor SK, Bernstein L, Peters JH, DeMeester SR, DeMeester tR, Skinner KA, Laird PW. epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res 2001;61:3410-3418. 11. Kang GH, Lee S, Kim WH, Lee HW, Kim JC, Rhyu MG, Ro JY. epstein-barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. Am J Pathol 2002;160:787-794. 12. van Diest PJ. No consent should be needed for using leftover body material for scientific purposes. BMJ 2002;325:648-651. 13. Riddell RH, Goldman H, Ransohoff DF, Appelman HD, Fenoglio CM, Haggitt RC, Ahren C, Correa P, Hamilton SR, Morson BC, . Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983;14:931-968. 14. Farraye FA, Odze RD, eaden J, Itzkowitz SH. AGA technical review on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology 2010;138:746-74, 774. 15. AJCC cancer staging manual. New York (NY): 2002. 16. Nygren AO, Ameziane N, Duarte HM, Vijzelaar RN, Waisfisz Q, Hess CJ, Schouten JP, errami A. Methylation-specific MLPA (MS-MLPA): simultaneous detection of CpG methylation and copy number changes of up to 40 sequences. Nucleic Acids Res 2005;33:e128. 17. Gylling A, bdel-Rahman WM, Juhola M, Nuorva K, Hautala e, Jarvinen HJ, Mecklin JP, Aarnio M, Peltomaki P. Is gastric cancer part of the tumour spectrum of hereditary non-polyposis colorectal cancer? A molecular genetic study. Gut 2007;56:926-933. 18. Suijkerbuijk KP, Fackler MJ, Sukumar S, van Gils CH, van Lt, van der We, Vooijs M, van Diest PJ. Methylation is less abundant in BRCA1-associated compared with sporadic breast cancer. Ann Oncol 2008;19:1870-1874. 19. Shen L, Ahuja N, Shen Y, Habib NA, toyota M, Rashid A, Issa JP. DNA methylation and environmental exposures in human hepatocellular carcinoma. J Natl Cancer Inst 2002;94:755-761. 20. toyooka S, Maruyama R, toyooka KO, McLerran D, Feng Z, Fukuyama Y, Virmani AK, ZochbauerMuller S, tsukuda K, Sugio K, Shimizu N, Shimizu K, Lee H, Chen CY, Fong KM, Gilcrease M, Roth JA, Minna JD, Gazdar AF. Smoke exposure, histologic type and geography-related differences in the methylation profiles of non-small cell lung cancer. Int J Cancer 2003;103:153-160.

proefschrift Marian.indd 101

10-10-2010 20:56:28

102

| CHAPteR 7
21. Konishi K, Shen L, Wang S, Meltzer SJ, Harpaz N, Issa JP. Rare CpG island methylator phenotype in ulcerative colitis-associated neoplasias. Gastroenterology 2007;132:1254-1260. 22. Issa JP. CpG-island methylation in aging and cancer. Curr top Microbiol Immunol 2000;249:101-118. 23. http://www.genecards.org/cgi-bin/carddisp.pl?gene=eSR1&search=eSR1. 2010. 24. Fujii S, tominaga K, Kitajima K, takeda J, Kusaka t, Fujita M, Ichikawa K, tomita S, Ohkura Y, Ono Y, Imura J, Chiba t, Fujimori t. Methylation of the oestrogen receptor gene in non-neoplastic epithelium as a marker of colorectal neoplasia risk in longstanding and extensive ulcerative colitis. Gut 2005;54:1287-1292. 25. tominaga K, Fujii S, Mukawa K, Fujita M, Ichikawa K, tomita S, Imai Y, Kanke K, Ono Y, terano A, Hiraishi H, Fujimori t. Prediction of colorectal neoplasia by quantitative methylation analysis of estrogen receptor gene in nonneoplastic epithelium from patients with ulcerative colitis. Clin Cancer Res 2005;11:8880-8885. 26. Jeffrey PD, tong L, Pavletich NP. Structural basis of inhibition of CDK-cyclin complexes by INK4 inhibitors. Genes Dev 2000;14:3115-3125. 27. Bai AH, tong JH, to KF, Chan MW, Man eP, Lo KW, Lee JF, Sung JJ, Leung WK. Promoter hypermethylation of tumor-related genes in the progression of colorectal neoplasia. Int J Cancer 2004;112:846-853. 28. Dong SM, Lee eJ, Jeon eS, Park CK, Kim KM. Progressive methylation during the serrated neoplasia pathway of the colorectum. Mod Pathol 2005;18:170-178. 29. Herman JG, Jen J, Merlo A, Baylin SB. Hypermethylation-associated inactivation indicates a tumor suppressor role for p15INK4B. Cancer Res 1996;56:722-727. 30. Lin SY, Yeh Kt, Chen Wt, Chen HC, Chen St, Chiou HY, Chang JG. Promoter CpG methylation of tumor suppressor genes in colorectal cancer and its relationship to clinical features. Oncol Rep 2004;11:341-348. 31. Ishiguro A, takahata t, Saito M, Yoshiya G, tamura Y, Sasaki M, Munakata A. Influence of methylated p15 and p16 genes on clinicopathological features in colorectal cancer. J Gastroenterol Hepatol 2006;21:1334-1339. 32. Xu XL, Yu J, Zhang HY, Sun MH, Gu J, Du X, Shi DR, Wang P, Yang ZH, Zhu JD. Methylation profile of the promoter CpG islands of 31 genes that may contribute to colorectal carcinogenesis. World J Gastroenterol 2004;10:3441-3454. 33. Jeuken JW, Cornelissen SJ, Vriezen M, Dekkers MM, errami A, Sijben A, Boots-Sprenger SH, Wesseling P. MS-MLPA: an attractive alternative laboratory assay for robust, reliable, and semiquantitative detection of MGMt promoter hypermethylation in gliomas. Lab Invest 2007;87:10551065.

proefschrift Marian.indd 102

10-10-2010 20:56:29

CHaPter 8
Wnt-pathway activation in IBD-associated colorectal carcinogenesis: potential biomarkers for surveillance

MMH Claessen1, MeI Schipper2, B Oldenburg1, PD Siersema1, GJA Offerhaus2, FP Vleggaar1


1

Department of Gastroenterology and Hepatology, 2Department of Pathology, University Medical Center Utrecht Cellular Oncology 2010; 32:303-310

proefschrift Marian.indd 103

10-10-2010 20:56:31

104

| CHAPteR 8 Abstract
Background the Wnt-pathway dominates the sporadic carcinogenesis whereas p53 plays a pivotal role in the colitis-associated counterpart. the expression of Wnt-signaling proteins and p53 during colitis-associated carcinogenesis was determined. Methods A tissue microarray was constructed with colonic samples from 5 groups of patients: controls (C, n=10), IBD without neoplasia (IBD, n=12), non-dysplastic IBD with neoplasia elsewhere in the colon (IBD-Ne, n=12), dysplastic lesion in IBD (IBD-DYS, n=12), and IBD-associated colorectal cancer (IBD-CRC, n=10). Immunohistochemistry was performed for -catenin, Cyclin D1, and p53. P53 sequence analysis was performed in some cases. results Nuclear -catenin expression was found in 0%, 0%, 50%, 55%, and 100% of the patients in the C-, IBD-, IBD-Ne-, IBD-DYS-, and IBD-CRC-groups, respectively. Nondysplastic IBD mucosa with neoplasia detected elsewhere showed nuclear expression in 50% of the cases compared to 0% in IBD mucosa without neoplasia (p=0.02). Cyclin D1 staining had similar expression patterns. Overexpression of p53 was only detected in the IBD-DYS (66.7%) and IBD-CRC groups (50%). Conclusion In contrast to previous findings, our results suggest activation of the Wnt- pathway in the early phase of colitis-associated carcinogenesis. Furthermore, as Wnt activation was observed in 50% of the IBD-Ne cases, nuclear -catenin may facilitate detection of neoplasia.

proefschrift Marian.indd 104

10-10-2010 20:56:31

Potential biomarkers for colonic surveillance | 105

Introduction
It is well established that patients with extensive and longstanding inflammatory bowel disease (IBD) have a higher life-time risk of developing colorectal cancer (CRC) than subjects in the general population 1. In order to prevent IBD-related CRC, patients are offered surveillance colonoscopies. However, colonoscopic surveillance in IBD is hampered by the fact that IBD-related carcinogenesis follows an inflammation-dysplasia-carcinoma sequence instead of the adenoma-carcinoma sequence as is seen in sporadic CRC, which makes endoscopic discerning of premalignant lesions far more difficult 2. Furthermore, colonoscopic surveillance in IBD is invasive, time consuming and its effectiveness has still to be proven. therefore, biomarker-assisted surveillance may potentially be a valuable additional diagnostic tool in the detection of premalignant lesions in IBD. the molecular pathogenesis of colitis-associated carcinogenesis differs from its sporadic counterpart. Previous data suggested that Wnt-pathway activation dominates early sporadic carcinogenesis, whereas Wnt activation occurs less frequently and, if occurring at all, at a late stage of colitis-associated carcinogenesis3. In contrast, mutations in the tumour suppressor gene p53 occur frequently in the early stages of colitis-associated carcinogenesis, whereas in sporadic CRC p53 mutations are only observed in advanced colonic neoplasia 4,5. An intriguing observation in this respect is that p53 mutations have been observed in nondysplastic IBD colonic mucosa adjacent to dysplastic areas, which may be a feature of colonic field cancerization 6,7. Comparable data on Wnt-pathway activation in nondysplastic colonic mucosa in patients with IBD-associated neoplastic lesions are not available. Recognition of these field related biomarkers of neoplastic progression may facilitate the detection of early neoplasia in case of surveillance. the aims of the current study were therefore to determine the expression of Wnt-signaling proteins and p53 during colitis-associated carcinogenesis and to assess their use as biomarkers of colonic field cancerization in longstanding colitis.

Methods
Patients and tissue samples Paraffin embedded tissue specimens from endoscopic biopsies and surgical resection material were obtained from the Department of Pathology of the University Medical Center Utrecht, the Netherlands. the study was carried out in accordance with the ethical guidelines of this centre concerning informed consent about the use of patients materials after surgical procedures. tissue samples of five groups of patients were collected. the first group (C) consisted of controls with complaints of

proefschrift Marian.indd 105

10-10-2010 20:56:31

106

| CHAPteR 8
abdominal pain or diarrhoea (n=10), in whom colonoscopy was performed to rule out pathology. Histological examination of these biopsies showed normal mucosa. In the second group (IBD) IBD-patients with chronically inflamed colonic mucosa without neoplasia were included (n=12). the third group (IBD-Ne) consisted of IBDpatients with neoplasia (eight CRC and four dysplasia) detected on colonoscopy. From these patients non-dyplastic mucosa was studied with a median distance between the biopsies and the neoplastic lesion of 11 cm (range: 2-78 cm). the fourth group (IBD-DYS) consisted of patients with IBD-related dysplastic lesions, six with low-grade dysplasia (LGD) and six with high-grade dysplasia (HGD). the fifth group (IBD-CRC) consisted of patients with IBD-related colorectal adenocarcinomas (n=10). the histological criteria by Riddell et al. 8 for colitis-associated dysplasia and carcinoma were applied. Indefinite for dysplasia cases were excluded. Since there is no robust evidence of a difference in carcinogenesis between Crohns disease (CD) and ulcerative colitis (UC), biopsies from patients with either disease were included in this study 2. tissue Microarray A tissue microarray (tMA) was constructed according to a standard protocol described by Kononen et al. 9. Specific parts of the samples were selected on corresponding haematoxylin and eosin (H&e) stained slides after which three core biopsies (diameter: 0.6 mm) were taken from each sample and punched into a recipient block using a manual tissue micro arrayer (Beecher instruments, Sun Prairie, USA). the slides were cut from the tMA with a microtome (Leica Microsystems, Wetzlar, Germany). Staining with antibodies was performed in triplicate, resulting in a maximum of nine tissue cores per patient. Immunohistochemistry Immunohistochemistry (IHC) was performed using monoclonal antibodies for p53 (Biogenex, San Ramon, USA, BP5312-1; 1:200, Citrate, automatically stained by Bond, Biovision Systems/Leica Microsystems, Wetzlar, Germany), -catenin (BD transduction Laboratories, Lexington, USA, clone 14; 1:5000, ARS, overnight incubation at 4 C), and Cyclin D1 (Neomarkers, Suffolk, UK, clone SP4 rabbitmonoclonal, 1:100, ARS, overnight incubation at 4 C). Briefly, 4m sections were deparaffinized, blocked for endogenous peroxidase activity by immersion with 0.3% H2O2 in methanol for 20 min. Antigen retrieval was performed in tris/eDtA buffer (10 mM/1 mM; pH 9.0) for 10 min at 120 C (using a bench autoclave). After cooling for 10 minutes and washing in PBS, non-specific binding sites were blocked in PBS with 5% normal goat serum for 10 minutes, followed by antibody incubation as indicated previously. Antibody binding was visualized using Powervision+ polyHRP detection system (Immuno Vision technologies Co., Brisbane, USA). the

proefschrift Marian.indd 106

10-10-2010 20:56:31

Potential biomarkers for colonic surveillance | 107

chromogen used for -catenin staining was 3.3-DiAmino Benzidine tetrachloride, DAB (Sigma, Missouri, USA) and for Cyclin D1 staining PowerDAB (Immuno Vision technologies Co., Brisbane,USA). Sections were counterstained with H&e. Staining interpretation the specimens were considered positive for p53 if the epithelial cells exhibited intense brown nuclear staining in the entire tissue sample or segments thereof (more than 20% positive cells) 10,11 A faint scattered staining pattern of p53 should not be considered as a marker of gene mutation or inactivation 11,12. Interpretation of cyclin D1 staining was based on intensity and divided into three categories: negative or weakly positive (-), positive (+), and strongly positive (++). Both the positive and strongly positive samples were regarded as cyclin D1 overexpression13. -Catenin staining was evaluated in the cell membrane and nucleus. A membranous staining pattern, as can be observed in normal cells was considered negative, whereas a reduced membranous expression accompanied by increased cytoplasmic and nuclear staining was deemed positive 14. the percentage of nuclear stained cells was semi-quantitatively analysed and divided into three categories: low (<10% of cells), moderate (10-50% of cells), or high (>50% of cells). If more than 10% of cells showed positive nuclear staining, this was interpreted as indicative of Wnt activation 15,16. All slides were evaluated by two observers conjointly (MS and MC). Discordant cases were reviewed by a second experienced gastrointestinal pathologist (JO). P53 Sequence analysis On rare occasions a stop codon (null) mutation of p53 can be associated with completely negative staining due to abrogation of translation of a stable protein fragment 17. When this was suspected, sequence analysis was performed to verify the mutation. Prior to DNA isolation, haematoxylin and eosin-stained histological slides were screened for the presence of at least 40% of tumour cells. DNA was extracted from formalin-fixed tissue sections of 15 m thickness. these were deparaffinized with xylene, rehydrated in serial-graded water-ethanol solution (100% and 70%, respectively) and rinsed in deionised water. Subsequently, DNA was extracted by incubation at 55 C in 1 ml extraction buffer overnight (100 mM NaCl, 10 mM trisHCl pH 8.0, 0.25 M eDtA pH 8.0, 0.5% SDS) to which 20 l proteinase K (10 mg/ ml) was added (Roche Diagnostics, Basel, Switzerland). Phenol-chloroform purification was performed and DNA precipitation was established by ethanol. exons 4 through 9 were amplified in seven different PCRs. For the amplification of DNA from paraffin sections, 1 U Ampli taq Gold DNA polymerase (Perkin elmer, Norwalk, Ct, USA) was used with 2 mM MgCl2. the first PCR was performed with

proefschrift Marian.indd 107

10-10-2010 20:56:31

108

| CHAPteR 8
primers without the sequencing templates. In the second PCR with 1l of the first amplified product, primers were used with sequencing templates. the PCR products were sequenced and were analysed subsequently as described previously 18. the mutated codons were numbered from the first coding nucleotides. Statistical analysis the Fishers exact test was used to determine the statistical significance of differences in expression between groups. Due to the heterogeneity within a tumour sample, the three cores taken from a biopsy were considered independently. the tissue core with the highest score of the three slides was selected, resulting in three values per patient 19. Statistical analysis was performed using SPSS Version 12.0 Statistical Software. A p-value <0.05 was considered statistically significant.

Results
Clinicopathological characteristics the clinicopathological characteristics of patients are listed in table 1. No statistically significant differences in age, gender or IBD phenotype and disease duration were observed between the five groups.

table 1. Clinical characteristics Variables Mean age, yrs (SD) Gender % male IBD, % UC* extent IBD, % pancolitis *
1

C (n=10) 45 (16) 50% na na na na na na

IBD (n=12) 48 (11) 42% 92% 67% 17 (7) 33% na na

IBD-Ne (n=12) 42 (12) 58% 50% 92% 15 (10) 33% 58% 11 (2-78)

IBD-DYS (n=12) 45 (14) 58% 58% 92% 13 (9) 25% 42% na

IBD-CRC (n=10) 44 (13) 60% 70% 80% 18 (10) 20% 90% na

Mean IBD duration, yrs (SD)* PSC, %* Location neoplasm, % distal 2 Distance to neoplasm, cm
3

C, controls; IBD, inflammatory bowel disease; IBD-Ne, inflammatory bowel disease and a neoplastic lesion (of which 67% CRC) elsewhere in the colon; DYS, colitis-related dysplasia; CRC, colitis-related colorectal cancer; PSC, primary sclerosing cholangitis; n/a, not applicable. 1 extent colitis grouped in pancolitis or left-sided colitis (below the splenic flexure), 2 Distal location: distal of the splenic flexure, 3 Distance expressed as median (range), * No statistically significant differences were observed between groups

proefschrift Marian.indd 108

10-10-2010 20:56:31

Potential biomarkers for colonic surveillance | 109

expression of -catenin Increased expression of nuclear -catenin was found in 50% of patients in the IBDNe group, in 55% of patients in the IBD-DYS group, and in 100% of the IBD-CRC cases, whereas in the C and IBD groups no nuclear -catenin staining was observed. Remarkably, an increased nuclear -catenin expression was detected in 50% of IBD-Ne cases, as compared to a 0% expression in IBD patients without neoplasia (p=0.02). the nuclear -catenin positive and negative tissue samples were located at a median distance of 7 cm (range 2-20 cm) and 13 cm (range 2-78 cm), respectively, from the neoplastic lesion. All positively staining non-dysplastic IBD mucosal samples adjacent to a (pre)malignant lesion were found in patients with neoplasia in the rectum. Percentages of overexpression per core (not individual patients) are depicted in Figure 1a, with an increased expression in 19%, 46%, and 77% of all IBD-Ne, IBD-DYS, and IBD-CRC cores, respectively. Membranous -catenin was highly expressed in the C and IBD groups, whereas nuclear staining was absent in these groups. As expected, membranous staining declined with increasing nuclear accumulation. taken all cores into account, a decreased membranous expression was observed in 5%, 15%, 21%, 40%, and 70% of the C, IBD, IBD-Ne, IBD-DYS, and IBD-CRC cores, respectively (Figure 1b). No difference in nuclear -catenin expression between samples with LGD or HGD was found in the IBD-DYS group. Positive nuclear and membranous staining is shown in Figure 2a and b, respectively. expression of Cyclin D1 the results of Cyclin D1 expression were in line with those of nuclear -catenin staining. Increased Cyclin D1 expression was found in 22%, 33%, 50%, 70%, and 80% of patients in the C, IBD, IBD-Ne, IBD-DYS, and IBD-CRC groups, respectively. In contrast to nuclear -catenin expression, no statistically significant difference in expression of Cyclin D1 was found between the IBD-Ne group and the IBD group. Both the IBD-DYS and IBD-CRC groups showed statistically significantly higher levels of Cyclin D1 expression compared to the IBD group (p=0.046 and p=0.002, respectively). In the IBD-Ne group, 75% of cases who were positive for nuclear -catenin also had increased Cyclin D1 expression, whereas 75% of cases who were negative for nuclear -catenin had no Cyclin D1 expression. Increased expression in all cores was found in 10%, 9%, 26%, 35%, and 61% of the C, IBD, IBD-Ne, IBD-DYS, and IBD-CRC cores, respectively (Figure 1c). Positive and negative staining is shown in Figures 2c and d, respectively.

proefschrift Marian.indd 109

10-10-2010 20:56:31

110

| CHAPteR 8
Figure 1a-D

proefschrift Marian.indd 110

10-10-2010 20:56:35

Potential biomarkers for colonic surveillance | 111

expression of p53 expression of p53 was only found in the IBD-DYS and IBD-CRC groups, with positive expression in 67% and 50%, respectively. All high-grade dysplastic lesions (n=6) and 2 out of 5 low-grade dysplastic lesions were p53 positive. Overexpression of p53 was found in 50% of the IBD-DYS cores and 45% of the IBD-CRC cores (Figure 1d). In five of the IBD-CRC patients, immunohistochemical expression of p53 was completely negative. A sequence analysis of the p53 gene was performed in these cases to search for a stop codon (null) mutation. Sequence analysis could not be performed in one patient due to a shortage of tissue. two out of the remaining four patients had a p53 mutation in the tumour, resulting in abrogation of protein production (insertion in exon 7 and mutation V197be), adding up to an overall abnormal p53 function in 70% of the IBD-CRC patients instead of 50%. Positive and negative expression for p53 is shown in Figure 2e and f, respectively.

Figure 1a-d: expression level per core a. Nuclear -catenin expression: determined by percentage of positively stained nuclei grouped in low (<10%), medium (10 -50%), and high (>50%). b. Membranous -catenin expression: grouped in positive (reduced of absent membranous staining, indicating an activated Wnt-pathway) or negative (presence of membranous staining, indicating an inactivated Wnt-pathway) c. Cyclin D1 expression: determined by the intensity of the expression grouped in negative (-), positive (+) or strongly positive (++). d. P53 expression: grouped in a positive expression or negative (no or <20% cells with expression of p53) C: controls, IBD: patients with inflammatory bowel disease, IBD-Ne: patients with IBD and neoplasia elsewhere in their colon, IBD-DYS: IBD-relateddysplasia, IBD-CRC: IBD-relatedcolorectal carcinoma

proefschrift Marian.indd 111

10-10-2010 20:56:35

112

| CHAPteR 8

Figure 2. Images illustrating immunohistochemical expression profiles of -catenin (a, b), Cyclin D1 (c, d), and p53 (e, f). Magnification 100x en 400x. Sample a shows a nuclear expression of -catenin and sample b a membranous staining pattern. Sample c demonstrates an increased expression of cyclin D1, whereas sample d shows a negative expression of cyclin D1. Sample e shows an increased expression of p53 and sample f a negative p53 staining pattern. the negative staining in f is caused by a stop codon mutation in dysplastic mucosa, see insert. the upper panel shows the dysplastic lesion with the arrow indicating the insertion causing the stop codon mutation. the lower panel shows the normal mucosa.

proefschrift Marian.indd 112

10-10-2010 20:56:37

Potential biomarkers for colonic surveillance | 113

Discussion
this study indicates that, in contrast to the general assumption, Wnt-pathway activation occurs early and frequently in IBD-related colorectal carcinogenesis. Moreover, even in non-dysplastic mucosa adjacent to neoplastic lesions nuclear -catenin and Cyclin D1, markers of an activated Wnt-pathway, could be detected, whereas p53 overexpression was not observed in these tissue specimens. the canonical Wnt-pathway (-catenin mediated Wnt- signaling) controls proliferation and differentiation in the colonic stem cell compartments and is involved in colon carcinogenesis 20,21. Loss of adenomatous polyposis coli (APC) gene function results in a shift from membranous to nuclear -catenin and activates transcription of target genes such as Cyclin D1 and C-myc 22. Previously published data on the role of the Wnt-pathway in colitis-associated colorectal carcinogenesis is conflicting. Van Dekken et al. 23 found increased nucleocytoplasmic -catenin staining in 88% and 79% of dysplastic and cancerous lesions, respectively, whereas Mikami et al. 24 demonstrated very low nuclear -catenin expression in high- and low-grade dysplasia and ulcerative colitis (UC)-associated CRC. In another study, increased nuclear -catenin staining was observed in 48% of UC-related cancers and abnormal APC expression in 67% of cases 25. However, other studies have shown that the frequency of APC gene mutations and -catenin gene mutations in UC-related cancers is lower than in sporadic carcinomas 3,22,26. Conflicting data about the expression of APC and -catenin could be due to a wide variation of employed methods and selected patient populations. A remarkable finding was the expression of nuclear -catenin in non-dysplastic tissue adjacent to neoplastic lesions in half of our patients, which may be regarded as a feature of colonic field cancerization. this observation is in line with previously published data showing pancolonic chromosomal instability in UC patients with a neoplastic lesion, DNA aneuploidy in non-dysplastic mucosa of IBD patients with neoplasia, and telomere shortening in non-dysplastic tissue of patients with neoplasia elsewhere 27-31. Our findings, however, are the first to suggest that the Wnt-pathway is already activated in the field surrounding a dysplastic or malignant lesion in IBD. Although these Wnt-proteins cannot be used as solitary markers of colorectal carcinogenesis, they may function as an additional diagnostic tool for a more biomarker-assisted colonic surveillance. Patients may be assigned to different risk groups, based on the expression level of Wnt-pathway proteins in the non-dysplastic biopsies obtained during surveillance colonoscopy. However, larger studies are needed to confirm our observations. Interestingly, all patients in the IBD-Ne group with nuclear -catenin staining had distal left-sided malignancies. Unfortunately the limited number of patients in this study impedes further speculation on a possible relation between tumour location and activation of the Wnt-pathway. the observation

proefschrift Marian.indd 113

10-10-2010 20:56:37

114

| CHAPteR 8
of nuclear -catenin expression in left-sided CRCs, however, seems to mimic the molecular pathogenesis of conventional distal CRCs. Positive p53 staining was found in two-third and half of the dysplastic and malignant lesions, respectively. this relatively low percentage of p53 positivity in the IBD-CRC group, compared to the IBD-DYS group, was at least in part explained by stop codon (null) mutations of p53 leading to a completely absent protein expression pattern (20%, n=2). Immunohistochemical overexpression of p53 has been reported in the majority of UC-associated carcinomas 27,32,33. Some studies have suggested that genetic mutations in p53, possibly due to oxidative injury, can already be detected in inflamed non-dysplastic mucosa of UC patients 34. However, immunohistochemical studies found only weak to moderately intense p53 expression in non-dysplastic tissue of UC patients 35. In our study, we also did not detect strong nuclear p53 staining in non-dysplastic IBD mucosa. therefore, in contrast to nuclear -catenin, p53 immunohistochemical staining of non-dysplastic mucosa does not serve as biomarker for malignant transformation elsewhere in the colon. Some limitations of our study have to be considered. First, the degree of expression of markers may be heterogeneously distributed within a single lesion. For that reason, three tissue cores were selected from each sample in order to minimize the chance of sampling error 36. this, however, was not always feasible, as the tissue core, which was punched out of the donor block, might not contain enough tissue. Second, the number of patients per group was relatively low, which requires a cautious interpretation of the results. Nevertheless, significant differences between groups were found. third, the patients included in this study were selected from tertiary centres; therefore the incidence of concurrent PSC was higher than in a normal IBD population. In conclusion, our results suggest an early role for the Wnt-pathway in colitisassociated colorectal carcinogenesis. Furthermore, nuclear -catenin may serve as a biomarker for colonic field cancerization, facilitating early detection of neoplasia during colonic surveillance.

Acknowledgements
the authors like to thank D. Castigliego and F. Morsink for their assistance with the immunohistochemistry and G. Bruck-Jansen for her help with the tissue microarray. In addition, we thank the Molecular Pathology Lab for carrying out the p53 mutation analysis. And finally, R. Stellato for her help with the statistical analyses.

proefschrift Marian.indd 114

10-10-2010 20:56:37

Potential biomarkers for colonic surveillance | 115

Reference list
1 2 3 eaden JA, Abrams KR, Mayberry JF. the risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut (2001),526-35. Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 287 (2004),G7-17. tarmin L, Yin J, Harpaz N et al. Adenomatous polyposis coli gene mutations in ulcerative colitis-associated dysplasias and cancers versus sporadic colon neoplasms. Cancer Res 55 (1995),2035-8. Vogelstein B, Fearon eR, Hamilton SR et al. Genetic alterations during colorectal-tumor development. N engl J Med 319 (1988),525-32. Risques RA, Rabinovitch PS, Brentnall tA. Cancer surveillance in inflammatory bowel disease: new molecular approaches. Curr Opin Gastroenterol 22 (2006),382-90. Braakhuis BJ, tabor MP, Kummer JA et al. A genetic explanation of Slaughters concept of field cancerization: evidence and clinical implications. Cancer Res 63 (2003),1727-30. Brentnall tA, Crispin DA, Rabinovitch PS et al. Mutations in the p53 gene: an early marker of neoplastic progression in ulcerative colitis. Gastroenterology 107 (1994),369-78. Riddell RH, Goldman H, Ransohoff DF et al.Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 14 (1983),931-68. Kononen J, Bubendorf L, Kallioniemi A et al. tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4 (1998),844-7. torhorst J, Bucher C, Kononen J et al. tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol 159 (2001),2249-56. Kaserer K, Schmaus J, Bethge U et al. Staining patterns of p53 immunohistochemistry and their biological significance in colorectal cancer. J Pathol 190 (2000),450-6. Krishna M, Woda B, Savas L et al. expression of p53 antigen in inflamed and regenerated mucosa in ulcerative colitis and Crohns disease. Mod Pathol 8 (1995),654-7. Wong NA, Morris RG, McCondochie A et al. Cyclin D1 overexpression in colorectal carcinoma in vivo is dependent on beta-catenin protein dysregulation, but not k-ras mutation. J Pathol 197 (2002),128-35. Joo Ye, Rew JS, Park CS et al. expression of e-cadherin, alpha- and beta-catenins in patients with pancreatic adenocarcinoma. Pancreatology 2 (2002),129-37. Kim MJ, Jang SJ, Yu e. Loss of e-cadherin and cytoplasmic-nuclear expression of beta-catenin are the most useful immunoprofiles in the diagnosis of solid-pseudopapillary neoplasm of the pancreas. Hum Pathol 39 (2008),251-8. Li CC, Xu B, Hirokawa M et al. Alterations of e-cadherin, alpha-catenin and beta-catenin expression in neuroendocrine tumors of the gastrointestinal tract. Virchows Arch 440 (2002),145-54. Alsner J, Jensen V, Kyndi M et al. A comparison between p53 accumulation determined by immunohistochemistry and tP53 mutations as prognostic variables in tumours from breast cancer patients. Acta Oncol 47 (2008),600-7. Kropveld A, Rozemuller eH, Leppers FG et al. Sequencing analysis of RNA and DNA of exons 1 through 11 shows p53 gene alterations to be present in almost 100% of head and neck squamous cell cancers. Lab Invest 79 (1999),347-53. Brosens LAA, Iacobuzio-Donahue CA, Keller JJ et al. Increased cyclooxygenase-2 expression in duodenal compared with colonic tissues in familial adenomatous polyposis and relationship to the -765G-> C cox-2 polymorphism. Clin Cancer Res 11 (2005), 4090-6 de Lau W, Barker N, Clevers H. WNt signaling in the normal intestine and colorectal cancer. Front Biosci 12 (2007),471-91. Pinto D, Clevers H. Wnt control of stem cells and differentiation in the intestinal epithelium. exp Cell Res 306 (2005),357-63.

4 5 6 7 8 9 10 11 12 13

14 15

16 17

18

19

20 21

proefschrift Marian.indd 115

10-10-2010 20:56:37

116

| CHAPteR 8
22 Aust De, terdiman JP, Willenbucher RF et al. the APC/beta-catenin pathway in ulcerative colitis-related colorectal carcinomas: a mutational analysis. Cancer 94 (2002),1421-7. 23 van Dekken H, Wink JC, Vissers KJ et al. Wnt pathway-related gene expression during malignant progression in ulcerative colitis. Acta Histochem 109 (2007),266-72. 24 Mikami t, Mitomi H, Hara A et al. Decreased expression of CD44, alpha-catenin, and deleted colon carcinoma and altered expression of beta-catenin in ulcerative colitis-associated dysplasia and carcinoma, as compared with sporadic colon neoplasms. Cancer 89 (2000),733-40. 25 Aust De, terdiman JP, Willenbucher RF et al. Altered distribution of beta-catenin, and its binding proteins e-cadherin and APC, in ulcerative colitis-related colorectal cancers. Mod Pathol 14 (2001)29-39. 26 Umetani N, Sasaki S, Watanabe t et al. Genetic alterations in ulcerative colitis-associated neoplasia focusing on APC, K-ras gene and microsatellite instability. Jpn J Cancer Res 90 (1999),1081-7. 27 Rabinovitch PS, Dziadon S, Brentnall tA et al. Pancolonic chromosomal instability precedes dysplasia and cancer in ulcerative colitis. Cancer Res 59 (1999),5148-53. 28 OSullivan JN, Bronner MP, Brentnall tA et al. Chromosomal instability in ulcerative colitis is related to telomere shortening. Nat Genet 32 (2002),280-4. 29 Porschen R, Robin U, Schumacher A et al. DNA aneuploidy in Crohns disease and ulcerative colitis: results of a comparative flow cytometric study. Gut 33 (1992),663-7. 30 Sjoqvist U, Befrits R, Soderlund S et al. Colorectal cancer in colonic Crohns disease--high frequency of DNA-aneuploidy. Anticancer Res 25 (2005),4393-7. 31 Rubin Ce, Haggitt RC, Burmer GC et al. DNA aneuploidy in colonic biopsies predicts future development of dysplasia in ulcerative colitis. Gastroenterology 103 (1992),1611-20. 32 Harpaz N, Peck AL, Yin J et al. p53 protein expression in ulcerative colitis-associated colorectal dysplasia and carcinoma. Hum Pathol 25 (1994),1069-74. 33 Bruwer M, Schmid KW, Senninger N et al. Immunohistochemical expression of P53 and oncogenes in ulcerative colitis-associated colorectal carcinoma. World J Surg 26 (2002),390-6. 34 Hussain SP, Amstad P, Raja K et al. Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res 60 (2000),3333-7. 35 Wong NA, Mayer NJ, MacKell S et al. Immunohistochemical assessment of Ki67 and p53 expression assists the diagnosis and grading of ulcerative colitis-related dysplasia. Histopathology 37 (2000),108-14. 36 Bubendorf L, Nocito A, Moch H et al. tissue microarray (tMA) technology: miniaturized pathology archives for high-throughput in situ studies. J Pathol 195 (2001),72-9.

proefschrift Marian.indd 116

10-10-2010 20:56:38

CHaPter 9
Summary and discussion

proefschrift Marian.indd 117

10-10-2010 20:56:40

118

| CHAPteR 9
Patients with inflammatory bowel disease (IBD) have an increased risk of developing colorectal cancer (CRC). Patients with concurrent primary sclerosing cholangitis (PSC) have an even higher risk to develop malignancies. the colitis-associated carcinogenesis is only slowly elucidated and more studies are required to fully understand this process. even less is known about the carcinogenetic pathways in patients with PSC and IBD-related CRC. An important issue to be resolved is whether, and if so, in what way the carcinogenesis resembles or just differs from that of IBD-related CRC. In this thesis, clinical and translational studies were reported that intended to get more insight in the molecular pathways leading to IBD-related CRC. the ultimate goal was to find potential biomarkers that could aid in improving surveillance protocols in patients with longstanding IBD. In particular, patients with PSC were included to determine possible differences in carcinogenetic pathways. In chapter 2, we provided a review on CRC and other tumours in patients with IBD. the risk of developing IBD-related CRC and known risk factors for developing IBDrelated CRC were discussed in this chapter. Furthermore, we considered issues concerning the diagnosis and presence of dysplasia in colonic mucosa of IBD patients. Finally, current knowledge on the molecular background of colitis-associated carcinogenesis was reviewed in order to identify potential biomarkers. Chapter 3 provided new data on the actuarial 10- and 20-years risk of developing CRC and cholangiocarcinoma (CCA) in a large cohort of Dutch PSC patients. In total, 211 patients with PSC were retrospectively reviewed to determine the risk of developing these malignancies. the estimated transplantation-free survival of this cohort was found to be 14 years. Patients being symptomatic when a diagnosis of PSC was made had a significantly lower transplantation-free survival than those who were asymptomatic. the 10- and 20-year risks of developing CRC were 14% and 31%, respectively. the risk of developing CCA was 9%. the conclusions in this chapter were that patients with PSC have a high risk of developing CRC and CCA. Furthermore, the overall survival was seriously shortened when these malignancies developed, but also by the development of liver failure due to progressive PSC. In chapter 4, differences in clinical, endoscopical, and histological characteristics of CRCs from patients with IBD and concurrent PSC (n=27) and patients with IBD alone (n=127) were determined. the most important finding of this study was a high frequency of right-sided tumours (proximal to the splenic flexure) in patients with concurrent PSC compared to IBD alone, 67% vs. 36%, respectively. After adjustment for age, gender and extent of IBD, this difference remained statistically significant. this may implicate a different carcinogenetic pathway in patients with IBD and

proefschrift Marian.indd 118

10-10-2010 20:56:40

Summary | 119

concurrent PSC compared to patients IBD alone. One of the hypotheses is that patients with PSC have an altered bile composition, resulting in an increased concentration of secondary bile acids with carcinogenic effect, causing a high frequency of proximally located tumours. Others have suggested that CRC in patients with PSC mimics Lynch syndrome-associated carcinogenesis, because Lynch syndrome-related CRCs are also frequently proximally located. this would imply a role for MisMatch Repair (MMR) genes. the role of the MMR genes in CRCs from patients with IBD and concurrent PSC was further analysed in chapter 5. A defect in one of the MMR genes may lead to microsatellite instability (MSI), which can easily be detected in tumour DNA. Apart from Lynch syndrome, which is caused by a germline mutation in one of the MMR genes, some sporadic CRCs may also show MSI. In sporadic cases, however, hypermethylation of the MMR genes causes MSI instead of a germline mutation as in Lynch syndrome. Because of the high frequency of proximally located tumours in patients with IBD and concurrent PSC we determined the microsatellite status in this specific subgroup of CRCs. the microsatellite status was analysed in DNA of various subgroups of CRCs including PSC-IBD-related CRC (n=10), IBD-related CRC (n=17), Lynch syndrome-related CRC (n=8), and sporadic CRC (n=37). Immunohistochemical expression of the MMR proteins was also determined. Results showed that despite the high frequency of right-sided tumours in the PSC-CRC group, no MSI was found in this specific group. the IBD-related CRC group showed MSI in 12% of the cases, whereas MSI was found in 32% of sporadic CRC samples. We concluded that the microsatellite instability pathway does not play a major role in PSC-IBD related CRCs. In chapter 6, an immunohistochemical expression and mutation analysis of several genes that are known to be involved in sporadic colorectal carcinogenesis was performed. this was done in two groups of CRC, inflammation-related CRCs (n=27), i.e., IBD-related CRCs with a subgroup of CRCs from patients with concurrent PSC, and sporadic CRCs (n=37) with a proximal and distal tumour location. A tissue microarray was constructed to determine immunohistochemical expression of p53, -catenin, Cyclin D1, SMAD4, COX-2, and CD44V6. In addition, DNA was isolated from all tumour samples to perform Kras and Braf mutation analysis. Results from this study showed less frequent expression of Cyclin D1 in inflammation-related CRC compared sporadic CRC, which remained statistically significant after adjustment for age and tumour stage. Furthermore, although not statistically significantly different, a trend towards a higher loss of SMAD4 expression was seen in the inflammation-related group. Finally, the inflammation-related CRC group showed fewer Kras and Braf mutations compared to the sporadic CRC group; however, this

proefschrift Marian.indd 119

10-10-2010 20:56:40

120

| CHAPteR 9
was no longer significant after adjustment for age and tumour location. We concluded that inflammation-related CRCs were only to a small extent different from sporadic CRCs when compared for various tumour suppressor genes. Another important pathway involved in colorectal carcinogenesis was studied in chapter 7. Besides genetic alterations, it is known that epigenetic alterations also play a major role in tumour development. therefore, hypermethylation of 24 tumour suppressor genes was studied again in inflammation-related tumours (n=15), i.e., IBD-related CRCs with a subgroup of CRCs from patients with concurrent PSC, and sporadic CRCs (n=17). DNA was isolated and a quantitative method known as Methylation-Specific Multiplex Ligation-dependent Probe Amplification (MS-MLPA) was used to determine methylation percentages in the tumour samples. the number of samples with methylation as well as the median methylation percentages for ESR1, CDKN2B, and CDH13 were significantly higher in the sporadic than in the inflammation-related CRC group (p<0.001). A borderline significance was also found for CD44 with 20% of the inflammation-related CRC samples showing >15% methylation compared to none of the sporadic group (p=0.092). the median cumulative methylation index was 197% and 326% in the IBD and sporadic tumour groups, respectively (p=0.051). this difference was however no longer found after age-adjustment (p=0.34). We concluded that epigenetic alterations occur less frequently in colitis-associated CRC compared to sporadic CRC. these results may well indicate a more important role for genetic than for epigenetic alterations in colitis-associated tumorigenesis Finally, in chapter 8, the role of the Wnt-pathway during colitis-associated carcinogenesis was determined. It is known that the Wnt-pathway, including APC and -catenin, plays a major role in sporadic carcinogenesis and is activated at an early stage. We studied the expression of Wnt-signaling proteins and p53 by immunohistochemical expression during different stages of the colitis-associated carcinogenesis. A tissue microarray was constructed with samples from 5 groups, i.e., healthy controls (n=10), patients with IBD without neoplasia (n=12), nondysplastic mucosa of patients with dysplasia elsewhere in their colon (n=12), samples with dysplasia (n=12) and finally, tumour samples (n=10). Immunohistochemical expression was determined for -catenin, Cyclin D1 (one of the target genes of the Wnt-pathway) and p53. In some of these cases, p53 sequence analysis could also be performed. Results showed that nuclear -catenin expression was already found in non-dysplastic mucosa from patients with neoplasia elsewhere in their colon. Cyclin D1 showed a similar pattern as -catenin. P53 expression was only found in groups with dysplastic and tumour samples. Our results suggest activation of the Wnt-pathway at an early stage during colitis-associated carcinogenesis, particularly

proefschrift Marian.indd 120

10-10-2010 20:56:40

Summary | 121

in mucosa which was microscopically not abnormal. therefore -catenin expression may facilitate detection of neoplasia at an early stage. Conclusive remarks and future studies Based on the first chapters of this thesis (Chapters 2,3, and 4), it appears that distinctive colorectal carcinogenetic pathways may exist in IBD patients with and without concurrent PSC, as illustrated by a higher frequency of proximally located tumours and a higher risk of developing CRC. Furthermore, presumably IBDassociated CRCs differ from conventional sporadic CRCs because IBD-related CRC arises against a background of chronic inflammation of the colorectal mucosa. Literature reports have suggested that in particular the timing of specific genetic alterations as well as the frequency of promoter hypermethylation may differ in IBD-associated colorectal carcinogenesis. In the last chapters of this thesis (Chapters 5,6, and 7), however, we were not able to detect significant differences between the molecular genetic alterations in IBDassociated CRC and conventional sporadic CRCs. Similarly, the molecular make up of the PSC-IBD-related CRCs, resembled that of the IBD-associated CRC and the conventional sporadic CRCs. Our findings suggest that once CRC has developed, whatever the underlying background (sporadic, IBD-related or PSC-IBD-related), the ultimate result of the molecular genetic aberrations, manifest in the tumour, is very similar. this suggests that the main differences between sporadic CRC, IBD-associated CRC and PSCIBD-related CRC may be related to the initial neoplastic stages, and such differences may include the genetic background of a patient as well as (micro)environmental factors like chronic inflammation. Our results suggest that the main road to the prevention of CRC, including IBDrelated and PSC-IBD-related CRC, lies in the timely recognition of subgroups of patients who are at an increased risk of developing CRC. this can probably be done by using a combination of advanced endoscopic and molecular techniques. In order to arrive there, we first should completely understand the molecular carcinogenetic pathway of these tumours. Following the natural history of this pathway in various patients is a precondition for understanding inflammation-related CRC and therefore the challenge will be to collaborate and in this fashion collect sufficient well documented patient materials to carry out these studies. the tissue samples need to be prospectively collected with the use of well organized bio banks; the Dutch Parelsnoer initiative may well play an important role in this progress.

proefschrift Marian.indd 121

10-10-2010 20:56:40

122

proefschrift Marian.indd 122

10-10-2010 20:56:41

Nederlandse samenvatting Acknowledgements About the author

proefschrift Marian.indd 123

10-10-2010 20:56:43

124

Nederlandse samenvatting

Inflammatoire darmziekten, in het engels Inflammatory Bowel Disease, afgekort IBD, is een verzamelnaam voor een chronische ontsteking van de darm. er zijn twee vormen te onderscheiden, colitis ulcerosa en de ziekte van Crohn. De oorzaak van IBD is niet goed bekend. Waarschijnlijk heeft IBD een multifactoriele oorzaak, waarbij zowel omgevingsfactoren als genetisch aanleg een rol spelen. Patinten met IBD hebben een hoger risico op het ontwikkelen van dikke darmkanker (ook wel colorectaal carcinoom, afgekort CRC) dan de rest van de bevolking. Gedacht wordt dat de chronische ontsteking van de darm hier een belangrijke rol in speelt. Deze ontsteking brengt over een langere periode schade toe aan het darmslijmvlies waardoor er keer op keer celvernieuwing plaatsvindt met als gevolg een hoger risico op het ontstaan van CRC. er zijn meerdere risicofactoren die de kans op CRC bij IBD verhogen, o.a. een langere ziekteduur, de uitgebreidheid van de darmontsteking, de ernst van de ontsteking en de aanwezigheid van primaire scleroserende cholangitis (PSC). PSC is een chronische aandoening die zich kenmerkt door een ontsteking van de galwegen. PSC komt voor bij ongeveer 5% van de patinten met IBD en omgekeerd heeft 90% van de patinten met PSC ook IBD. PSC is een progressieve aandoening die kan leiden tot leverfalen. een levertransplantatie is momenteel de enige beschikbare therapeutische optie met kans op genezing. er is m.n. geen effectieve medicatie die PSC kan genezen. Naast het risico op leverfalen wordt de levensverwachting van patinten met PSC ook negatief benvloedt door een verhoogd risico op het ontwikkelen van maligniteiten, in het bijzonder van de galwegen (cholangiocarcinoom, afgekort CCA) en CRC. Vanwege het verhoogde risico op CRC zijn er internationale richtlijnen (surveillance guidelines) die adviseren om patinten met IBD regelmatig te controleren. Deze controles hebben tot doel om CRC in een vroeg stadium, m.n. als er slechts dysplasie is, of in een vroeg tumorstadium te diagnosticeren en daarmee de prognose van patinten gunstig te benvloeden. Deze controles vinden plaats door middel van colonoscopie. Hierbij worden biopsien van het slijmvlies genomen die histologisch worden beoordeeld. Nadelen van dit onderzoek zijn enerzijds het ondergaan van een ingrijpend onderzoek voor de patint en anderzijds is het voor een patholoog lastig om de juiste stadia van dysplasie te onderscheiden. Op dit moment wordt alle aan IBD patinten, die de aandoening langere tijd hebben, geadviseerd om met regelmaat een surveillance colonoscopie te ondergaan. Idealiter zou het wenselijk zijn om patinten in te delen in risicogroepen, waardoor patinten met een hoger risico om CRC te krijgen frequenter gecontroleerd zouden kunnen worden dan patinten met een lager risico. Om in de toekomst een betere surveillance op maat te kunnen bieden wordt er momenteel veel onderzoek gedaan naar de moleculaire ontstaanswijze van darmkanker. De achterliggende gedachte hierbij is dat bepaalde afwijkingen in het

proefschrift Marian.indd 124

10-10-2010 20:56:43

Nederlandse samenvatting | 125

DNA al vroeg zouden kunnen wijzen op een (verhoogd) risico op het ontstaan van kanker. Deze afwijkingen, ook wel biomarkers genoemd, zouden dan gedetecteerd kunnen worden in weefsel dat er onder de microscoop nog niet afwijkend uitziet, waardoor maligne ontaarding in een zeer vroeg stadium gedetecteerd kan worden. Langzamerhand wordt er steeds meer bekend over de ontstaanswijze van darmtumoren, zowel van de sporadische variant (voorkomend bij de bevolking zonder bekende risicofactoren) als van de IBD-gerelateerde variant. In grote lijnen weten we dat de meeste genveranderingen bij beide groepen voorkomen, maar dat er toch ook verschillen zijn tussen beide groepen. In dit proefschrift is er op verschillende manieren gekeken naar de moleculaire achtergrond van IBD-gerelateerde tumoren, zowel op eiwit- als op genniveau, met als achterliggende gedachte meer kennis te vergaren over de carcinogenese (ontstaan van kanker) met als uiteindelijk doel bij te dragen aan een meer individueel gerichte surveillance strategie bij patinten met IBD. In hoofdstuk 2 wordt een overzicht gegeven van alle recente literatuur over surveillance bij IBD patinten. Hierin werd duidelijk dat er momenteel nog geen plaats is voor moleculaire diagnostiek bij surveillance omdat er nog onvoldoende bekend is over het moleculaire proces om deze informatie te kunnen gebruiken voor surveillance doeleinden. Het vinden van afwijkingen heeft belangrijke consequenties voor patinten. Indien dat het geval is zal de patint een operatie moeten ondergaan waarbij het colon wordt verwijderd (colectomie). Prospectieve studies met langdurige follow-up zijn noodzakelijk om te bepalen of het toevoegen van nieuw gevonden biomarkers aan het bestaande surveillance programma een positief effect heeft op de overleving van IBD patinten. Hoofdstuk 3 gaat dieper in op de incidentie van CRC en CCA bij patinten met zowel PSC als IBD. Van 1980 tot 2006 werden 211 Nederlandse PSC patinten (143 mannen (68%)) gevolgd. De follow-up bedroeg 9 jaar (range 0.3-25). tijdens followup overleden 45 (21%) patinten en ondergingen 49 (23%) patinten een levertransplantatie. De meest voorkomende oorzaken van overlijden waren een maligniteit (44%) en leverfalen (29%). De meest voorkomende maligniteiten waren CCA en CRC. De berekende 10- en 20-jaars risicos op CRC bij patinten met IBD en PSC waren respectievelijk 14% en 31%. Het risico op een CCA was 9%. Daarnaast werd vastgesteld dat het optreden van een CCA of CRC, een hogere leeftijd en een symptomatische presentatie ten tijde van de diagnose PSC een negatieve invloed hadden op de overleving. Conclusies die uit dit hoofdstuk kunnen worden getrokken is dat patinten met PSC en IBD een verhoogd risico hebben om een CCA of CRC te ontwikkelen. Het is echter niet duidelijk waarom patinten die naast IBD ook PSC hebben een nog hoger risico hebben op CRC. Mogelijk zou een

proefschrift Marian.indd 125

10-10-2010 20:56:43

126

Nederlandse samenvatting

afwijkende galsamenstelling een rol kunnen spelen. een andere hypothese berust op de observatie dat IBD vaak asymptomatisch verloopt in patinten met PSC. Hierdoor zou er een vertraging in de diagnostiek (het verrichten van surveillance colonoscopie) kunnen optreden en ondergaan patinten minder vaak een colectomie voordat ze kanker ontwikkelen. Het is dus van groot belang om bij deze patintengroep op zeer regelmatige basis endoscopisch onderzoek te verrichten om tijdig te kunnen handelen. In hoofdstuk 4 werden darmtumoren van patinten met zowel PSC en IBD (n=27) en darmtumoren van patinten met alleen IBD (n=127) vergeleken. De belangrijkste bevinding van deze studie was de hoge frequentie van rechtszijdige darmtumoren (proximaal van de flexura lienalis) in de groep patinten met PSC en IBD. In totaal, was 67% van de tumoren rechtszijdig gelokaliseerd versus 36% in de IBD-groep (p=0.006). Ook na multivariate analyse met correctie voor leeftijd, geslacht en uitgebreidheid van de colitis bleef PSC een risicofactor voor het ontwikkelen van rechtszijdige tumoren met een odds ratio van 4.8 (95% CI: 2.0-11.8). een duidelijke verklaring voor de hogere frequentie rechtszijdige tumoren is er niet. een hypothese hiervoor zou kunnen zijn dat een veranderde galsamenstelling bij PSC patinten een mogelijk kankerverwekkende invloed heeft op het proximale deel van de dikke darm. een andere hypothese is dat de carcinogenese lijkt op die van een erfelijke vorm van CRC passend bij Lynch syndroom, waarbij ook vaak proximale tumoren worden gevonden. Bij dit syndroom is er een defect opgetreden in de genen die normaliter kleine replicatiefouten in het DNA herstellen. In hoofdstuk 5 wordt dieper ingegaan op de laatst genoemde hypothese en worden PSC-gerelateerde CRCs vergeleken met andere typen darmtumoren, onder andere Lynch-gerelateerde tumoren. Bij het Lynch-syndroom is er een mutatie opgetreden in n van de zogenaamde mismatch repair genen (MMR genen), die normaliter fouten, die ontstaan zijn tijdens de normale celdeling (DNA replicatie), repareren. Op het moment dat deze genen niet meer goed functioneren, zal er een ophoping van fouten in de genen ontstaan, waarbij uiteindelijk ook genen worden aangedaan die de celdeling controleren, met als gevolg het ontstaan van een ongeremde celdeling en kanker. Bepaalde stukken van het genoom, de zogenaamde microsatellieten zijn extra gevoelig voor fouten en veranderen van lengte wanneer ze niet worden gerepareerd door de MMR genen. Het opgetreden lengte verschil wordt ook wel beschreven als microsatelliet instabiliteit, afgekort MSI. MSI wordt in het laboratorium relatief eenvoudig aangetoond en vormt dan ook een van de diagnostische pijlers om het Lynch syndroom te diagnosticeren. Bij het Lynch syndroom ligt er een kiembaanmutatie ten grondslag aan het ontstaan van MSI. Bij sporadische darmtumoren wordt echter ook bij 15% van de tumoren MSI geconstateerd, meestal

proefschrift Marian.indd 126

10-10-2010 20:56:43

Nederlandse samenvatting | 127

in proximale tumoren. Deze MSI berust dan niet op een kiembaanmutatie maar op uitschakeling van de MMR genen op een andere manier, namelijk door methylering. De term methylering beschrijft de toevoeging van een methylgroep aan het DNA. Het DNA is georganiseerd in duizenden genen met allemaal hun eigen functie. Op het moment dat een methylgroep aan het DNA vastzit, kan het gen zijn functie niet meer uitoefenen. Zo kan methylering er voor zorgen dat genen hun (beschermende of controlerende) functie verliezen en op deze manier bijdragen aan het ontstaan van kanker. De achterliggende gedachte van de studie in dit hoofdstuk was dat we bij patinten met PSC en CRC ook MSI zouden constateren vanwege de hoge frequentie rechtszijdige tumoren in deze groep. Mogelijk zou dan methylering een belangrijke rol hierin spelen. De resultaten van deze studie lieten echter zien dat er geen MSI werd gevonden bij deze patinten groep. Waarschijnlijk worden tumoren in deze patinten groep dus niet veroorzaakt door een defect in de MMR genen, maar spelen andere genen c.q. factoren een rol. In hoofdstuk 6 wordt dieper in gegaan op andere genen die mogelijk van invloed kunnen zijn. Zoals hierboven beschreven is het DNA georganiseerd in genen (20.00030.000 genen) die allemaal hun eigen functie hebben. Normaal gesproken wordt het DNA gekopieerd naar RNA (transcriptie) waarna het RNA wordt vertaald naar eiwit (translatie), dat vervolgens de functie van het gen uitvoert. De mate van eiwitexpressie zegt dus iets over de functie van het gen. In dit hoofdstuk werd de eiwitexpressie van verschillende genen bestudeerd waarvan bekend is dat deze een rol spelen in verschillende vormen van kanker. De eiwitexpressie in sporadische en IBD-gerelateerde darmtumoren werd vergeleken, met in de laatste groep ook tumoren afkomstig van patinten met IBD en PSC. Naast de eiwitexpressie werden ook mutaties in twee belangrijke tumor suppressor genen bepaald, Kras en Braf. In deze studie werden geen duidelijke significante verschillen gevonden tussen beide groepen, behalve in de eiwitexpressie van Cyclin D1, een gen betrokken bij de regulering van de cel cyclus dat bij IBD-gerelateerde tumoren minder tot expressie kwam dan bij sporadische tumoren. Daarnaast viel ook op dat er meer eiwitverlies van SMAD4, een tumor suppressor gen, werd gezien in de IBD-gerelateerde tumoren. Ook leken er duidelijk minder Kras en Braf mutaties voor te komen in IBDgerelateerde tumoren, echter na correctie voor leeftijd en tumorlocatie was dit verschil niet meer significant. Deze bevindingen lijken te wijzen op slechts subtiele verschillen in de bestudeerde genen tussen IBD-gerelateerde tumoren en sporadische tumoren, in ieder geval in een laat stadium. Het zou goed mogelijk kunnen zijn dat de mutaties die optreden gedurende de carcinogenese wel verschillen laten zien. Hoofdstuk 7 beschrijft de rol van methylering in diezelfde groepen van IBDgerelateerde tumoren en sporadische darmtumoren als in hoofdstuk 6 beschreven.

proefschrift Marian.indd 127

10-10-2010 20:56:43

128

Nederlandse samenvatting

De functie van een gen kan worden uitgeschakeld door een mutatie in het gen zelf of door invloed van buitenaf, zoals methylering (ook wel epigenetische inactivatie genoemd). er werd gekeken naar het voorkomen van methylatie in 24 verschillende genen. De conclusie van dit hoofdstuk is dat een aantal genen meer methylatie liet zien in de sporadische tumorgroep dan in de IBD-gerelateerde tumorgroep. Ook het totale percentage methylatie was beduidend lager in de IBD-gerelateerde groep. De conclusie van dit hoofdstuk is dat met betrekking tot methylatie er verschillen lijken te zijn tussen IBD-gerelateerde en sporadische tumoren. In hoofdstuk 8 hebben we in verschillende stadia van de carcinogenese van IBDgerelateerde tumoren gekeken naar de eiwitexpressie van genen die in sporadische tumoren uitgebreid betrokken zijn bij het ontstaan van kanker. Deze genen maken allemaal deel uit van de Wnt-pathway, een schakel tussen verschillende genen die met nauw verbonden zijn met elkaar. De Wnt-pathway is al vroeg, in een voorstadium van kanker, betrokken bij het ontstaan van sporadische tumoren. Bij IBD-gerelateerde tumoren was nog niet goed bekend wat de rol is van deze pathway en in het bijzonder of en in welke stadium deze pathway actief is. De verschillende groepen bestonden uit weefsel van patinten met IBD maar zonder kanker, normaal weefsel van IBD patinten met kanker, weefsel met dysplasie en tumorweefsel van IBD patinten. De resultaten lieten zien dat een verhoogde eiwitexpressie, wijzend op een actieve Wnt-pathway, werd gevonden in normaal weefsel van IBD patinten die op een andere locatie in hun darm een tumor hebben. Dit wijst op een rol van de Wnt-pathway vroeg in de carcinogenese. Daarnaast is het bijzonder dat er al afwijkingen op eiwit niveau kunnen worden gevonden in weefsel van patinten dat histologisch normaal is, maar die op een andere locatie plek in de darm al wel een tumor hebben. Dit wijst op een zogenaamd field effect: genetische afwijkingen in normaal weefsel die wijzen op een ontregeling op een andere locatie. Dit principe zou een belangrijke rol kunnen spelen bij de surveillance van patinten. Hoofdstuk 9 vat de bevindingen van dit proefschrift samen en plaatst ze in de context van de bestaande literatuur.

proefschrift Marian.indd 128

10-10-2010 20:56:43

Acknowledgements | 129

Acknowledgements
Dit proefschrift is tot stand gekomen met de hulp van velen. Iedereen die op wat voor wijze dan ook heeft bijgedragen aan dit proefschrift wil ik hartelijk bedanken. In het bijzonder wil ik de volgende mensen bedanken. Prof P.D.Siersema, beste Peter. Je enthousiaste ideen over nieuwe onderzoekslijnen, het bewaren van overzicht met het uiteindelijke doel helder voor ogen maken jou tot een fijne promotor. Onze maandelijkse bijeenkomsten heb ik als zeer prettig ervaren. De enorme hoeveelheid werk leek altijd weer mee te vallen als ik jouw kamer verliet, helaas duurde dat gevoel nooit lang. toen de afrondende fase van dit proefschrift toch drukker bleek te zijn dan verwacht bleef je mij vanuit je vakantieadres in Amerika voorzien van heldere correcties en aanwijzingen. Bedankt. Prof G.J.A. Offerhaus, beste Johan. Als ik weer met een berg onoverzichtelijke resultaten uit het lab kwam aanzetten, reageerde jij steevast enthousiast. Jij gaf een heldere en duidelijke interpretatie aan de resultaten zodat ik vol goede moed weer verder kon. Ik heb de verdieping in de pathologie en moleculaire technieken als zeer bruikbaar ervaren. Deze kennis zal me goed van pas komen als toekomstig mdl-arts. Dr. F.P. Vleggaar, beste Frank. Bedankt voor je open deur waar ik altijd mocht komen binnenvallen voor zinvolle en minder zinvolle mededelingen. Je grote relativeringsvermogen had een geruststellende uitwerking op mij, dat bleek ook bij de voltooiing van mijn proefschrift. Ik vind het ontzettend leuk je eerste promovendus te mogen zijn. Prof M. Samsom, beste Melvin. Hartelijk dank voor het vertrouwen dat je in mij hebt gesteld door me aan te nemen als arts-onderzoeker. Destijds had ik niet durven hopen dat ik vier jaar later, gepromoveerd en wel, bezig zou zijn met de opleiding tot mdl-arts. Drs M.e.I. Schipper, beste Marguerite. De vele uren die wij achter een microscoop hebben doorgebracht, de hoeveelheid engelse drop die daarbij is weggewerkt en alle gesprekken die we voerden zijn het allemaal waard geweest. Bedankt voor de vele vrije tijd die je in mijn onderzoek hebt willen steken. Stafleden en arts-assistenten van de afdeling MDL-ziekten: bedankt voor het kritisch meedenken bij de research meetings, de gezelligheid, en de borrels; dat er nog vele mogen volgen. Afleiding was er altijd te vinden op het secretariaat, de endoscopie balie, het functie lab of de koffiekamer.

proefschrift Marian.indd 129

10-10-2010 20:56:43

130 | Acknowledgements

Iedereen van het moleculaire lab, PRL en de afdeling pathologie wil ik hartelijk danken voor de handige tips, geduldige uitleg en hulp bij het uitvoeren van mijn onderzoek. Folkert, jij bent befaamd om je immuno-kunsten, ook ik heb daarvan mogen meegenieten. Collega arts-onderzoekers: Met veel plezier ging ik de afgelopen drie jaar naar mijn werk, mede door jullie aanwezigheid. Ik heb genoten van de borrels, briljante feesten en congressen. Maarten en Durk: hanen in het kippenhok. Bedankt voor jullie culinaire uitspattingen, altijd fijn om bij jullie te mogen komen eten. Lieve Ofke en Petra: samen de drie Wijzen uit het Oosten. Mooie herinneringen heb ik aan de sky bingo, onze vriend timo, de strop, alle denkbeeldige koffers die zijn gepakt en natuurlijk de Overkant met bizarre hoeveelheden appelbollen en Napoleons. Ik heb genoten! Lieve vrienden: wat fijn dat ik jullie mag kennen! Bedankt voor alle afleiding en gezelligheid, ik verheug me op vele nieuwe Haagse sessies, Yalikavak the sequel en nog veel meer. Lieve Marion, het kan niet anders dat jij vandaag naast me staat. en dan nu familie. Lieve papa en mama, ontzettend bedankt voor jullie onvoorwaardelijke steun en vertrouwen in mij. Jullie zorgzaamheid is grenzeloos, na een weekendje Limburg voel ik me herboren. Lieve Bart & Joyce, Susan & Martijn en natuurlijk Josephine, Marius en Leander: aan aandacht en interesse kom ik niet tekort, bedankt hiervoor. Lieve Wim en Shosho: bedankt voor de warme gastvrijheid en oprechte interesse die jullie in mij tonen. een weekendje Bergen voelt als thuiskomen. SW en tiny: Nu jullie zo dicht in de buurt komen wonen ontkom ik niet meer aan een spelletje, gezellig! tenslotte, allerliefste Robin. Bedankt dat je het hebt volgehouden met mij de afgelopen maanden. Met jou is het leven pas echt genieten! Volgend jaar wordt ons jaar!

proefschrift Marian.indd 130

10-10-2010 20:56:43

About the author | 131

Curriculum Vitae
Marian Claessen werd geboren op 3 november 1981 te Heumen en groeide op in Heerlen. Haar middelbare schoolperiode bracht zij door op het Bernardinuscollege in Heerlen. In 1999 begon zij met de studie medische biologie te Utrecht. Na het behalen van haar propedeuse begon zij in 2000 met de studie geneeskunde aan de universiteit te Utrecht. tijdens haar studie verbleef zij gedurende 3 maanden in Manzini, Swaziland, waar zij een co-schap kindergeneeskunde deed in het Mbabane Government Hospital. In het laatste jaar van haar studie verrichtte zij onderzoek op de afdeling Maag-, Darm-, en Leverziekten onder begeleiding van dr F.P. Vleggaar naar het voorkomen van maligniteiten bij patinten met primaire scleroserende cholangitis. Dit resulteerde in haar eerste wetenschappelijke publicatie en was mede de start van een drie jaar durend promotietraject onder begeleiding van prof P.D. Siersema, prof G.J.A. Offerhaus en dr F.P Vleggaar. In februari 2010 is zij begonnen met de opleiding tot Maag-Darm-Leverarts. thans doorloopt zij de vooropleiding interne geneeskunde in het Meander Medisch Centrum te Amersfoort (opleider: dr. C.A.J.M Gaillard).

proefschrift Marian.indd 131

10-10-2010 20:56:43

132

proefschrift Marian.indd 132

10-10-2010 20:56:43

You might also like